1
|
Escriche-Navarro B, Garrido E, Sancenón F, García-Fernández A, Martínez-Máñez R. A navitoclax-loaded nanodevice targeting matrix metalloproteinase-3 for the selective elimination of senescent cells. Acta Biomater 2024; 176:405-416. [PMID: 38185231 DOI: 10.1016/j.actbio.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/18/2023] [Accepted: 01/01/2024] [Indexed: 01/09/2024]
Abstract
Cellular senescence is implicated in the occurrence and progression of multiple age-related disorders. In this context, the selective elimination of senescent cells, senolysis, has emerged as an effective therapeutic strategy. However, the heterogeneous senescent phenotype hinders the discovery of a universal and robust senescence biomarker that limits the effective of senolytic with off-target toxic effects. Therefore, the development of more selective strategies represents a promising approach to increase the specificity of senolytic therapy. In this study, we have developed an innovative nanodevice for the selective elimination of senescent cells (SCs) based on the specific enzymatic activity of the senescent secretome. The results revealed that when senescence is induced in proliferating WI-38 by ionizing radiation (IR), the cells secrete high levels of matrix metalloproteinase-3 (MMP-3). Based on this result, mesoporous silica nanoparticles (MSNs) were loaded with the senolytic navitoclax (Nav) and coated with a specific peptide which is substrate of MMP-3 (NPs(Nav)@MMP-3). Studies in cells confirmed the preferential release of cargo in IR-induced senescent cells compared to proliferating cells, depending on MMP-3 levels. Moreover, treatment with NPs(Nav)@MMP-3 induced a selective decrease in the viability of SCs as well as a protective effect on non-proliferating cells. These results demonstrate the potential use of NPs to develop enhanced senolytic therapies based on specific enzymatic activity in the senescent microenvironment, with potential clinical relevance. STATEMENT OF SIGNIFICANCE: The common β-galactosidase activity has been exploited to develop nanoparticles for the selective elimination of senescent cells. However, the identification of new senescent biomarkers is a key factor for the development of improved strategies. In this scenario, we report for the first time the development of NPs targeting senescent cells based on specific enzymatic activity of the senescent secretome. We report a navitoclax-loaded nanodevice responsive to the matrix metalloproteinase-3 (MMP-3) associated with the senescent phenotype. Our nanosystem achieves the selective release of navitoclax in an MMP-3-dependent manner while limiting off-target effects on non-senescent cells. This opens the possibility of using nanoparticles able to detect an altered senescent environment and selectively release its content, thus enhancing the efficacy of senolytic therapies.
Collapse
Affiliation(s)
- Blanca Escriche-Navarro
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026, Valencia, Spain
| | - Eva Garrido
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026, Valencia, Spain
| | - Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain.
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM) Universitat Politècnica de València, Universitat de València, Camino de Vera, s/n. 46022, Valencia, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, 46012, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029 Madrid, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València, IIS La Fe. Av. Fernando Abril Martorell, 106 Torre A 7ª planta, 46026, Valencia, Spain.
| |
Collapse
|
2
|
Abdul-Aziz A, Devine RD, Lyberger JM, Chang H, Kovacs A, Lerma JR, Rogers AM, Byrd JC, Hertlein E, Behbehani GK. Mass Cytometry as a Tool for Investigating Senescence in Multiple Model Systems. Cells 2023; 12:2045. [PMID: 37626855 PMCID: PMC10453346 DOI: 10.3390/cells12162045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Cellular senescence is a durable cell cycle arrest as a result of the finite proliferative capacity of cells. Senescence responds to both intrinsic and extrinsic cellular stresses, such as aging, mitochondrial dysfunction, irradiation, and chemotherapy. Here, we report on the use of mass cytometry (MC) to analyze multiple model systems and demonstrate MC as a platform for senescence analysis at the single-cell level. We demonstrate changes to p16 expression, cell cycling fraction, and histone tail modifications in several established senescent model systems and using isolated human T cells. In bone marrow mesenchymal stromal cells (BMSCs), we show increased p16 expression with subsequent passage as well as a reduction in cycling cells and open chromatin marks. In WI-38 cells, we demonstrate increased p16 expression with both culture-induced senescence and oxidative stress-induced senescence (OSIS). We also use Wanderlust, a trajectory analysis tool, to demonstrate how p16 expression changes with histone tail modifications and cell cycle proteins. Finally, we demonstrate that repetitive stimulation of human T cells with CD3/CD28 beads induces an exhausted phenotype with increased p16 expression. This p16-expressing population exhibited higher expression of exhaustion markers such as EOMES and TOX. This work demonstrates that MC is a useful platform for studying senescence at a single-cell protein level, and is capable of measuring multiple markers of senescence at once with high confidence, thereby improving our understanding of senescent pathways.
Collapse
Affiliation(s)
- Amina Abdul-Aziz
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45221, USA; (A.A.-A.)
| | - Raymond D. Devine
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Justin M. Lyberger
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Hsiaochi Chang
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Amy Kovacs
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - James R. Lerma
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45221, USA; (A.A.-A.)
| | - Andrew M. Rogers
- Maine Medical Center, Portland, ME 04102, USA
- Tufts University School of Medicine, Boston, MA 02111, USA
| | - John C. Byrd
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45221, USA; (A.A.-A.)
| | - Erin Hertlein
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45221, USA; (A.A.-A.)
| | - Gregory K. Behbehani
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Goy E, Tomezak M, Facchin C, Martin N, Bouchaert E, Benoit J, de Schutter C, Nassour J, Saas L, Drullion C, Brodin PM, Vandeputte A, Molendi-Coste O, Pineau L, Goormachtigh G, Pluquet O, Pourtier A, Cleri F, Lartigau E, Penel N, Abbadie C. The out-of-field dose in radiation therapy induces delayed tumorigenesis by senescence evasion. eLife 2022; 11:67190. [PMID: 35302491 PMCID: PMC8933005 DOI: 10.7554/elife.67190] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
A rare but severe complication of curative-intent radiation therapy is the induction of second primary cancers. These cancers preferentially develop not inside the planning target volume (PTV) but around, over several centimeters, after a latency period of 1–40 years. We show here that normal human or mouse dermal fibroblasts submitted to the out-of-field dose scattering at the margin of a PTV receiving a mimicked patient’s treatment do not die but enter in a long-lived senescent state resulting from the accumulation of unrepaired DNA single-strand breaks, in the almost absence of double-strand breaks. Importantly, a few of these senescent cells systematically and spontaneously escape from the cell cycle arrest after a while to generate daughter cells harboring mutations and invasive capacities. These findings highlight single-strand break-induced senescence as the mechanism of second primary cancer initiation, with clinically relevant spatiotemporal specificities. Senescence being pharmacologically targetable, they open the avenue for second primary cancer prevention.
Collapse
Affiliation(s)
- Erwan Goy
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Maxime Tomezak
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France.,Univ. Lille, CNRS, UMR8520, Institut d'Electronique, Microélectronique et Nanotechnologie, F-59652 Villeneuve d'Ascq, France
| | - Caterina Facchin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Nathalie Martin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Emmanuel Bouchaert
- Oncovet Clinical Research, Plateforme PRECI, F-59120 Loos, France.,Oncovet, Plateforme PRECI, F-59650 Villeneuve d'Ascq, France
| | - Jerome Benoit
- Oncovet Clinical Research, Plateforme PRECI, F-59120 Loos, France.,Oncovet, Plateforme PRECI, F-59650 Villeneuve d'Ascq, France
| | - Clementine de Schutter
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Joe Nassour
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Laure Saas
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Claire Drullion
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Priscille M Brodin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Alexandre Vandeputte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - CIIL - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Olivier Molendi-Coste
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000 Lille, France
| | - Laurent Pineau
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011 - EGID, F-59000 Lille, France
| | - Gautier Goormachtigh
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Olivier Pluquet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Albin Pourtier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| | - Fabrizio Cleri
- Univ. Lille, CNRS, UMR8520, Institut d'Electronique, Microélectronique et Nanotechnologie, F-59652 Villeneuve d'Ascq, France
| | - Eric Lartigau
- Lille University, Medical School and Centre Oscar Lambret, Lille, France
| | - Nicolas Penel
- Lille University, Medical School and Centre Oscar Lambret, Lille, France
| | - Corinne Abbadie
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies, F-59000 Lille, France
| |
Collapse
|
4
|
Abdelgawad IY, Sadak KT, Lone DW, Dabour MS, Niedernhofer LJ, Zordoky BN. Molecular mechanisms and cardiovascular implications of cancer therapy-induced senescence. Pharmacol Ther 2021; 221:107751. [PMID: 33275998 PMCID: PMC8084867 DOI: 10.1016/j.pharmthera.2020.107751] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Cancer treatment has been associated with accelerated aging that can lead to early-onset health complications typically experienced by older populations. In particular, cancer survivors have an increased risk of developing premature cardiovascular complications. In the last two decades, cellular senescence has been proposed as an important mechanism of premature cardiovascular diseases. Cancer treatments, specifically anthracyclines and radiation, have been shown to induce senescence in different types of cardiovascular cells. Additionally, clinical studies identified increased systemic markers of senescence in cancer survivors. Preclinical research has demonstrated the potential of several approaches to mitigate cancer therapy-induced senescence. However, strategies to prevent and/or treat therapy-induced cardiovascular senescence have not yet been translated to the clinic. In this review, we will discuss how therapy-induced senescence can contribute to cardiovascular complications. Thereafter, we will summarize the current in vitro, in vivo, and clinical evidence regarding cancer therapy-induced cardiovascular senescence. Then, we will discuss interventional strategies that have the potential to protect against therapy-induced cardiovascular senescence. To conclude, we will highlight challenges and future research directions to mitigate therapy-induced cardiovascular senescence in cancer survivors.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
| | - Karim T Sadak
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA; University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Diana W Lone
- University of Minnesota Masonic Children's Hospital, Minneapolis, MN 55455, USA
| | - Mohamed S Dabour
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| |
Collapse
|
5
|
Chai X, Han HH, Sedgwick AC, Li N, Zang Y, James TD, Zhang J, Hu XL, Yu Y, Li Y, Wang Y, Li J, He XP, Tian H. Photochromic Fluorescent Probe Strategy for the Super-resolution Imaging of Biologically Important Biomarkers. J Am Chem Soc 2020; 142:18005-18013. [DOI: 10.1021/jacs.0c05379] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Xianzhi Chai
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Road, Shanghai 201203, P. R. China
| | - Adam C. Sedgwick
- Department of Chemistry, The University of Texas at Austin, 105 East 24th Street A5300, Austin, Texas 78712-1224, United States
| | - Na Li
- National Center for Protein Science Shanghai, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Road, Shanghai 201203, P. R. China
| | - Tony D. James
- Department of Chemistry, University of Bath, Bath BA2 7AY, U.K
| | - Junji Zhang
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Xi-Le Hu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - Yang Yu
- National Center for Protein Science Shanghai, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Yao Li
- National Center for Protein Science Shanghai, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Yan Wang
- National Center for Protein Science Shanghai, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, P. R. China
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 189 Guo Shoujing Road, Shanghai 201203, P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, Frontiers Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, P. R. China
| |
Collapse
|
6
|
Senoptosis: non-lethal DNA cleavage as a route to deep senescence. Oncotarget 2018; 8:30656-30671. [PMID: 28427150 PMCID: PMC5458157 DOI: 10.18632/oncotarget.15693] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
DNA-damage-induced apoptosis and cellular senescence are perceived as two distinct cell fates. We found that after ionizing radiation (IR)-induced DNA damage the majority (up to 70 %) of senescent human diploid fibroblasts (HDFs) were subjected to controlled cleavage of DNA, resulting in the establishment of a viable and stable sub-G1 population, i.e. deeply senescent cells. We show that in senescent HDFs this DNA cleavage is triggered by modest loss of the mitochondrial membrane potential, which is not sufficient to activate caspases, but strong enough to release mitochondrial endonuclease G (EndoG). We demonstrate that upon γ-irradiation in HDFs EndoG translocates into the nucleus playing an essential role in the non-lethal cleavage of damaged DNA. Notably, the established sub-G1 cell population does not contribute to the senescence-associated secretory phenotype (SASP), however, it exhibits increased senescence-associated β-galactosidase activity. We show that EndoG knockdown causes an increase in DNA damage, indicating a role of this enzyme in DNA repair. Thus, we conclude that IR-induced deep senescence of HDFs exhibits features of both senescence, such as cell cycle arrest and viability, and apoptosis like reduced DNA content and no SASP, and, resembles uncomplete or stalled apoptosis, a phenomenon we term senoptosis.
Collapse
|
7
|
Effect of sulfated galactan from Porphyra haitanensis on H 2O 2-induced premature senescence in WI-38 cells. Int J Biol Macromol 2017; 106:1235-1239. [PMID: 28860061 DOI: 10.1016/j.ijbiomac.2017.08.123] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 02/01/2023]
Abstract
Porphyran sulfated galactan extracted from red algae Porphyra haitanensis is a sulfated polysaccharide, which possesses excellent activities. In the present study, WI-38 cells were treated with H2O2 to induce premature senescence and then the protection of porphyran against aging in vitro and associated molecular mechanisms were investigated. The protection occurred in a dose-dependent manner, offering an optimal efficacy starting at 10μg/mL. The proportion of SA-β-gal positive cells in porphyran group decreases from 53% to 23% in the cultures at 30 PDs. Porphyran has been detected specifically reducing SAHF-like foci formation in senescent cells. In addition, porphyran significantly affected the p53-p21 pathways in H2O2-treated WI-38 cells. Our data suggest the promising role of porphyran as an attractive and bio-safe agent with the potential to retard senescence and attenuate senescence-related diseases.
Collapse
|
8
|
Triana-Martínez F, Pedraza-Vázquez G, Maciel-Barón LA, Königsberg M. Reflections on the role of senescence during development and aging. Arch Biochem Biophys 2016; 598:40-9. [PMID: 27059850 DOI: 10.1016/j.abb.2016.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/02/2016] [Accepted: 04/04/2016] [Indexed: 01/07/2023]
Abstract
New and stimulating results have challenged the concept that cellular senescence might not be synonymous with aging. It is indisputable that during aging, senescent cell accumulation has an impact on organismal health. Nevertheless, senescent cells are now known to display physiological roles during embryonic development, during wound healing repair and as a cellular response to stress. The fact that senescence has been found in cells that did not attain their maximal round of replications, nor have metabolic alterations or DNA damage, also challenges the paradigm that senescence is cellular aging, and it is in favor of the idea that cellular senescence is a phenomenon that has a function by itself. Therefore, in order to understand this phenomenon it is important to analyze the relationship between senescence and other cellular responses that have many features in common, such as apoptosis, cancer and autophagy, particularly highlighting their role during development and adulthood.
Collapse
Affiliation(s)
- F Triana-Martínez
- Dept. Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México D.F. 09340, Mexico
| | - G Pedraza-Vázquez
- Dept. Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México D.F. 09340, Mexico
| | - L A Maciel-Barón
- Dept. Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México D.F. 09340, Mexico
| | - M Königsberg
- Dept. Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana Iztapalapa, México D.F. 09340, Mexico.
| |
Collapse
|
9
|
Appukuttan A, Flacke JP, Flacke H, Posadowsky A, Reusch HP, Ladilov Y. Inhibition of soluble adenylyl cyclase increases the radiosensitivity of prostate cancer cells. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2656-63. [PMID: 25257405 DOI: 10.1016/j.bbadis.2014.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 09/15/2014] [Accepted: 09/16/2014] [Indexed: 11/16/2022]
Abstract
Pharmacological modulation of tumor radiosensitivity is a promising strategy for enhancing the outcome of radiotherapy. cAMP signaling plays an essential role in modulating the proliferation and apoptosis of different cell types, including cancer cells. Until now, the regulation of this pathway was restricted to the transmembrane class of adenylyl cyclases. In the present study, the role of an alternative source of cAMP, the intracellular localized soluble adenylyl cyclase (sAC), in the radiosensitivity of prostate cancer cells was investigated. Pharmacological inhibition of sAC activity led to marked suppression of proliferation, lactate dehydrogenase release, and induction of apoptosis. The combination of ionizing radiation with partial suppression of sAC activity (~50%) immediately after irradiation synergistically inhibited proliferation and induced apoptosis. Overexpression of sAC in normal prostate epithelial PNT2 cells increased the cAMP content and accelerated cell proliferation under control conditions. The effects of radiation were significantly reduced in transformed PNT2 cells compared with control cells. Analysis of the underlying cellular mechanisms of sAC-induced radioresistance revealed the sAC-dependent activation of B-Raf/ERK1/2 signaling. In agreement with this finding, inhibition of ERK1/2 in prostate cancer cells enhanced the cytotoxic effect of irradiation. In conclusion, the present study suggests that sAC-dependent signaling plays an important role in the radioresistance of prostate cancer cells. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.
Collapse
Affiliation(s)
| | - Jan-Paul Flacke
- Department of Clinical Pharmacology, Ruhr-University Bochum, Germany
| | - Hanna Flacke
- Department of Clinical Pharmacology, Ruhr-University Bochum, Germany
| | | | - H Peter Reusch
- Department of Clinical Pharmacology, Ruhr-University Bochum, Germany
| | - Yury Ladilov
- Department of Clinical Pharmacology, Ruhr-University Bochum, Germany; Center for Cardiovascular Research, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
10
|
Liu Q, Chen W, Jiao Y, Hou J, Wu Q, Liu Y, Qi X. Pulsatilla saponin A, an active molecule from Pulsatilla chinensis, induces cancer cell death and inhibits tumor growth in mouse xenograft models. J Surg Res 2014; 188:387-95. [PMID: 24576780 DOI: 10.1016/j.jss.2014.01.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/05/2014] [Accepted: 01/16/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Many natural compounds possess antitumor growth activities. Pulsatilla chinensis is an herb used in traditional Chinese medicine to treat infectious diseases. More recently, extracts from P chinensis have been shown to contain antitumor activities. MATERIALS AND METHODS In this study, we isolated Pulsatilla saponin A as an active compound from P chinensis extracts and tested its anticancer activity in vitro and in vivo. RESULTS In cell culture, Pulsatilla saponin A significantly inhibited the growth of human hepatocellular carcinoma SMCC-7721 cells and pancreatic BXPC3 and SW1990 cancer cells. Similar inhibitory activities were observed when the compound was tested in mouse xenograft tumor models using human hepatocellular carcinoma Bel-7402 and pancreatic cancer SW1990 cells. In Comet assay and flow cytometric analysis of cell cycle distribution and annexin V expression, DNA damage, G2 arrest, and apoptosis were identified in Pulsatilla saponin A-treated cancer cells. Based on the results of Western blotting, p53 and cyclin B protein levels were higher, whereas Bcl-2 protein levels were lower in Pulsatilla saponin A-treated cancer cells than in vehicle-treated cells. CONCLUSIONS Pulsatilla saponin A may exert its antitumor effect by inducing DNA damage and causing G2 arrest and apoptosis in cancer cells. Pulsatilla saponin A and its derivatives may be developed as a new class of anticancer agents.
Collapse
Affiliation(s)
- Qiang Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Jiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanli Liu
- College of Pharmacy, Medical College of Soochow University, Suzhou, China.
| | - Xiaofei Qi
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Lab Medicine and Pharmacy, Suzhou Health College, Suzhou, China.
| |
Collapse
|
11
|
Yang S, Wang H, Guo Y, Chen S, Zhang MY, Shen J, Yu H, Miao J, Wang HY, Wei W. RMP plays distinct roles in the proliferation of hepatocellular carcinoma cells and normal hepatic cells. Int J Biol Sci 2013; 9:637-48. [PMID: 23847445 PMCID: PMC3708043 DOI: 10.7150/ijbs.6439] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 06/17/2013] [Indexed: 12/25/2022] Open
Abstract
RMP has been shown to function in the transcription regulation through association with RNA polymerase (RNAP) II subunit RPB5. It also has been shown to be required for the proliferation of hepatocellular carcinoma (HCC) cells with an antiapoptotic property. In this article, we further demonstrate that RMP displays distinct features in HCC cells compared with normal hepatic cells. RMP expression is remarkably increased in various cancer cell lines including HCC cells when compared with normal cells. Depletion of RMP could inhibit the proliferation of HCC cells, but not the normal hepatic cells. RMP significantly prevented apoptosis of HCC cells in SMMC-7721 and HepG2, but had little effect on apoptosis in the normal hepatic cells. The mechanisms of RMP's distinct features rely on different responsive expressions of apoptosis factors induced by RMP in HCC and hepatic cells. Either overexpression or depletion of RMP significantly affected the expression of apoptosis factors in HCC cells. However, normal hepatic cells showed a tendency to resist RMP for the regulation of apoptosis. In the clinical samples, the increased expression of RMP in HCCs was also observed when compared with the matched non-tumor tissues from 30 HCC patients. The different expression levels of and distinct responses to RMP between HCC and hepatic cells suggest that RMP might serve as not only a biomarker for the diagnosis of HCC, but also a potential target for the HCC therapy.
Collapse
Affiliation(s)
- Sijun Yang
- Department of Cell Biology, School of Medicine, Soochow University, Suzhou, 215123 China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Padberg I, Janßen S, Meyer TF. Chlamydia trachomatis inhibits telomeric DNA damage signaling via transient hTERT upregulation. Int J Med Microbiol 2013; 303:463-74. [PMID: 23830072 DOI: 10.1016/j.ijmm.2013.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 05/27/2013] [Accepted: 06/02/2013] [Indexed: 01/30/2023] Open
Abstract
Epidemiological data exist to support a positive association between Chlamydia trachomatis (Ctr) infection and gynecological cancers; however, putative cellular mechanisms for this association are lacking. Here, we identified Ctr-induced perturbations to host cell phenotypes in vitro that persisted after clearance of infection and could directly contribute to host cell transformation. In particular, human telomerase catalytic subunit (hTERT) mRNA expression and catalytic subunit activity were increased in acute infected late passage IMR90E1A cells. hTERT upregulation was accompanied by recruitment of ceramide, a known regulator of hTERT, to the chlamydial inclusion and was abrogated following doxycycline-mediated infection clearance. In cells cleared of Ctr infection, average telomere length was slightly increased and immunofluorescence staining of the DNA damage marker γH2A.X was reduced after clearance of infection compared with cells that had not been infected. Reduced p53 binding to the promoter of the cell cycle checkpoint regulator p21 was also detected in cells cleared of infection and p21 levels were reduced; moreover, this cell population exhibited increased resistance to etoposide-induced DNA damage. Thus, Ctr infection altered cell aging and survival pathways, which persisted after infection clearance. Cells that survive infection are likely to exhibit altered physiology, as evidenced by an increased resistance to DNA damage-induced apoptosis, which may support cellular transformation.
Collapse
Affiliation(s)
- Inken Padberg
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany
| | | | | |
Collapse
|
13
|
Cmielova J, Havelek R, Kohlerova R, Soukup T, Bruckova L, Suchanek J, Vavrova J, Mokry J, Rezacova M. The effect of ATM kinase inhibition on the initial response of human dental pulp and periodontal ligament mesenchymal stem cells to ionizing radiation. Int J Radiat Biol 2013; 89:501-11. [PMID: 23425510 DOI: 10.3109/09553002.2013.775530] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
PURPOSE This study evaluates early changes in human mesenchymal stem cells (MSC) isolated from dental pulp and periodontal ligament after γ-irradiation and the effect of ataxia-telangiectasia mutated (ATM) inhibition. METHODS MSC were irradiated with 2 and 20 Gy by (60)Co. For ATM inhibition, specific inhibitor KU55933 was used. DNA damage was measured by Comet assay and γH2AX detection. Cell cycle distribution and proteins responding to DNA damage were analyzed 2-72 h after the irradiation. RESULTS The irradiation of MSC causes an increase in γH2AX; the phosphorylation was ATM-dependent. Irradiation activates ATM kinase, and the level of p53 protein is increased due to its phosphorylation on serine15. While this phosphorylation of p53 is ATM-dependent in MSC, the increase in p53 was not prevented by ATM inhibition. A similar trend was observed for Chk1 and Chk2. The increase in p21 is greater without ATM inhibition. ATM inhibition also does not fully abrogate the accumulation of irradiated MSC in the G2-phase of the cell-cycle. CONCLUSIONS In irradiated MSC, double-strand breaks are tagged quickly by γH2AX in an ATM-dependent manner. Although phosphorylations of p53(ser15), Chk1(ser345) and Chk2(thr68) are ATM-dependent, the overall amount of these proteins increases when ATM is inhibited. In both types of MSC, ATM-independent mechanisms for cell-cycle arrest in the G2-phase are triggered.
Collapse
Affiliation(s)
- Jana Cmielova
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove,Charles University in Prague, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gelam honey protects against gamma-irradiation damage to antioxidant enzymes in human diploid fibroblasts. Molecules 2013; 18:2200-11. [PMID: 23434870 PMCID: PMC6270489 DOI: 10.3390/molecules18022200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/18/2013] [Accepted: 01/22/2013] [Indexed: 01/24/2023] Open
Abstract
The present study was designed to determine the radioprotective effects of Malaysian Gelam honey on gene expression and enzyme activity of superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) of human diploid fibroblasts (HDFs) subjected to gamma-irradiation. Six groups of HDFs were studied: untreated control, irradiated HDFs, Gelam honey-treated HDFs and HDF treated with Gelam honey pre-, during- and post-irradiation. HDFs were treated with 6 mg/mL of sterilized Gelam honey (w/v) for 24 h and exposed to 1 Gray (Gy) of gamma rays at the dose rate of 0.25 Gy/min. Gamma-irradiation was shown to down-regulate SOD1, SOD2, CAT and GPx1 gene expressions (p < 0.05). Conversely, HDFs treated with Gelam honey alone showed up-regulation of all genes studied. Similarly, SOD, CAT and GPx enzyme activities in HDFs decreased with gamma-irradiation and increased when cells were treated with Gelam honey (p < 0.05). Furthermore, of the three different stages of study treatment, pre-treatment with Gelam honey caused up-regulation of SOD1, SOD2 and CAT genes expression and increased the activity of SOD and CAT. As a conclusion, Gelam honey modulates the expression of antioxidant enzymes at gene and protein levels in irradiated HDFs indicating its potential as a radioprotectant agent.
Collapse
|
15
|
Alekseenko LL, Zemelko VI, Zenin VV, Pugovkina NA, Kozhukharova IV, Kovaleva ZV, Grinchuk TM, Fridlyanskaya II, Nikolsky NN. Heat shock induces apoptosis in human embryonic stem cells but a premature senescence phenotype in their differentiated progeny. Cell Cycle 2012; 11:3260-9. [PMID: 22895173 DOI: 10.4161/cc.21595] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Embryonic stem cells (ESC) are able to self-renew and to differentiate into any cell type. To escape error transmission to future cell progeny, ESC require robust mechanisms to ensure genomic stability. It was stated that stress defense of mouse and human ESC against oxidative stress and irradiation is superior compared with differentiated cells. Here, we investigated heat shock response of human ESC (hESC) and their differentiated progeny. Fibroblast-like cells were generated by spontaneous hESC differentiation via embryoid bodies. Like normal human diploid fibroblasts, these cells have a finite lifespan in culture, undergo replicative senescence and die. We found that sublethal heat shock affected survival of both cell types, but in hESC it induced apoptosis, whereas in differentiated cells it produced cell cycle arrest and premature senescence phenotype. Heat shock survived hESC and differentiated cells restored the properties of initial cells. Heated hESC progeny exhibited pluripotent markers and the capacity to differentiate into the cells of three germ layers. Fibroblast-like cells resisted heat shock, proliferated for a limited number of passages and entered replicative senescence as unheated parental cells. Taken together, these results show for the first time that both hESC and their differentiated derivatives are sensitive to heat shock, but the mechanisms of their stress response are different: hESC undergo apoptosis, whereas differentiated cells under the same conditions exhibit stress-induced premature senescence (SIPS) phenotype. Both cell types that survived sublethal heat shock sustain parental cell properties.
Collapse
|
16
|
Kumar MG, Patel NM, Nicholson AM, Kalen AL, Sarsour EH, Goswami PC. Reactive oxygen species mediate microRNA-302 regulation of AT-rich interacting domain 4a and C-C motif ligand 5 expression during transitions between quiescence and proliferation. Free Radic Biol Med 2012; 53:974-82. [PMID: 22732186 PMCID: PMC3418417 DOI: 10.1016/j.freeradbiomed.2012.06.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 06/12/2012] [Accepted: 06/14/2012] [Indexed: 01/17/2023]
Abstract
Normal cell growth consists of two distinct phases, quiescence and proliferation. Quiescence, or G(0), is a reversible growth arrest in which cells retain the ability to reenter the proliferative cycle (G(1), S, G(2), and M). Although not actively dividing, quiescent cells are metabolically active and quiescence is actively maintained. Our results from microRNA PCR arrays and Taqman PCR assays showed a significant decrease (4-fold) in miR-302 levels during quiescence compared to proliferating normal human fibroblasts, suggesting that miR-302 could regulate cellular proliferation. Results from a Q-RT-PCR and dual-luciferase-3'-UTR reporter assays identified ARID4a (AT-rich interacting domain 4a, also known as RBP1) and CCL5 (C-C motif ligand 5) as targets for miR-302. Ionizing radiation decreased miR-302 levels, which was associated with an increase in its target mRNA levels, ARID4a and CCL5. Such an inverse correlation was also observed in cells treated with hydrogen peroxide as well as SOD2-overexpressing cells. Overexpression of miR-302 suppresses ARID4a and CCL5 mRNA levels, and increased the percentage of S-phase cells. These results identified miR-302 as an ROS-sensitive regulator of ARID4a and CCL5 mRNAs as well as demonstrate a regulatory role of miR-302 during quiescence and proliferation.
Collapse
Affiliation(s)
- Maneesh G. Kumar
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Neil M. Patel
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Adam M. Nicholson
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Amanda L. Kalen
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Ehab H. Sarsour
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Prabhat C. Goswami
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
17
|
Abstract
EHD {EH [Eps15 (epidermal growth factor receptor substrate 15) homology]-domain-containing} proteins participate in several endocytic events, such as the internalization and the recycling processes. There are four EHD proteins in mammalian cells, EHD1–EHD4, each with diverse roles in the recycling pathway of endocytosis. EHD2 is a plasma-membrane-associated member of the EHD family that regulates internalization. Since several endocytic proteins have been shown to undergo nucleocytoplasmic shuttling and have been assigned roles in regulation of gene expression, we tested the possibility that EHD proteins also shuttle to the nucleus. Our results showed that, among the three EHD proteins (EHD1–EHD3) that were tested, only EHD2 accumulates in the nucleus under nuclear export inhibition treatment. Moreover, the presence of a NLS (nuclear localization signal) was essential for its entry into the nucleus. Nuclear exit of EHD2 depended partially on its NES (nuclear export signal). Elimination of a potential SUMOylation site in EHD2 resulted in a major accumulation of the protein in the nucleus, indicating the involvement of SUMOylation in the nuclear exit of EHD2. We confirmed the SUMOylation of EHD2 by employing co-immunoprecipitation and the yeast two-hybrid system. Using GAL4-based transactivation assay as well as a KLF7 (Krüppel-like factor 7)-dependent transcription assay of the p21WAF1/Cip1 [CDKN1A (cyclin-dependent kinase inhibitor 1A)] gene, we showed that EHD2 represses transcription. qRT-PCR (quantitative real-time PCR) of RNA from cells overexpressing EHD2 or of RNA from cells knocked down for EHD2 confirmed that EHD2 represses transcription of the p21WAF1/Cip1 gene.
Collapse
|
18
|
Li L, Yue GGL, Lau CBS, Sun H, Fung KP, Leung PC, Han Q, Leung PS. Eriocalyxin B induces apoptosis and cell cycle arrest in pancreatic adenocarcinoma cells through caspase- and p53-dependent pathways. Toxicol Appl Pharmacol 2012; 262:80-90. [PMID: 22561874 DOI: 10.1016/j.taap.2012.04.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/11/2012] [Accepted: 04/17/2012] [Indexed: 01/06/2023]
Abstract
Pancreatic cancer is difficult to detect early and responds poorly to chemotherapy. A breakthrough in the development of new therapeutic agents is urgently needed. Eriocalyxin B (EriB), isolated from the Isodon eriocalyx plant, is an ent-kaurane diterpenoid with promise as a broad-spectrum anti-cancer agent. The anti-leukemic activity of EriB, including the underlying mechanisms involved, has been particularly well documented. In this study, we demonstrated for the first time EriB's potent cytotoxicity against four pancreatic adenocarcinoma cell lines, namely PANC-1, SW1990, CAPAN-1, and CAPAN-2. The effects were comparable to that of the chemotherapeutic camptothecin (CAM), but with much lower toxicity against normal human liver WRL68 cells. EriB's cytoxicity against CAPAN-2 cells was found to involve caspase-dependent apoptosis and cell cycle arrest at the G2/M phase. Moreover, the p53 pathway was found to be activated by EriB in these cells. Furthermore, in vivo studies showed that EriB inhibited the growth of human pancreatic tumor xenografts in BALB/c nude mice without significant secondary adverse effects. These results suggest that EriB should be considered a candidate for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Lin Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Cmielová J, Rezáčová M. p21Cip1/Waf1 protein and its function based on a subcellular localization [corrected]. J Cell Biochem 2012; 112:3502-6. [PMID: 21815189 DOI: 10.1002/jcb.23296] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protein p21(Cip1/Waf1) is a cyclin-dependent kinase inhibitor, which is important in the response of cells to genotoxic stress and a major transcriptional target of p53 protein. Based on the localization, p21(Cip1/Waf1) protein executes various functions in the cell. In the nucleus p21(Cip1/Waf1) binds to and inhibits the activity of cyclin dependent kinases Cdk1 and Cdk2 and blocks the transition from G1 phase into S phase or from G2 phase into mitosis after DNA damage. This enables the repair of damaged DNA. p21(Cip1/Waf1) was also found as an important protein for the induction of replication senescence as well as stress-induced premature senescence. In the cytoplasm, p21(Cip1/Waf1) protein has an anti-apoptotic effect. It is able to bind to and inhibit caspase 3, as well as the apoptotic kinases ASK1 and JNK. The function of p21(Cip1/Waf1) in response to a DNA damage probably depends on the extent of the damage. In the case of low-level DNA damage, the expression of p21(Cip1/Waf1) is increased, it induces cell cycle arrest, and performs also anti-apoptotic activities. However, after extensive DNA damage the amount of p21(Cip1/Waf1) protein is decreased and the cell undergoes apoptosis. Dual function of p21(Cip1/Waf1) was also observed in cancerogenesis. On the one hand, p21(Cip1/Waf1) acts as a tumor suppressor; on the other hand it prevents apoptosis and acts as an oncogene. Better understanding of the role of p21(Cip1/Waf1) in various conditions would help to develop better cancer-treatment strategies.
Collapse
Affiliation(s)
- Jana Cmielová
- Department of Medical Biochemistry, Charles University in Prague, Medical Faculty in Hradec Králové, Šimkova 870, 500 38 Hradec Králové 1, Czech Republic.
| | | |
Collapse
|
20
|
Pathologic characteristics of pediatric intracranial pilocytic astrocytomas and their impact on outcome in 3 countries: a multi-institutional study. Am J Surg Pathol 2012; 36:43-55. [PMID: 21989351 DOI: 10.1097/pas.0b013e3182329480] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pilocytic astrocytoma (PA) is one of the most common glial neoplasms in the pediatric population, and its gross total resection can be curative. Treatment of partially resected or recurrent tumors is challenging, and the factors associated with increased recurrence risk are not well defined. Identification of favorable and unfavorable factors can lead to a better understanding and management of patients with PA. We studied the pathologic characteristics of 116 intracranial PAs from 4 institutions representing 3 distinct geographic regions to identify factors that may be associated with biological behavior. The study included 65 boys and 51 girls with a median age of 6 years. Median follow-up for all patients was 101 months, during which time 38 patients had recurrence. Progression-free and overall survivals were better in patients who underwent gross total resection. We were not able to identify any differences in pathologic and molecular markers among the 4 institutions from 3 different countries. However, progression-free survival varied significantly among institutions. Sox-2 was the most prevalent stem cell marker in PA, and many tumors showed synaptophysin positivity. BRAF immunostaining was not useful in determining BRAF duplication. BRAF duplication was more typical of posterior fossa tumors. There was a strong correlation between BRAF duplication and pERK immunostaining, suggesting that the RAF/MEK/ERK pathway is active in these tumors. This finding has significant implications given its role in oncogen-induced senescence and possible influence on treatment decisions of subtotally resected tumors.
Collapse
|