1
|
Park M, Ha J, Lee Y, Kwon Y, Choi SH, Kim BS, Jeong YK. BR101801 enhances the radiosensitivity of p53-deficient colorectal cancer cells by inducing G2/M arrest, apoptosis, and senescence in a p53-independent manner. Am J Cancer Res 2023; 13:5887-5900. [PMID: 38187039 PMCID: PMC10767343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 10/28/2023] [Indexed: 01/09/2024] Open
Abstract
Inhibition of DNA-dependent protein kinase (DNA-PK) in the non-homologous end-joining repair pathway reportedly increases the radiation sensitivity of cancer cells. We have recently reported that BR101801, a novel triple inhibitor of PI3K-gamma (γ), delta (δ), and DNA-PK, functions as an efficient sensitizer of radiation-induced DNA damage in various human solid cancer cells and a xenograft mouse model. Given that the p53 tumor suppressor gene plays an important role in radiotherapeutic efficacy, in the current study, we focused on the impact of the p53 status on BR101801-induced radiosensitization using isogenic HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cell lines. In vitro, HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cells were pretreated with 1 μM BR101801 for 24 h before exposure to ionizing radiation (IR), followed by assays to analyze colony formation, DNA damage, cell cycle changes, senescence, autophagy, apoptosis, and DNA damage response-related proteins. Xenograft mouse models were constructed to examine the potential synergistic effects of BR101801 (50 mg/kg, orally administered once daily) and fractionated IR (2 Gy × 3 days) on tumor growth inhibition in vivo. BR101801 inhibited cell proliferation and prolonged DNA damage in both HCT116 p53+/+ and HCT116 p53-/- human colorectal cancer cells. Combined treatment with BR101801 and IR robustly induced G2/M phase cell cycle arrest, apoptosis, and cellular senescence in HCT116 p53-/- cells when compared with treatment with IR alone. Furthermore, BR101801 synergistically inhibited tumor growth in the HCT116 p53-/- xenograft mouse model. BR101801 enhanced the radiosensitivity of HCT116 human colorectal cancer cells regardless of their p53 status. Moreover, BR101801 exerted robust synergistic effects on IR-induced cell cycle arrest, apoptosis, and tumor growth inhibition, even in radioresistant HCT116 p53-/- cells. Overall, these findings provide a scientific rationale for combining BR101801 with IR as a new therapeutic strategy to overcome radioresistance induced by p53 deficiency.
Collapse
Affiliation(s)
- Mijeong Park
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans UniversitySeoul 03760, Republic of Korea
| | - Jimin Ha
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Yuri Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans UniversitySeoul 03760, Republic of Korea
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Youngjoo Kwon
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans UniversitySeoul 03760, Republic of Korea
| | - Sang Hyun Choi
- Research Team of Medical Physics and Engineering, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Byoung Soo Kim
- Division of Applied RI, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| | - Youn Kyoung Jeong
- Radiological and Medical Support Center, Korea Institute of Radiological and Medical SciencesSeoul 01812, Republic of Korea
| |
Collapse
|
2
|
Bu LL, Yuan HH, Xie LL, Guo MH, Liao DF, Zheng XL. New Dawn for Atherosclerosis: Vascular Endothelial Cell Senescence and Death. Int J Mol Sci 2023; 24:15160. [PMID: 37894840 PMCID: PMC10606899 DOI: 10.3390/ijms242015160] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Endothelial cells (ECs) form the inner linings of blood vessels, and are directly exposed to endogenous hazard signals and metabolites in the circulatory system. The senescence and death of ECs are not only adverse outcomes, but also causal contributors to endothelial dysfunction, an early risk marker of atherosclerosis. The pathophysiological process of EC senescence involves both structural and functional changes and has been linked to various factors, including oxidative stress, dysregulated cell cycle, hyperuricemia, vascular inflammation, and aberrant metabolite sensing and signaling. Multiple forms of EC death have been documented in atherosclerosis, including autophagic cell death, apoptosis, pyroptosis, NETosis, necroptosis, and ferroptosis. Despite this, the molecular mechanisms underlying EC senescence or death in atherogenesis are not fully understood. To provide a comprehensive update on the subject, this review examines the historic and latest findings on the molecular mechanisms and functional alterations associated with EC senescence and death in different stages of atherosclerosis.
Collapse
Affiliation(s)
- Lan-Lan Bu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.-L.B.); (D.-F.L.)
| | - Huan-Huan Yuan
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
| | - Ling-Li Xie
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Min-Hua Guo
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (H.-H.Y.); (L.-L.X.); (M.-H.G.)
| | - Duan-Fang Liao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (L.-L.B.); (D.-F.L.)
| | - Xi-Long Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
3
|
Nasiri L, Vaez-Mahdavi MR, Hassanpour H, Ghazanfari T, Kaboudanian Ardestani S, Askari N, Mohseni Majd MA, Rahimlou B. Increased serum lipofuscin associated with leukocyte telomere shortening in veterans: a possible role for sulfur mustard exposure in delayed-onset accelerated cellular senescence. Int Immunopharmacol 2023; 114:109549. [PMID: 36508921 DOI: 10.1016/j.intimp.2022.109549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Sulfur mustard (SM) is a toxic gas that causes chronic inflammation and oxidative stress leading to cell senescence. This study aimed to evaluate two indicators of biological aging (i.e., serum lipofuscin level and leukocyte telomere length) and assess their relationship based on the severity of SM exposure in the long term. METHODS The study was performed on two groups of male participants. 1) SM-exposed group (exposed to SM once in 1987), 73 volunteers. 2) Non-exposed group, 16 healthy volunteers. The SM-exposed group was categorized into three subgroups based on the severity of SM exposure and body damage (asymptom, mild, and severe). The blood sample was prepared from members of each group. The serum lipofuscin, TGF-β, malondialdehyde (MDA), c-reactive protein (CRP), and leukocyte telomere length (TL) were measured in all participants. RESULTS The MDA level was increased in the SM-exposed group (mean = 39.6 µM, SD = 16.5) compared to the non-exposed group (mean = 21.1 µM, SD = 10.3) (P < 0.05). The CRP level was also increased in the SM-exposed group (mean = 5.12 mg/l, SD = 3.36) compared to the non-exposed group (mean = 3.51 mg/l, SD = 1.21), while the TGF-β level was decreased (P < 0.05) in the SM-exposed group (mean = 52.6 pg/ml, SD = 18.7) compared to the non-exposed group (mean = 68.9 pg/ml, SD = 13.8). The relative TL was shorter in the SM-exposed group (mean = 0.40, SD = 0.28) than in the non-exposed group (mean = 2.25, SD = 1.41) (P < 0.05). The lipofuscin level was higher in the total SM-exposed group (mean = 1.44 ng/ml, SD = 0.685) than in the non-exposed group (mean = 0.88 ng/ml, SD = 0.449) (P < 0.05). The MDA and CRP levels were increased in the SM-exposed subgroups of asymptom, mild, and severe than the non-exposed group, while TGF-β level and TL were decreased in those subgroups. The lipofuscin level was higher in the SM-exposed subgroups of mild and severe than in the non-exposed group. The regression analysis determined a negative correlation between lipofuscin level and TL. The lipofuscin/TL ratio was higher in the total SM-exposed group (mean = 6.36, SD = 5.342) than in the non-exposed group (mean = 0.51, SD=0.389). This ratio was also higher in the SM-exposed subgroups of asymptom, mild, and severe than in the non-exposed group. The lipofuscin/TL ratio did not differ between mild and severe subgroups. CONCLUSION The delayed toxicity of SM is associated with chronic oxidative stress, continuous inflammatory stimulation, increased lipofuscin, and telomere shortening. Future studies are needed to verify the suitability of serum lipofuscin to telomere length ratio in determining the severity of SM toxicity.
Collapse
Affiliation(s)
- Leila Nasiri
- Department of Health Equity, Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Mohammad-Reza Vaez-Mahdavi
- Department of Health Equity, Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Physiology, Medical Faculty, Shahed University, Tehran, Iran.
| | - Hossein Hassanpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| | - Sussan Kaboudanian Ardestani
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Nayere Askari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of biology, Faculty of Basic Sciences, Shahid Bahonar University, Kerman, Iran
| | | | - Bahman Rahimlou
- Department of Immunology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
4
|
Onset of Senescence and Steatosis in Hepatocytes as a Consequence of a Shift in the Diacylglycerol/Ceramide Balance at the Plasma Membrane. Cells 2021; 10:cells10061278. [PMID: 34064003 PMCID: PMC8224046 DOI: 10.3390/cells10061278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/06/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Ceramide and diacylglycerol (DAG) are bioactive lipids and mediate many cellular signaling pathways. Sphingomyelin synthase (SMS) is the single metabolic link between the two, while SMS2 is the only SMS form located at the plasma membrane. SMS2 functions were investigated in HepG2 cell lines stably expressing SMS2. SMS2 overexpression did not alter sphingomyelin (SM), phosphatidylcholine (PC), or ceramide levels. DAG content increased by approx. 40% and led to downregulation of DAG-dependent protein kinase C (PKC). SMS2 overexpression also induced senescence, characterized by positivity for β-galactosidase activity and heterochromatin foci. HepG2-SMS2 cells exhibited protruded mitochondria and suppressed mitochondrial respiration rates. ATP production and the abundance of Complex V were substantially lower in HepG2-SMS2 cells as compared to controls. SMS2 overexpression was associated with inflammasome activation based on increases in IL-1β and nlpr3 mRNA levels. HepG2-SMS2 cells exhibited lipid droplet accumulation, constitutive activation of AMPK based on elevated 172Thr phosphorylation, increased AMPK abundance, and insensitivity to insulin suppression of AMPK. Thus, our results show that SMS2 regulates DAG homeostasis and signaling in hepatocytes and also provide proof of principle for the concept that offset in bioactive lipids’ production at the plasma membrane can drive the senescence program in association with steatosis and, seemingly, by cell-autonomous mechanisms.
Collapse
|
5
|
Lex K, Maia Gil M, Lopes-Bastos B, Figueira M, Marzullo M, Giannetti K, Carvalho T, Ferreira MG. Telomere shortening produces an inflammatory environment that increases tumor incidence in zebrafish. Proc Natl Acad Sci U S A 2020; 117:15066-15074. [PMID: 32554492 PMCID: PMC7334448 DOI: 10.1073/pnas.1920049117] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer incidence increases exponentially with age when human telomeres are shorter. Similarly, telomerase reverse transcriptase (tert) mutant zebrafish have premature short telomeres and anticipate cancer incidence to younger ages. However, because short telomeres constitute a road block to cell proliferation, telomere shortening is currently viewed as a tumor suppressor mechanism and should protect from cancer. This conundrum is not fully understood. In our current study, we report that telomere shortening promotes cancer in a noncell autonomous manner. Using zebrafish chimeras, we show increased incidence of invasive melanoma when wild-type (WT) tumors are generated in tert mutant zebrafish. Tissues adjacent to melanoma lesions (skin) and distant organs (intestine) in tert mutants exhibited higher levels of senescence and inflammation. In addition, we transferred second generation (G2) tert blastula cells into WT to produce embryo chimeras. Cells with very short telomeres induced increased tumor necrosis factor1-α (TNF1-α) expression and senescence in larval tissues in a noncell autonomous manner, creating an inflammatory environment. Considering that inflammation is protumorigenic, we transplanted melanoma-derived cells into G2 tert zebrafish embryos and observed that tissue environment with short telomeres leads to increased tumor development. To test if inflammation was necessary for this effect, we treated melanoma transplants with nonsteroid anti-inflammatory drugs and show that higher melanoma dissemination can be averted. Thus, apart from the cell autonomous role of short telomeres in contributing to genome instability, we propose that telomere shortening with age causes systemic chronic inflammation leading to increased tumor incidence.
Collapse
Affiliation(s)
- Kirsten Lex
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Mariana Maia Gil
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Bruno Lopes-Bastos
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
- Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, UMR7284 U1081 UNS, 06107 Nice, France
| | - Margarida Figueira
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Marta Marzullo
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Kety Giannetti
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Tânia Carvalho
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Av Brasilia, 1400-038 Lisbon, Portugal
| | - Miguel Godinho Ferreira
- Telomere and Genome Stability Laboratory, Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal;
- Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, UMR7284 U1081 UNS, 06107 Nice, France
| |
Collapse
|
6
|
Wieczór M, Czub J. Telomere uncapping by common oxidative guanine lesions: Insights from atomistic models. Free Radic Biol Med 2020; 148:162-169. [PMID: 31926882 DOI: 10.1016/j.freeradbiomed.2020.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/02/2020] [Accepted: 01/05/2020] [Indexed: 12/15/2022]
Abstract
Oxidative damage to DNA is widely known to contribute to aging and disease. This relationship has been extensively studied for telomeres - structures that cap chromosome ends - due to their role in cell proliferation and senescence, and exceptional susceptibility to oxidation. Indeed, the repetitive telomeric DNA sequence contains the 5'-GGG-3' motif that has the lowest ionization potential of all trinucleotides. Accordingly, experiments consistently show that telomeric oxidative lesions are more abundant and persistent than elsewhere in the genome. This led to a hypothesis that telomeres act as sensors of prolonged oxidative stress and prevent carcinogenesis, as disruption of telomeric integrity triggers senescence or apoptosis. Here, we use atomistic alchemical Molecular Dynamics simulations to perform a combinatorial assessment of changes in DNA binding affinity of telomeric proteins induced by oxidative guanine lesions. We rank lesions by their effect on telomere integrity, as well as telomeric proteins by their sensitivity to DNA oxidation. While the binding of most proteins is abolished by DNA oxidation, HOT1 emerges as a notable exception, suggesting its potential role in sensing of oxidative damage. Through statistical analysis and free energy decomposition, we also identify common trends in structural responses of protein-DNA complexes that contribute to decreased binding affinity.
Collapse
Affiliation(s)
- Miłosz Wieczór
- Department of Physical Chemistry, Gdansk University of Technology, Gdańsk, Poland.
| | - Jacek Czub
- Department of Physical Chemistry, Gdansk University of Technology, Gdańsk, Poland.
| |
Collapse
|
7
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a group of liver disorders encompassing simple hepatic steatosis and its more aggressive forms of nonalcoholic steatohepatitis and cirrhosis. It is a rapidly growing health concern and the major cause for the increasing incidence of primary liver tumors. Unequivocal evidence shows that sphingolipid metabolism is altered in the course of the disease and these changes might contribute to NAFLD progression. Recent data provide solid support to the notion that deregulated ceramide and sphingosine-1-phosphate metabolism are present at all stages of NAFLD, i.e., steatosis, nonalcoholic steatohepatitis, advanced fibrosis, and hepatocellular carcinoma (HCC). Insulin sensitivity, de novo lipogenesis, and the resulting lipotoxicity, fibrosis, and angiogenesis are all seemingly regulated in a manner that involves either ceramide and/or sphingosine-1-phosphate. Sphingolipids might also participate in the onset of hepatocellular senescence. The latter has been shown to contribute to the advancement of cirrhosis to HCC in the classical cases of end-stage liver disease, i.e., viral- or alcohol-induced; however, emerging evidence suggests that senescence is also involved in the pathogenicity of NAFLD possibly via changes in ceramide metabolism.
Collapse
|
8
|
Telomere Biology and Thoracic Aortic Aneurysm. Int J Mol Sci 2017; 19:ijms19010003. [PMID: 29267201 PMCID: PMC5795955 DOI: 10.3390/ijms19010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 12/27/2022] Open
Abstract
Ascending aortic aneurysms are mostly asymptomatic and present a great risk of aortic dissection or perforation. Consequently, ascending aortic aneurysms are a source of lethality with increased age. Biological aging results in progressive attrition of telomeres, which are the repetitive DNA sequences at the end of chromosomes. These telomeres play an important role in protection of genomic DNA from end-to-end fusions. Telomere maintenance and telomere attrition-associated senescence of endothelial and smooth muscle cells have been indicated to be part of the pathogenesis of degenerative vascular diseases. This systematic review provides an overview of telomeres, telomere-associated proteins and telomerase to the formation and progression of aneurysms of the thoracic ascending aorta. A better understanding of telomere regulation in the vascular pathology might provide new therapeutic approaches. Measurements of telomere length and telomerase activity could be potential prognostic biomarkers for increased risk of death in elderly patients suffering from an aortic aneurysm.
Collapse
|
9
|
Zhou G, Liu X, Li Y, Xu S, Ma C, Wu X, Cheng Y, Yu Z, Zhao G, Chen Y. Telomere targeting with a novel G-quadruplex-interactive ligand BRACO-19 induces T-loop disassembly and telomerase displacement in human glioblastoma cells. Oncotarget 2017; 7:14925-39. [PMID: 26908447 PMCID: PMC4924762 DOI: 10.18632/oncotarget.7483] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/29/2016] [Indexed: 12/12/2022] Open
Abstract
Interference with telomerase and telomere maintenance is emerging as an attractive target for anticancer therapies. Ligand-induced stabilization of G-quadruplex formation by the telomeric DNA 3'-overhang inhibits telomerase from catalyzing telomeric DNA synthesis and from capping telomeric ends, making these ligands good candidates for chemotherapeutic purposes. BRACO-19 is one of the most effective and specific ligand for telomeric G4. It is shown here that BRACO-19 suppresses proliferation and reduces telomerase activity in human glioblastoma cells, paralleled by the displacement of telomerase from nuclear to cytoplasm. Meanwhile, BRACO-19 triggers extensive DNA damage response at telomere, which may result from uncapping and disassembly of telomeric T-loop structure, characterized by the formation of anaphase bridge and telomere fusion, as well as the release of telomere-binding protein from telomere. The resulting dysfunctional telomere ultimately provokes p53 and p21-mediated cell cycle arrest, apoptosis and senescence. Notably, normal primary astrocytes do not respond to the treatment of BRACO-19, suggesting the agent's good selectivity for cancer cells. These results reinforce the notion that G-quadruplex binding compounds can act as broad inhibitors of telomere-related processes and have potential as selective antineoplastic drugs for various tumors including malignant gliomas.
Collapse
Affiliation(s)
- Guangtong Zhou
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Xinrui Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Yunqian Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Chengyuan Ma
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Xinmin Wu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Ye Cheng
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Zhiyun Yu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Gang Zhao
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Yong Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Resveratrol Ameliorates Mitochondrial Elongation via Drp1/Parkin/PINK1 Signaling in Senescent-Like Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4175353. [PMID: 29201272 PMCID: PMC5671746 DOI: 10.1155/2017/4175353] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/26/2017] [Accepted: 09/05/2017] [Indexed: 01/26/2023]
Abstract
Resveratrol is widely known for its antiaging properties and exerts cardiovascular protective effects in different experimental models. The role of resveratrol in regulating mitochondrial functions and dynamics during the cardiac aging process remains poorly understood. In this study, the effects of resveratrol on mitochondrial morphology and mitochondrial depolarization and on expressions of Drp1, parkin, PINK1, and LC3 were investigated in H9c2 cells after D-galactose treatment that induced senescent-like cardiomyocytes. The results show that downregulation of Drp1 markedly increased mitochondrial elongation. Senescent-like cardiomyocytes were more resistant to CCCP-induced mitochondrial depolarization, which was accompanied by suppressed expression of parkin, PINK1, and LC3-II. Resveratrol treatment significantly increased Drp1 expression, ameliorated mitochondrial elongation, and increased the mitochondrial translocations of parkin and PINK1. In addition, resveratrol significantly enhanced LC3-II expression and decreased TOM20-labeled mitochondrial content. Resveratrol also suppressed the phosphorylation of parkin and PINK1, which may relate to its abilities to degrade the impaired mitochondria in senescent-like cardiomyocytes. These findings show that suppressing mitochondrial elongation in a Drp1-dependent manner is involved in the effect of resveratrol on attenuating the development of aging cardiomyocytes. Activation of parkin and PINK1 may be a potential mechanism of resveratrol for treating cardiovascular complications related to aging.
Collapse
|
11
|
Zhong HH, Hu SJ, Yu B, Jiang SS, Zhang J, Luo D, Yang MW, Su WY, Shao YL, Deng HL, Hong FF, Yang SL. Apoptosis in the aging liver. Oncotarget 2017; 8:102640-102652. [PMID: 29254277 PMCID: PMC5731987 DOI: 10.18632/oncotarget.21123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/15/2017] [Indexed: 12/11/2022] Open
Abstract
Various changes in the liver during aging can reduce hepatic function and promote liver injury. Aging is associated with high morbidity and a poor prognosis in patients with various liver diseases, including nonalcoholic fatty liver disease, hepatitis C and liver cancer, as well as with surgeries such as partial hepatectomy and liver transplantation. In addition, apoptosis increases with liver aging. Because apoptosis is involved in regeneration, fibrosis and cancer prevention during liver aging, and restoration of the appropriate level of apoptosis can alleviate the adverse effects of liver aging, it is important to understand the mechanisms underlying this process. Herein, we elaborate on the causes of apoptosis during liver aging, with a focus on oxidative stress, genomic instability, lipotoxicity, endoplasmic reticulum stress, dysregulation of nutrient sensing, and liver stem/progenitor cell activity.
Collapse
Affiliation(s)
- Hua-Hua Zhong
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Shao-Jie Hu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Bo Yu
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Sha-Sha Jiang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Jin Zhang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Dan Luo
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Mei-Wen Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Wan-Ying Su
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Ya-Lan Shao
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Hao-Lin Deng
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Fen-Fang Hong
- Department of Experimental Teaching Center, Nanchang University, Nanchang 330031, China
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| |
Collapse
|
12
|
Transcription regulation of CDKN1A (p21/CIP1/WAF1) by TRF2 is epigenetically controlled through the REST repressor complex. Sci Rep 2017; 7:11541. [PMID: 28912501 PMCID: PMC5599563 DOI: 10.1038/s41598-017-11177-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/20/2017] [Indexed: 12/13/2022] Open
Abstract
We observed extra-telomeric binding of the telomere repeat binding factor TRF2 within the promoter of the cyclin-dependent kinase CDKNIA (p21/CIP1/WAF1). This result in TRF2 induced transcription repression of p21. Interestingly, p21 repression was through engagement of the REST-coREST-LSD1-repressor complex and altered histone marks at the p21 promoter in a TRF2-dependent fashion. Furthermore, mutational analysis shows p21 repression requires interaction of TRF2 with a p21 promoter G-quadruplex. Physiologically, TRF2-mediated p21 repression attenuated drug-induced activation of cellular DNA damage response by evading G2/M arrest in cancer cells. Together these reveal for the first time role of TRF2 in REST- repressor complex mediated transcription repression.
Collapse
|
13
|
Nakayama K, Rahman M, Rahman MT, Nakamura K, Sato E, Katagiri H, Ishibashi T, Ishikawa M, Iida K, Razia S, Ishikawa N, Kyo S. Nucleus accumbens-1/GADD45GIP1 axis mediates cisplatin resistance through cellular senescence in ovarian cancer. Oncol Lett 2017; 13:4713-4719. [PMID: 28599472 PMCID: PMC5453174 DOI: 10.3892/ol.2017.6099] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 01/04/2017] [Indexed: 01/25/2023] Open
Abstract
Nucleus accumbens-1 (NAC1), a nuclear factor belonging to the bric-a-brac-tramtrack-broad complex/pox virus and zinc finger gene family, is known to serve important roles in the proliferation and growth of tumor cells, and in chemotherapy resistance. However, the underlying molecular mechanisms through which NAC1 contributes to drug resistance remain unclear. In the present study, the role of NAC1 in drug resistance in ovarian cancer was investigated. NAC1 expression was markedly negatively associated with growth arrest and DNA-damage-inducible 45γ-interacting protein 1 (GADD45GIP1) expression in ovarian cancer. Increased NAC1 expression or decreased GADD45GIP1 expression was significantly associated with decreased progression-free survival (P=0.0041). Multivariate analysis demonstrated that NAC1/GADD45GIP1 expression was an independent prognostic factor of progression-free survival (P=0.0405). It was investigated whether cellular senescence was involved in NAC1-mediated resistance to cisplatin, a commonly used chemotherapeutic drug in the treatment of ovarian cancer. Treatment with cisplatin activated cellular senescence in ovarian cancer cell lines (SKOV3 and TOV-21G cells). Furthermore, knockdown of NAC1 by RNA interference significantly increased GADD45GIP1 expression and inhibited cisplatin-induced cellular senescence, resulting in increased cisplatin cytotoxicity in SKOV3 cells, which express increased levels of NAC1. To investigate whether the sensitizing effect of NAC1 inhibition on cisplatin-induced cytotoxicity may be attributed to the suppression of cellular senescence, the effects of NAC1 overexpression were assessed in TOV-21G cells, which do not express endogenous NAC1. Transfection with NAC1 in TOV-21G cells reduced the sensitivity of TOV-21G cells to cisplatin, indicating that suppression of cellular senescence was induced by GADD45GP1 activation. The results of the present study suggest that NAC1 is a negative regulator of cellular senescence and that NAC1-dependent suppression of senescence, mediated through GADD45GIP1, serves an important role in promoting cisplatin resistance. Therefore, the NAC1/GADD45GIP1 axis may be a potential target for the treatment of ovarian cancer, particularly in platinum-resistant cancers.
Collapse
Affiliation(s)
- Kentaro Nakayama
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Munmun Rahman
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Mohammed Tanjimur Rahman
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Kohei Nakamura
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Emi Sato
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Hiroshi Katagiri
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Tomoka Ishibashi
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Masako Ishikawa
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Kouji Iida
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Sultana Razia
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Noriyuki Ishikawa
- Department of Organ Pathology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo, Shimane 6938501, Japan
| |
Collapse
|
14
|
García-Prat L, Muñoz-Cánoves P. Aging, metabolism and stem cells: Spotlight on muscle stem cells. Mol Cell Endocrinol 2017; 445:109-117. [PMID: 27531569 DOI: 10.1016/j.mce.2016.08.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/11/2016] [Indexed: 12/15/2022]
Abstract
All tissues and organs undergo a progressive regenerative decline as they age. This decline has been mainly attributed to loss of stem cell number and/or function, and both stem cell-intrinsic changes and alterations in local niches and/or systemic environment over time are known to contribute to the stem cell aging phenotype. Advancing in the molecular understanding of the deterioration of stem cell cells with aging is key for targeting the specific causes of tissue regenerative dysfunction at advanced stages of life. Here, we revise exciting recent findings on why stem cells age and the consequences on tissue regeneration, with a special focus on regeneration of skeletal muscle. We also highlight newly identified common molecular pathways affecting diverse types of aging stem cells, such as altered proteostasis, metabolism, or senescence entry, and discuss the questions raised by these findings. Finally, we comment on emerging stem cell rejuvenation strategies, principally emanating from studies on muscle stem cells, which will surely burst tissue regeneration research for future benefit of the increasing human aging population.
Collapse
Affiliation(s)
- Laura García-Prat
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra; University (UPF) y CIBERNED, Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra; University (UPF) y CIBERNED, Barcelona, Spain; ICREA, Barcelona, Spain.
| |
Collapse
|
15
|
ROS, Cell Senescence, and Novel Molecular Mechanisms in Aging and Age-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3565127. [PMID: 27247702 PMCID: PMC4877482 DOI: 10.1155/2016/3565127] [Citation(s) in RCA: 702] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/02/2016] [Accepted: 04/06/2016] [Indexed: 12/15/2022]
Abstract
The aging process worsens the human body functions at multiple levels, thus causing its gradual decrease to resist stress, damage, and disease. Besides changes in gene expression and metabolic control, the aging rate has been associated with the production of high levels of Reactive Oxygen Species (ROS) and/or Reactive Nitrosative Species (RNS). Specific increases of ROS level have been demonstrated as potentially critical for induction and maintenance of cell senescence process. Causal connection between ROS, aging, age-related pathologies, and cell senescence is studied intensely. Senescent cells have been proposed as a target for interventions to delay the aging and its related diseases or to improve the diseases treatment. Therapeutic interventions towards senescent cells might allow restoring the health and curing the diseases that share basal processes, rather than curing each disease in separate and symptomatic way. Here, we review observations on ROS ability of inducing cell senescence through novel mechanisms that underpin aging processes. Particular emphasis is addressed to the novel mechanisms of ROS involvement in epigenetic regulation of cell senescence and aging, with the aim to individuate specific pathways, which might promote healthy lifespan and improve aging.
Collapse
|
16
|
Altieri P, Barisione C, Lazzarini E, Garuti A, Bezante GP, Canepa M, Spallarossa P, Tocchetti CG, Bollini S, Brunelli C, Ameri P. Testosterone Antagonizes Doxorubicin-Induced Senescence of Cardiomyocytes. J Am Heart Assoc 2016; 5:JAHA.115.002383. [PMID: 26746999 PMCID: PMC4859360 DOI: 10.1161/jaha.115.002383] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Chronic cardiotoxicity is less common in male than in female patients receiving doxorubicin and other anthracyclines at puberty and adolescence. We hypothesized that this sex difference might be secondary to distinct activities of sex hormones on cardiomyocyte senescence, which is thought to be central to the development of long‐term anthracycline cardiomyopathy. Methods and Results H9c2 cells and neonatal mouse cardiomyocytes were exposed to doxorubicin with or without prior incubation with testosterone or 17β‐estradiol, the main androgen and estrogen, respectively. Testosterone, but not 17β‐estradiol, counteracted doxorubicin‐elicited senescence. Downregulation of telomere binding factor 2, which has been pinpointed previously as being pivotal to doxorubicin‐induced senescence, was also prevented by testosterone, as were p53 phosphorylation and accumulation. Pretreatment with the androgen receptor antagonist flutamide, the phosphatidylinositol 3 kinase inhibitor LY294002, and the nitric oxide synthase inhibitor L‐NG‐nitroarginine methyl ester abrogated the reduction in senescence and the normalization of telomere binding factor 2 levels attained by testosterone. Consistently, testosterone enhanced the phosphorylation of AKT and nitric oxide synthase 3. In H9c2 cells, doxorubicin‐stimulated senescence was still observed up to 21 days after treatment and increased further when cells were rechallenged with doxorubicin 14 days after the first exposure to mimic the schedule of anthracycline‐containing chemotherapy. Remarkably, these effects were also inhibited by testosterone. Conclusions Testosterone protects cardiomyocytes against senescence caused by doxorubicin at least in part by modulating telomere binding factor 2 via a pathway involving the androgen receptor, phosphatidylinositol 3 kinase, AKT, and nitric oxide synthase 3. This is a potential mechanism by which pubescent and adolescent boys are less prone to chronic anthracycline cardiotoxicity than girls.
Collapse
Affiliation(s)
- Paola Altieri
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Chiara Barisione
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Edoardo Lazzarini
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Anna Garuti
- Laboratory of Cellular TherapiesDepartment of Internal MedicineUniversity of GenovaItaly
| | - Gian Paolo Bezante
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Marco Canepa
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Paolo Spallarossa
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Carlo Gabriele Tocchetti
- Division of Internal MedicineDepartment of Translational Medical SciencesFederico II UniversityNapoliItaly
| | - Sveva Bollini
- Regenerative Medicine LaboratoryDepartment of Experimental MedicineUniversity of GenovaItaly
| | - Claudio Brunelli
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| | - Pietro Ameri
- Laboratory of Cardiovascular BiologyDepartment of Internal MedicineUniversity of GenovaItaly
| |
Collapse
|
17
|
Wang J, Uryga AK, Reinhold J, Figg N, Baker L, Finigan A, Gray K, Kumar S, Clarke M, Bennett M. Vascular Smooth Muscle Cell Senescence Promotes Atherosclerosis and Features of Plaque Vulnerability. Circulation 2015; 132:1909-19. [DOI: 10.1161/circulationaha.115.016457] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/17/2015] [Indexed: 12/28/2022]
Abstract
Background—
Although vascular smooth muscle cell (VSMC) proliferation is implicated in atherogenesis, VSMCs in advanced plaques and cultured from plaques show evidence of VSMC senescence and DNA damage. In particular, plaque VSMCs show shortening of telomeres, which can directly induce senescence. Senescence can have multiple effects on plaque development and morphology; however, the consequences of VSMC senescence or the mechanisms underlying VSMC senescence in atherosclerosis are mostly unknown.
Methods and Results—
We examined the expression of proteins that protect telomeres in VSMCs derived from human plaques and normal vessels. Plaque VSMCs showed reduced expression and telomere binding of telomeric repeat-binding factor-2 (TRF2), associated with increased DNA damage. TRF2 expression was regulated by p53-dependent degradation of the TRF2 protein. To examine the functional consequences of loss of TRF2, we expressed TRF2 or a TRF2 functional mutant (T188A) as either gain- or loss-of-function studies in vitro and in apolipoprotein E
–/–
mice. TRF2 overexpression bypassed senescence, reduced DNA damage, and accelerated DNA repair, whereas TRF2
188A
showed opposite effects. Transgenic mice expressing VSMC-specific TRF2
T188A
showed increased atherosclerosis and necrotic core formation in vivo, whereas VSMC-specific TRF2 increased the relative fibrous cap and decreased necrotic core areas. TRF2 protected against atherosclerosis independent of secretion of senescence-associated cytokines.
Conclusions—
We conclude that plaque VSMC senescence in atherosclerosis is associated with loss of TRF2. VSMC senes cence promotes both atherosclerosis and features of plaque vulnerability, identifying prevention of senescence as a potential target for intervention.
Collapse
Affiliation(s)
- Julie Wang
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Anna K. Uryga
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Johannes Reinhold
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Nichola Figg
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Lauren Baker
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Alison Finigan
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Kelly Gray
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Sheetal Kumar
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Murray Clarke
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Martin Bennett
- From Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
18
|
|
19
|
Yi W, Hu X, Chen Z, Liu L, Tian Y, Chen H, Cong YS, Yang F, Zhang L, Rudolph KL, Zhang Z, Zhao Y, Ju Z. Phosphatase Wip1 controls antigen-independent B-cell development in a p53-dependent manner. Blood 2015; 126:620-8. [PMID: 26012568 PMCID: PMC4520877 DOI: 10.1182/blood-2015-02-624114] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 05/06/2015] [Indexed: 01/08/2023] Open
Abstract
Wild-type p53-induced phosphatase 1 (Wip1), a phosphatase previously considered as an oncogene, has been implicated in the regulation of thymus homeostasis and neutrophil maturation. However, the role of Wip1 in B-cell development is unknown. We show that Wip1-deficient mice exhibit a significant reduction of B-cell numbers in the bone marrow, peripheral blood, and spleen. A reciprocal transplantation approach revealed a cell-intrinsic defect in early B-cell precursors caused by Wip1 deficiency. Further experiments revealed that Wip1 deficiency led to a sustained activation of p53 in B cells, which led to increased level of apoptosis in the pre-B-cell compartment. Notably, the impairment of B-cell development in Wip1-deficient mice was completely rescued by genetic ablation of p53, but not p21. Therefore, loss of Wip1 phosphatase induces a p53-dependent, but p21-independent, mechanism that impairs B-cell development by enhancing apoptosis in early B-cell precursors. Moreover, Wip1 deficiency exacerbated a decline in B-cell development caused by aging as evidenced in mice with aging and mouse models with serial competitive bone marrow transplantation, respectively. Our present data indicate that Wip1 plays a critical role in maintaining antigen-independent B-cell development in the bone marrow and preventing an aging-related decline in B-cell development.
Collapse
Affiliation(s)
- Weiwei Yi
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Xuelian Hu
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhiyang Chen
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China; Leibniz Institute for Age Research-Fritz Lipmann Institute, Jena, Germany
| | - Leiming Liu
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China; Sir Runrun Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Tian
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hui Chen
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yu-Sheng Cong
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Fan Yang
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases Comparative Medicine, Ministry of Health; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; and
| | | | - Zhixin Zhang
- Department of Pediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Ministry of Education Key Laboratory of Birth Defects, Sichuan University, Chengdu, China
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhenyu Ju
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| |
Collapse
|
20
|
Zhang L, Liu L, He Z, Li G, Liu J, Song Z, Jin H, Rudolph KL, Yang H, Mao Y, Zhang L, Zhang H, Xiao Z, Ju Z. Inhibition of wild-type p53-induced phosphatase 1 promotes liver regeneration in mice by direct activation of mammalian target of rapamycin. Hepatology 2015; 61:2030-41. [PMID: 25704606 DOI: 10.1002/hep.27755] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/14/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED The liver possesses extraordinary regenerative capacity in response to injury. However, liver regeneration (LR) is often impaired in disease conditions. Wild-type p53-induced phosphatase 1 (Wip1) is known as a tumor promoter and enhances cell proliferation, mainly by deactivating antioncogenes. However, in this work, we identified an unexpected role of Wip1 in LR. In contrast to its known role in promoting cell proliferation in extrahepatic tissue, we found that Wip1 suppressed hepatocyte proliferation after partial hepatectomy (PHx). Deletion of Wip1 increased the rate of LR after PHx. Enhanced LR in Wip1-deficient mice was a result of the activation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) pathway. Furthermore, we showed that Wip1 physically interacted with and dephosphorylated mTOR. Interestingly, inhibition of Wip1 also activated the p53 pathway during LR. Disruption of the p53 pathway further enhanced LR in Wip1-deficient mice. Therefore, inhibition of Wip1 has a dual role in LR, i.e., promoting hepatocyte proliferation through activation of the mTORC1 pathway, meanwhile suppressing LR through activation of the p53 pathway. However, the proregenerative role of mTORC1 overwhelms the antiproliferative role of p53. Furthermore, CCT007093, a Wip1 inhibitor, enhanced LR and increased the survival rate of mice after major hepatectomy. CONCLUSION mTOR is a new direct target of Wip1. Wip1 inhibition can activate the mTORC1 pathway and enhance hepatocyte proliferation after hepatectomy. These findings have clinical applications in cases where LR is critical, including acute liver failure, cirrhosis, or small-for-size liver transplantations.
Collapse
Affiliation(s)
- Lingling Zhang
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Leiming Liu
- Sir Runrun Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Zhiyong He
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China
| | - Guangbing Li
- Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junping Liu
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Zhangfa Song
- Sir Runrun Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Hongchuan Jin
- Sir Runrun Shaw Hospital, Medical School, Zhejiang University, Hangzhou, China
| | | | - Huayu Yang
- Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yilei Mao
- Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lianfeng Zhang
- Institute of Laboratory Animal Sciences, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hongbing Zhang
- Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhicheng Xiao
- The Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Molecular and Clinical Medicine, Kunming Medical University, Kunming, China
| | - Zhenyu Ju
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, School of Medicine, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
21
|
Sklirou AD, Ralli M, Dominguez M, Papassideri I, Skaltsounis AL, Trougakos IP. Hexapeptide-11 is a novel modulator of the proteostasis network in human diploid fibroblasts. Redox Biol 2015; 5:205-215. [PMID: 25974626 PMCID: PMC4434199 DOI: 10.1016/j.redox.2015.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 04/20/2015] [Accepted: 04/26/2015] [Indexed: 01/01/2023] Open
Abstract
Despite the fact that several natural products (e.g. crude extracts or purified compounds) have been found to activate cell antioxidant responses and/or delay cellular senescence the effect(s) of small peptides on cell viability and/or modulation of protective mechanisms (e.g. the proteostasis network) remain largely elusive. We have thus studied a hexapeptide (Hexapeptide-11) of structure Phe-Val-Ala-Pro-Phe-Pro (FVAPFP) originally isolated from yeast extracts and later synthesized by solid state synthesis to high purity. We show herein that Hexapeptide-11 exhibits no significant toxicity in normal human diploid lung or skin fibroblasts. Exposure of fibroblasts to Hexapeptide-11 promoted dose and time-dependent activation of proteasome, autophagy, chaperones and antioxidant responses related genes. Moreover, it promoted increased nuclear accumulation of Nrf2; higher expression levels of proteasomal protein subunits and increased proteasome peptidase activities. In line with these findings we noted that Hexapeptide-11 conferred significant protection in fibroblasts against oxidative-stress-mediated premature cellular senescence, while at in vivo skin deformation assays in human subjects it improved skin elasticity. Finally, Hexapeptide-11 was found to induce the activity of extracellular MMPs and it also suppressed cell migration. Our presented findings indicate that Hexapeptide-11 is a promising anti-ageing agent.
Collapse
Affiliation(s)
- Aimilia D Sklirou
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Marianna Ralli
- Korres S.A. Natural Products, 57th Athens-Lamia National Road, 32011 Inofyta, Greece
| | | | - Issidora Papassideri
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| | - Alexios-Leandros Skaltsounis
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Ioannis P Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Panepistimiopolis, Athens 15784, Greece
| |
Collapse
|
22
|
Comparative Meta-Analysis of Transcriptomics Data during Cellular Senescence and In Vivo Tissue Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:732914. [PMID: 25977747 PMCID: PMC4419258 DOI: 10.1155/2015/732914] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/22/2015] [Accepted: 03/23/2015] [Indexed: 02/06/2023]
Abstract
Several studies have employed DNA microarrays to identify gene expression signatures that mark human ageing; yet the features underlying this complicated phenomenon remain elusive. We thus conducted a bioinformatics meta-analysis on transcriptomics data from human cell- and biopsy-based microarrays experiments studying cellular senescence or in vivo tissue ageing, respectively. We report that coregulated genes in the postmitotic muscle and nervous tissues are classified into pathways involved in cancer, focal adhesion, actin cytoskeleton, MAPK signalling, and metabolism regulation. Genes that are differentially regulated during cellular senescence refer to pathways involved in neurodegeneration, focal adhesion, actin cytoskeleton, proteasome, cell cycle, DNA replication, and oxidative phosphorylation. Finally, we revealed genes and pathways (referring to cancer, Huntington's disease, MAPK signalling, focal adhesion, actin cytoskeleton, oxidative phosphorylation, and metabolic signalling) that are coregulated during cellular senescence and in vivo tissue ageing. The molecular commonalities between cellular senescence and tissue ageing are also highlighted by the fact that pathways that were overrepresented exclusively in the biopsy- or cell-based datasets are modules either of the same reference pathway (e.g., metabolism) or of closely interrelated pathways (e.g., thyroid cancer and melanoma). Our reported meta-analysis has revealed novel age-related genes, setting thus the basis for more detailed future functional studies.
Collapse
|
23
|
A switch-like dynamic mechanism for the initiation of replicative senescence. FEBS Lett 2014; 588:4369-74. [DOI: 10.1016/j.febslet.2014.09.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 09/15/2014] [Indexed: 11/23/2022]
|
24
|
Xu S, Cai Y, Wei Y. mTOR Signaling from Cellular Senescence to Organismal Aging. Aging Dis 2014; 5:263-73. [PMID: 25110610 DOI: 10.14336/ad.2014.0500263] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/17/2022] Open
Abstract
The TOR (target of rapamycin) pathway has been convincingly shown to promote aging in various model organisms. In mice, inhibiting mTOR (mammalian TOR) by rapamycin treatment later in life can significantly extend lifespan and mitigate multiple age-related diseases. However, the underlying mechanisms are poorly understood. Cellular senescence is strongly correlated to organismal aging therefore providing an attractive model to examine the mechanisms by which mTOR inhibition contributes to longevity and delaying the onset of related diseases. In this review, we examine the connections between mTOR and cellular senescence and discuss how understanding cellular senescence on the aspect of mTOR signaling may help to fully appreciate its role in the organismal aging. We also highlight the opposing roles of senescence in various human diseases and discuss the caveats in interpreting the emerging experimental data.
Collapse
Affiliation(s)
| | - Ying Cai
- No.3 People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yuehua Wei
- No.3 People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China ; Gladstone Institute of Cardiovascular Disease, University of California San Francisco, San Francisco, CA94102, USA
| |
Collapse
|
25
|
A comparison of replicative senescence and doxorubicin-induced premature senescence of vascular smooth muscle cells isolated from human aorta. Biogerontology 2013; 15:47-64. [PMID: 24243065 PMCID: PMC3905196 DOI: 10.1007/s10522-013-9477-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 11/04/2013] [Indexed: 11/13/2022]
Abstract
Senescence of vascular smooth muscle cells (VSMCs) contributes to aging as well as age-related diseases of the cardiovascular system. Senescent VSMCs have been shown to be present in atherosclerotic plaques. Both replicative (RS) and stress-induced premature senescence (SIPS) accompany cardiovascular diseases. We aimed to establish the signature of RS and SIPS of VSMCs, induced by a common anticancer drug, doxorubicin, and to discover the so far undisclosed features of senescent cells that are potentially harmful to the organism. Most of the senescence hallmarks were common for both RS and SIPS; however, some differences were observed. 32 % of doxorubicin-treated cells were arrested in the G2/M phase of the cell cycle, while 73 % of replicatively senescing cells were arrested in the G1 phase. Moreover, on the basis of alkaline phosphatase activity measurements, we show that a 7-day treatment with doxorubicin (dox), does not cause precocious cell calcification, which is a characteristic feature of RS. We did not observe calcification even though after 7 days of dox-treatment many other markers characteristic for senescent cells were present. It can suggest that dox-induced SIPS does not accelerate the mineralization of vessels. We consider that detailed characterization of the two types of cellular senescence can be useful in in vitro studies of potential anti-aging factors.
Collapse
|
26
|
Her YR, Chung IK. p300-mediated acetylation of TRF2 is required for maintaining functional telomeres. Nucleic Acids Res 2013; 41:2267-83. [PMID: 23307557 PMCID: PMC3575801 DOI: 10.1093/nar/gks1354] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The human telomeric protein TRF2 is required to protect chromosome ends by facilitating their organization into the protective capping structure. Post-translational modifications of TRF2 such as phosphorylation, ubiquitination, SUMOylation, methylation and poly(ADP-ribosyl)ation have been shown to play important roles in telomere function. Here we show that TRF2 specifically interacts with the histone acetyltransferase p300, and that p300 acetylates the lysine residue at position 293 of TRF2. We also report that p300-mediated acetylation stabilizes the TRF2 protein by inhibiting its ubiquitin-dependent proteolysis and is required for efficient telomere binding of TRF2. Furthermore, overexpression of the acetylation-deficient mutant, K293R, induces DNA-damage response foci at telomeres, thereby leading to induction of impaired cell growth, cellular senescence and altered cell cycle distribution. A small but significant number of metaphase chromosomes show no telomeric signals at chromatid ends, suggesting an aberrant telomere structure. These findings demonstrate that acetylation of TRF2 by p300 plays a crucial role in the maintenance of functional telomeres as well as in the regulation of the telomere-associated DNA-damage response, thus providing a new route for modulating telomere protection function.
Collapse
Affiliation(s)
- Yoon Ra Her
- Departments of Systems Biology and Integrated Omics for Biomedical Science, WCU Program of Graduate School, Yonsei University, Seoul 120-749, Korea
| | | |
Collapse
|
27
|
Hodjat M, Haller H, Dumler I, Kiyan Y. Urokinase receptor mediates doxorubicin-induced vascular smooth muscle cell senescence via proteasomal degradation of TRF2. J Vasc Res 2012; 50:109-23. [PMID: 23172421 DOI: 10.1159/000343000] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 08/23/2012] [Indexed: 11/19/2022] Open
Abstract
The anthracycline doxorubicin is a widely used effective anti-cancer drug. However, its application and dosage are severely limited due to its cardiotoxicity. The exact mechanisms of doxorubicin-induced cardiotoxic side effects remain poorly understood. Even less is known about the impact of doxorubicin treatment on vascular damage. We found that low doses of doxorubicin induced a senescent response in human primary vascular smooth muscle cells (VSMC). We observed that expression of urokinase receptor (uPAR) was upregulated in response to doxorubicin. Furthermore, the level of uPAR expression played a decisive role in developing doxorubicin-induced senescence. uPAR silencing in human VSMC by means of RNA interference as well as uPAR knockout in mouse VSMC resulted in abrogation of doxorubicin-induced cellular senescence. On the contrary, uPAR overexpression promoted VSMC senescence. We further found that proteasomal degradation of telomeric repeat binding factor 2 (TRF2) mediates doxorubicin-induced VSMC senescence. Our results demonstrate that uPAR controls the ubiquitin-proteasome system in VSMC and regulates doxorubicin-induced TRF2 ubiquitination and proteasomal degradation via this mechanism. Therefore, VSMC senescence induced by low doses of doxorubicin may contribute to vascular damage upon doxorubicin treatment. uPAR-mediated TRF2 ubiquitination and proteasomal degradation are further identified as a molecular mechanism underlying this process.
Collapse
Affiliation(s)
- Mahshid Hodjat
- Nephrology Department, Hannover Medical School, Hannover, Germany
| | | | | | | |
Collapse
|
28
|
Aini W, Miyagawa-Hayashino A, Tsuruyama T, Hashimoto S, Sumiyoshi S, Ozeki M, Tamaki K, Uemoto S, Haga H. Telomere shortening and karyotypic alterations in hepatocytes in long-term transplanted human liver allografts. Transpl Int 2012; 25:956-66. [PMID: 22775391 DOI: 10.1111/j.1432-2277.2012.01523.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The long-term fate of aged liver allografts in young recipients who received grafts from older donors is unknown. We evaluated graft aging by analyzing hepatocytic telomere length and karyotypic changes. Seventeen pediatric individuals who underwent living-donor liver transplantation for congenital biliary diseases were selected. At a median of 10.4 years post-transplant, ten had tolerated grafts with weaned off immunosuppressants, and seven had idiopathic post-transplantation hepatitis. Fluorescence in situ hybridization was used to evaluate the telomere signal intensity (TI) and karyotypic changes. First, we measured predictive age-dependent TI decline with regression analysis of donor livers. The mean TI at the earliest (within a year) and latest biopsies was significantly lower than the predicted TI of the studied allografts. With univariate analysis, a higher abnormal karyotype ratio in the donor liver was correlated with development of idiopathic post-transplantation hepatitis. With multivariate analysis that included clinical parameters, a greater TI decline at the earliest biopsy was correlated with the development of idiopathic post-transplantation hepatitis. In conclusion, graft aging as measured by TI decline and donor abnormal karyotype ratio was associated with idiopathic post-transplantation hepatitis of long-term transplanted liver allografts.
Collapse
Affiliation(s)
- Wulamujiang Aini
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Begus-Nahrmann Y, Hartmann D, Kraus J, Eshraghi P, Scheffold A, Grieb M, Rasche V, Schirmacher P, Lee HW, Kestler HA, Lechel A, Rudolph KL. Transient telomere dysfunction induces chromosomal instability and promotes carcinogenesis. J Clin Invest 2012; 122:2283-8. [PMID: 22622037 DOI: 10.1172/jci61745] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 04/06/2012] [Indexed: 01/23/2023] Open
Abstract
Telomere shortening limits the proliferative capacity of a cell, but perhaps surprisingly, shortening is also known to be associated with increased rates of tumor initiation. A current hypothesis suggests that telomere dysfunction increases tumor initiation by induction of chromosomal instability, but that initiated tumors need to reactivate telomerase for genome stabilization and tumor progression. This concept has not been tested in vivo, since appropriate mouse models were lacking. Here, we analyzed hepatocarcinogenesis in a mouse model of inducible telomere dysfunction on a telomerase-proficient background, in telomerase knockout mice with chronic telomere dysfunction (G3 mTerc-/-), and in WT mice with functional telomeres and telomerase. Transient or chronic telomere dysfunction enhanced the rates of chromosomal aberrations during hepatocarcinogenesis, but only telomerase-proficient mice exhibited significantly increased rates of macroscopic tumor formation in response to telomere dysfunction. In contrast, telomere dysfunction resulted in pronounced accumulation of DNA damage, cell-cycle arrest, and apoptosis in telomerase-deficient liver tumors. Together, these data provide in vivo evidence that transient telomere dysfunction during early or late stages of tumorigenesis promotes chromosomal instability and carcinogenesis in telomerase-proficient mice.
Collapse
|
30
|
A stochastic step model of replicative senescence explains ROS production rate in ageing cell populations. PLoS One 2012; 7:e32117. [PMID: 22359661 PMCID: PMC3281103 DOI: 10.1371/journal.pone.0032117] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 01/23/2012] [Indexed: 11/30/2022] Open
Abstract
Increases in cellular Reactive Oxygen Species (ROS) concentration with age have been observed repeatedly in mammalian tissues. Concomitant increases in the proportion of replicatively senescent cells in ageing mammalian tissues have also been observed. Populations of mitotic human fibroblasts cultured in vitro, undergoing transition from proliferation competence to replicative senescence are useful models of ageing human tissues. Similar exponential increases in ROS with age have been observed in this model system. Tracking individual cells in dividing populations is difficult, and so the vast majority of observations have been cross-sectional, at the population level, rather than longitudinal observations of individual cells. One possible explanation for these observations is an exponential increase in ROS in individual fibroblasts with time (e.g. resulting from a vicious cycle between cellular ROS and damage). However, we demonstrate an alternative, simple hypothesis, equally consistent with these observations which does not depend on any gradual increase in ROS concentration: the Stochastic Step Model of Replicative Senescence (SSMRS). We also demonstrate that, consistent with the SSMRS, neither proliferation-competent human fibroblasts of any age, nor populations of hTERT overexpressing human fibroblasts passaged beyond the Hayflick limit, display high ROS concentrations. We conclude that longitudinal studies of single cells and their lineages are now required for testing hypotheses about roles and mechanisms of ROS increase during replicative senescence.
Collapse
|
31
|
Melnik BC. Milk signalling in the pathogenesis of type 2 diabetes. Med Hypotheses 2011; 76:553-9. [PMID: 21251764 DOI: 10.1016/j.mehy.2010.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Revised: 10/04/2010] [Accepted: 12/23/2010] [Indexed: 01/02/2023]
Abstract
The presented hypothesis identifies milk consumption as an environmental risk factor of Western diet promoting type 2 diabetes (T2D). Milk, commonly regarded as a valuable nutrient, exerts important endocrine functions as an insulinotropic, anabolic and mitogenic signalling system supporting neonatal growth and development. The presented hypothesis substantiates milk's physiological role as a signalling system for pancreatic β-cell proliferation by milk's ability to increase prolactin-, growth hormone and incretin-signalling. The proposed mechanism of milk-induced postnatal β-cell mass expansion mimics the adaptive prolactin-dependent proliferative changes observed in pregnancy. Milk signalling down-regulates the key transcription factor FoxO1 leading to up-regulation of insulin promoter factor-1 which stimulates β-cell proliferation, insulin secretion as well as coexpression of islet amyloid polypeptide (IAPP). The recent finding that adult rodent β-cells only proliferate by self-duplication is of crucial importance, because permanent milk consumption beyond the weaning period may continuously over-stimulate β-cell replication thereby accelerating the onset of replicative β-cell senescence. The long-term use of milk may thus increase endoplasmic reticulum (ER) stress and toxic IAPP oligomer formation by overloading the ER with cytotoxic IAPPs thereby promoting β-cell apoptosis. Both increased β-cell proliferation and β-cell apoptosis are hallmarks of T2D. This hypothesis gets support from clinical states of hyperprolactinaemia and progeria syndromes with early onset of cell senescence which are both associated with an increased incidence of T2D and share common features of milk signalling. Furthermore, the presented milk hypothesis of T2D is compatible with the concept of high ER stress in T2D and the toxic oligomer hypothesis of T2D and may explain the high association of T2D and Alzheimer disease.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany.
| |
Collapse
|
32
|
Xu Y. Chemistry in human telomere biology: structure, function and targeting of telomere DNA/RNA. Chem Soc Rev 2011; 40:2719-40. [DOI: 10.1039/c0cs00134a] [Citation(s) in RCA: 249] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Fujita K, Horikawa I, Mondal AM, Miller Jenkins LM, Appella E, Vojtesek B, Bourdon JC, Lane DP, Harris CC. Positive feedback between p53 and TRF2 during telomere-damage signalling and cellular senescence. Nat Cell Biol 2010; 12:1205-12. [PMID: 21057505 PMCID: PMC3470109 DOI: 10.1038/ncb2123] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 09/24/2010] [Indexed: 12/12/2022]
Abstract
The telomere-capping complex shelterin protects functional telomeres and prevents the initiation of unwanted DNA-damage-response pathways. At the end of cellular replicative lifespan, uncapped telomeres lose this protective mechanism and DNA-damage signalling pathways are triggered that activate p53 and thereby induce replicative senescence. Here, we identify a signalling pathway involving p53, Siah1 (a p53-inducible E3 ubiquitin ligase) and TRF2 (telomere repeat binding factor 2; a component of the shelterin complex). Endogenous Siah1 and TRF2 were upregulated and downregulated, respectively, during replicative senescence with activated p53. Experimental manipulation of p53 expression demonstrated that p53 induces Siah1 and represses TRF2 protein levels. The p53-dependent ubiquitylation and proteasomal degradation of TRF2 are attributed to the E3 ligase activity of Siah1. Knockdown of Siah1 stabilized TRF2 and delayed the onset of cellular replicative senescence, suggesting a role for Siah1 and TRF2 in p53-regulated senescence. This study reveals that p53, a downstream effector of telomere-initiated damage signalling, also functions upstream of the shelterin complex.
Collapse
Affiliation(s)
- Kaori Fujita
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| | - Abdul M. Mondal
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| | - Lisa M. Miller Jenkins
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4256, USA
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4256, USA
| | - Borivoj Vojtesek
- Masaryk Memorial Cancer Institute, Zluty Kopec 7, 65653 Brno, Czech Republic
| | - Jean-Christophe Bourdon
- University of Dundee, Ninewells Hospital, Dept. of Surgery and Molecular Oncology, Inserm-European Associated Laboratory, Dundee, DD1 9SY, UK
| | - David P. Lane
- University of Dundee, Ninewells Hospital, Dept. of Surgery and Molecular Oncology, Inserm-European Associated Laboratory, Dundee, DD1 9SY, UK
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| |
Collapse
|
34
|
Xu Y, Ishizuka T, Kurabayashi K, Komiyama M. Consecutive formation of G-quadruplexes in human telomeric-overhang DNA: a protective capping structure for telomere ends. Angew Chem Int Ed Engl 2009; 48:7833-6. [PMID: 19757477 DOI: 10.1002/anie.200903858] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yan Xu
- Research Center for Advanced Science and Technology, The University of Tokyo, Komaba, Meguro-ku, Tokyo 153-8904, Japan.
| | | | | | | |
Collapse
|
35
|
Westhoff JH, Schildhorn C, Jacobi C, Hömme M, Hartner A, Braun H, Kryzer C, Wang C, von Zglinicki T, Kränzlin B, Gretz N, Melk A. Telomere shortening reduces regenerative capacity after acute kidney injury. J Am Soc Nephrol 2009; 21:327-36. [PMID: 19959722 DOI: 10.1681/asn.2009010072] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Telomeres of most somatic cells progressively shorten, compromising the regenerative capacity of human tissues during aging and chronic diseases and after acute injury. Whether telomere shortening reduces renal regeneration after acute injury is unknown. Here, renal ischemia-reperfusion injury led to greater impairment of renal function and increased acute and chronic histopathologic damage in fourth-generation telomerase-deficient mice compared with both wild-type and first-generation telomerase-deficient mice. Critically short telomeres, increased expression of the cell-cycle inhibitor p21, and more apoptotic renal cells accompanied the pronounced damage in fourth-generation telomerase-deficient mice. These mice also demonstrated significantly reduced proliferative capacity in tubular, glomerular, and interstitial cells. These data suggest that critical telomere shortening in the kidney leads to increased senescence and apoptosis, thereby limiting regenerative capacity in response to injury.
Collapse
Affiliation(s)
- Jens H Westhoff
- Children's Hospital, Medical School Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Xu Y, Ishizuka T, Kurabayashi K, Komiyama M. Consecutive Formation of G-Quadruplexes in Human Telomeric-Overhang DNA: A Protective Capping Structure for Telomere Ends. Angew Chem Int Ed Engl 2009. [DOI: 10.1002/ange.200903858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
37
|
Spallarossa P, Altieri P, Aloi C, Garibaldi S, Barisione C, Ghigliotti G, Fugazza G, Barsotti A, Brunelli C. Doxorubicin induces senescence or apoptosis in rat neonatal cardiomyocytes by regulating the expression levels of the telomere binding factors 1 and 2. Am J Physiol Heart Circ Physiol 2009; 297:H2169-81. [PMID: 19801496 DOI: 10.1152/ajpheart.00068.2009] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Low or high doses of doxorubicin induce either senescence or apoptosis, respectively, in cardiomyocytes. The mechanism by which different doses of doxorubicin may induce different stress-response cellular programs is not well understood. A recent study showed that the level of telomere dysfunction may induce senescence or apoptosis. We investigated the pathways to both apoptosis and senescence in neonatal rat cardiomyocytes and in H9c2 cells exposed to a single pulsed incubation with low or high doses of doxorubicin. High-dose doxorubicin strongly reduces TRF2 expression while enhancing TRF1 expression, and it determines early apoptosis. Low-dose doxorubicin induces downregulation of both TRF2 and TRF1, and it also increases the senescence-associated-beta-galactosidase activity, downregulates the checkpoint kinase Chk2, induces chromosomal abnormalities, and alters the cell cycle. The involvement of TRF1 and TRF2 with apoptosis and senescence was assessed by short interfering RNA interference. The cells maintain telomere dysfunction and a senescent phenotype over time and undergo late death. The increase in the phase>4N and the presence of micronuclei and anaphase bridges indicate that cells die by mitotic catastrophe. p38 modulates TRF2 expression, whereas JNK and cytoplasmic p53 regulate TRF1. Pretreatment with specific inhibitors of MAPKs and p53 may either attenuate the damage induced by doxorubicin or shift the cellular response to stress from senescence to apoptosis. In conclusion, various doses of doxorubicin induce differential regulation of TRF1 and TRF2 through p53 and MAPK, which is responsible for inducing either early apoptosis or senescence and late death due to mitotic catastrophe.
Collapse
Affiliation(s)
- Paolo Spallarossa
- Research Center of Cardiovascular Biology, Division of Cardiology, University of Genoa, Viale Benedetto XV, 6, 16132 Genoa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
von Figura G, Hartmann D, Song Z, Rudolph KL. Role of telomere dysfunction in aging and its detection by biomarkers. J Mol Med (Berl) 2009; 87:1165-71. [PMID: 19669107 DOI: 10.1007/s00109-009-0509-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2009] [Revised: 07/07/2009] [Accepted: 07/17/2009] [Indexed: 01/27/2023]
Abstract
Aging is a complex process that has been shown to be linked to accumulation of DNA damage. Telomere shortening represents a cell-intrinsic mechanism leading to DNA damage accumulation and activation of DNA damage checkpoints in aging cells. Activation of DNA damage checkpoints in response to telomere dysfunction results in induction of cellular senescence-a permanent cell cycle arrest. Senescence represents a tumor suppressor mechanism protecting cells from evolution of genomic instability and transformation. As a drawback, telomere shortening may also limit tissue renewal and regenerative capacity of tissues in response to aging and chronic disease. In aged organs, telomere shortening may also increase the cancer risk by initiation of chromosomal instability, loss of proliferative competition of aging stem cells, and selection of aberrant growing clones. Consequently, aged individuals are more susceptible and vulnerable to various diseases and show an increased cancer risk. Recently, proteins were discovered, which are induced by telomere dysfunction and DNA damage. It was shown that these proteins represent new biomarkers of human aging and disease. Here, we review the scientific background and experimental data on these newly discovered biomarkers.
Collapse
Affiliation(s)
- Guido von Figura
- Department of Molecular Medicine and Max-Planck-Research Group on Stem Cell Aging, University of Ulm, 89081 Ulm, Germany
| | | | | | | |
Collapse
|
39
|
Pang LY, Argyle DJ. Using naturally occurring tumours in dogs and cats to study telomerase and cancer stem cell biology. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1792:380-91. [PMID: 19254761 DOI: 10.1016/j.bbadis.2009.02.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 02/15/2009] [Accepted: 02/17/2009] [Indexed: 01/06/2023]
Abstract
The recently described cancer stem cell theory opens up many new challenges and opportunities to identify targets for therapeutic intervention. However, the majority of cancer related therapeutic studies rely upon rodent models of human cancer that rarely translate into clinical success in human patients. Naturally occurring cancers in dogs, cats and humans share biological features, including molecular targets, telomerase biology and tumour genetics. Studying cancer stem cell biology and telomere/telomerase dynamics in the cancer bearing pet population may offer the opportunity to develop a greater understanding of cancer biology in the natural setting and evaluate the development of novel therapies targeted at these systems.
Collapse
Affiliation(s)
- Lisa Y Pang
- University of Edinburgh, Midlothian EH25 9RG, Scotland, UK
| | | |
Collapse
|
40
|
Folini M, Gandellini P, Zaffaroni N. Targeting the telosome: therapeutic implications. Biochim Biophys Acta Mol Basis Dis 2009; 1792:309-16. [PMID: 19419699 DOI: 10.1016/j.bbadis.2009.01.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 01/28/2009] [Accepted: 01/30/2009] [Indexed: 12/12/2022]
Abstract
Since telomere integrity is required to guarantee the unlimited replicative potential of cancer cells, telomerase, the enzyme responsible for telomere length maintenance in most human tumors, and lately also telomeres themselves have become extremely attractive targets for new anticancer interventions. At the current status of knowledge, it is still not possible to define the best therapeutic target between telomerase and telomeres. It is noteworthy that interfering with telomeres, through direct targeting of telomeric DNA or proteins involved in the telosome complex, could negatively affect the proliferative potential not only of tumors expressing telomerase activity but also of those that maintain their telomeres through alternative lengthening or still unknown mechanisms. This review presents the different therapeutic approaches proposed thus far and developed in preclinical tumor models and discusses the perspectives for their use in the clinical setting.
Collapse
Affiliation(s)
- Marco Folini
- Department of Experimental Oncology and Laboratories, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | | | | |
Collapse
|
41
|
Senescence and immortality in hepatocellular carcinoma. Cancer Lett 2008; 286:103-13. [PMID: 19070423 DOI: 10.1016/j.canlet.2008.10.048] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 06/23/2008] [Accepted: 10/29/2008] [Indexed: 12/31/2022]
Abstract
Cellular senescence is a process leading to terminal growth arrest with characteristic morphological features. This process is mediated by telomere-dependent, oncogene-induced and ROS-induced pathways, but persistent DNA damage is the most common cause. Senescence arrest is mediated by p16(INK4a)- and p21(Cip1)-dependent pathways both leading to retinoblastoma protein (pRb) activation. p53 plays a relay role between DNA damage sensing and p21(Cip1) activation. pRb arrests the cell cycle by recruiting proliferation genes to facultative heterochromatin for permanent silencing. Replicative senescence that occurs in hepatocytes in culture and in liver cirrhosis is associated with lack of telomerase activity and results in telomere shortening. Hepatocellular carcinoma (HCC) cells display inactivating mutations of p53 and epigenetic silencing of p16(INK4a). Moreover, they re-express telomerase reverse transcriptase required for telomere maintenance. Thus, senescence bypass and cellular immortality is likely to contribute significantly to HCC development. Oncogene-induced senescence in premalignant lesions and reversible immortality of cancer cells including HCC offer new potentials for tumor prevention and treatment.
Collapse
|
42
|
Marie-Egyptienne DT, Brault ME, Nimmo GAM, Londoño-Vallejo JA, Autexier C. Growth defects in mouse telomerase RNA-deficient cells expressing a template-mutated mouse telomerase RNA. Cancer Lett 2008; 275:266-76. [PMID: 19056167 DOI: 10.1016/j.canlet.2008.10.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/15/2008] [Accepted: 10/17/2008] [Indexed: 01/03/2023]
Abstract
Cellular viability requires telomere maintenance, which, in mammals, is mainly mediated by the reverse transcriptase telomerase. Telomerase core components are a catalytic subunit TERT and an RNA subunit TR (hTR in humans, mTR in mouse) that carries the template to generate telomeres de novo. Telomere dysfunction can lead to senescence or apoptosis and impairs the continued growth of immortal cancerous cell lines. The introduction of a template-mutated hTR in telomerase-positive and telomerase-negative human cell lines results in dramatic growth defects. No study has addressed the consequences of expressing a template-mutated mTR in mouse immortal cell lines. Therefore, we analyzed the effects of long-term expression of a template-mutated mTR in the telomerase-positive and telomerase-negative murine cell lines CB17 and DKO301, respectively. Whereas the CB17 clones expressing the template-mutated mTR did not demonstrate any growth impairment, many of the DKO301 clones expressing the template-mutated mTR underwent growth and cell cycle defects and eventual cell death. These results suggest that in the absence of wild-type telomerase, the expression of the template-mutated mTR likely perturbs telomere function, leading to decreased cellular viability. Furthermore, whereas the expression of template-mutated hTR in telomerase-negative human cell lines leads to immediate cellular toxicity, the expression of the template-mutated mTR in the telomerase-negative mouse cell line did not.
Collapse
|
43
|
Jeyapalan JC, Sedivy JM. Cellular senescence and organismal aging. Mech Ageing Dev 2008; 129:467-74. [PMID: 18502472 PMCID: PMC3297662 DOI: 10.1016/j.mad.2008.04.001] [Citation(s) in RCA: 262] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2007] [Revised: 03/27/2008] [Accepted: 04/06/2008] [Indexed: 01/22/2023]
Abstract
Cellular senescence, first observed and defined using in vitro cell culture studies, is an irreversible cell cycle arrest which can be triggered by a variety of factors. Emerging evidence suggests that cellular senescence acts as an in vivo tumor suppression mechanism by limiting aberrant proliferation. It has also been postulated that cellular senescence can occur independently of cancer and contribute to the physiological processes of normal organismal aging. Recent data have demonstrated the in vivo accumulation of senescent cells with advancing age. Some characteristics of senescent cells, such as the ability to modify their extracellular environment, could play a role in aging and age-related pathology. In this review, we examine current evidence that links cellular senescence and organismal aging.
Collapse
Affiliation(s)
- Jessie C. Jeyapalan
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - John M. Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| |
Collapse
|
44
|
Panda S, Isbatan A, Adami GR. Modification of the ATM/ATR directed DNA damage response state with aging and long after hepatocyte senescence induction in vivo. Mech Ageing Dev 2008; 129:332-40. [PMID: 18440596 PMCID: PMC2532062 DOI: 10.1016/j.mad.2008.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 02/26/2008] [Accepted: 02/29/2008] [Indexed: 01/24/2023]
Abstract
The cellular DNA damage response (DDR) entails the activation of ATM, ATR and/or DNA PK protein kinases that causes modifications of proteins including Chk1, Chk2 and 53BP1, aggregation of DDR proteins into foci, and activation of p53. The DDR is thought to be required for initiation and maintenance of cellular senescence. Potentially senescent cells with DNA damage foci occur in large numbers in vivo with many diseases, but, with the exception of mammalian dermis, there is little evidence for that with normal aging. After experimental induction of cellular senescence in the livers of juvenile mice, there was robust expression of DDR markers in hepatocytes at 1 week; however, by 7 weeks, activation of ATM/ATR kinase targets was limited, although cells with DNA damage foci were present. An analysis of hepatocytes of aged, 22-month-old mice, not experimentally exposed to genotoxins, showed limited activation of ATM/ATR targets, though high numbers of cells with DNA damage foci were found, similar to that seen many weeks after artificial senescence induction in young mice. Based on senescence heterochromatin and SA ss Gal assays of the 22-month-old mouse liver, more than 20% of hepatocytes were potentially senescent, though only some components of the DDR were enriched.
Collapse
Affiliation(s)
| | | | - Guy R. Adami
- Dept. of Oral Medicine and Diagnostic Sciences, Center for Molecular Biology of Oral Diseases, College of Dentistry, University of Illinois at Chicago, 801 S Paulina St., Chicago, IL 60612
| |
Collapse
|
45
|
Spyridopoulos I, Erben Y, Brummendorf TH, Haendeler J, Dietz K, Seeger F, Kissel CK, Martin H, Hoffmann J, Assmus B, Zeiher AM, Dimmeler S. Telomere Gap Between Granulocytes and Lymphocytes Is a Determinant for Hematopoetic Progenitor Cell Impairment in Patients With Previous Myocardial Infarction. Arterioscler Thromb Vasc Biol 2008; 28:968-74. [DOI: 10.1161/atvbaha.107.160846] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Objective—
We have previously demonstrated that ischemic cardiomyopathy is associated with selective impairment of progenitor cell function in the bone marrow and in the peripheral blood, which may contribute to an unfavorable left ventricular remodeling process.
Methods and Results—
With this study, we intended to identify the influence of telomere length on bone marrow functionality in 50 patients with coronary artery disease (CAD) and previous myocardial infarction. Mean telomere length (mTL) was measured simultaneously in peripheral blood leukocytes and mononuclear bone marrow cells (BMC), using the flow-FISH method. Telomere erosion already occurred at the bone marrow level, whereby age (39 bp/yr,
P
=0.025) and the number of affected vessels (434 bp/vessel,
P
=0.029) were the only independent predictors. Lymphocytes demonstrated significant TL shortening between BMCs and peripheral blood in CAD patients (−1011±897 bp) as opposed to an increase in a young control group (+235±459 bp,
P
<0.001). SDF- and VEGF-specific migration of BMCs correlated with mTL of lymphocytes (
r
=0.42,
P
<0.001) and was significantly reduced in CAD patients. Finally, the telomere length difference between granulocytes and lymphocytes was the most determinant for telomere-associated bone marrow impairment (
P
<0.001).
Conclusion—
In patients with CAD, telomere shortening of BMCs is dependent on both age and the extent of CAD and correlates with bone marrow cell functionality.
Collapse
Affiliation(s)
- Ioakim Spyridopoulos
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Young Erben
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Tim H. Brummendorf
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Judith Haendeler
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Klaus Dietz
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Florian Seeger
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Christine K. Kissel
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Hans Martin
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Jedrzej Hoffmann
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Birgit Assmus
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Andreas M. Zeiher
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| | - Stefanie Dimmeler
- From the Departments of Cardiology and Molecular Cardiology (I.S., Y.E., J.H., F.S., C.K., J.H., B.A., A.M.Z., S.D.), Johann Wolfgang Goethe University of Frankfurt, Germany; the Department of Oncology and Hematology (T.H.B.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; the Department of Medical Biometry (K.D.), University of Tübingen, Germany; and the Department of Hematology (H.M.), Johann Wolfgang Goethe University of Frankfurt, Germany
| |
Collapse
|
46
|
Zhang YW, Zhang ZX, Miao ZH, Ding J. The telomeric protein TRF2 is critical for the protection of A549 cells from both telomere erosion and DNA double-strand breaks driven by salvicine. Mol Pharmacol 2008; 73:824-32. [PMID: 18025071 DOI: 10.1124/mol.107.039081] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Telomere repeat binding factor 2 (TRF2) has been increasingly recognized to be involved in DNA damage response and telomere maintenance. Our previous report found that salvicine (SAL), a novel topoisomerase II poison, elicited DNA double-strand breaks and telomere erosion in separate experimental systems. However, it remains to be clarified whether they share a common response to these two events and in particular whether TRF2 is involved in this process. In this study, we found that SAL concurrently induced DNA double-strand breaks, telomeric DNA damage, and telomere erosion in lung carcinoma A549 cells. It was unexpected to find that SAL led to disruption of TRF2, independently of either its transcription or proteasome-mediated degradation. By overexpressing the full-length trf2 gene and transfecting TRF2 small interfering RNAs, we showed that TRF2 protein protected both telomeric and genomic DNA from the SAL-elicited events. It is noteworthy that although both the Ataxia-telangiectasia-mutated (ATM) and the ATM- and Rad3-related (ATR) kinases responded to the SAL-induced DNA damages, only ATR was essential for the telomere erosion. The study also showed that the activated ATR augmented the SAL-triggered TRF2 disruption, whereas TRF2 reduction in turn enhanced ATR function. All of these findings suggest the emerging significance of TRF2 protecting both telomeric DNA and genomic DNA on the one hand and reveal the mutual modulation between ATR and TRF2 in sensing DNA damage signaling during cancer development on the other hand.
Collapse
Affiliation(s)
- Yong-Wei Zhang
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | | | | | | |
Collapse
|
47
|
Ju Z, Rudolph L. Telomere dysfunction and stem cell ageing. Biochimie 2008; 90:24-32. [DOI: 10.1016/j.biochi.2007.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2007] [Accepted: 09/10/2007] [Indexed: 02/08/2023]
|
48
|
Abstract
Adult stem cells have become the focus of intense research in recent years as a result of their role in the maintenance and repair of tissues. They exert this function through their extensive expansion (self-renewal) and multipotent differentiation capacity. Understanding whether adult stem cells retain this capacity throughout the lifespan of the individual, or undergo a process of ageing resulting in a decreased stem cell pool, is an important area of investigation. Progress in this area has been hampered by lack of suitable models and of appropriate markers and assays to identify stem cells. However, recent data suggest that an understanding of the mechanisms governing stem cell ageing can give insight into the mechanism of tissue ageing and, most importantly, advance our ability to use stem cells in cell and gene therapy strategies.
Collapse
|
49
|
Schaetzlein S, Kodandaramireddy N, Ju Z, Lechel A, Stepzynska A, Lilli DR, Clark AB, Rudolph C, Wei K, Schlegelberger B, Schirmacher P, Kunkel TA, Greenberg RA, Edelmann W, Rudolph KL. Exonuclease-1 deletion impairs DNA damage signaling and prolongs lifespan of telomere-dysfunctional mice. Cell 2007; 130:863-77. [PMID: 17803909 PMCID: PMC2658812 DOI: 10.1016/j.cell.2007.08.029] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 05/17/2007] [Accepted: 08/20/2007] [Indexed: 12/26/2022]
Abstract
Exonuclease-1 (EXO1) mediates checkpoint induction in response to telomere dysfunction in yeast, but it is unknown whether EXO1 has similar functions in mammalian cells. Here we show that deletion of the nuclease domain of Exo1 reduces accumulation of DNA damage and DNA damage signal induction in telomere-dysfunctional mice. Exo1 deletion improved organ maintenance and lifespan of telomere-dysfunctional mice but did not increase chromosomal instability or cancer formation. Deletion of Exo1 also ameliorated the induction of DNA damage checkpoints in response to gamma-irradiation and conferred cellular resistance to 6-thioguanine-induced DNA damage. Exo1 deletion impaired upstream induction of DNA damage responses by reducing ssDNA formation and the recruitment of Replication Protein A (RPA) and ATR at DNA breaks. Together, these studies provide evidence that EXO1 contributes to DNA damage signal induction in mammalian cells, and deletion of Exo1 can prolong survival in the context of telomere dysfunction.
Collapse
Affiliation(s)
- Sonja Schaetzlein
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Germany
| | - N.R Kodandaramireddy
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Germany
| | - Zhenyu Ju
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Germany
| | - Andre Lechel
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Germany
| | - Anna Stepzynska
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Germany
| | - Dana R. Lilli
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104-6160, U.S.A
| | - Alan B. Clark
- Laboratory of Structural Biology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Cornelia Rudolph
- Institute of Cell and Molecular Pathology, Medical School Hannover, Germany
| | - Kaichun Wei
- Department of Molecular Biology, Albert Einstein College of Medicine, New York, U.S.A
| | | | - Peter Schirmacher
- Institute of Pathology, University Hospital, Im Neuenheimer Feld 220/221, 69120 Heidelberg, Germany
| | - Thomas A. Kunkel
- Laboratory of Structural Biology, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA
| | - Roger A. Greenberg
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA, 19104-6160, U.S.A
| | - Winfried Edelmann
- Department of Molecular Biology, Albert Einstein College of Medicine, New York, U.S.A
| | - K. Lenhard Rudolph
- Department of Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Germany
| |
Collapse
|
50
|
Jennings P, Koppelstaetter C, Aydin S, Abberger T, Wolf AM, Mayer G, Pfaller W. Cyclosporine A induces senescence in renal tubular epithelial cells. Am J Physiol Renal Physiol 2007; 293:F831-8. [PMID: 17596534 DOI: 10.1152/ajprenal.00005.2007] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The nephrotoxic potential of the widely used immunosuppressive agent cyclosporine A (CsA) is well recognized. However, the mechanism of renal tubular toxicity is not yet fully elucidated. Chronic CsA nephropathy and renal organ aging share some clinical features, such as renal fibrosis and tubular atrophy, raising the possibility that CsA may exert some of its deleterious effects via induction of a stress-induced senescent phenotype. We investigated this hypothesis in HK-2 cells and primary proximal tubular cells in vitro. CsA induced the production of H2O2, caused cell cycle arrest in the G0/G1 phase, and inhibited DNA synthesis. Furthermore, CsA exposure lead to a reduction of telomere length, increased p53 serine 15 phosphorylation, and caused an upregulation of the cell cycle inhibitor p21Kip1 (CDKN1A) mRNA levels. CsA caused an increase in p16INK4a (CDKN2A) expression after a 13-day exposure in primary proximal tubular cells but not in HK-2 cells. Coincubation of cells with CsA and catalase was able to prevent telomere shortening and partially restored DNA synthesis. In summary, CsA induces cellular senescence in human renal tubular epithelial cells, which can be attenuated by scavenging reactive oxygen species.
Collapse
Affiliation(s)
- Paul Jennings
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria
| | | | | | | | | | | | | |
Collapse
|