101
|
Kawka E, Witowski J, Sandoval P, Rudolf A, Vidal AR, Cabrera ML, Jörres A. Epithelial-to-Mesenchymal Transition and Migration of Human Peritoneal Mesothelial Cells Undergoing Senescence. Perit Dial Int 2018; 39:35-41. [PMID: 30478141 DOI: 10.3747/pdi.2017.00244] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 07/17/2018] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Epithelial-to-mesenchymal transition (EMT) of human peritoneal mesothelial cells (HPMCs) contributes to fibrotic thickening of the peritoneum that develops in patients on peritoneal dialysis (PD). The process is thought to be largely mediated by transforming growth factor-beta (TGF-β). As TGF-β has also been implicated in senescence of HPMCs, we have performed an exploratory study to examine if senescent HPMCs can undergo EMT. METHODS Omentum-derived HPMCs were rendered senescent by repeated passages in culture. Features of EMT were assessed by immunostaining and quantitative polymerase chain reaction (qPCR) at various stages of the HPMC lifespan and after treatment with or without TGF-β. The motility of HPMCs was assessed in a scratch wound migration assay. RESULTS Replicative senescence of HPMCs was associated with a gradual increase in the constitutive expression of EMT markers, including increased production of extracellular matrix proteins. However, senescent HPMCs also retained epithelial cell features such as cytokeratin, calretinin, and E-cadherin and showed decreased, rather than increased, motility. In contrast, exposure to TGF-β resulted in an up-regulation of mesenchymal markers and down-regulation of epithelial markers. Such effects of TGF-β occurred both in young and senescent cells, although they were less pronounced in senescence. CONCLUSIONS Senescence of HPMCs is associated with spontaneous development of several EMT features. At the same time, senescent HPMCs preserve epithelial cell-like characteristics and are less prone to develop a full EMT phenotype in response to TGF-β. These observations may support the concept of cellular senescence being antagonistically pleiotropic with regard to EMT.
Collapse
Affiliation(s)
- Edyta Kawka
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Janusz Witowski
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Department of Pathophysiology, Poznan University of Medical Sciences, Poznań, Poland
| | - Pilar Sandoval
- Centro de Biología Molecular-Severo Ochoa, Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Andras Rudolf
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznań, Poland
| | - Angela Rynne Vidal
- Centro de Biología Molecular-Severo Ochoa, Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Manuel Lopez Cabrera
- Centro de Biología Molecular-Severo Ochoa, Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Achim Jörres
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany .,Department of Medicine I - Nephrology, Transplantation & Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Cologne, Germany
| |
Collapse
|
102
|
Callender LA, Carroll EC, Bober EA, Henson SM. Divergent mechanisms of metabolic dysfunction drive fibroblast and T-cell senescence. Ageing Res Rev 2018; 47:24-30. [PMID: 29902528 DOI: 10.1016/j.arr.2018.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/22/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
The impact of cellular senescence during ageing is well established, however senescence is now recognised to play a role in a variety of age related and metabolic diseases, such as cancer, autoimmune and cardiovascular diseases. It is therefore crucial to gain a better understanding of the mechanisms that control cellular senescence. In recent years our understanding of the intimate relationship between cell metabolism, cell signalling and cellular senescence has greatly improved. In this review we discuss the differing roles of glucose and protein metabolism in both senescent fibroblast and CD8+ T-cells, and explore the impact cellular metabolism has on the senescence-associated secretory phenotype (SASP) of these cell types.
Collapse
|
103
|
Roles of long noncoding RNAs in aging and aging complications. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1763-1771. [PMID: 31109449 DOI: 10.1016/j.bbadis.2018.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/23/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
Aging is a universal and time dependent complex biological process, characterized by a progressive physiological dysfunction and an increased vulnerability to death. Though the physiological process of aging is still not fully understood, several cellular and molecular mechanisms have been identified. Long noncoding RNAs is a class of regulatory ncRNAs with transcript lengths >200 nucleotides. Discovery of this vast pool of regulators in mammalian genome supplies a new dimension to study and explore the aging process. In this review, we discuss the contribution of lncRNAs in aging and aging complications, and raise interest of serving lncRNAs as biomarkers and potential therapeutic targets to prolong health and ameliorate age-associated diseases. We hope understanding the roles of these high specificity and low conservation regulators in generating age-associated phenotypes might benefit human lifespan.
Collapse
|
104
|
Zhu Y, Liu X, Ding X, Wang F, Geng X. Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology 2018; 20:1-16. [PMID: 30229407 DOI: 10.1007/s10522-018-9769-1] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/12/2018] [Indexed: 01/10/2023]
Abstract
Aging is a biological process characterized by a progressive functional decline in tissues and organs, which eventually leads to mortality. Telomeres, the repetitive DNA repeat sequences at the end of linear eukaryotic chromosomes protecting chromosome ends from degradation and illegitimate recombination, play a crucial role in cell fate and aging. Due to the mechanism of replication, telomeres shorten as cells proliferate, which consequently contributes to cellular senescence and mitochondrial dysfunction. Cells are the basic unit of organismal structure and function, and mitochondria are the powerhouse and metabolic center of cells. Therefore, cellular senescence and mitochondrial dysfunction would result in tissue or organ degeneration and dysfunction followed by somatic aging through multiple pathways. In this review, we summarized the main mechanisms of cellular senescence, mitochondrial malfunction and aging triggered by telomere attrition. Understanding the molecular mechanisms involved in the aging process may elicit new strategies for improving health and extending lifespan.
Collapse
Affiliation(s)
- Yukun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, China
| | - Xuewen Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, China
| | - Xuelu Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, China
| | - Fei Wang
- Department of Neurology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Xin Geng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China. .,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
105
|
Oost W, Talma N, Meilof JF, Laman JD. Targeting senescence to delay progression of multiple sclerosis. J Mol Med (Berl) 2018; 96:1153-1166. [PMID: 30229272 PMCID: PMC6208951 DOI: 10.1007/s00109-018-1686-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/18/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic and often progressive, demyelinating disease of the central nervous system (CNS) white and gray matter and the single most common cause of disability in young adults. Age is one of the factors most strongly influencing the course of progression in MS. One of the hallmarks of aging is cellular senescence. The elimination of senescent cells with senolytics has very recently been shown to delay age-related dysfunction in animal models for other neurological diseases. In this review, the possible link between cellular senescence and the progression of MS is discussed, and the potential use of senolytics as a treatment for progressive MS is explored. Currently, there is no cure for MS and there are limited treatment options to slow the progression of MS. Current treatment is based on immunomodulatory approaches. Various cell types present in the CNS can become senescent and thus potentially contribute to MS disease progression. We propose that, after cellular senescence has indeed been shown to be directly implicated in disease progression, administration of senolytics should be tested as a potential therapeutic approach for the treatment of progressive MS.
Collapse
Affiliation(s)
- Wendy Oost
- University of Groningen, Groningen, The Netherlands
| | - Nynke Talma
- European Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan F Meilof
- Department of Neurology, Martini Hospital, Groningen, The Netherlands.,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. .,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
106
|
Nrf2-Mediated Fibroblast Reprogramming Drives Cellular Senescence by Targeting the Matrisome. Dev Cell 2018; 46:145-161.e10. [PMID: 30016619 DOI: 10.1016/j.devcel.2018.06.012] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/14/2018] [Accepted: 06/14/2018] [Indexed: 01/06/2023]
Abstract
Nrf2 is a key regulator of the antioxidant defense system, and pharmacological Nrf2 activation is a promising strategy for cancer prevention and promotion of tissue repair. Here we show, however, that activation of Nrf2 in fibroblasts induces cellular senescence. Using a combination of transcriptomics, matrix proteomics, chromatin immunoprecipitation and bioinformatics we demonstrate that fibroblasts with activated Nrf2 deposit a senescence-promoting matrix, with plasminogen activator inhibitor-1 being a key inducer of the senescence program. In vivo, activation of Nrf2 in fibroblasts promoted re-epithelialization of skin wounds, but also skin tumorigenesis. The pro-tumorigenic activity is of general relevance, since Nrf2 activation in skin fibroblasts induced the expression of genes characteristic for cancer-associated fibroblasts from different mouse and human tumors. Therefore, activated Nrf2 qualifies as a marker of the cancer-associated fibroblast phenotype. These data highlight the bright and the dark sides of Nrf2 and the need for time-controlled activation of this transcription factor.
Collapse
|
107
|
Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology 2018; 19:447-459. [PMID: 30054761 PMCID: PMC6223730 DOI: 10.1007/s10522-018-9763-7] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/21/2018] [Indexed: 12/13/2022]
Abstract
Cellular senescence is now considered as a major mechanism in the development and progression of various diseases and this may include metabolic diseases such as obesity and type-2 diabetes. The presence of obesity and diabetes is a major risk factor in the development of additional health conditions, such as cardiovascular disease, kidney disease and cancer. Since senescent cells can drive disease development, obesity and diabetes can potentially create an environment that accelerates cell senescence within other tissues of the body. This can consequently manifest as age-related biological impairments and secondary diseases. Cell senescence in cell types linked with obesity and diabetes, namely adipocytes and pancreatic beta cells will be explored, followed by a discussion on the role of obesity and diabetes in accelerating ageing through induction of premature cell senescence mediated by high glucose levels and oxidised low-density lipoproteins. Particular emphasis will be placed on accelerated cell senescence in endothelial progenitor cells, endothelial cells and vascular smooth muscle cells with relation to cardiovascular disease and proximal tubular cells with relation to kidney disease. A summary of the potential strategies for therapeutically targeting senescent cells for improving health is also presented.
Collapse
|
108
|
Senescent tumor cells building three-dimensional tumor clusters. Sci Rep 2018; 8:10503. [PMID: 30002435 PMCID: PMC6043561 DOI: 10.1038/s41598-018-28963-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/03/2018] [Indexed: 01/26/2023] Open
Abstract
Cellular senescence, a permanent cell-cycle arrest, is a common yet intriguing phenomenon, in which its beneficial significance for biological organisms has only begun to be explored. Among others, senescent cells are able to transform tissue structures around them. Tumor cells, whose hallmark is their ability to proliferate indefinitely, are not free from the phenomenon. Here, we report a remarkable observation where senescent cells in a dense mono-layer of breast cancer colony act as aggregating centers for non-senescent cells in their vicinity. Consequently, the senescent cells actively form localized 3D cell-clusters in a confluent 2D tumor layer. The biophysical mechanism underpinning the surprising phenomenon primarily involves mitotic cell-rounding, dynamic and differential cell attachments, and cellular chemotaxis. By incorporating these few biophysical factors, we were able to recapitulate the experimental observation via a cellular Potts Model.
Collapse
|
109
|
Abstract
Senescence is a double-edged sword that can function in opposite directions. It is a potential mechanism for a cell to avoid malignant transformation. However, senescence can also promote cancer development by altering the cellular microenvironment through a senescence-associated secretory phenotype (SASP). At least, three types of cellular stress such as activation of oncogenes, loss of tumor suppressor genes, and chemo/radiotherapy can induce cell senescence. Oncogene-induced senescence can be intertwiningly associated with the replicative senescence. Early-stage senescence may protect cell from transformation, while prolonged senescence often promotes cancer development. This review will focus on the characteristics of senescence, discuss the regulation of senescence during cancer development, and highlight the complexity of senescence that makes cancer treatment challenging.
Collapse
Affiliation(s)
- Sulin Zeng
- Department of Microbiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China.,Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY, USA
| | - Wen H Shen
- Department of Radiation Oncology, Weill Medical College of Cornell University, New York, NY, USA
| | - Li Liu
- Department of Microbiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
110
|
Wu X, Zhu L, Ma PC. Next-Generation Novel Noninvasive Cancer Molecular Diagnostics Platforms Beyond Tissues. Am Soc Clin Oncol Educ Book 2018; 38:964-977. [PMID: 30231325 PMCID: PMC6381937 DOI: 10.1200/edbk_199767] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In recent years, there has been a revolutionary expansion in technologic advances and therapeutic innovations in cancer medicine. Cancer diagnostics has begun to move away from a sole dependence on direct tumor tissue biopsy for cancer detection, diagnosis, and treatment monitoring. The need for improvement in molecular cancer diagnostics has never been more important, with not only the advent of cancer genomics and genomics-guided precision medicine but also the recent arrival of cancer immunotherapies. Owing to the practical limitations and risks associated with tissue-based biopsy diagnostics, novel noninvasive cancer diagnostics platforms have continued to evolve and expand in recent years. Examples of these platforms include the liquid biopsy, which is used to interrogate ctDNA or circulating tumor cells, proteomics, metabolomics, and exosomes; the urine biopsy, which is used to assay ctDNAs; saliva and stool biopsies, which are used for molecular genomics assays; and the breath biopsy, which measures volatile organic compounds. These next-generation noninvasive molecular diagnostics assays beyond tissues fundamentally transform the potential utilities of cancer diagnostics to enable repeat, prospective, and serial longitudinal "biopsies" to monitor disease response resistance and progression on therapies. Moreover, they allow continual interrogation and molecular in-depth analysis of the evolving tumor's pan-canceromics under therapeutic stress. These technological and diagnostic advances have already brought about paradigm-changing next-generation cancer therapeutic strategies to enhance overall treatment efficacies. This article reviews the key noninvasive next-generation molecular diagnostics platforms beyond tissues, with emphasis on clinical utilities and applications.
Collapse
Affiliation(s)
- Xiaoliang Wu
- From the West Virginia University Cancer Institute, West Virginia University Medicine, West Virginia University, Morgantown, WA; West Virginia Clinical and Translational Institute, Morgantown, WV
| | - Lin Zhu
- From the West Virginia University Cancer Institute, West Virginia University Medicine, West Virginia University, Morgantown, WA; West Virginia Clinical and Translational Institute, Morgantown, WV
| | - Patrick C Ma
- From the West Virginia University Cancer Institute, West Virginia University Medicine, West Virginia University, Morgantown, WA; West Virginia Clinical and Translational Institute, Morgantown, WV
| |
Collapse
|
111
|
Saleh T, Tyutynuk-Massey L, Cudjoe EK, Idowu MO, Landry JW, Gewirtz DA. Non-Cell Autonomous Effects of the Senescence-Associated Secretory Phenotype in Cancer Therapy. Front Oncol 2018; 8:164. [PMID: 29868482 PMCID: PMC5968105 DOI: 10.3389/fonc.2018.00164] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 04/30/2018] [Indexed: 12/24/2022] Open
Abstract
In addition to promoting various forms of cell death, most conventional anti-tumor therapies also promote senescence. There is now extensive evidence that therapy-induced senescence (TIS) might be transient, raising the concern that TIS could represent an undesirable outcome of therapy by providing a mechanism for tumor dormancy and eventual disease recurrence. The senescence-associated secretory phenotype (SASP) is a hallmark of TIS and may contribute to aberrant effects of cancer therapy. Here, we propose that the SASP may also serve as a major driver of escape from senescence and the re-emergence of proliferating tumor cells, wherein factors secreted from the senescent cells contribute to the restoration of tumor growth in a non-cell autonomous fashion. Accordingly, anti-SASP therapies might serve to mitigate the deleterious outcomes of TIS. In addition to providing an overview of the putative actions of the SASP, we discuss recent efforts to identify and eliminate senescent tumor cells.
Collapse
Affiliation(s)
- Tareq Saleh
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Liliya Tyutynuk-Massey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Emmanuel K Cudjoe
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, United States
| | - Michael O Idowu
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, United States
| | - Joseph W Landry
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States.,Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, United States.,Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
112
|
Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G, Cacciatore F, Bonaduce D, Abete P. Oxidative stress, aging, and diseases. Clin Interv Aging 2018; 13:757-772. [PMID: 29731617 PMCID: PMC5927356 DOI: 10.2147/cia.s158513] [Citation(s) in RCA: 2272] [Impact Index Per Article: 324.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen and nitrogen species (RONS) are produced by several endogenous and exogenous processes, and their negative effects are neutralized by antioxidant defenses. Oxidative stress occurs from the imbalance between RONS production and these antioxidant defenses. Aging is a process characterized by the progressive loss of tissue and organ function. The oxidative stress theory of aging is based on the hypothesis that age-associated functional losses are due to the accumulation of RONS-induced damages. At the same time, oxidative stress is involved in several age-related conditions (ie, cardiovascular diseases [CVDs], chronic obstructive pulmonary disease, chronic kidney disease, neurodegenerative diseases, and cancer), including sarcopenia and frailty. Different types of oxidative stress biomarkers have been identified and may provide important information about the efficacy of the treatment, guiding the selection of the most effective drugs/dose regimens for patients and, if particularly relevant from a pathophysiological point of view, acting on a specific therapeutic target. Given the important role of oxidative stress in the pathogenesis of many clinical conditions and aging, antioxidant therapy could positively affect the natural history of several diseases, but further investigation is needed to evaluate the real efficacy of these therapeutic interventions. The purpose of this paper is to provide a review of literature on this complex topic of ever increasing interest.
Collapse
Affiliation(s)
- Ilaria Liguori
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Gennaro Russo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Francesco Curcio
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giulia Bulli
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Luisa Aran
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.,San Raffaele Roma Open University, Rome, Italy
| | - Gaetano Gargiulo
- Division of Internal Medicine, AOU San Giovanni di Dio e Ruggi di Aragona, Salerno, Italy
| | - Gianluca Testa
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Francesco Cacciatore
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Azienda Ospedaliera dei Colli, Monaldi Hospital, Heart Transplantation Unit, Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
113
|
Abstract
Resistance of solid tumors to chemo- and radiotherapy remains a major obstacle in anti-cancer treatment. Herein, the membrane protein caveolin-1 (CAV1) came into focus as it is highly expressed in many tumors and high CAV1 levels were correlated with tumor progression, invasion and metastasis, and thus a worse clinical outcome. Increasing evidence further indicates that the heterogeneous tumor microenvironment, also known as the tumor stroma, contributes to therapy resistance resulting in poor clinical outcome. Again, CAV1 seems to play an important role in modulating tumor host interactions by promoting tumor growth, metastasis, therapy resistance and cell survival. However, the mechanisms driving stroma-mediated tumor growth and radiation resistance remain to be clarified. Understanding these interactions and thus, targeting CAV1 may offer a novel strategy for preventing cancer therapy resistance and improving clinical outcomes. In this review, we will summarize the resistance-promoting effects of CAV1 in tumors, and emphasize its role in the tumor-stroma communication as well as the resulting malignant phenotype of epithelial tumors.
Collapse
Affiliation(s)
- Julia Ketteler
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute for Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
114
|
Liu Y, Zhang J, Sun X, Su Q, You C. Down-regulation of miR-29b in carcinoma associated fibroblasts promotes cell growth and metastasis of breast cancer. Oncotarget 2018; 8:39559-39570. [PMID: 28465475 PMCID: PMC5503632 DOI: 10.18632/oncotarget.17136] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 02/27/2017] [Indexed: 01/08/2023] Open
Abstract
Carcinoma associated fibroblasts (CAFs) play important roles in breast cancer development and progression. Recent studies show that microRNAs (miRNAs) are the main regulators in CAFs. MiR-29b is one of the significant down-regulated miRNAs in CAFs from the miRNA screening. The role of miR-29b in the interaction between CAFs and breast cancer is still unclear. In the present study, we investigated the effects of CAFs on breast cancer cell proliferation and metastasis regulated by miR-29b. We found that fibroblasts activated by co-cultured breast cancer cells produced higher levels of some chemokines like CCL11, CXCL14, which accelerated breast cancer cell growth and induced drug resistance and metastasis. Increased miR-29b expression in activated fibroblasts could suppress the activating p38-STAT1 signal pathway in breast cancer cells. We also found that the expression of CCL11 and CXCL14 could be regulated by miR-29b in CAFs. Our results illustrate that down-regulation of miR-29b in CAFs plays an important role in tumor stroma by activating p38-STAT1 in breast cancer cells. The study indicates that cancer cells and fibroblasts interaction promotes breast cancer cell growth, drug resistance, migration and invasion due to the lack of miR-29b expression in CAFs.
Collapse
Affiliation(s)
- Yonglei Liu
- Research Center, Linyi People's Hospital, Shandong, China.,Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingling Zhang
- Research Center, Linyi People's Hospital, Shandong, China
| | - Xiangjun Sun
- Department of Surgery, Linyi People's Hospital, Shandong, China
| | - Quanping Su
- Research Center, Linyi People's Hospital, Shandong, China
| | - Cuiping You
- Research Center, Linyi People's Hospital, Shandong, China
| |
Collapse
|
115
|
Senoptosis: non-lethal DNA cleavage as a route to deep senescence. Oncotarget 2018; 8:30656-30671. [PMID: 28427150 PMCID: PMC5458157 DOI: 10.18632/oncotarget.15693] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
DNA-damage-induced apoptosis and cellular senescence are perceived as two distinct cell fates. We found that after ionizing radiation (IR)-induced DNA damage the majority (up to 70 %) of senescent human diploid fibroblasts (HDFs) were subjected to controlled cleavage of DNA, resulting in the establishment of a viable and stable sub-G1 population, i.e. deeply senescent cells. We show that in senescent HDFs this DNA cleavage is triggered by modest loss of the mitochondrial membrane potential, which is not sufficient to activate caspases, but strong enough to release mitochondrial endonuclease G (EndoG). We demonstrate that upon γ-irradiation in HDFs EndoG translocates into the nucleus playing an essential role in the non-lethal cleavage of damaged DNA. Notably, the established sub-G1 cell population does not contribute to the senescence-associated secretory phenotype (SASP), however, it exhibits increased senescence-associated β-galactosidase activity. We show that EndoG knockdown causes an increase in DNA damage, indicating a role of this enzyme in DNA repair. Thus, we conclude that IR-induced deep senescence of HDFs exhibits features of both senescence, such as cell cycle arrest and viability, and apoptosis like reduced DNA content and no SASP, and, resembles uncomplete or stalled apoptosis, a phenomenon we term senoptosis.
Collapse
|
116
|
Varela-Eirin M, Loureiro J, Fonseca E, Corrochano S, Caeiro JR, Collado M, Mayan MD. Cartilage regeneration and ageing: Targeting cellular plasticity in osteoarthritis. Ageing Res Rev 2018; 42:56-71. [PMID: 29258883 DOI: 10.1016/j.arr.2017.12.006] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/20/2017] [Accepted: 12/15/2017] [Indexed: 01/15/2023]
Abstract
Ageing processes play a major contributing role for the development of Osteoarthritis (OA). This prototypic degenerative condition of ageing is the most common form of arthritis and is accompanied by a general decline, chronic pain and mobility deficits. The disease is primarily characterized by articular cartilage degradation, followed by subchondral bone thickening, osteophyte formation, synovial inflammation and joint degeneration. In the early stages, osteoarthritic chondrocytes undergo phenotypic changes that increase cell proliferation and cluster formation and enhance the production of matrix-remodelling enzymes. In fact, chondrocytes exhibit differentiation plasticity and undergo phenotypic changes during the healing process. Current studies are focusing on unravelling whether OA is a consequence of an abnormal wound healing response. Recent investigations suggest that alterations in different proteins, such as TGF-ß/BMPs, NF-Kß, Wnt, and Cx43, or SASP factors involved in signalling pathways in wound healing response, could be directly implicated in the initiation of OA. Several findings suggest that osteoarthritic chondrocytes remain in an immature state expressing stemness-associated cell surface markers. In fact, the efficacy of new disease-modifying OA drugs that promote chondrogenic differentiation in animal models indicates that this may be a drug-sensible state. In this review, we highlight the current knowledge regarding cellular plasticity in chondrocytes and OA. A better comprehension of the mechanisms involved in these processes may enable us to understand the molecular pathways that promote abnormal repair and cartilage degradation in OA. This understanding would be advantageous in identifying novel targets and designing therapies to promote effective cartilage repair and successful joint ageing by preventing functional limitations and disability.
Collapse
Affiliation(s)
- Marta Varela-Eirin
- CellCOM research group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | - Jesus Loureiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Eduardo Fonseca
- CellCOM research group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | | | - Jose R Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Manuel Collado
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Maria D Mayan
- CellCOM research group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, 84, 15006 A Coruña, Spain.
| |
Collapse
|
117
|
Transcriptional response to organic compounds from diverse gasoline and biogasoline fuel emissions in human lung cells. Toxicol In Vitro 2018; 48:329-341. [PMID: 29432896 DOI: 10.1016/j.tiv.2018.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 01/15/2023]
Abstract
Modern vehicles equipped with Gasoline Direct Injection (GDI) engine have emerged as an important source of particulate emissions potentially harmful to human health. We collected and characterized gasoline exhaust particles (GEPs) produced by neat gasoline fuel (E0) and its blends with 15% ethanol (E15), 25% n-butanol (n-But25) and 25% isobutanol (i-But25). To study the toxic effects of organic compounds extracted from GEPs, we analyzed gene expression profiles in human lung BEAS-2B cells. Despite the lowest GEP mass, n-But25 extract contained the highest concentration of polycyclic aromatic hydrocarbons (PAHs), while i-But25 extract the lowest. Gene expression analysis identified activation of the DNA damage response and other subsequent events (cell cycle arrest, modulation of extracellular matrix, cell adhesion, inhibition of cholesterol biosynthesis) following 4 h exposure to all GEP extracts. The i-But25 extract induced the most distinctive gene expression pattern particularly after 24 h exposure. Whereas E0, E15 and n-But25 extract treatments resulted in persistent stress signaling including DNA damage response, MAPK signaling, oxidative stress, metabolism of PAHs or pro-inflammatory response, i-But25 induced changes related to the metabolism of the cellular nutrients required for cell recovery. Our results indicate that i-But25 extract possessed the weakest genotoxic potency possibly due to the low PAH content.
Collapse
|
118
|
Haston S, Manshaei S, Martinez-Barbera JP. Stem/progenitor cells in pituitary organ homeostasis and tumourigenesis. J Endocrinol 2018; 236:R1-R13. [PMID: 28855316 PMCID: PMC5744558 DOI: 10.1530/joe-17-0258] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/30/2017] [Indexed: 01/06/2023]
Abstract
Evidence for the presence of pituitary gland stem cells has been provided over the last decade using a combination of approaches including in vitro clonogenicity assays, flow cytometric side population analysis, immunohistochemical analysis and genetic approaches. These cells have been demonstrated to be able to self-renew and undergo multipotent differentiation to give rise to all hormonal lineages of the anterior pituitary. Furthermore, evidence exists for their contribution to regeneration of the organ and plastic responses to changing physiological demand. Recently, stem-like cells have been isolated from pituitary neoplasms raising the possibility that a cytological hierarchy exists, in keeping with the cancer stem cell paradigm. In this manuscript, we review the evidence for the existence of pituitary stem cells, their role in maintaining organ homeostasis and the regulation of their differentiation. Furthermore, we explore the emerging concept of stem cells in pituitary tumours and their potential roles in these diseases.
Collapse
Affiliation(s)
- Scott Haston
- Developmental Biology and Cancer Research ProgrammeBirth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Saba Manshaei
- Developmental Biology and Cancer Research ProgrammeBirth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Research ProgrammeBirth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
119
|
Bayraktar R, Van Roosbroeck K, Calin GA. Cell-to-cell communication: microRNAs as hormones. Mol Oncol 2017; 11:1673-1686. [PMID: 29024380 PMCID: PMC5709614 DOI: 10.1002/1878-0261.12144] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/24/2017] [Indexed: 12/11/2022] Open
Abstract
Mammalian cells can release different types of extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies. Accumulating evidence suggests that EVs play a role in cell-to-cell communication within the tumor microenvironment. EVs' components, such as proteins, noncoding RNAs [microRNAs (miRNAs), and long noncoding RNAs (lncRNAs)], messenger RNAs (mRNAs), DNA, and lipids, can mediate paracrine signaling in the tumor microenvironment. Recently, miRNAs encapsulated in secreted EVs have been identified in the extracellular space. Mature miRNAs that participate in intercellular communication are released from most cells, often within EVs, and disseminate through the extracellular fluid to reach remote target cells, including tumor cells, whose phenotypes they can influence by regulating mRNA and protein expression either as tumor suppressors or as oncogenes, depending on their targets. In this review, we discuss the roles of miRNAs in intercellular communication, the biological function of extracellular miRNAs, and their potential applications for diagnosis and therapeutics. We will give examples of miRNAs that behave as hormones.
Collapse
Affiliation(s)
- Recep Bayraktar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katrien Van Roosbroeck
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
120
|
Dolivo D, Hernandez S, Dominko T. Cellular lifespan and senescence: a complex balance between multiple cellular pathways. Bioessays 2017; 38 Suppl 1:S33-44. [PMID: 27417120 DOI: 10.1002/bies.201670906] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/14/2015] [Accepted: 09/17/2015] [Indexed: 11/09/2022]
Abstract
The study of cellular senescence and proliferative lifespan is becoming increasingly important because of the promises of autologous cell therapy, the need for model systems for tissue disease and the implication of senescent cell phenotypes in organismal disease states such as sarcopenia, diabetes and various cancers, among others. Here, we explain the concepts of proliferative cellular lifespan and cellular senescence, and we present factors that have been shown to mediate cellular lifespan positively or negatively. We review much recent literature and present potential molecular mechanisms by which lifespan mediation occurs, drawing from the fields of telomere biology, metabolism, NAD(+) and sirtuin biology, growth factor signaling and oxygen and antioxidants. We conclude that cellular lifespan and senescence are complex concepts that are governed by multiple independent and interdependent pathways, and that greater understanding of these pathways, their interactions and their convergence upon specific cellular phenotypes may lead to viable therapies for tissue regeneration and treatment of age-related pathologies, which are caused by or exacerbated by senescent cells in vivo.
Collapse
Affiliation(s)
- David Dolivo
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Sarah Hernandez
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Tanja Dominko
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| |
Collapse
|
121
|
Trivanović D, Krstić J, Jauković A, Bugarski D, Santibanez JF. Mesenchymal stromal cell engagement in cancer cell epithelial to mesenchymal transition. Dev Dyn 2017; 247:359-367. [PMID: 28850772 DOI: 10.1002/dvdy.24583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 07/04/2017] [Accepted: 08/10/2017] [Indexed: 12/14/2022] Open
Abstract
Due to coexistence of stromal and epithelial tumor cells, their dynamic interactions have been widely recognized as significant cellular components to the tumor tissue integrity. Initiation and outcome of epithelial to mesenchymal transition (EMT) in tumor cells are dependent on their interaction with adjacent or recruited mesenchymal stromal cells (MSCs). A plethora of mechanisms are involved in MSCs-controlled employment of the developmental processes of EMT that contribute to loss of epithelial cell phenotype and acquisition of stemness, invasiveness and chemoresistance of tumor cells. Interplay of MSCs with tumor cells, including interchange of soluble biomolecules, plasma membrane structures, cytoplasmic content, and organelles, is established through cell-cell contact and/or by means of paracrine signaling. The main focus of this review is to summarize knowledge about involvement of MSCs in cancer cell EMT. Understanding the underlying cellular and molecular mechanism involved in the interplay between MSCs and cancer EMT is essential for development of effective therapy approaches, which in combination with current treatments may improve the control of tumor progression. Developmental Dynamics 247:359-367, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Jelena Krstić
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| |
Collapse
|
122
|
Kastenhuber ER, Lowe SW. Putting p53 in Context. Cell 2017; 170:1062-1078. [PMID: 28886379 DOI: 10.1016/j.cell.2017.08.028] [Citation(s) in RCA: 1359] [Impact Index Per Article: 169.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/09/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
TP53 is the most frequently mutated gene in human cancer. Functionally, p53 is activated by a host of stress stimuli and, in turn, governs an exquisitely complex anti-proliferative transcriptional program that touches upon a bewildering array of biological responses. Despite the many unveiled facets of the p53 network, a clear appreciation of how and in what contexts p53 exerts its diverse effects remains unclear. How can we interpret p53's disparate activities and the consequences of its dysfunction to understand how cell type, mutation profile, and epigenetic cell state dictate outcomes, and how might we restore its tumor-suppressive activities in cancer?
Collapse
Affiliation(s)
- Edward R Kastenhuber
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Scott W Lowe
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, New York, NY 10065, USA.
| |
Collapse
|
123
|
MicroRNA Profiling Reveals Distinct Profiles for Tissue-Derived and Cultured Endothelial Cells. Sci Rep 2017; 7:10943. [PMID: 28887500 PMCID: PMC5591252 DOI: 10.1038/s41598-017-11487-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/25/2017] [Indexed: 12/19/2022] Open
Abstract
Endothelial plasticity enables the cells to switch their phenotype according to the surrounding vascular microenvironment. MicroRNAs (miRNAs) are small noncoding RNAs that control endothelial plasticity. The objective of this study was to investigate the differences in miRNA profiles of tissue-derived cells and cultured endothelial cells. To this end, miRNA expression was profiled from freshly isolated tissue-derived human vascular endothelial cells and endothelial cells cultured until cellular senescence using miRNA sequencing. In addition, the data was searched for putative novel endothelial miRNAs and miRNA isoforms. The data analysis revealed a striking change in endothelial miRNA profile as the cells adapted from tissue to cell culture environment and the overall miRNA expression decreased significantly in cultured compared to tissue-derived endothelial cells. In addition to changes in mechanosensitive miRNA expression, alterations in senescence-associated and endothelial-to-mesenchymal-transition-associated miRNAs were observed in aging cells. Collectively, the data illustrates the adaptability of endothelial cell miRNA expression that mirrors prevailing cellular environment.
Collapse
|
124
|
Wang WJ, Cai GY, Chen XM. Cellular senescence, senescence-associated secretory phenotype, and chronic kidney disease. Oncotarget 2017; 8:64520-64533. [PMID: 28969091 PMCID: PMC5610023 DOI: 10.18632/oncotarget.17327] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022] Open
Abstract
Chronic kidney disease (CKD) is increasingly being accepted as a type of renal ageing. The kidney undergoes age-related alterations in both structure and function. To date, a comprehensive analysis of cellular senescence and senescence-associated secretory phenotype (SASP) in CKD is lacking. Hence, this review mainly discusses the relationship between the two phenomena to show the striking similarities between SASP and CKD-associated secretory phenotype (CASP). It has been reported that replicative senescence, stress-induced premature ageing, and epigenetic abnormalities participate in the occurrence and development of CKD. Genomic damage and external environmental stimuli cause increased levels of oxidative stress and a chronic inflammatory state as a result of irreversible cell cycle arrest and low doses of SASP. Similar to SASP, CASP factors activate tissue repair by multiple mechanisms. Once tissue repair fails, the accumulated SASP or CASP species aggravate DNA damage response (DDR) and cause the senescent cells to secrete more SASP factors, accelerating the process of cellular ageing and eventually leading to various ageing-related changes. It is concluded that cellular senescence and SASP participate in the pathological process of CKD, and correspondingly CKD accelerated the progression of cell senescence and the secretion of SASP. These results will facilitate the integration of these mechanisms into the care and management of CKD and other age-related diseases.
Collapse
Affiliation(s)
- Wen-Juan Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
- Department of Nephrology, Beijing Changping Hospital, Beijing 102200, China
| | - Guang-Yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| |
Collapse
|
125
|
Yang L, Xing S, Wang K, Yi H, Du B. Paeonol attenuates aging MRC-5 cells and inhibits epithelial–mesenchymal transition of premalignant HaCaT cells induced by aging MRC-5 cell-conditioned medium. Mol Cell Biochem 2017; 439:117-129. [DOI: 10.1007/s11010-017-3141-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 08/02/2017] [Indexed: 12/15/2022]
|
126
|
Jin J, Tao J, Gu X, Yu Z, Wang R, Zuo G, Li Q, Lv X, Miao D. P16 INK4a Deletion Ameliorated Renal Tubulointerstitial Injury in a Stress-induced Premature Senescence Model of Bmi-1 Deficiency. Sci Rep 2017; 7:7502. [PMID: 28790310 PMCID: PMC5548892 DOI: 10.1038/s41598-017-06868-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
To determine whether p16 INK4a deletion ameliorated renal tubulointerstitial injury by inhibiting a senescence-associated secretory phenotype (SASP) in Bmi-1-deficient (Bmi-1 -/-) mice, renal phenotypes were compared among 5-week-old Bmi-1 and p16 INK4a double-knockout, and Bmi-1 -/- and wild-type mice. Fifth-passage renal interstitial fibroblasts (RIFs) from the three groups were analyzed for senescence and proliferation. The effect of Bmi-1 deficiency on epithelial-to-mesenchymal transition (EMT) was examined in Bmi-1-knockdown human renal proximal tubular epithelial (HK2) cells, which were treated with concentrated conditioned medium (CM) from the fifth-passage renal interstitial fibroblasts (RIFs) of above three group mice or with exogenous TGF-β1. Our results demonstrated that p16 INK4a deletion largely rescued renal aging phenotypes caused by Bmi-1 deficiency, including impaired renal structure and function, decreased proliferation, increased apoptosis, senescence and SASP, DNA damage, NF-κB and TGF-β1/Smad signal activation, inflammatory cell infiltration, and tubulointerstitial fibrosis and tubular atrophy. P16 INK4a deletion also promoted proliferation, reduced senescence and SASP of RIFs and subsequently inhibited EMT of Bmi-1-knockdown HK2 cells. TGF-β1 further induced the EMT of Bmi-1-knockdown HK2 cells. Thus, p16 INK4a positive senescent cells would be a therapeutic target for preventing renal tubulointerstitial injury.
Collapse
MESH Headings
- Acute Kidney Injury/genetics
- Acute Kidney Injury/metabolism
- Acute Kidney Injury/pathology
- Acute Kidney Injury/prevention & control
- Animals
- Cell Line, Transformed
- Cell Proliferation
- Cellular Senescence
- Coculture Techniques
- Culture Media, Conditioned/pharmacology
- Cyclin-Dependent Kinase Inhibitor p16/deficiency
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Epithelial-Mesenchymal Transition/genetics
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Gene Expression Regulation
- Humans
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Mice
- Mice, Knockout
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/metabolism
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/prevention & control
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Polycomb Repressive Complex 1/deficiency
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Smad Proteins/genetics
- Smad Proteins/metabolism
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/pharmacology
Collapse
Affiliation(s)
- Jianliang Jin
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianguo Tao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xin Gu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhenzhen Yu
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Rong Wang
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Guoping Zuo
- Laboratory Centre for Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qing Li
- Department of Science and Technology, Jiangsu Jiankang Vocational College, Nanjing, Jiangsu, 210029, China
| | - Xianhui Lv
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Dengshun Miao
- The State Key Laboratory of Reproductive Medicine; Key Laboratory for Aging & Disease, Research Centre for Bone and Stem Cells, Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
127
|
Capece D, Verzella D, Tessitore A, Alesse E, Capalbo C, Zazzeroni F. Cancer secretome and inflammation: The bright and the dark sides of NF-κB. Semin Cell Dev Biol 2017; 78:51-61. [PMID: 28779979 DOI: 10.1016/j.semcdb.2017.08.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/29/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022]
Abstract
Tumour promoting inflammation is widely recognized as a hallmark of cancer. The source of this chronic inflammation in cancer has been ascribed to the progressive activation over time of immune cells, mostly of the innate arm of the immune system. However, recent evidence has shown that chronic inflammation may also derive, at least in part, from senescent cells. Hence, due to the prominent role of inflammation in cancer, the cancer secretome definition includes all the secretory factors ensuing from the crosstalk between the cancer cell and the tumour microenvironment. The mechanistic basis underlying the paracrine signalling between the cancer cell and the surrounding tumour microenvironment in malignancy have been widely investigated by using in vivo models of cancers, thus identifying the NF-κB transcription factor as the molecular hub linking inflammation and cancer. In this review, we highlight the roles of NF-κB in regulating the inflammation-derived secretome emanating from immune and senescent cells, with a special focus on the bright and the dark sides of their pro-inflammatory signalling on tumorigenesis.
Collapse
Affiliation(s)
- Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy; Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy; Centre for Cell Signalling and Inflammation, Department of Medicine, Imperial College London, London W12 0NN, UK.
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy.
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy.
| | - Carlo Capalbo
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
128
|
Cohen J, Torres C. HIV-associated cellular senescence: A contributor to accelerated aging. Ageing Res Rev 2017; 36:117-124. [PMID: 28017881 DOI: 10.1016/j.arr.2016.12.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/28/2016] [Accepted: 12/12/2016] [Indexed: 01/15/2023]
Abstract
Due to the advent of antiretroviral therapy HIV is no longer a terminal disease and the HIV infected patients are becoming increasingly older. While this is a major success, with increasing age comes an increased risk for disease. The age-related comorbidities that HIV infected patients experience suggest that they suffer from accelerated aging. One possible contributor to this accelerated aging is cellular senescence, an age-associated response that can occur prematurely in response to stress, and that is emerging as a contributor to disease and aging. HIV patients experience several stressors such as the virus itself, antiretroviral drugs and to a lesser extent, substance abuse that can induce cellular senescence. This review summarizes the current knowledge of senescence induction in response to these stressors and their relation to the comorbidities in HIV patients. Cellular senescence may be a possible therapeutic target for these comorbidities.
Collapse
|
129
|
Srdic-Rajic T, Santibañez JF, Kanjer K, Tisma-Miletic N, Cavic M, Galun D, Jevric M, Kardum N, Konic-Ristic A, Zoranovic T. Iscador Qu inhibits doxorubicin-induced senescence of MCF7 cells. Sci Rep 2017. [PMID: 28630419 PMCID: PMC5476621 DOI: 10.1038/s41598-017-03898-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chemotherapy in patients with inoperable or advanced breast cancer inevitably results in low-dose exposure of tumor-cell subset and senescence. Metabolically active senescent cells secrete multiple tumor promoting factors making their elimination a therapeutic priority. Viscum album is one of the most widely used alternative anti-cancer medicines facilitating chemotherapy tolerance of breast cancer patients. The aim of this study was to model and investigate how Viscum album extracts execute additive anti-tumor activity with low-dose Dox using ER + MCF7 breast cancer cells. We report that cotreatment of MCF7 with Viscum album and Dox abrogates G2/M cycle arrest replacing senescence with intrinsic apoptotic program. Mechanistically, this switch was associated with down-regulation of p21, p53/p73 as well as Erk1/2 and p38 activation. Our findings, therefore, identify a novel mechanistic axis of additive antitumor activity of Viscum album and low dose-Dox. In conclusion, ER + breast cancer patients may benefit from addition of Viscum album to low-dose Dox chemotherapy due to suppression of cancer cell senescence and induction of apoptosis.
Collapse
Affiliation(s)
- Tatjana Srdic-Rajic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Juan F Santibañez
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia.,Laboratorio de Bionanotecnologia, Universidad Bernardo O Higgins, General Gana 1780, 8370854, Santiago, Chile
| | - Ksenija Kanjer
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Nevena Tisma-Miletic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Milena Cavic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia
| | - Daniel Galun
- University Clinic for Digestive Surgery, Clinical center of Serbia, Belgrade, Serbia.,Medical School, University of Belgrade, Belgrade, Serbia
| | - Marko Jevric
- Department of Surgery, National Cancer Research Center, Belgrade, Serbia
| | - Nevena Kardum
- Institute for Medical Research, Center of Research Excellence in Nutrition and Metabolism, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Konic-Ristic
- Institute for Medical Research, Center of Research Excellence in Nutrition and Metabolism, University of Belgrade, Belgrade, Serbia
| | - Tamara Zoranovic
- Department of Experimental Oncology, National Cancer Research Center, Belgrade, Serbia. .,Max Plank Institute for Infection Biology, Berlin Area, Germany.
| |
Collapse
|
130
|
Liu Y, Yu C, Wu Y, Sun X, Su Q, You C, Xin H. CD44 + fibroblasts increases breast cancer cell survival and drug resistance via IGF2BP3-CD44-IGF2 signalling. J Cell Mol Med 2017; 21:1979-1988. [PMID: 28523716 PMCID: PMC5571562 DOI: 10.1111/jcmm.13118] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022] Open
Abstract
CD44, a cell adhesion protein, involves in various process in cancer such as cell survival and metastasis. Most researches on CD44 in cancer focus on cancer cells. Recently, it is found that CD44 expression is high in fibroblasts of tumour microenvironment. However, its role in communication between fibroblasts and breast cancer cells is seldom known. In this study, CD44-positive (CD44+ Fbs) and CD44-negative carcinoma-associated fibroblasts (CD44- Fbs) were isolated and cocultured with breast cancer cells for analysis of cell survival and drug resistance. We found that CD44+ Fbs promoted breast cancer cell survival and paclitaxel resistance and inhibited paclitaxel-induced apoptosis. Our further research for the molecular mechanism showed that IGF2BP3 bound to CD44 mRNA and enhanced CD44 expression, which increased IGF2 levels of fibroblasts and then stimulated breast cancer cell proliferation and drug resistance. IGF2 was found to activate Hedgehog signal pathway in breast cancer cells. In conclusion, the results illustrated that in CD44+ Fbs, binding of IGF2BP3 and CD44 promotes IGF2 expression and then accelerates breast cancer cell proliferation, survival and induced chemotherapy resistance likely by activating Hedgehog signal pathways.
Collapse
Affiliation(s)
- Yonglei Liu
- Research Center, Linyi People's Hospital, Shandong, China.,Zhongshan Hospital, Fudan University, Shanghai, China.,The First Peoples' Hospital, School of Clinical Medicine, Yangtze University, Jingzhou, Hubei, China.,Laboratory of Oncology, Center for Molecular Medicine, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Conghui Yu
- Department of Hepatobilinary Surgery, The General Hospital of Beijing Military Region of China, Beijing, China
| | - Yonggang Wu
- Department of Orthopedics, Bayannaoer City Hospital, Bayannaoer City, Inner Mongolia, China
| | - Xiangjun Sun
- Department of Surgery, Linyi People's Hospital, Shandong, China
| | - Quanping Su
- Research Center, Linyi People's Hospital, Shandong, China
| | - Cuiping You
- Research Center, Linyi People's Hospital, Shandong, China
| | - Hongwu Xin
- The First Peoples' Hospital, School of Clinical Medicine, Yangtze University, Jingzhou, Hubei, China.,Laboratory of Oncology, Center for Molecular Medicine, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
131
|
Samaraweera L, Adomako A, Rodriguez-Gabin A, McDaid HM. A Novel Indication for Panobinostat as a Senolytic Drug in NSCLC and HNSCC. Sci Rep 2017; 7:1900. [PMID: 28507307 PMCID: PMC5432488 DOI: 10.1038/s41598-017-01964-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 04/07/2017] [Indexed: 01/07/2023] Open
Abstract
Panobinostat (pano) is an FDA-approved histone deacetylase inhibitor. There is interest in evaluating alternate dosing schedules and novel combinations of pano for the treatment of upper aerodigestive and lung malignancies; thus we evaluated it in combination with Taxol, a chemotherapeutic with activity in both diseases. Dose-dependent synergy was observed in Non-Small Cell Lung Cancer (NSCLC) and Head and Neck Squamous Cell Carcinoma (HNSCC) cell lines and was due to senescence rather than potentiation of cell death. Senescence occurred following cisplatin- or Taxol-treatment in cell lines from both cancer types and was associated with decreased histone 3 (H3) acetylation and increased Bcl-xL expression: the latter a biomarker of senescence and target of anti-senescence therapeutics, or senolytics. Since H3 acetylation and Bcl-xL expression were altered in senescence, we subsequently evaluated pano as a senolytic in chemotherapy-treated cancer cells enriched for senescent cells. Pano caused cell death at significantly higher rates compared to repeat dosing with chemotherapy. This was associated with decreased expression of Bcl-xL and increased acetylated H3, reversing the expression patterns observed in senescence. These data support evaluating pano as a post-chemotherapy senolytic with the potential to kill persistent senescent cells that accumulate during standard chemotherapy in NSCLC and HNSCC.
Collapse
Affiliation(s)
- Leleesha Samaraweera
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alfred Adomako
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Alicia Rodriguez-Gabin
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Hayley M McDaid
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
132
|
Perrott KM, Wiley CD, Desprez PY, Campisi J. Apigenin suppresses the senescence-associated secretory phenotype and paracrine effects on breast cancer cells. GeroScience 2017; 39:161-173. [PMID: 28378188 PMCID: PMC5411372 DOI: 10.1007/s11357-017-9970-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/17/2017] [Indexed: 12/16/2022] Open
Abstract
Apigenin (4',5,7,-trihydroxyflavone) is a flavonoid found in certain herbs, fruits, and vegetables. Apigenin can attenuate inflammation, which is associated with many chronic diseases of aging. Senescent cells-stressed cells that accumulate with age in mammals-display a pro-inflammatory senescence-associated secretory phenotype (SASP) that can drive or exacerbate several age-related pathologies, including cancer. Flavonoids, including apigenin, were recently shown to reduce the SASP of a human fibroblast strain induced to senesce by bleomycin. Here, we confirm that apigenin suppresses the SASP in three human fibroblast strains induced to senesce by ionizing radiation, constitutive MAPK (mitogen-activated protein kinase) signaling, oncogenic RAS, or replicative exhaustion. Apigenin suppressed the SASP in part by suppressing IL-1α signaling through IRAK1 and IRAK4, p38-MAPK, and NF-κB. Apigenin was particularly potent at suppressing the expression and secretion of CXCL10 (IP10), a newly identified SASP factor. Further, apigenin-mediated suppression of the SASP substantially reduced the aggressive phenotype of human breast cancer cells, as determined by cell proliferation, extracellular matrix invasion, and epithelial-mesenchymal transition. Our results support the idea that apigenin is a promising natural product for reducing the impact of senescent cells on age-related diseases such as cancer.
Collapse
Affiliation(s)
- Kevin M Perrott
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Christopher D Wiley
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA
| | - Pierre-Yves Desprez
- California Pacific Medical Center Research Institute, 475 Brannan Street, San Francisco, CA, 94107, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.
- Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA, 94720, USA.
| |
Collapse
|
133
|
Chandrasekaran A, Idelchik MDPS, Melendez JA. Redox control of senescence and age-related disease. Redox Biol 2017; 11:91-102. [PMID: 27889642 PMCID: PMC5126126 DOI: 10.1016/j.redox.2016.11.005] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 11/10/2016] [Indexed: 12/17/2022] Open
Abstract
The signaling networks that drive the aging process, associated functional deterioration, and pathologies has captured the scientific community's attention for decades. While many theories exist to explain the aging process, the production of reactive oxygen species (ROS) provides a signaling link between engagement of cellular senescence and several age-associated pathologies. Cellular senescence has evolved to restrict tumor progression but the accompanying senescence-associated secretory phenotype (SASP) promotes pathogenic pathways. Here, we review known biological theories of aging and how ROS mechanistically control senescence and the aging process. We also describe the redox-regulated signaling networks controlling the SASP and its important role in driving age-related diseases. Finally, we discuss progress in designing therapeutic strategies that manipulate the cellular redox environment to restrict age-associated pathology.
Collapse
Affiliation(s)
- Akshaya Chandrasekaran
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, Albany, NY 12203, USA
| | | | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, 257 Fuller Road, Albany, NY 12203, USA.
| |
Collapse
|
134
|
Yang X, Liu S, Huang C, Wang H, Luo Y, Xu W, Huang K. Ochratoxin A induced premature senescence in human renal proximal tubular cells. Toxicology 2017; 382:75-83. [PMID: 28286205 DOI: 10.1016/j.tox.2017.03.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 03/07/2017] [Accepted: 03/07/2017] [Indexed: 12/12/2022]
Abstract
Ochratoxin A (OTA) has many nephrotoxic effects and is a promising compound for the study of nephrotoxicity. Human renal proximal tubular cells (HKC) are an important model for the study of renal reabsorption, renal physiology and pathology. Since the induction of OTA in renal senescence is largely unknown, whether OTA can induce renal senescence, especially at a sublethal dose, and the mechanism of OTA toxicity remain unclear. In our study, a sublethal dose of OTA led to an enhanced senescent phenotype, β-galactosidase staining and senescence associated secretory phenotype (SASP). Cell cycle arrest and cell shape alternations also confirmed senescence. In addition, telomere analysis by RT-qPCR allowed us to classify OTA-induced senescence as a premature senescence. Western blot assays showed that the p53-p21 and the p16-pRB pathways and the ezrin-associated cell spreading changes were activated during the OTA-induced senescence of HKC. In conclusion, our results demonstrate that OTA promotes the senescence of HKC through the p53-p21 and p16-pRB pathways. The understanding of the mechanisms of OTA-induced senescence is critical in determining the role of OTA in cytotoxicity and its potential carcinogenicity.
Collapse
Affiliation(s)
- Xuan Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Sheng Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Chuchu Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Haomiao Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yunbo Luo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Wentao Xu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Kunlun Huang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China.
| |
Collapse
|
135
|
Abstract
Post-translational protein modification by small ubiquitin-like modifier (SUMO), termed sumoylation, is an important mechanism in cellular responses to stress and one that appears to be upregulated in many cancers. Here, we examine the role of sumoylation in tumorigenesis as a possibly necessary safeguard that protects the stability and functionality of otherwise easily misregulated gene expression programmes and signalling pathways of cancer cells.
Collapse
Affiliation(s)
- Jacob-Sebastian Seeler
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Institut Pasteur, 28 rue de Dr Roux, 75724 Paris Cedex 15, France
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, INSERM U993, Institut Pasteur, 28 rue de Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
136
|
Yang M, Liu P, Wang K, Glorieux C, Hu Y, Wen S, Jiang W, Huang P. Chemotherapy induces tumor immune evasion by upregulation of programmed cell death ligand 1 expression in bone marrow stromal cells. Mol Oncol 2017; 11:358-372. [PMID: 28218497 PMCID: PMC5527486 DOI: 10.1002/1878-0261.12032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) is a negative regulator of the immune response that enables tumor cells to escape T-cell immunity. Although PD-L1 expression in cancer cells has been extensively studied, the expression of PD-L1 in stromal cells and its clinical significance remain largely unknown. Here, we show that bone marrow stromal cells express a low level of PD-L1 and that this molecule is significantly upregulated by key drugs used in the treatment of lymphoma at clinically relevant concentrations. Mechanistically, chemotherapeutic drugs induce PD-L1 expression in stromal cells through upregulation of granulocyte macrophage colony-stimulating factor and activation of the extracellular signal-regulated kinase (ERK) 1/2 signaling pathway. Suppression of ERK by a chemical inhibitor or genetic silencing of ERK2 expression prevents drug-induced PD-L1 expression. PD-L1 expression is upregulated in the bone marrow stromal cells of mice treated with doxorubicin and in drug-treated bone marrow specimens from lymphoma patients. Drug-induced PD-L1 expression in stromal cells can cause significant impairment of T-cell functions. Overall, our study reveals a previously unrecognized mechanism by which chemotherapy induces tumor immune evasion by upregulation of PD-L1 in bone marrow stromal cells, and provides new evidence for the combination of chemotherapy and anti-PD-L1/PD-1 as an effective strategy for treatment of lymphoma and other cancers.
Collapse
Affiliation(s)
- Mengqi Yang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Panpan Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Kefeng Wang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Christophe Glorieux
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yumin Hu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shijun Wen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenqi Jiang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Peng Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
137
|
Thiele JA, Bethel K, Králíčková M, Kuhn P. Circulating Tumor Cells: Fluid Surrogates of Solid Tumors. ANNUAL REVIEW OF PATHOLOGY 2017; 12:419-447. [PMID: 28135562 PMCID: PMC7846475 DOI: 10.1146/annurev-pathol-052016-100256] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Evaluation of circulating tumor cells (CTCs) has demonstrated clinical validity as a prognostic tool based on enumeration, but since the introduction of this tool to the clinic in 2004, further clinical utility and widespread adoption have been limited. However, immense efforts have been undertaken to further the understanding of the mechanisms behind the biology and kinetics of these rare cells, and progress continues toward better applicability in the clinic. This review describes recent advances within the field, with a particular focus on understanding the biological significance of CTCs, and summarizes emerging methods for identifying, isolating, and interrogating the cells that may provide technical advantages allowing for the discovery of more specific clinical applications. Included is an atlas of high-definition images of CTCs from various cancer types, including uncommon CTCs captured only by broadly inclusive nonenrichment techniques.
Collapse
Affiliation(s)
- J-A Thiele
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University in Prague, 323 00 Pilsen, Czech Republic
| | - K Bethel
- Scripps Clinic Medical Group, Scripps Clinic, La Jolla, California 92121
| | - M Králíčková
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University in Prague, 301 00 Pilsen, Czech Republic
| | - P Kuhn
- Bridge Institute, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California 90089;
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
138
|
Morgans AK, Dale W, Briganti A. Screening and Treating Prostate Cancer in the Older Patient: Decision Making Across the Clinical Spectrum. Am Soc Clin Oncol Educ Book 2017; 37:370-381. [PMID: 28561697 DOI: 10.1200/edbk_175491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Treatment of the growing geriatric patient population is increasingly being recognized as a necessary priority of the oncology community. As the most common cancer among men in developed countries, prostate cancer afflicts a sizable portion of elderly men. Caring for this population requires knowledge of aspects of disease presentation, screening strategies, treatment approaches, and survivorship care considerations unique to the geriatric population. In this article, we review characteristics of prostate cancer screening and treatment decision making for localized disease in elderly men, including a discussion of the biology of disease in the elderly population. We also review best practices for providing treatment for localized and recurrent disease in an elderly population, including engaging in a basic geriatric assessment to determine fitness for treatment, eliciting information about patient preferences and support systems, and balancing treatment decisions in the context of these factors using the resources of a multidisciplinary care team. We then consider complications of prostate cancer survivorship related to systemic treatment in the elderly population of men with this disease. Finally, we emphasize the importance of engaging patients in treatment decision making across the spectrum of disease to personalize treatment plans and provide optimal care.
Collapse
Affiliation(s)
- Alicia K Morgans
- From the Vanderbilt University Medical Center, Nashville, TN; The University of Chicago, Chicago, IL; Division of Oncology/Unit of Urology, Urological Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Osperdale San Faffaele, Milan, Italy
| | - William Dale
- From the Vanderbilt University Medical Center, Nashville, TN; The University of Chicago, Chicago, IL; Division of Oncology/Unit of Urology, Urological Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Osperdale San Faffaele, Milan, Italy
| | - Alberto Briganti
- From the Vanderbilt University Medical Center, Nashville, TN; The University of Chicago, Chicago, IL; Division of Oncology/Unit of Urology, Urological Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Osperdale San Faffaele, Milan, Italy
| |
Collapse
|
139
|
Davaapil H, Brockes JP, Yun MH. Conserved and novel functions of programmed cellular senescence during vertebrate development. Development 2017; 144:106-114. [PMID: 27888193 PMCID: PMC5278627 DOI: 10.1242/dev.138222] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 11/10/2017] [Indexed: 12/14/2022]
Abstract
Cellular senescence, a form of stable cell cycle arrest that is traditionally associated with tumour suppression, has been recently found to occur during mammalian development. Here, we show that cell senescence is an intrinsic part of the developmental programme in amphibians. Programmed senescence occurs in specific structures during defined time windows during amphibian development. It contributes to the physiological degeneration of the amphibian pronephros and to the development of the cement gland and oral cavity. In both contexts, senescence depends on TGFβ but is independent of ERK/MAPK activation. Furthermore, elimination of senescent cells through temporary TGFβ inhibition leads to developmental defects. Our findings uncover conserved and new roles of senescence in vertebrate organogenesis and support the view that cellular senescence may have arisen in evolution as a developmental mechanism.
Collapse
Affiliation(s)
- Hongorzul Davaapil
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Jeremy P Brockes
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Maximina H Yun
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK
| |
Collapse
|
140
|
Urbanelli L, Buratta S, Sagini K, Tancini B, Emiliani C. Extracellular Vesicles as New Players in Cellular Senescence. Int J Mol Sci 2016; 17:ijms17091408. [PMID: 27571072 PMCID: PMC5037688 DOI: 10.3390/ijms17091408] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 02/07/2023] Open
Abstract
Cell senescence is associated with the secretion of many factors, the so-called "senescence-associated secretory phenotype", which may alter tissue microenvironment, stimulating the organism to clean up senescent cells and replace them with newly divided ones. Therefore, although no longer dividing, these cells are still metabolically active and influence the surrounding tissue. Much attention has been recently focused not only on soluble factors released by senescent cells, but also on extracellular vesicles as conveyors of senescence signals outside the cell. Here, we give an overview of the role of extracellular vesicles in biological processes and signaling pathways related to senescence and aging.
Collapse
Affiliation(s)
- Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Krizia Sagini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Brunella Tancini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy.
| |
Collapse
|
141
|
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
|
142
|
Sosa Peña MDP, Lopez-Soler R, Melendez JA. Senescence in chronic allograft nephropathy. Am J Physiol Renal Physiol 2016; 315:F880-F889. [PMID: 27306980 DOI: 10.1152/ajprenal.00195.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite increasing numbers of patients on dialysis, the numbers of renal transplants performed yearly have remained relatively static. During the last 50 years, there have been many advances in the pharmacology of prevention of organ rejection. However, most patients will suffer from a slow but steady decline in renal function leading to graft loss. The most common cause of long-term graft loss is chronic allograft nephropathy (CAN). Therefore, elucidating and understanding the mechanisms involved in CAN is crucial for achieving better posttransplant outcomes. It is thought that the development of epithelial to mesenchymal transition (EMT) in proximal tubules is one of the first steps towards CAN, and has been shown to be a result of cellular senescence. Cells undergoing senescence acquire a senescence associated secretory phenotype (SASP) leading to the production of interleukin-1 alpha (IL-1α), which has been implicated in several degenerative and inflammatory processes including renal disease. A central mediator in SASP activation is the production of reactive oxygen species (ROS), which are produced in response to numerous physiological and pathological stimuli. This review explores the connection between SASP and the development of EMT/CAN in an effort to suggest future directions for research leading to improved long-term graft outcomes.
Collapse
Affiliation(s)
| | - Reynold Lopez-Soler
- Albany Medical Center, Department of Surgery, Division of Transplantation, Albany, New York
| | - J Andrés Melendez
- SUNY Polytechnic Institute, Colleges of Nanoscale Science and Engineering, Albany, New York
| |
Collapse
|
143
|
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
|
144
|
Lecot P, Alimirah F, Desprez PY, Campisi J, Wiley C. Context-dependent effects of cellular senescence in cancer development. Br J Cancer 2016; 114:1180-4. [PMID: 27140310 PMCID: PMC4891501 DOI: 10.1038/bjc.2016.115] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence is an established tumour-suppressive mechanism that prevents the proliferation of premalignant cells. However, several lines of evidence show that senescent cells, which often persist in vivo, can also promote tumour progression in addition to other age-related pathologies via the senescence-associated secretory phenotype (SASP). Moreover, new insights suggest the SASP can facilitate tissue repair. Here, we review the beneficial and detrimental roles of senescent cells, highlighting conditions under which the senescence response does and does not promote pathology, particularly cancer. By better understanding the context-dependent effects of cellular senescence, it may be feasible to limit its detrimental properties while preserving its beneficial effects, and develop novel therapeutic strategies to prevent or treat cancer and possibly other age-associated diseases.
Collapse
Affiliation(s)
- Pacome Lecot
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Fatouma Alimirah
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA.,California Pacific Medical Center, 475 Brannan Street, San Francisco, CA 94107, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA.,Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Christopher Wiley
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| |
Collapse
|
145
|
Filippi-Chiela EC, Bueno e Silva MM, Thomé MP, Lenz G. Single-cell analysis challenges the connection between autophagy and senescence induced by DNA damage. Autophagy 2016; 11:1099-113. [PMID: 25701485 PMCID: PMC4590630 DOI: 10.1080/15548627.2015.1009795] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Autophagy and senescence have been described as central features of cell biology, but the interplay between these mechanisms remains obscure. Using a therapeutically relevant model of DNA damage-induced senescence in human glioma cells, we demonstrated that acute treatment with temozolomide induces DNA damage, a transitory activation of PRKAA/AMPK-ULK1 and MAPK14/p38 and the sustained inhibition of AKT-MTOR. This produced a transient induction of autophagy, which was followed by senescence. However, at the single cell level, this coordinated transition was not observed, and autophagy and senescence were triggered in a very heterogeneous manner. Indeed, at a population level, autophagy was highly negatively correlated with senescence markers, while in single cells this correlation did not exist. The inhibition of autophagy triggered apoptosis and decreased senescence, while its activation increased temozolomide-induced senescence, showing that DNA damage-induced autophagy acts by suppressing apoptosis.
Collapse
Key Words
- 3MA, 3-methyladenine
- AMP-activated
- AO, acridine orange
- BafA1, bafilomycin A1
- CDKN1A/p21, cyclin-dependent kinase inhibitor 1A (p21 Cip1)
- CPD, cumulative population doubling
- DDR, DNA damage response
- DFM, drug-free medium
- DNA damage
- H2AFX, H2A histone family
- MAP1LC3A/LC3, microtubule-associated protein 1 light chain 3 α
- MTOR, mechanistic target of rapamycin
- MTORC1, MTOR complex 1
- NA, nuclear area
- NMA, nuclear morphometric analysis
- PRKAA/AMPKα, protein kinase
- RAPA, rapamycin
- RPTOR/RAPTOR, regulatory-associated protein of MTOR
- SA-β-gal, senescence associated β-galactosidase assay
- SQSTM1/p62, sequestosome 1
- TMZ, temozolomide
- autophagy
- cP1-4, cellular population 1 to 4
- complex 1
- member X
- nP1–5, nuclear population 1 to 5
- senescence
- single cell
- temozolomide
Collapse
|
146
|
Single-cell analysis challenges the connection between autophagy and senescence induced by DNA damage. Autophagy 2016. [PMID: 25701485 DOI: 10.0180/15548627.2015.1009795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Autophagy and senescence have been described as central features of cell biology, but the interplay between these mechanisms remains obscure. Using a therapeutically relevant model of DNA damage-induced senescence in human glioma cells, we demonstrated that acute treatment with temozolomide induces DNA damage, a transitory activation of PRKAA/AMPK-ULK1 and MAPK14/p38 and the sustained inhibition of AKT-MTOR. This produced a transient induction of autophagy, which was followed by senescence. However, at the single cell level, this coordinated transition was not observed, and autophagy and senescence were triggered in a very heterogeneous manner. Indeed, at a population level, autophagy was highly negatively correlated with senescence markers, while in single cells this correlation did not exist. The inhibition of autophagy triggered apoptosis and decreased senescence, while its activation increased temozolomide-induced senescence, showing that DNA damage-induced autophagy acts by suppressing apoptosis.
Collapse
Key Words
- 3MA, 3-methyladenine
- AMP-activated
- AO, acridine orange
- BafA1, bafilomycin A1
- CDKN1A/p21, cyclin-dependent kinase inhibitor 1A (p21 Cip1)
- CPD, cumulative population doubling
- DDR, DNA damage response
- DFM, drug-free medium
- DNA damage
- H2AFX, H2A histone family
- MAP1LC3A/LC3, microtubule-associated protein 1 light chain 3 α
- MTOR, mechanistic target of rapamycin
- MTORC1, MTOR complex 1
- NA, nuclear area
- NMA, nuclear morphometric analysis
- PRKAA/AMPKα, protein kinase
- RAPA, rapamycin
- RPTOR/RAPTOR, regulatory-associated protein of MTOR
- SA-β-gal, senescence associated β-galactosidase assay
- SQSTM1/p62, sequestosome 1
- TMZ, temozolomide
- autophagy
- cP1-4, cellular population 1 to 4
- complex 1
- member X
- nP1–5, nuclear population 1 to 5
- senescence
- single cell
- temozolomide
Collapse
|
147
|
Manojlovic-Gacic E, Skender-Gazibara M, Popovic V, Soldatovic I, Boricic N, Raicevic S, Pekic S, Doknic M, Miljic D, Alafuzoff I, Pontén F, Casar-Borota O. Oncogene-Induced Senescence in Pituitary Adenomas--an Immunohistochemical Study. Endocr Pathol 2016; 27:1-11. [PMID: 26573928 DOI: 10.1007/s12022-015-9405-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Oncogene-induced senescence (OIS) serves as an initial barrier to cancer development, being proposed as a possible explanation for the usually benign behavior of the pituitary adenomas. We aimed to explore the immunohistochemical expression of the OIS markers, senescence-associated lysosomal β-galactosidase (SA-β-GAL), p16, and p21 in different types of 345 pituitary adenomas and compared it with the expression in the normal pituitary and in the specimens from the repeated surgeries. SA-β-GAL was overexpressed in the pituitary adenomas, compared to the normal pituitaries. Growth hormone (GH) producing adenomas showed the strongest SA-β-GAL, with densely granulated (DG)-GH adenomas more reactive than the sparsely granulated (SG). Nuclear p21 was decreased in the adenomas, except for the SG-GH adenomas that had higher p21 than the normal pituitaries and the other adenomas. p16 was significantly lower in the adenomas, without type-related differences. SA-β-GAL was slightly lower and p16 slightly higher in the recurrences. Our findings indicate alterations of the senescence program in the different types of pituitary adenomas. Activation of senescence in the pituitary adenomas presents one possible explanation for their usually benign behavior, at least in the GH adenomas that show a synchronous increase of two OIS markers. However, subdivision into GH adenoma subtypes reveals differences that reflect complex regulatory mechanisms influenced by the interplay between the granularity pattern and the hormonal factors, with possible impact on the different clinical behavior of the SG- and DG-GH adenoma subtypes. p16 seems to have a more prominent role in the pituitary tumorigenesis than in the senescence. Recurrent growth in a subset of the pituitary adenomas is not associated with consistent changes in the senescence pattern.
Collapse
Affiliation(s)
- Emilija Manojlovic-Gacic
- Institute of Pathology, Medical Faculty, University of Belgrade, Dr Subotica 1, Belgrade, Serbia.
| | - Milica Skender-Gazibara
- Institute of Pathology, Medical Faculty, University of Belgrade, Dr Subotica 1, Belgrade, Serbia
| | - Vera Popovic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Ivan Soldatovic
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Novica Boricic
- Institute of Pathology, Medical Faculty, University of Belgrade, Dr Subotica 1, Belgrade, Serbia
| | - Savo Raicevic
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovica 4, Belgrade, Serbia
| | - Sandra Pekic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Mirjana Doknic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Dragana Miljic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
- Department of Clinical Pathology and Cytology, Uppsala University Hospital, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
| | - Olivera Casar-Borota
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
- Department of Clinical Pathology and Cytology, Uppsala University Hospital, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
| |
Collapse
|
148
|
Chemotherapy induces the cancer-associated fibroblast phenotype, activating paracrine Hedgehog-GLI signalling in breast cancer cells. Oncotarget 2016; 6:10728-45. [PMID: 25915429 PMCID: PMC4484415 DOI: 10.18632/oncotarget.3828] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 03/26/2015] [Indexed: 12/19/2022] Open
Abstract
Cancer cells recruit normal cells such as fibroblasts to establish reactive microenvironments. Via metabolic stress, catabolism and inflammation, these cancer-associated fibroblasts set up a synergistic relationship with tumour cells, that contributes to their malignancy and resistance to therapy. Given that chemotherapy is a systemic treatment, the possibility that healthy cell damage affects the metastatic risk or the prospect of developing a second malignancy becomes relevant. Here, we demonstrate that standard chemotherapies phenotypically and metabolically transform stromal fibroblasts into cancer-associated fibroblasts, leading to the emergence of a highly glycolytic, autophagic and pro-inflammatory microenvironment. This catabolic microenvironment, in turn, activates stemness (Sonic hedgehog/GLI signalling), antioxidant response and interferon-mediated signalling, in adjacent breast cancer cells. Thus, we propose a model by which chemotherapy-induced catabolism in healthy fibroblasts constitutes a source of energy-rich nutrients and inflammatory cytokines that would activate stemness in adjacent epithelial cells, possibly triggering new tumorigenic processes. In this context, immune cell recruitment would be also stimulated to further support malignancy.
Collapse
|
149
|
Homeostatic Signaling by Cell-Cell Junctions and Its Dysregulation during Cancer Progression. J Clin Med 2016; 5:jcm5020026. [PMID: 26901232 PMCID: PMC4773782 DOI: 10.3390/jcm5020026] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 12/16/2022] Open
Abstract
The transition of sessile epithelial cells to a migratory, mesenchymal phenotype is essential for metazoan development and tissue repair, but this program is exploited by tumor cells in order to escape the confines of the primary organ site, evade immunosurveillance, and resist chemo-radiation. In addition, epithelial-to-mesenchymal transition (EMT) confers stem-like properties that increase efficiency of colonization of distant organs. This review evaluates the role of cell–cell junctions in suppressing EMT and maintaining a quiescent epithelium. We discuss the conflicting data on junctional signaling in cancer and recent developments that resolve some of these conflicts. We focus on evidence from breast cancer, but include other organ sites where appropriate. Current and potential strategies for inhibition of EMT are discussed.
Collapse
|
150
|
Chen P, Guo H, Chen J, Fu Y. The chemotherapeutic drug boanmycin induces cell senescence and senescence-associated secretory phenotype factors, thus acquiring the potential to remodel the tumor microenvironment. Anticancer Drugs 2016; 27:84-8. [DOI: 10.1097/cad.0000000000000304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|