1
|
Han Z, Wang K, Ding S, Zhang M. Cross-talk of inflammation and cellular senescence: a new insight into the occurrence and progression of osteoarthritis. Bone Res 2024; 12:69. [PMID: 39627227 PMCID: PMC11615234 DOI: 10.1038/s41413-024-00375-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 12/06/2024] Open
Abstract
Osteoarthritis (OA) poses a significant challenge in orthopedics. Inflammatory pathways are regarded as central mechanisms in the onset and progression of OA. Growing evidence suggests that senescence acts as a mediator in inflammation-induced OA. Given the lack of effective treatments for OA, there is an urgent need for a clearer understanding of its pathogenesis. In this review, we systematically summarize the cross-talk between cellular senescence and inflammation in OA. We begin by focusing on the mechanisms and hallmarks of cellular senescence, summarizing evidence that supports the relationship between cellular senescence and inflammation. We then discuss the mechanisms of interaction between cellular senescence and inflammation, including senescence-associated secretory phenotypes (SASP) and the effects of pro- and anti-inflammatory interventions on cellular senescence. Additionally, we focus on various types of cellular senescence in OA, including senescence in cartilage, subchondral bone, synovium, infrapatellar fat pad, stem cells, and immune cells, elucidating their mechanisms and impacts on OA. Finally, we highlight the potential of therapies targeting senescent cells in OA as a strategy for promoting cartilage regeneration.
Collapse
Affiliation(s)
- Zeyu Han
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China
| | - Ketao Wang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China
| | - Shenglong Ding
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China
| | - Mingzhu Zhang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, PR China.
| |
Collapse
|
2
|
Wagner KD, Safwan-Zaiter H, Wagner N. A Dual Role of the Senescence Marker P16Ink4a in Liver Endothelial Cell Function. Cells 2024; 13:1929. [PMID: 39682678 PMCID: PMC11640762 DOI: 10.3390/cells13231929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
P16Ink4a is a well-established marker of senescence. Although P16Ink4a is expressed in endothelial cells, little is known about its function in these cells. Using isolated liver endothelial cells with silencing or overexpression of P16Ink4a, we show here that dependent on P16Ink4a levels, different pathways and functions are affected. High levels of P16Ink4a reduce proliferation and induce senescence, while low levels have the opposite effects. Only high P16Ink4a expression reduces in vitro angiogenesis. Expression profiling reveals an inflammatory phenotype upon silencing of P16Ink4a, while P16Ink4a overexpression is associated with a profile associated with DNA damage, repair and senescence. Low levels of P16Ink4a induce reactive oxygen species (ROS) generation and increase endothelial cell leakage. Collectively, P16Ink4a represents an "antagonistic pleiotropy" gene, which is, on the one hand, required to prevent ROS generation and endothelial damage and, on the other hand, inhibits angiogenesis through induction of senescence at high levels.
Collapse
Affiliation(s)
| | | | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France;
| |
Collapse
|
3
|
Wang X, Zhang C, Su J, Ren S, Wang X, Zhang Y, Yuan Z, He X, Wu X, Li M, Du F, Chen Y, Deng S, Zhao Y, Wang X, Sun Y, Shen J, Ji H, Hou Y, Xiao Z. Rejuvenation Strategy for Inducing and Enhancing Autoimmune Response to Eliminate Senescent Cells. Aging Dis 2024:AD.2024.0579. [PMID: 39122450 DOI: 10.14336/ad.2024.0579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024] Open
Abstract
The process of aging, which involves progressive changes in the body over time, is closely associated with the development of age-related diseases. Cellular senescence is a pivotal hallmark and mechanism of the aging process. The accumulation of senescent cells can significantly contribute to the onset of age-related diseases, thereby compromising overall health. Conversely, the elimination of senescent cells enhances the body's regenerative and reparative capacity, thereby retarding the aging process. Here, we present a brief overview of 12 Hallmarks of aging and subsequently emphasize the potential of immune checkpoint blockade, innate immune cell therapy (including T cells, iNKT cells, macrophages, and NK cells), as well as CAR-T cell therapy for inducing and augmenting immune responses aimed at eliminating senescent cells. In addition to CAR-T cells, we also explore the possibility of engineered immune cells such as CAR-NK and CAR-M cells to eliminate senescent cells. In summary, immunotherapy, as an emerging strategy for the treatment of aging, offers new prospects for age-related research.
Collapse
Affiliation(s)
- Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Chengyu Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zijun Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Xiaodong Wang
- Department of Hepatobiliary Disease, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yunqing Hou
- LongmaTan District People's Hospital of Luzhou City, Luzhou 646600, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy &;amp Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang 621000, China
| |
Collapse
|
4
|
Yan H, Miranda EAD, Jin S, Wilson F, An K, Godbee B, Zheng X, Brau-Rodríguez AR, Lei L. Primary oocytes with cellular senescence features are involved in ovarian aging in mice. Sci Rep 2024; 14:13606. [PMID: 38871781 PMCID: PMC11176158 DOI: 10.1038/s41598-024-64441-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024] Open
Abstract
In mammalian females, quiescent primordial follicles serve as the ovarian reserve and sustain normal ovarian function and egg production via folliculogenesis. The loss of primordial follicles causes ovarian aging. Cellular senescence, characterized by cell cycle arrest and production of the senescence-associated secretory phenotype (SASP), is associated with tissue aging. In the present study, we report that some quiescent primary oocytes in primordial follicles become senescent in adult mouse ovaries. The senescent primary oocytes share senescence markers characterized in senescent somatic cells. The senescent primary oocytes were observed in young adult mouse ovaries, remained at approximately 15% of the total primary oocytes during ovarian aging from 6 to 12 months, and accumulated in aged ovaries. Administration of a senolytic drug ABT263 to 3-month-old mice reduced the percentage of senescent primary oocytes and the transcription of the SASP factors in the ovary, in addition, led to increased numbers of primordial and total follicles and a higher rate of oocyte maturation. Our study provides experimental evidence that primary oocytes, a germline cell type that is arrested in meiosis, become senescent in adult mouse ovaries and that senescent cell clearance reduced primordial follicle loss and mitigated ovarian aging phenotypes.
Collapse
Affiliation(s)
- Hao Yan
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Edgar Andres Diaz Miranda
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA
| | - Shiying Jin
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA
| | - Faith Wilson
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA
- Division of Biological Sciences, College of Arts and Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Kang An
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA
| | - Brooke Godbee
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA
- College of Health Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Xiaobin Zheng
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
| | - Astrid Roshealy Brau-Rodríguez
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA
| | - Lei Lei
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, 65211, USA.
- Division of Biological Sciences, College of Arts and Sciences, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
5
|
Yan H, Miranda EAD, Jin S, Wilson F, An K, Godbee B, Zheng X, Brau-Rodríguez AR, Lei L. Primary oocytes with cellular senescence features are involved in ovarian aging in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574768. [PMID: 38260383 PMCID: PMC10802418 DOI: 10.1101/2024.01.08.574768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
In mammalian females, quiescent primordial follicles serve as the ovarian reserve and sustain normal ovarian function and egg production via folliculogenesis. The loss of primordial follicles causes ovarian aging. Cellular senescence, characterized by cell cycle arrest and production of the senescence-associated secretory phenotype (SASP), is associated with tissue aging. In the present study, we report that some quiescent primary oocytes in primordial follicles become senescent in adult mouse ovaries. The senescent primary oocytes share senescence markers characterized in senescent somatic cells. The senescent primary oocytes were observed in young adult mouse ovaries, remained at approximately 15% of the total primary oocytes during ovarian aging from 6 months to 12 months, and accumulated in aged ovaries. Administration of a senolytic drug ABT263 to 3-month-old mice reduced the percentage of senescent primary oocytes and the transcription of the SASP cytokines in the ovary. In addition, led to increased numbers of primordial and total follicles and a higher rate of oocyte maturation and female fertility. Our study provides experimental evidence that primary oocytes, a germline cell type that is arrested in meiosis, become senescent in adult mouse ovaries and that senescent cell clearance reduced primordial follicle loss and mitigated ovarian aging phenotypes.
Collapse
Affiliation(s)
- Hao Yan
- Buck Institute for Research on Aging, Novato, California, 94945
- Carnegie Institution for Science, Department of Embryology, Baltimore, Maryland, 21218
| | - Edgar Andres Diaz Miranda
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
| | - Shiying Jin
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
| | - Faith Wilson
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
- Division of Biological Sciences, College of Arts and Sciences, University of Missouri, Columbia, Missouri, 65211
| | - Kang An
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
| | - Brooke Godbee
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
- College of Health Sciences, University of Missouri, Columbia, Missouri, 65211
| | - Xiaobin Zheng
- Carnegie Institution for Science, Department of Embryology, Baltimore, Maryland, 21218
| | - Astrid Roshealy Brau-Rodríguez
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
| | - Lei Lei
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, Missouri, 65211
- Division of Biological Sciences, College of Arts and Sciences, University of Missouri, Columbia, Missouri, 65211
| |
Collapse
|
6
|
Erichsen L, Adjaye J. Crosstalk between age accumulated DNA-damage and the SIRT1-AKT-GSK3ß axis in urine derived renal progenitor cells. Aging (Albany NY) 2022; 14:8179-8204. [PMID: 36170022 PMCID: PMC9648809 DOI: 10.18632/aging.204300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/08/2022] [Indexed: 12/02/2022]
Abstract
The aging process is manifested by a multitude of inter-linked biological processes. These processes contribute to genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, de-regulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. The mammalian ortholog of the yeast silent information regulator (Sir2) SIRT1 is a NAD+-dependent class III histone deacetylase and has been recognized to be involved in many of the forementioned processes. Furthermore, the physiological activity of several Sirtuin family members has been connected to the regulation of life span of lower organisms (Caenorhabditis elegans and Drosophila melanogaster) as well as mammals. In the present study, we provide evidence that SIX2-positive urine derived renal progenitor cells-UdRPCs isolated directly from human urine show typical hallmarks of aging. This includes the subsequent transcriptional downregulation of SIRT1 and its downstream targets AKT and GSK3ß with increased donor age. This transcriptional downregulation is accompanied by an increase in DNA damage and transcriptional levels of several cell cycle inhibitors such as P16. We provide evidence that the renal progenitor transcription factor SIX2 binds to the coding sequence of SIRT1. Furthermore, we show that the SIRT1 promoter region is methylation sensitive and becomes methylated during aging, dividing them into SIRT1-high and -low expressing UdRPCs. Our results highlight the importance of SIRT1 in DNA damage repair recognition in UdRPCs and the control of differentiation by regulating the activation of GSK3β through AKT.
Collapse
Affiliation(s)
- Lars Erichsen
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine- University Düsseldorf, Düsseldorf 40225, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine- University Düsseldorf, Düsseldorf 40225, Germany
| |
Collapse
|
7
|
Safwan-Zaiter H, Wagner N, Wagner KD. P16INK4A-More Than a Senescence Marker. Life (Basel) 2022; 12:1332. [PMID: 36143369 PMCID: PMC9501954 DOI: 10.3390/life12091332] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is a biological feature that is characterized by gradual degeneration of function in cells, tissues, organs, or an intact organism due to the accumulation of environmental factors and stresses with time. Several factors have been attributed to aging such as oxidative stress and augmented production or exposure to reactive oxygen species, inflammatory cytokines production, telomere shortening, DNA damage, and, importantly, the deposit of senescent cells. These are irreversibly mitotically inactive, yet metabolically active cells. The reason underlying their senescence lies within the extrinsic and the intrinsic arms. The extrinsic arm is mainly characterized by the expression and the secretory profile known as the senescence-associated secretory phenotype (SASP). The intrinsic arm results from the impact of several genes meant to regulate the cell cycle, such as tumor suppressor genes. P16INK4A is a tumor suppressor and cell cycle regulator that has been linked to aging and senescence. Extensive research has revealed that p16 expression is significantly increased in senescent cells, as well as during natural aging or age-related pathologies. Based on this fact, p16 is considered as a specific biomarker for detecting senescent cells and aging. Other studies have found that p16 is not only a senescence marker, but also a protein with many functions outside of senescence and aging. In this paper, we discuss and shed light on several studies that show the different functions of p16 and provide insights in its role in several biological processes besides senescence and aging.
Collapse
Affiliation(s)
| | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France
| | | |
Collapse
|
8
|
Marzullo M, Maï ME, Ferreira MG. Whole-mount Senescence-Associated Beta-Galactosidase (SA-β-GAL) Activity Detection Protocol for Adult Zebrafish. Bio Protoc 2022; 12:e4457. [PMID: 35937931 PMCID: PMC9303820 DOI: 10.21769/bioprotoc.4457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 12/29/2022] Open
Abstract
Senescence-associated beta-galactosidase (SA-β-GAL) is an enzyme that accumulates in the lysosomes of senescent cells, where it hydrolyses β-galactosides. With p16, it represents a well-recognized biomarker used to assess senescence both in vivo and in cell culture. The use of a chromogenic substrate, such as 5-bromo-4-chloro-3-indoyl-β-d-galactopyranoside (X-Gal), allows the detection of SA-β-GAL activity at pH 6.0 by the release of a visible blue product. Senescence occurs during aging and is part of the aging process itself. We have shown that prematurely aged zebrafish accumulate senescent cells detectable by SA-β-GAL staining in different tissues, including testis and gut. Here, we report a detailed protocol to perform an SA-β-GAL assay to detect senescent cell accumulation across the entire adult zebrafish organism ( Danio rerio ). We also identify previously unreported organs that show increased cell senescence in telomerase mutants, including the liver and the spinal cord.
Collapse
Affiliation(s)
- Marta Marzullo
- IBPM CNR c/o Department of Biology and Biotechnology, Sapienza University of Rome, Rome, Italy
,
Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
,
*For correspondence:
;
;
| | - Mounir El Maï
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
,
Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284 INSERM U1081 Université Côte d’Azur, 06107 Nice, France
,
*For correspondence:
;
;
| | - Miguel Godinho Ferreira
- Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
,
Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284 INSERM U1081 Université Côte d’Azur, 06107 Nice, France
,
*For correspondence:
;
;
| |
Collapse
|
9
|
Dynamic Spatiotemporal Expression Pattern of the Senescence-Associated Factor p16Ink4a in Development and Aging. Cells 2022; 11:cells11030541. [PMID: 35159350 PMCID: PMC8833900 DOI: 10.3390/cells11030541] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
A plethora of factors have been attributed to underly aging, including oxidative stress, telomere shortening and cellular senescence. Several studies have shown a significant role of the cyclin-dependent kinase inhibitor p16ink4a in senescence and aging. However, its expression in development has been less well documented. Therefore, to further clarify a potential role of p16 in development and aging, we conducted a developmental expression study of p16, as well as of p19ARF and p21, and investigated their expression on the RNA level in brain, heart, liver, and kidney of mice at embryonic, postnatal, adult, and old ages. P16 expression was further assessed on the protein level by immunohistochemistry. Expression of p16 was highly dynamic in all organs in embryonic and postnatal stages and increased dramatically in old mice. Expression of p19 and p21 was less variable and increased to a moderate extent at old age. In addition, we observed a predominant expression of p16 mRNA and protein in liver endothelial cells versus non-endothelial cells of old mice, which suggests a functional role specifically in liver endothelium of old subjects. Thus, p16 dynamic spatiotemporal expression might implicate p16 in developmental and physiological processes in addition to its well-known function in the build-up of senescence.
Collapse
|
10
|
Fathi I, Miki T. Human Amniotic Epithelial Cells Secretome: Components, Bioactivity, and Challenges. Front Med (Lausanne) 2022; 8:763141. [PMID: 35083233 PMCID: PMC8784524 DOI: 10.3389/fmed.2021.763141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
Human amniotic epithelial cells (hAECs) derived from placental tissue have received significant attention as a promising tool in regenerative medicine. Several studies demonstrated their anti-inflammatory, anti-fibrotic, and tissue repair potentials. These effects were further shown to be retained in the conditioned medium of hAECs, suggesting their paracrine nature. The concept of utilizing the hAEC-secretome has thus evolved as a therapeutic cell-free option. In this article, we review the different components and constituents of hAEC-secretome and their influence as demonstrated through experimental studies in the current literature. Studies examining the effects of conditioned medium, exosomes, and micro-RNA (miRNA) derived from hAECs are included in this review. The challenges facing the application of this cell-free approach will also be discussed based on the current evidence.
Collapse
Affiliation(s)
- Ibrahim Fathi
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
11
|
Benítez S, Cordero A, Santamaría PG, Redondo-Pedraza J, Rocha AS, Collado-Solé A, Jimenez M, Sanz-Moreno A, Yoldi G, Santos JC, De Benedictis I, Gómez-Aleza C, Da Silva-Álvarez S, Troulé K, Gómez-López G, Alcazar N, Palmero I, Collado M, Serrano M, Gonzalez-Suarez E. RANK links senescence to stemness in the mammary epithelia, delaying tumor onset but increasing tumor aggressiveness. Dev Cell 2021; 56:1727-1741.e7. [PMID: 34004159 PMCID: PMC8221814 DOI: 10.1016/j.devcel.2021.04.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 03/25/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022]
Abstract
Rank signaling enhances stemness in mouse and human mammary epithelial cells (MECs) and mediates mammary tumor initiation. Mammary tumors initiated by oncogenes or carcinogen exposure display high levels of Rank and Rank pathway inhibitors have emerged as a new strategy for breast cancer prevention and treatment. Here, we show that ectopic Rank expression in the mammary epithelia unexpectedly delays tumor onset and reduces tumor incidence in the oncogene-driven Neu and PyMT models. Mechanistically, we have found that ectopic expression of Rank or exposure to Rankl induces senescence, even in the absence of other oncogenic mutations. Rank leads to DNA damage and senescence through p16/p19. Moreover, RANK-induced senescence is essential for Rank-driven stemness, and although initially translates into delayed tumor growth, eventually promotes tumor progression and metastasis. We uncover a dual role for Rank in the mammary epithelia: Rank induces senescence and stemness, delaying tumor initiation but increasing tumor aggressiveness.
Collapse
Affiliation(s)
- Sandra Benítez
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Alex Cordero
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain; Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Patricia G Santamaría
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red, Área de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid 28029, Spain
| | | | - Ana S Rocha
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Alejandro Collado-Solé
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain; Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Maria Jimenez
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Adrian Sanz-Moreno
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain; German Mouse Clinic, Institute of Experimental Genetics, HMGU, Neuherberg, 85764, Germany
| | - Guillermo Yoldi
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Juliana C Santos
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Ilaria De Benedictis
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Clara Gómez-Aleza
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain
| | - Sabela Da Silva-Álvarez
- Health Research Institute of Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), E15706 Santiago de Compostela, Spain
| | - Kevin Troulé
- Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | | | - Noelia Alcazar
- Institute for Research in Biomedicine (IRB), 08028 Barcelona, Spain
| | - Ignacio Palmero
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, 28029 Madrid, Spain
| | - Manuel Collado
- Health Research Institute of Santiago de Compostela (IDIS), Xerencia de Xestión Integrada de Santiago (XXIS/SERGAS), E15706 Santiago de Compostela, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB), 08028 Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Eva Gonzalez-Suarez
- Oncobell, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain; Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.
| |
Collapse
|
12
|
Kowald A, Passos JF, Kirkwood TBL. On the evolution of cellular senescence. Aging Cell 2020; 19:e13270. [PMID: 33166065 PMCID: PMC7744960 DOI: 10.1111/acel.13270] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 10/09/2020] [Indexed: 01/10/2023] Open
Abstract
The idea that senescent cells are causally involved in aging has gained strong support from findings that the removal of such cells alleviates many age‐related diseases and extends the life span of mice. While efforts proceed to make therapeutic use of such discoveries, it is important to ask what evolutionary forces might have been behind the emergence of cellular senescence, in order better to understand the biology that we might seek to alter. Cellular senescence is often regarded as an anti‐cancer mechanism, since it limits the division potential of cells. However, many studies have shown that senescent cells often also have carcinogenic properties. This is difficult to reconcile with the simple idea of an anti‐cancer mechanism. Furthermore, other studies have shown that cellular senescence is involved in wound healing and tissue repair. Here, we bring these findings and ideas together and discuss the possibility that these functions might be the main reason for the evolution of cellular senescence. Furthermore, we discuss the idea that senescent cells might accumulate with age because the immune system had to strike a balance between false negatives (overlooking some senescent cells) and false positives (destroying healthy body cells).
Collapse
Affiliation(s)
- Axel Kowald
- Campus for Ageing and Vitality Newcastle University Institute for Ageing Newcastle upon Tyne UK
- Rostock University Medical Center Institute for Biostatistics and Informatics in Medicine and Aging Research (IBIMA) Rostock Germany
| | - João F. Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester Minnesota USA
| | - Thomas B. L. Kirkwood
- Campus for Ageing and Vitality Newcastle University Institute for Ageing Newcastle upon Tyne UK
- Center for Healthy Aging Department of Cellular and Molecular Medicine University of Copenhagen Copenhagen Denmark
| |
Collapse
|
13
|
Uyar B, Palmer D, Kowald A, Murua Escobar H, Barrantes I, Möller S, Akalin A, Fuellen G. Single-cell analyses of aging, inflammation and senescence. Ageing Res Rev 2020; 64:101156. [PMID: 32949770 PMCID: PMC7493798 DOI: 10.1016/j.arr.2020.101156] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 02/09/2023]
Abstract
Single-cell gene expression (transcriptomics) data are becoming robust and abundant, and are increasingly used to track organisms along their life-course. This allows investigation into how aging affects cellular transcriptomes, and how changes in transcriptomes may underlie aging, including chronic inflammation (inflammaging), immunosenescence and cellular senescence. We compiled and tabulated aging-related single-cell datasets published to date, collected and discussed relevant findings, and inspected some of these datasets ourselves. We specifically note insights that cannot (or not easily) be based on bulk data. For example, in some datasets, the fraction of cells expressing p16 (CDKN2A), one of the most prominent markers of cellular senescence, was reported to increase, in addition to its upregulated mean expression over all cells. Moreover, we found evidence for inflammatory processes in most datasets, some of these driven by specific cells of the immune system. Further, single-cell data are specifically useful to investigate whether transcriptional heterogeneity (also called noise or variability) increases with age, and many (but not all) studies in our review report an increase in such heterogeneity. Finally, we demonstrate some stability of marker gene expression patterns across closely similar studies and suggest that single-cell experiments may hold the key to provide detailed insights whenever interventions (countering aging, inflammation, senescence, disease, etc.) are affecting cells depending on cell type.
Collapse
Affiliation(s)
- Bora Uyar
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniel Palmer
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Axel Kowald
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Hugo Murua Escobar
- Rostock University Medical Center, Department of Hematology, Oncology and Palliative Medicine, Department of Medicine III, Rostock, Germany
| | - Israel Barrantes
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Steffen Möller
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Altuna Akalin
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany.
| |
Collapse
|
14
|
Pinto C, Ninfole E, Gaggiano L, Benedetti A, Marzioni M, Maroni L. Aging and the Biological Response to Liver Injury. Semin Liver Dis 2020; 40:225-232. [PMID: 31887774 DOI: 10.1055/s-0039-3402033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Interest in understanding the aging process has recently risen in the scientific community. Aging, commonly defined as the functional decline in the function of organs and tissues, is indeed the major risk factor for the development of many chronic diseases, such as cardiovascular diseases, pathologies of nervous system, or cancer. To date, the influence of aging in the pathophysiology of liver and biliary diseases is not fully understood. Although liver cells have a high regenerative capacity, hepatocytes and cholangiocytes undergo extensive molecular changes in response to aging. Following time-dependent damage induced by aging, liver cells initially activate compensatory mechanisms that, if hyperstimulated, may lead to the decline of regenerative capacity and the development of pathologies. A deeper understanding of molecular aging has undoubtedly the potential to improve the clinical management of patients, possibly unveiling new pathways for selective drug treatment.
Collapse
Affiliation(s)
- Claudio Pinto
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Elisabetta Ninfole
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Gaggiano
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Benedetti
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Marco Marzioni
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Luca Maroni
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
15
|
You J, Dong R, Ying M, He Q, Cao J, Yang B. Cellular Senescence and Anti-Cancer Therapy. Curr Drug Targets 2020; 20:705-715. [PMID: 30556499 DOI: 10.2174/1389450120666181217100833] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cellular senescence is generally understood as a permanent cell cycle arrest stemming from different causes. The mechanism of cellular senescence-induced cell cycle arrest is complex, involving interactions between telomere shortening, inflammations and cellular stresses. In recent years, a growing number of studies have revealed that cellular senescence could mediate the cancer progression of neighboring cells, but this idea is controversial and contradictory evidence argues that cellular senescence also contributes to tumor suppression. OBJECTIVE Given that the complicated role of senescence in various physiological and pathological scenarios, we try to clarify the precise contribution role of cellular senescence to tumor progression. METHODS Search for the information in a large array of relevant articles to support our opinion. RESULTS We discuss the relatively widespread occurrence of cellular senescence in cancer treatment and identify the positive and negative side of senescence contributed to tumor progression. CONCLUSION We argue that the availability of pro-senescence therapy could represent as a promising regimen for managing cancer disease, particularly with regard to the poor clinical outcome obtained with other anticancer therapies.
Collapse
Affiliation(s)
- Jieqiong You
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Rong Dong
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
16
|
Lee YY, Choi YS, Kim DW, Cheong JY, Song KY, Ryu MS, Lim IK. Mitochondrial nucleoid remodeling and biogenesis are regulated by the p53-p21 WAF1-PKCζ pathway in p16 INK4a-silenced cells. Aging (Albany NY) 2020; 12:6700-6732. [PMID: 32330121 PMCID: PMC7202532 DOI: 10.18632/aging.103029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/22/2020] [Indexed: 11/25/2022]
Abstract
Mitochondrial dysfunction is linked to age-related senescence phenotypes. We report here the pathway increasing nucleoid remodeling and biogenesis in mitochondria during the senescence of foreskin human diploid fibroblasts (fs-HDF) and WI-38 cells. Replicative senescence in fs-HDF cells increased mitochondrial nucleoid remodeling as indicated by 5-bromo-2'-deoxyuridine (BrdU) incorporation and mitochondrial transcription factor A (TFAM) expression in enlarged and fused mitochondria. Mitochondrial nucleoid remodeling was accompanied by mitochondrial biogenesis in old cells, and the expression levels of OXPHOS complex-I, -IV and -V subunits, PGC-1α and NRF1 were greatly increased compared to young cells. Activated protein kinase C zeta (PKCζ) increased mitochondrial activity and expressed phenotypes of delayed senescence in fs-HDF cells, but not in WI-38 cells. The findings were reproduced in the doxorubicin-induced senescence of young fs-HDF and WI-38 cells via the PKCζ-LKB1-AMPK signaling pathway, which was regulated by the p53-p21WAF1 pathway when p16INK4a was silenced. The signaling enhanced PGC-1α-NRF1-TFAM axis in mitochondria, which was demonstrated by Ingenuity Pathway Analysis of young and old fs-HDF cells. Activation of the p53-p21WAF1 pathway and silencing of p16INK4a are responsible for mitochondrial reprogramming in senescent cells, which may be a compensatory mechanism to promote cell survival under senescence stress.
Collapse
Affiliation(s)
- Yun Yeong Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Otolaryngology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Yeon Seung Choi
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon 16499, Korea
| | - Do Wan Kim
- Omics Center, Ajou University School of Medicine, Suwon 16499, Korea
| | - Jae Youn Cheong
- Omics Center, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Gastroenterology, Ajou University of Medicine, Suwon 16499, Korea
| | - Kye Yong Song
- Department of Pathology, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Min Sook Ryu
- Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon 16499, Korea
| | - In Kyoung Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Biomedical Sciences, The Graduate School, Ajou University, Suwon 16499, Korea
| |
Collapse
|
17
|
Zhang X, Koga N, Suzuki H, Kato M. Promotion of cellular senescence by THG-1/TSC22D4 knockout through activation of JUNB. Biochem Biophys Res Commun 2020; 522:897-902. [DOI: 10.1016/j.bbrc.2019.11.145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/21/2019] [Indexed: 10/25/2022]
|
18
|
Integrating context of tumor biology and vaccine design to shape multidimensional immunotherapies. FUTURE DRUG DISCOVERY 2020. [DOI: 10.4155/fdd-2019-0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Advances in cancer therapy have offered great promise but only modest clinical benefits as monotherapies to date. Patients usually respond well to therapies targeted at specific mutations, but only for a short time. Conversely, immunotherapies help fewer patients, but increase survival. Combination therapies, which could offer the best of both worlds, are currently limited by substantial toxicity. While recent advances in genomics and proteomics have yielded an unprecedented depth of enabling datasets, it has also shifted the focus toward in silico predictions. Designing the next wave of multidimensional immunotherapies will require leveraging this knowledge while providing a renewed emphasis on tumor biology and vaccine design. This includes careful selection of tumor clinical stage in the context of pre-existing tumor microenvironments, target antigen and technology platform selections to maximize their effect, and treatment staging. Here, we review strategies on how to approach an increasingly complex landscape of immunotherapeutic agents for use in combination therapies.
Collapse
|
19
|
lncRNA miat functions as a ceRNA to upregulate sirt1 by sponging miR-22-3p in HCC cellular senescence. Aging (Albany NY) 2019; 11:7098-7122. [PMID: 31503007 PMCID: PMC6756895 DOI: 10.18632/aging.102240] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/22/2019] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer related deaths and lacks effective therapies. Cellular senescence acts as a barrier against cancer progression and plays an important role in tumor suppression. Senescence associated long noncoding RNAs (SAL-RNAs) are thought to be critical regulators of cancer development. Here, the long noncoding RNA (lncRNA) myocardial infarction-associated transcript (miat) was first identified as an HCC specific SALncRNA. Knockdown of miat significantly promoted cellular senescence and inhibited HCC progression. Mechanistic study revealed that SAL-miat acted as a competitive endogenous RNA (ceRNA) that upregulated the expression of sirt1 by sponging miR-22-3p. Moreover, miat downregulation activated the tumor suppressor pathway (p53/p21 and p16/pRb) and stimulated senescent cancer cells to secrete senescence-associated secretory phenotype (SASP), which contributed to inhibition of tumor cell proliferation, and resulted in the suppression of HCC tumorigenesis. Together, our study provided mechanistic insights into a critical role of miat as a miRNA sponge in HCC cellular senescence, which might offer a potential therapeutic strategy for HCC treatment.
Collapse
|
20
|
Dai L, Qureshi AR, Witasp A, Lindholm B, Stenvinkel P. Early Vascular Ageing and Cellular Senescence in Chronic Kidney Disease. Comput Struct Biotechnol J 2019; 17:721-729. [PMID: 31303976 PMCID: PMC6603301 DOI: 10.1016/j.csbj.2019.06.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by progressive vascular disease, systemic inflammation, muscle wasting and frailty. The predominant early vascular ageing (EVA) process mediated by medial vascular calcification (VC) results in a marked discrepancy between chronological and biological vascular age in CKD. Though the exact underlying mechanisms of VC and EVA are not fully elucidated, accumulating evidence indicates that cellular senescence - and subsequent chronic inflammation through the senescence-associated secretary phenotype (SASP) - plays a fundamental role in its initiation and progression. In this review, we discuss the pathophysiological links between senescence and the EVA process in CKD, with focus on cellular senescence and media VC, and potential anti-ageing therapeutic strategies of senolytic drugs targeting cellular senescence and EVA in CKD.
Collapse
Affiliation(s)
| | | | | | | | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
21
|
Sui A, Xu Y, Pan B, Guo T, Wu J, Shen Y, Yang J, Guo X. Histone demethylase KDM6B regulates 1,25‐dihydroxyvitamin D3‐induced senescence in glioma cells. J Cell Physiol 2019; 234:17990-17998. [PMID: 30825201 DOI: 10.1002/jcp.28431] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/04/2019] [Accepted: 02/14/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Aixia Sui
- Department of Oncology Hebei General Hospital Shijiazhuang China
| | - Yongbing Xu
- Department of Oncology Hebei General Hospital Shijiazhuang China
- Graduate School, Hebei Medical University Shijiazhuang China
| | - Baogen Pan
- Department of Neurosurgery Hebei General Hospital Shijiazhuang China
| | - Tao Guo
- Department of Oncology Hebei General Hospital Shijiazhuang China
| | - Jiang Wu
- Department of Neurosurgery Hebei General Hospital Shijiazhuang China
| | - Yongqing Shen
- Department of Nursing Hebei University of Chinese Medicine Shijiazhuang China
| | - Junjie Yang
- Department of Oncology Hebei General Hospital Shijiazhuang China
| | - Xiaoqiang Guo
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics Peking University Shenzhen Hospital Shenzhen China
| |
Collapse
|
22
|
Abstract
Conventional root canal therapies yield high success rates. The treatment outcomes are negatively affected by the presence of apical periodontitis (AP), which reflects active root canal infection and inflammatory responses. Also, cross-sectional studies revealed surprisingly high prevalence of AP in the general population, especially in those with prior endodontic treatments. Hence, AP is an ongoing disease entity in endodontics that needs further understanding of the pathogenesis and disease progression. The current Chapter will discuss the basic mechanisms of AP with emphasis on emerging role of epigenetic regulators in regulation of inflammatory mediators.
Collapse
|
23
|
Abstract
Aging is characterized by progressive deterioration of physiological integrity, decline in homeostasis, and degeneration of the tissues that occurs after the reproductive phase of life is complete, leading to impaired function. This deterioration is an important risk factor for chronic lung pathologies such as chronic obstructive pulmonary disease (COPD). COPD is a disease that develops gradually. Emphysematous changes in the lung take years to develop after exposure to cigarette smoke; hence, the vast majority of patients are elderly. There has been a dramatic increase in the life expectancy of the general population, resulting in an increased burden of chronic lung diseases. There is growing evidence that molecular mechanisms involved in aging may also play a role in COPD pathogenesis. Recently, the nine hallmarks of aging were identified. In this article, we will review the nine hallmarks of aging and how each hallmark contributes to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Shweta P Kukrety
- Department of Internal Medicine, Creighton University, Omaha, NE, USA
| | - Jai D Parekh
- Department of Internal Medicine, Creighton University, Omaha, NE, USA
| | - Kristina L Bailey
- Department of Internal Medicine, University of Nebraska Medical Center; Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
24
|
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
|
25
|
The evolution of Sex-linked barring alleles in chickens involves both regulatory and coding changes in CDKN2A. PLoS Genet 2017; 13:e1006665. [PMID: 28388616 PMCID: PMC5384658 DOI: 10.1371/journal.pgen.1006665] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 03/01/2017] [Indexed: 02/03/2023] Open
Abstract
Sex-linked barring is a fascinating plumage pattern in chickens recently shown to be associated with two non-coding and two missense mutations affecting the ARF transcript at the CDKN2A tumor suppressor locus. It however remained a mystery whether all four mutations are indeed causative and how they contribute to the barring phenotype. Here, we show that Sex-linked barring is genetically heterogeneous, and that the mutations form three functionally different variant alleles. The B0 allele carries only the two non-coding changes and is associated with the most dilute barring pattern, whereas the B1 and B2 alleles carry both the two non-coding changes and one each of the two missense mutations causing the Sex-linked barring and Sex-linked dilution phenotypes, respectively. The data are consistent with evolution of alleles where the non-coding changes occurred first followed by the two missense mutations that resulted in a phenotype more appealing to humans. We show that one or both of the non-coding changes are cis-regulatory mutations causing a higher CDKN2A expression, whereas the missense mutations reduce the ability of ARF to interact with MDM2. Caspase assays for all genotypes revealed no apoptotic events and our results are consistent with a recent study indicating that the loss of melanocyte progenitors in Sex-linked barring in chicken is caused by premature differentiation and not apoptosis. Our results show that CDKN2A is a major locus driving the differentiation of avian melanocytes in a temporal and spatial manner.
Collapse
|
26
|
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
|
27
|
Kang MK, Mehrazarin S, Park NH, Wang CY. Epigenetic gene regulation by histone demethylases: emerging role in oncogenesis and inflammation. Oral Dis 2016; 23:709-720. [PMID: 27514027 DOI: 10.1111/odi.12569] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/05/2016] [Accepted: 08/09/2016] [Indexed: 12/11/2022]
Abstract
Histone N-terminal tails of nucleosomes are the sites of complex regulation of gene expression through post-translational modifications. Among these modifications, histone methylation had long been associated with permanent gene inactivation until the discovery of Lys-specific demethylase (LSD1), which is responsible for dynamic gene regulation. There are more than 30 members of the Lys demethylase (KDM) family, and with exception of LSD1 and LSD2, all other KDMs possess the Jumonji C (JmjC) domain exhibiting demethylase activity and require unique cofactors, for example, Fe(II) and α-ketoglutarate. These cofactors have been targeted when devising KDM inhibitors, which may yield therapeutic benefit. KDMs and their counterpart Lys methyltransferases (KMTs) regulate multiple biological processes, including oncogenesis and inflammation. KDMs' functional interactions with retinoblastoma (Rb) and E2 factor (E2F) target promoters illustrate their regulatory role in cell cycle progression and oncogenesis. Recent findings also demonstrate the control of inflammation and immune functions by KDMs, such as KDM6B that regulates the pro-inflammatory gene expression and CD4+ T helper (Th) cell lineage determination. This review will highlight the mechanisms by which KDMs and KMTs regulate the target gene expression and how epigenetic mechanisms may be applied to our understanding of oral inflammation.
Collapse
Affiliation(s)
- M K Kang
- Shapiro Laboratory of Viral Oncology and Aging Research, Los Angeles, CA, USA
| | - S Mehrazarin
- Shapiro Laboratory of Viral Oncology and Aging Research, Los Angeles, CA, USA
| | - N-H Park
- Shapiro Laboratory of Viral Oncology and Aging Research, Los Angeles, CA, USA.,David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - C-Y Wang
- Laboratory of Molecular Signaling, UCLA School of Dentistry, Los Angeles, CA, USA
| |
Collapse
|
28
|
Menon R, Papaconstantinou J. p38 Mitogen activated protein kinase (MAPK): a new therapeutic target for reducing the risk of adverse pregnancy outcomes. Expert Opin Ther Targets 2016; 20:1397-1412. [PMID: 27459026 DOI: 10.1080/14728222.2016.1216980] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Spontaneous preterm birth (PTB) and preterm premature rupture of the membranes (pPROM) remain as a major clinical and therapeutic problem for intervention and management. Current strategies, based on our knowledge of pathways of preterm labor, have only been effective, in part, due to major gaps in our existing knowledge of risks and risk specific pathways. Areas covered: Recent literature has identified physiologic aging of fetal tissues as a potential mechanistic feature of normal parturition. This process is affected by telomere dependent and p38 mitogen activated protein kinase (MAPK) induced senescence activation. Pregnancy associated risk factors can cause pathologic activation of this pathway that can cause oxidative stress induced p38 MAPK activation leading to senescence and premature aging of fetal tissues. Premature aging is associated with sterile inflammation capable of triggering preterm labor or preterm premature rupture of membranes. Preterm activation of p38MAPK can be considered as a key contributor to adverse pregnancies. Expert opinion: This review considers p38MAPK activation as a potential target for therapeutic interventions to prevent adverse pregnancy outcomes mediated by stress factors. In this review, we propose multiple strategies to prevent p38MAPK activation.
Collapse
Affiliation(s)
- Ramkumar Menon
- a Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology , The University of Texas Medical Branch at Galveston , Galveston , TX , USA
| | - John Papaconstantinou
- b Department of Biochemistry and Molecular Biology , The University of Texas Medical Branch at Galveston , Galveston , TX , USA
| |
Collapse
|
29
|
Papaconstantinou J, Wang CZ, Zhang M, Yang S, Deford J, Bulavin DV, Ansari NH. Attenuation of p38α MAPK stress response signaling delays the in vivo aging of skeletal muscle myofibers and progenitor cells. Aging (Albany NY) 2016; 7:718-33. [PMID: 26423835 PMCID: PMC4600628 DOI: 10.18632/aging.100802] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Functional competence and self-renewal of mammalian skeletal muscle myofibers and progenitor cells declines with age. Progression of the muscle aging phenotype involves the decline of juvenile protective factors i.e., proteins whose beneficial functions translate directly to the quality of life, and self-renewal of progenitor cells. These characteristics occur simultaneously with the age-associated increase of p38α stress response signaling. This suggests that the maintenance of low levels of p38α activity of juvenile tissues may delay or attenuate aging. We used the dominant negative haploinsufficient p38α mouse (DN-p38αAF/+) to demonstrate that in vivo attenuation of p38α activity in the gastrocnemius of the aged mutant delays age-associated processes that include: a) the decline of the juvenile protective factors, BubR1, aldehyde dehydrogenase 1A (ALDH1A1), and aldehyde dehydrogenase 2 (ALDH2); b) attenuated expression of p16Ink4a and p19Arf tumor suppressor genes of the Cdkn2a locus; c) decreased levels of hydroxynonenal protein adducts, expression of COX2 and iNOS; d) decline of the senescent progenitor cell pool level and d) the loss of gastrocnemius muscle mass. We propose that elevated P-p38α activity promotes skeletal muscle aging and that the homeostasis of p38α impacts the maintenance of a beneficial healthspan.
Collapse
Affiliation(s)
- John Papaconstantinou
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - Chen Z Wang
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - Min Zhang
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - San Yang
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - James Deford
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| | - Dmitry V Bulavin
- Institute for Research on Cancer and Ageing of Nice, INSERM, U1081-UMR CNRS 7284, University of Nice - Sophia Antipolis, Centre Antoine Lacassagne, Nice, France
| | - Naseem H Ansari
- The Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX 77551-06743, USA
| |
Collapse
|
30
|
Yun MH. Changes in Regenerative Capacity through Lifespan. Int J Mol Sci 2015; 16:25392-432. [PMID: 26512653 PMCID: PMC4632807 DOI: 10.3390/ijms161025392] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 12/14/2022] Open
Abstract
Most organisms experience changes in regenerative abilities through their lifespan. During aging, numerous tissues exhibit a progressive decline in homeostasis and regeneration that results in tissue degeneration, malfunction and pathology. The mechanisms responsible for this decay are both cell intrinsic, such as cellular senescence, as well as cell-extrinsic, such as changes in the regenerative environment. Understanding how these mechanisms impact on regenerative processes is essential to devise therapeutic approaches to improve tissue regeneration and extend healthspan. This review offers an overview of how regenerative abilities change through lifespan in various organisms, the factors that underlie such changes and the avenues for therapeutic intervention. It focuses on established models of mammalian regeneration as well as on models in which regenerative abilities do not decline with age, as these can deliver valuable insights for our understanding of the interplay between regeneration and aging.
Collapse
Affiliation(s)
- Maximina H Yun
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
31
|
Zhou L, Chen X, Liu T, Gong Y, Chen S, Pan G, Cui W, Luo ZP, Pei M, Yang H, He F. Melatonin reverses H2 O2 -induced premature senescence in mesenchymal stem cells via the SIRT1-dependent pathway. J Pineal Res 2015; 59:190-205. [PMID: 25975679 PMCID: PMC4523475 DOI: 10.1111/jpi.12250] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/08/2015] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) represent an attractive source for stem cell-based regenerative therapy, but they are vulnerable to oxidative stress-induced premature senescence in pathological conditions. We previously reported antioxidant and antiarthritic effects of melatonin on MSCs against proinflammatory cytokines. In this study, we hypothesized that melatonin could protect MSCs from premature senescence induced by hydrogen peroxide (H2 O2 ) via the silent information regulator type 1 (SIRT1)-dependent pathway. In response to H2 O2 at a sublethal concentration of 200 μm, human bone marrow-derived MSCs (BM-MSCs) underwent growth arrest and cellular senescence. Treatment with melatonin before H2 O2 exposure cannot significantly prevent premature senescence; however, treatment with melatonin subsequent to H2 O2 exposure successfully reversed the senescent phenotypes of BM-MSCs in a dose-dependent manner. This result was made evident by improved cell proliferation, decreased senescence-associated β-galactosidase activity, and the improved entry of proliferating cells into the S phase. In addition, treatment with 100 μm melatonin restored the osteogenic differentiation potential of BM-MSCs that was inhibited by H2 O2 -induced premature senescence. We also found that melatonin attenuated the H2 O2 -stimulated phosphorylation of p38 mitogen-activated protein kinase, decreased expression of the senescence-associated protein p16(INK) (4α) , and increased SIRT1. Further molecular experiments revealed that luzindole, a nonselective antagonist of melatonin receptors, blocked melatonin-mediated antisenescence effects. Inhibition of SIRT1 by sirtinol counteracted the protective effects of melatonin, suggesting that melatonin reversed the senescence in cells through the SIRT1-dependent pathway. Together, these findings lay new ground for understanding oxidative stress-induced premature senescence and open perspectives for therapeutic applications of melatonin in stem cell-based regenerative medicine.
Collapse
Affiliation(s)
- Long Zhou
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi Chen
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yihong Gong
- School of Engineering, Sun Yat-sen University, Guangzhou, China
| | - Sijin Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoqing Pan
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenguo Cui
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zong-Ping Luo
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, USA
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
32
|
p16(INK4A) mediates age-related changes in mesenchymal stem cells derived from human dental pulp through the DNA damage and stress response. Mech Ageing Dev 2014; 141-142:46-55. [PMID: 25304494 DOI: 10.1016/j.mad.2014.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 09/21/2014] [Accepted: 09/26/2014] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells derived from human dental pulp (DP-MSCs) are characterized by self-renewal and multi-lineage differentiation, which play important roles in regenerative medicine. Autologous transfers, as non-immunogenic, constitute the safest approach in cellular transplantations. However, their use may be limited by age-related changes. In the study, we compared DP-MSCs isolated from human in five age groups: 5-12 y, 12-20 y, 20-35 y, 35-50 y, and >50 y. We tested the effect of age on proliferation, differentiation, senescence-associated β-galactosidase (SA-β-gal), cell cycle and programmed cell death. DP-MSCs showed characteristics of senescence as a function of age. Meanwhile, the expression of p16(INK4A) and γ-H2A.X significantly increased with age, whereas heat shock protein 60 (HSP60) was decreased in the senescent DP-MSCs. Reactive oxygen species (ROS) staining showed the number of ROS-stained cells and the DCFH fluorescent level were higher in the aged group. Further we examined the senescence of DP-MSCs after modulating p16(INK4A) signaling. The results indicated the dysfunction of DP-MSCs was reversed by p16(INK4A) siRNA. In summary, our study indicated p16(INK4A) pathway may play a critical role in DP-MSCs age-related changes and the DNA damage response (DDR) and stress response may be the main mediators of DP-MSCs senescence induced by excessive activation of p16(INK4A) signaling.
Collapse
|
33
|
Campisi J, Robert L. Cell senescence: role in aging and age-related diseases. ACTA ACUST UNITED AC 2014; 39:45-61. [PMID: 24862014 DOI: 10.1159/000358899] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cell senescence is one of the major paradigms of aging research. It started with the demonstration by L. Hayflick of the limited number of divisions by normal, nontransformed cells, not shown by transformed malignant cells, this processes being largely regulated by the telomere-telomerase system. A complete renewal of this discipline came from the demonstration that cells can enter senescence at any time by an anti-oncogene-triggered pathway, enabling them to escape malignancy. The senescent cell became a major actor of the aging process, among others, by the acquisition of the senescence-associated secretory phenotype. This chapter is devoted to the regulatory process involved in the acquisition of the senescent cell phenotype and its role in organismal aging.
Collapse
Affiliation(s)
- Judith Campisi
- Buck Institute for Research on Aging, Novato, Calif., USA
| | | |
Collapse
|
34
|
Feng X, Feng G, Xing J, Shen B, Tan W, Huang D, Lu X, Tao T, Zhang J, Li L, Gu Z. Repeated lipopolysaccharide stimulation promotes cellular senescence in human dental pulp stem cells (DPSCs). Cell Tissue Res 2014; 356:369-80. [DOI: 10.1007/s00441-014-1799-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/09/2014] [Indexed: 01/09/2023]
|
35
|
Levav-Cohen Y, Goldberg Z, Tan KH, Alsheich-Bartok O, Zuckerman V, Haupt S, Haupt Y. The p53-Mdm2 loop: a critical juncture of stress response. Subcell Biochem 2014; 85:161-86. [PMID: 25201194 DOI: 10.1007/978-94-017-9211-0_9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The presence of a functional p53 protein is a key factor for the proper suppression of cancer development. A loss of p53 activity, by mutations or inhibition, is often associated with human malignancies. The p53 protein integrates various stress signals into a growth restrictive cellular response. In this way, p53 eliminates cells with a potential to become cancerous. Being a powerful decision maker, it is imperative that p53 will be activated properly, efficiently and temporarily in response to stress. Equally important is that p53 activation will be extinguished upon recovery from stress, and that improper activation of p53 will be avoided. Failure to achieve these aims is likely to have catastrophic consequences for the organism. The machinery that governs this tight regulation is largely based on the major inhibitor of p53, Mdm2, which both blocks p53 activities and promotes its destabilization. The interplay between p53 and Mdm2 involves a complex network of positive and negative feedback loops. Relief from Mdm2 suppression is required for p53 to be stabilized and activated in response to stress. Protection from Mdm2 entails a concerted action of modifying enzymes and partner proteins. The association of p53 with the PML-nuclear bodies may provide an infrastructure in which this complex regulatory network can be orchestrated. In this chapter we use examples to illustrate the regulatory machinery that drives this network.
Collapse
Affiliation(s)
- Yaara Levav-Cohen
- Lautenberg Center, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
36
|
Han J, Kim YL, Lee KW, Her NG, Ha TK, Yoon S, Jeong SI, Lee JH, Kang MJ, Lee MG, Ryu BK, Baik JH, Chi SG. ZNF313 is a novel cell cycle activator with an E3 ligase activity inhibiting cellular senescence by destabilizing p21(WAF1.). Cell Death Differ 2013; 20:1055-67. [PMID: 23645206 DOI: 10.1038/cdd.2013.33] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 03/21/2013] [Accepted: 03/27/2013] [Indexed: 02/02/2023] Open
Abstract
ZNF313 encoding a zinc-binding protein is located at chromosome 20q13.13, which exhibits a frequent genomic amplification in multiple human cancers. However, the biological function of ZNF313 remains largely undefined. Here we report that ZNF313 is an ubiquitin E3 ligase that has a critical role in the regulation of cell cycle progression, differentiation and senescence. In this study, ZNF313 is initially identified as a XIAP-associated factor 1 (XAF1)-interacting protein, which upregulates the stability and proapoptotic effect of XAF1. Intriguingly, we found that ZNF313 activates cell cycle progression and suppresses cellular senescence through the RING domain-mediated degradation of p21(WAF1). ZNF313 ubiquitinates p21(WAF1) and also destabilizes p27(KIP1) and p57(KIP2), three members of the CDK-interacting protein (CIP)/kinase inhibitor protein (KIP) family of cyclin-dependent kinase inhibitors, whereas it does not affect the stability of the inhibitor of CDK (INK4) family members, such as p16(INK4A) and p15(INK4B). ZNF313 expression is tightly controlled during the cell cycle and its elevation at the late G1 phase is crucial for the G1-to-S phase transition. ZNF313 is induced by mitogenic growth factors and its blockade profoundly delays cell cycle progression and accelerates p21(WAF1)-mediated senescence. Both replicative and stress-induced senescence are accompanied with ZNF313 reduction. ZNF313 is downregulated during cellular differentiation process in vitro and in vivo, while it is commonly upregulated in many types of cancer cells. ZNF313 shows both the nuclear and cytoplasmic localization in epithelial cells of normal tissues, but exhibits an intense cytoplasmic distribution in carcinoma cells of tumor tissues. Collectively, ZNF313 is a novel E3 ligase for p21(WAF1), whose alteration might be implicated in the pathogenesis of several human diseases, including cancers.
Collapse
Affiliation(s)
- J Han
- School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fu L, Kettner NM. The circadian clock in cancer development and therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 119:221-82. [PMID: 23899600 PMCID: PMC4103166 DOI: 10.1016/b978-0-12-396971-2.00009-9] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most aspects of mammalian function display circadian rhythms driven by an endogenous clock. The circadian clock is operated by genes and comprises a central clock in the brain that responds to environmental cues and controls subordinate clocks in peripheral tissues via circadian output pathways. The central and peripheral clocks coordinately generate rhythmic gene expression in a tissue-specific manner in vivo to couple diverse physiological and behavioral processes to periodic changes in the environment. However, with the industrialization of the world, activities that disrupt endogenous homeostasis with external circadian cues have increased. This change in lifestyle has been linked to an increased risk of diseases in all aspects of human health, including cancer. Studies in humans and animal models have revealed that cancer development in vivo is closely associated with the loss of circadian homeostasis in energy balance, immune function, and aging, which are supported by cellular functions important for tumor suppression including cell proliferation, senescence, metabolism, and DNA damage response. The clock controls these cellular functions both locally in cells of peripheral tissues and at the organismal level via extracellular signaling. Thus, the hierarchical mammalian circadian clock provides a unique system to study carcinogenesis as a deregulated physiological process in vivo. The asynchrony between host and malignant tissues in cell proliferation and metabolism also provides new and exciting options for novel anticancer therapies.
Collapse
Affiliation(s)
- Loning Fu
- Department of Pediatrics/U.S. Department of Agriculture/Agricultural Research Service/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Nicole M. Kettner
- Department of Pediatrics/U.S. Department of Agriculture/Agricultural Research Service/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
38
|
Methods for assessing effects of Wnt/β-catenin signaling in senescence of mesenchymal stem cells. Methods Mol Biol 2013; 976:111-30. [PMID: 23400438 DOI: 10.1007/978-1-62703-317-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs) represent a main population of stem cells and can differentiate into multiple cell lineages. Recently, MSC transplantation has been applied to repair the malfunctioned tissues. However, increasing evidences show that some MSCs expanded in vitro and in the aged individuals become senescent. Capacity of senescent MSCs in repairing the tissues may decrease significantly. Interestingly, preventing MSC senescence is a powerful potential strategy to delay aging of individuals and promote application of cell therapy for treating aging-related diseases. Therefore, it is necessary to explore mechanisms of MSC senescence in detail. Methods to assess MSC senescence in vitro include induction of senescence, detection of senescent changes and investigation of the molecules involved in senescence. Here we describe the methods to detect MSC senescence induced with old serum and investigate effects of Wnt/β-catenin signaling on MSC senescence.
Collapse
|
39
|
Abstract
For most species, aging promotes a host of degenerative pathologies that are characterized by debilitating losses of tissue or cellular function. However, especially among vertebrates, aging also promotes hyperplastic pathologies, the most deadly of which is cancer. In contrast to the loss of function that characterizes degenerating cells and tissues, malignant (cancerous) cells must acquire new (albeit aberrant) functions that allow them to develop into a lethal tumor. This review discusses the idea that, despite seemingly opposite characteristics, the degenerative and hyperplastic pathologies of aging are at least partly linked by a common biological phenomenon: a cellular stress response known as cellular senescence. The senescence response is widely recognized as a potent tumor suppressive mechanism. However, recent evidence strengthens the idea that it also drives both degenerative and hyperplastic pathologies, most likely by promoting chronic inflammation. Thus, the senescence response may be the result of antagonistically pleiotropic gene action.
Collapse
Affiliation(s)
- Judith Campisi
- Buck Institute for Research on Aging, Novato, California 94945, USA.
| |
Collapse
|
40
|
Yoruker EE, Mert U, Bugra D, Yamaner S, Dalay N. Promoter and histone methylation and p16(INK4A) gene expression in colon cancer. Exp Ther Med 2012; 4:865-870. [PMID: 23226740 PMCID: PMC3493785 DOI: 10.3892/etm.2012.683] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/06/2012] [Indexed: 01/08/2023] Open
Abstract
The inactivation of the cyclin-dependent kinase inhibitor p16INK4A gene by hypermethylation is observed in numerous types of cancer. New findings indicate that DNA and histone methylation act in concert in gene silencing. In this study, we investigated the methylation status of the p16INK4A gene promoter and the histone 3 lysine 9 residue in the tumors and matched normal tissue samples from patients with colorectal cancer and analyzed their association with gene expression. The methylation and expression of the p16INK4A gene were analyzed by real-time PCR, and histone methylation was analyzed by chromatin immunoprecipitation followed by real-time PCR. p16INK4A expression was significantly higher in the tumors compared to normal tissue. Mono-, di- and trimethylation levels of the H3K9 residue were similar in the tumor and normal tissue samples. We did not observe any significant correlation between p16INK4A methylation or expression and clinical parameters. Our results suggest that epigenetic modifications of the p16INK4A gene and histone lysine methylation do not play a major role in colon carcinogenesis.
Collapse
|
41
|
Dekker P, Gunn D, McBryan T, Dirks RW, van Heemst D, Lim FL, Jochemsen AG, Verlaan-de Vries M, Nagel J, Adams PD, Tanke HJ, Westendorp RG, Maier AB. Microarray-based identification of age-dependent differences in gene expression of human dermal fibroblasts. Mech Ageing Dev 2012; 133:498-507. [DOI: 10.1016/j.mad.2012.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 02/07/2012] [Accepted: 06/10/2012] [Indexed: 10/28/2022]
|
42
|
Abstract
Aging is a dominant risk factor for most forms of cardiovascular disease. Impaired angiogenesis and endothelial dysfunction likely contribute to the increased prevalence of both cardiovascular diseases and their adverse sequelae in the elderly. Angiogenesis is both an essential adaptive response to physiological stress and an endogenous repair mechanism after ischemic injury. In addition, induction of angiogenesis is a promising therapeutic approach for ischemic diseases. For these reasons, understanding the basis of age-related impairment of angiogenesis and endothelial function has important implications for understanding and managing cardiovascular disease. In this review, we discuss the molecular mechanisms that contribute to impaired angiogenesis in the elderly and potential therapeutic approaches to improving vascular function and angiogenesis in aging patients.
Collapse
Affiliation(s)
- Johanna Lähteenvuo
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
43
|
Kong Y, Cui H, Zhang H. Smurf2-mediated ubiquitination and degradation of Id1 regulates p16 expression during senescence. Aging Cell 2011; 10:1038-46. [PMID: 21933340 DOI: 10.1111/j.1474-9726.2011.00746.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The inhibitor of differentiation or DNA binding (Id) family of transcription regulators plays an important role in cell proliferation, differentiation, and senescence. However, regulation of Id expression during these processes is poorly understood. Id proteins are known to undergo rapid turnover mediated by the ubiquitin-proteasome pathway. Anaphase-promoting complex has been shown to ubiquitinate Id2, but E3 ubiquitin ligase(s) that ubiquitinate other Id family members are not known. Here, we report for the first time the identification of Smurf2 as the E3 ligase that ubiquitinates Id1 and Id3. Smurf2-mediated ubiquitination and consequent degradation of Id1 or Id3 plays an important role in the regulation of Id expression in senescent cells. Furthermore, we found that Id1 is the mediator through which Smurf2 regulates p16 expression, providing a mechanistic link between Smurf2 and p16 expression during senescence.
Collapse
Affiliation(s)
- Yahui Kong
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | |
Collapse
|
44
|
Kim HJ, Ham SA, Kim MY, Hwang JS, Lee H, Kang ES, Yoo T, Woo IS, Yabe-Nishimura C, Paek KS, Kim JH, Seo HG. PPARδ coordinates angiotensin II-induced senescence in vascular smooth muscle cells through PTEN-mediated inhibition of superoxide generation. J Biol Chem 2011; 286:44585-93. [PMID: 22072715 DOI: 10.1074/jbc.m111.222562] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence-associated changes in blood vessels have been implicated in aging and age-related cardiovascular disorders. Here, we demonstrate that peroxisome proliferator-activated receptor (PPAR) δ coordinates angiotensin (Ang) II-induced senescence of human vascular smooth muscle cells (VSMCs). Activation of PPARδ by GW501516, a specific ligand for PPARδ, significantly attenuated Ang II-induced generation of superoxides and suppressed senescence of VSMCs. A marked increase in the levels of p53 and p21 induced by Ang II was blunted by the treatment with GW501516. Ligand-activated PPARδ up-regulated expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and suppressed the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Knockdown of PTEN with siRNA abrogated the effects of PPARδ on cellular senescence, on PI3K/Akt signaling, and on generation of ROS in VSMCs treated with Ang II. Finally, administration of GW501516 to apoE-deficient mice treated with Ang II significantly reduced the number of senescent cells in the aorta, where up-regulation of PTEN with reduced levels of phosphorylated Akt and ROS was demonstrated. Thus, ligand-activated PPARδ confers resistance to Ang II-induced senescence by up-regulation of PTEN and ensuing modulation of the PI3K/Akt signaling to reduce ROS generation in vascular cells.
Collapse
Affiliation(s)
- Hyo Jung Kim
- Department of Animal Biotechnology, Konkuk University, Seoul 143-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chen IP, Henning S, Faust A, Boukamp P, Volkmer B, Greinert R. UVA-induced epigenetic regulation of P16(INK4a) in human epidermal keratinocytes and skin tumor derived cells. Photochem Photobiol Sci 2011; 11:180-90. [PMID: 21986889 DOI: 10.1039/c1pp05197k] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
UVA-radiation (315-400 nm) has been demonstrated to be capable of inducing DNA damage and is regarded as a carcinogen. While chromosomal aberrations found in UVA-irradiated cells and skin tumors provided evidence of the genetic involvement in UVA-carcinogenesis, its epigenetic participation is still illusive. We thus analysed the epigenetic patterns of 5 specific genes that are involved in stem cell fate (KLF4, NANOG), telomere maintenance (hTERT) and tumor suppression in cell cycle control (P16(INK4a), P21(WAFI/CIPI)) in chronically UVA-irradiated HaCaT human keratinocytes. A striking reduction of the permissive histone mark H3K4me3 has been detected in the promoter of P16(INK4a) (4-fold and 9-fold reduction for 10 and 15 weeks UVA-irradiated cells, respectively), which has often been found deregulated in skin cancers. This alteration in histone modification together with a severe promoter hypermethylation strongly impaired the transcription of P16(INK4a) (20-fold and 40-fold for 10 weeks and 15 weeks UVA-irradiation, respectively). Analysis of the skin tumor-derived cells revealed the same severe impairment of the P16(INK4a) transcription attributed to promoter hypermethylation and enrichment of the heterochromatin histone mark H3K9me3 and the repressive mark H3K27me3. Less pronounced UVA-induced epigenetic alterations were also detected for the other genes, demonstrating for the first time that UVA is able to modify transcription of skin cancer associated genes by means of epigenetic DNA and histone alterations.
Collapse
Affiliation(s)
- I-Peng Chen
- Dept. Mol. Cell Biology, Center of Dermatology, Elbekliniken, Stade/Buxtehude, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Vandenberk B, Brouwers B, Hatse S, Wildiers H. p16INK4a: A central player in cellular senescence and a promising aging biomarker in elderly cancer patients. J Geriatr Oncol 2011. [DOI: 10.1016/j.jgo.2011.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Sabin RJ, Anderson RM. Cellular Senescence - its role in cancer and the response to ionizing radiation. Genome Integr 2011; 2:7. [PMID: 21834983 PMCID: PMC3169443 DOI: 10.1186/2041-9414-2-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 08/11/2011] [Indexed: 12/11/2022] Open
Abstract
Cellular senescence is a normal biological process that is initiated in response to a range of intrinsic and extrinsic factors that functions to remove irreparable damage and therefore potentially harmful cells, from the proliferative pool. Senescence can therefore be thought of in beneficial terms as a tumour suppressor. In contrast to this, there is a growing body of evidence suggesting that senescence is also associated with the disruption of the tissue microenvironment and development of a pro-oncogenic environment, principally via the secretion of senescence-associated pro-inflammatory factors. The fraction of cells in a senescent state is known to increase with cellular age and from exposure to various stressors including ionising radiation therefore, the implications of the detrimental effects of the senescent phenotype are important to understand within the context of the increasing human exposure to ionising radiation. This review will discuss what is currently understood about senescence, highlighting possible associations between senescence and cancer and, how exposure to ionising radiation may modify this.
Collapse
Affiliation(s)
- Rebecca J Sabin
- Centre for Cell and Chromosome Biology and Centre for Infection, Immunity and Disease Mechanisms, Division of Biosciences, Brunel University, West London, UB8 3PH, UK.
| | | |
Collapse
|
48
|
Zhang DY, Wang HJ, Tan YZ. Wnt/β-catenin signaling induces the aging of mesenchymal stem cells through the DNA damage response and the p53/p21 pathway. PLoS One 2011; 6:e21397. [PMID: 21712954 PMCID: PMC3119703 DOI: 10.1371/journal.pone.0021397] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 05/27/2011] [Indexed: 11/19/2022] Open
Abstract
Recent studies have demonstrated the importance of cellular extrinsic factors in the aging of adult stem cells. However, the effects of an aged cell–extrinsic environment on mesenchymal stem cell (MSC) aging and the factors involved remain unclear. In the current study, we examine the effects of old rat serum (ORS) on the aging of MSCs, and explore the effects and mechanisms of Wnt/β-catenin signaling on MSC aging induced by ORS treatment. Senescence-associated changes in the cells are examined with SA-β-galactosidase staining and ROS staining. The proliferation ability is detected by MTT assay. The surviving and apoptotic cells are determined using AO/EB staining. The results suggest that ORS promotes MSC senescence and reduces the proliferation and survival of cells. The immunofluorescence staining shows that the expression of β-catenin increases in MSCs of old rats. To identify the effects of Wnt/β-catenin signaling on MSC aging induced with ORS, the expression of β-catenin, GSK-3β, and c-myc are detected. The results show that the Wnt/β-catenin signaling in the cells is activated after ORS treatment. Then we examine the aging, proliferation, and survival of MSCs after modulating Wnt/β-catenin signaling. The results indicate that the senescence and dysfunction of MSCs in the medium containing ORS is reversed by the Wnt/β-catenin signaling inhibitor DKK1 or by β-catenin siRNA. Moreover, the expression of γ-H2A.X, a molecular marker of DNA damage response, p16INK4a, p53, and p21 is increased in senescent MSCs induced with ORS, and is also reversed by DKK1 or by β-catenin siRNA. In summary, our study indicates the Wnt/β-catenin signaling may play a critical role in MSC aging induced by the serum of aged animals and suggests that the DNA damage response and p53/p21 pathway may be the main mediators of MSC aging induced by excessive activation of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Da-yong Zhang
- Department of Anatomy and Histology and Embryology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Hai-jie Wang
- Department of Anatomy and Histology and Embryology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
| | - Yu-zhen Tan
- Department of Anatomy and Histology and Embryology, Shanghai Medical College of Fudan University, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
49
|
p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J 2011; 30:1536-48. [PMID: 21399611 PMCID: PMC3102277 DOI: 10.1038/emboj.2011.69] [Citation(s) in RCA: 744] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 02/18/2011] [Indexed: 12/11/2022] Open
Abstract
Cellular senescence suppresses cancer by forcing potentially oncogenic cells into a permanent cell cycle arrest. Senescent cells also secrete growth factors, proteases, and inflammatory cytokines, termed the senescence-associated secretory phenotype (SASP). Much is known about pathways that regulate the senescence growth arrest, but far less is known about pathways that regulate the SASP. We previously showed that DNA damage response (DDR) signalling is essential, but not sufficient, for the SASP, which is restrained by p53. Here, we delineate another crucial SASP regulatory pathway and its relationship to the DDR and p53. We show that diverse senescence-inducing stimuli activate the stress-inducible kinase p38MAPK in normal human fibroblasts. p38MAPK inhibition markedly reduced the secretion of most SASP factors, constitutive p38MAPK activation was sufficient to induce an SASP, and p53 restrained p38MAPK activation. Further, p38MAPK regulated the SASP independently of the canonical DDR. Mechanistically, p38MAPK induced the SASP largely by increasing NF-κB transcriptional activity. These findings assign p38MAPK a novel role in SASP regulation--one that is necessary, sufficient, and independent of previously described pathways.
Collapse
|
50
|
Kong Y, Cui H, Ramkumar C, Zhang H. Regulation of senescence in cancer and aging. J Aging Res 2011; 2011:963172. [PMID: 21423549 PMCID: PMC3056284 DOI: 10.4061/2011/963172] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/12/2011] [Indexed: 12/12/2022] Open
Abstract
Senescence is regarded as a physiological response of cells to stress, including telomere dysfunction, aberrant oncogenic activation, DNA damage, and oxidative stress. This stress response has an antagonistically pleiotropic effect to organisms: beneficial as a tumor suppressor, but detrimental by contributing to aging. The emergence of senescence as an effective tumor suppression mechanism is highlighted by recent demonstration that senescence prevents proliferation of cells at risk of neoplastic transformation. Consequently, induction of senescence is recognized as a potential treatment of cancer. Substantial evidence also suggests that senescence plays an important role in aging, particularly in aging of stem cells. In this paper, we will discuss the molecular regulation of senescence its role in cancer and aging. The potential utility of senescence in cancer therapeutics will also be discussed.
Collapse
Affiliation(s)
- Yahui Kong
- Department of Cell Biology, University of Massachusetts Medical School, 55 Lake Avenue North, S7-125, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|