1001
|
Keta O, Bulat T, Golić I, Incerti S, Korać A, Petrović I, Ristić-Fira A. The impact of autophagy on cell death modalities in CRL-5876 lung adenocarcinoma cells after their exposure to γ-rays and/or erlotinib. Cell Biol Toxicol 2016; 32:83-101. [PMID: 27026538 DOI: 10.1007/s10565-016-9319-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/21/2016] [Indexed: 01/27/2023]
Abstract
In most patients with lung cancer radiation treatment is used either as single agent or in combination with radiosensitizing drugs. However, the mechanisms underlying combined therapy and its impact on different modes of cell death have not yet been fully elucidated. We aimed to examine effects of single and combined treatments with γ-rays and erlotinib on radioresistant CRL-5876 human lung adenocarcinoma cells with particular emphasis on cell death. CRL-5876 cells were treated with γ-rays and/or erlotinib and changes in cell cycle, DNA repair dynamics, ultrastructure, nuclear morphology and protein expression were monitored at different time points. To reveal the relationship between types of cell death that arise after these treatments, autophagy was blocked with chloroquine. We found that higher dose of γ-rays causes G2/M arrest while adding of erlotinib to this treatment decreases the number of cells in S phase. Impact of erlotinib on kinetics of disappearance of irradiation-induced DNA double strand breaks is reflected in the increase of residual γ-H2AX foci after 24 h. γ-rays provoke cytoprotective autophagy which precedes development of senescence. Erlotinib predominantly induces apoptosis and enlarges the number of apoptotic cells in the irradiated CRL-5876 cells. Chloroquine improved cytotoxicity induced by radiation and erlotinib, increased apoptosis and decreased senescence in the CRL-5876 cells. The results obtained on CRL-5876 cells indicate significant radiosensitizing effect of erlotinib and suggest that chloroquine in the combination with the above treatments may have an additional antitumor effect in lung adenocarcinoma.
Collapse
Affiliation(s)
- Otilija Keta
- Vinča Institute of Nuclear Sciences, University of Belgrade, PO Box 522, 11001, Belgrade, Serbia
| | - Tanja Bulat
- Vinča Institute of Nuclear Sciences, University of Belgrade, PO Box 522, 11001, Belgrade, Serbia
| | - Igor Golić
- Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Sebastien Incerti
- CNRS/IN2P3, Centre d'Etudes Nucléaires de Bordeaux Gradignan, CENBG, Université Bordeaux 1, 33175, Gradignan, France
| | | | - Ivan Petrović
- Vinča Institute of Nuclear Sciences, University of Belgrade, PO Box 522, 11001, Belgrade, Serbia
| | - Aleksandra Ristić-Fira
- Vinča Institute of Nuclear Sciences, University of Belgrade, PO Box 522, 11001, Belgrade, Serbia.
| |
Collapse
|
1002
|
Shi W, Tang MK, Yao Y, Tang C, Chui YL, Lee KKH. BRE plays an essential role in preventing replicative and DNA damage-induced premature senescence. Sci Rep 2016; 6:23506. [PMID: 27001068 PMCID: PMC4802329 DOI: 10.1038/srep23506] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/08/2016] [Indexed: 11/20/2022] Open
Abstract
The BRE gene, alias BRCC45, produces a 44 kDa protein that is normally distributed in both cytoplasm and nucleus. In this study, we used adult fibroblasts isolated from wild-type (WT) and BRE knockout (BRE−/−) mice to investigate the functional role of BRE in DNA repair and cellular senescence. We compared WT with BRE−/− fibroblasts at different cell passages and observed that the mutant fibroblasts entered replicative senescence earlier than the WT fibroblasts. With the use of gamma irradiation to induce DNA damage in fibroblasts, the percentage of SA-β-Gal+ cells was significantly higher in BRE−/− fibroblasts compared with WT cells, suggesting that BRE is also associated with DNA damage-induced premature senescence. We also demonstrated that the gamma irradiation induced γ-H2AX foci, a DNA damage marker, persisted significantly longer in BRE−/− fibroblasts than in WT fibroblasts, confirming that the DNA repair process is impaired in the absence of BRE. In addition, the BRCA1-A complex recruitment and homologous recombination (HR)-dependent DNA repair process upon DNA damage were impaired in BRE−/− fibroblasts. Taken together, our results demonstrate a role for BRE in both replicative senescence and DNA damage-induced premature senescence. This can be attributed to BRE being required for BRCA1-A complex-driven HR DNA repair.
Collapse
Affiliation(s)
- Wenting Shi
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Mei Kuen Tang
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yao Yao
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Chengcheng Tang
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yiu Loon Chui
- Department of Chemical Pathology, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Kenneth Ka Ho Lee
- Stem Cell and Regeneration Thematic Research Programme, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
1003
|
Tyagi A, Vishnoi K, Mahata S, Verma G, Srivastava Y, Masaldan S, Roy BG, Bharti AC, Das BC. Cervical Cancer Stem Cells Selectively Overexpress HPV Oncoprotein E6 that Controls Stemness and Self-Renewal through Upregulation of HES1. Clin Cancer Res 2016; 22:4170-84. [PMID: 26988248 DOI: 10.1158/1078-0432.ccr-15-2574] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/19/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Perturbation of keratinocyte differentiation by E6/E7 oncoproteins of high-risk human papillomaviruses that drive oncogenic transformation of cells in squamocolumnar junction of the uterine cervix may confer "stem-cell like" characteristics. However, the crosstalk between E6/E7 and stem cell signaling during cervical carcinogenesis is not well understood. We therefore examined the role of viral oncoproteins in stem cell signaling and maintenance of stemness in cervical cancer. EXPERIMENTAL DESIGN Isolation and enrichment of cervical cancer stem-like cells (CaCxSLCs) was done from cervical primary tumors and cancer cell lines by novel sequential gating using a set of functional and phenotypic markers (ABCG2, CD49f, CD71, CD133) in defined conditioned media for assessing sphere formation and expression of self-renewal and stemness markers by FACS, confocal microscopy, and qRT-PCR. Differential expression level and DNA-binding activity of Notch1 and its downstream targets in CaCxSLCs as well as silencing of HPVE6/Hes1 by siRNA was evaluated by gel retardation assay, FACS, immunoblotting, and qRT-PCR followed by in silico and in vivo xenograft analysis. RESULTS CaCxSLCs showed spheroid-forming ability, expressed self-renewal and stemness markers Oct4, Sox2, Nanog, Lrig1, and CD133, and selectively overexpressed E6 and HES1 transcripts in both cervical primary tumors and cancer cell lines. The enriched CaCxSLCs were highly tumorigenic and did recapitulate primary tumor histology in nude mice. siRNA silencing of HPVE6 or Hes1 abolished sphere formation, downregulated AP-1-STAT3 signaling, and induced redifferentiation. CONCLUSIONS Our findings suggest the possible mechanism by which HPVE6 potentially regulate and maintain stem-like cancer cells through Hes1. Clin Cancer Res; 22(16); 4170-84. ©2016 AACR.
Collapse
Affiliation(s)
- Abhishek Tyagi
- Stem Cell and Cancer Research Laboratory, Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Campus, Noida, Uttar Pradesh, India. Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India. Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | - Kanchan Vishnoi
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Sutapa Mahata
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Gaurav Verma
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Yogesh Srivastava
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Shashank Masaldan
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Bal Gangadhar Roy
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi, India
| | - Alok C Bharti
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India. Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India.
| | - Bhudev C Das
- Stem Cell and Cancer Research Laboratory, Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Campus, Noida, Uttar Pradesh, India. Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India.
| |
Collapse
|
1004
|
Helman A, Klochendler A, Azazmeh N, Gabai Y, Horwitz E, Anzi S, Swisa A, Condiotti R, Granit RZ, Nevo Y, Fixler Y, Shreibman D, Zamir A, Tornovsky-Babeay S, Dai C, Glaser B, Powers AC, Shapiro AMJ, Magnuson MA, Dor Y, Ben-Porath I. p16(Ink4a)-induced senescence of pancreatic beta cells enhances insulin secretion. Nat Med 2016; 22:412-20. [PMID: 26950362 DOI: 10.1038/nm.4054] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/02/2016] [Indexed: 12/15/2022]
Abstract
Cellular senescence is thought to contribute to age-associated deterioration of tissue physiology. The senescence effector p16(Ink4a) is expressed in pancreatic beta cells during aging and limits their proliferative potential; however, its effects on beta cell function are poorly characterized. We found that beta cell-specific activation of p16(Ink4a) in transgenic mice enhances glucose-stimulated insulin secretion (GSIS). In mice with diabetes, this leads to improved glucose homeostasis, providing an unexpected functional benefit. Expression of p16(Ink4a) in beta cells induces hallmarks of senescence--including cell enlargement, and greater glucose uptake and mitochondrial activity--which promote increased insulin secretion. GSIS increases during the normal aging of mice and is driven by elevated p16(Ink4a) activity. We found that islets from human adults contain p16(Ink4a)-expressing senescent beta cells and that senescence induced by p16(Ink4a) in a human beta cell line increases insulin secretion in a manner dependent, in part, on the activity of the mechanistic target of rapamycin (mTOR) and the peroxisome proliferator-activated receptor (PPAR)-γ proteins. Our findings reveal a novel role for p16(Ink4a) and cellular senescence in promoting insulin secretion by beta cells and in regulating normal functional tissue maturation with age.
Collapse
Affiliation(s)
- Aharon Helman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Agnes Klochendler
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Narmen Azazmeh
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yael Gabai
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Elad Horwitz
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shira Anzi
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Avital Swisa
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Reba Condiotti
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Roy Z Granit
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yuval Nevo
- Computation Center, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yaakov Fixler
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Dorin Shreibman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Amit Zamir
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Sharona Tornovsky-Babeay
- Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Chunhua Dai
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alvin C Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Veteran Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada.,Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | - Mark A Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.,Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ittai Ben-Porath
- Department of Developmental Biology and Cancer Research, Institute for Medical Research-Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
1005
|
Durbin KR, Fornelli L, Fellers RT, Doubleday PF, Narita M, Kelleher NL. Quantitation and Identification of Thousands of Human Proteoforms below 30 kDa. J Proteome Res 2016; 15:976-82. [PMID: 26795204 PMCID: PMC4794255 DOI: 10.1021/acs.jproteome.5b00997] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Top-down proteomics is capable of identifying and quantitating unique proteoforms through the analysis of intact proteins. We extended the coverage of the label-free technique, achieving differential analysis of whole proteins <30 kDa from the proteomes of growing and senescent human fibroblasts. By integrating improved control software with more instrument time allocated for quantitation of intact ions, we were able to collect protein data between the two cell states, confidently comparing 1577 proteoform levels. To then identify and characterize proteoforms, our advanced acquisition software, named Autopilot, employed enhanced identification efficiency in identifying 1180 unique Swiss-Prot accession numbers at 1% false-discovery rate. This coverage of the low mass proteome is equivalent to the largest previously reported but was accomplished in 23% of the total acquisition time. By maximizing both the number of quantified proteoforms and their identification rate in an integrated software environment, this work significantly advances proteoform-resolved analyses of complex systems.
Collapse
Affiliation(s)
- Kenneth R. Durbin
- Departments of Chemistry and Molecular Biosciences, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| | - Luca Fornelli
- Departments of Chemistry and Molecular Biosciences, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| | - Ryan T. Fellers
- Departments of Chemistry and Molecular Biosciences, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| | - Peter F. Doubleday
- Departments of Chemistry and Molecular Biosciences, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Robinson Way, Cambridge CB2 0RE, U.K
| | - Neil L. Kelleher
- Departments of Chemistry and Molecular Biosciences, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| |
Collapse
|
1006
|
Ercilla A, Llopis A, Feu S, Aranda S, Ernfors P, Freire R, Agell N. New origin firing is inhibited by APC/CCdh1 activation in S-phase after severe replication stress. Nucleic Acids Res 2016; 44:4745-62. [PMID: 26939887 PMCID: PMC4889930 DOI: 10.1093/nar/gkw132] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/23/2016] [Indexed: 01/28/2023] Open
Abstract
Defects in DNA replication and repair are known to promote genomic instability, a hallmark of cancer cells. Thus, eukaryotic cells have developed complex mechanisms to ensure accurate duplication of their genomes. While DNA damage response has been extensively studied in tumour cells, the pathways implicated in the response to replication stress are less well understood especially in non-transformed cells. Here we show that in non-transformed cells, APC/C(Cdh1) is activated upon severe replication stress. Activation of APC/C(Cdh1) prevents new origin firing and induces permanent arrest in S-phase. Moreover, Rad51-mediated homologous recombination is also impaired under these conditions. APC/C(Cdh1) activation in S-phase occurs after replication forks have been processed into double strand breaks. Remarkably, this activation, which correlates with decreased Emi1 levels, is not prevented by ATR/ATM inhibition, but it is abrogated in cells depleted of p53 or p21. Importantly, we found that the lack of APC/C(Cdh1) activity correlated with an increase in genomic instability. Taken together, our results define a new APC/C(Cdh1) function that prevents cell cycle resumption after prolonged replication stress by inhibiting origin firing, which may act as an additional mechanism in safeguarding genome integrity.
Collapse
Affiliation(s)
- Amaia Ercilla
- Departament de Biologia Cellular, Immunologia i Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, C/ Casanova 143, 08036 Barcelona, Spain
| | - Alba Llopis
- Departament de Biologia Cellular, Immunologia i Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, C/ Casanova 143, 08036 Barcelona, Spain
| | - Sonia Feu
- Departament de Biologia Cellular, Immunologia i Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, C/ Casanova 143, 08036 Barcelona, Spain
| | - Sergi Aranda
- Center for Genomic Regulation (CRG), C/ Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Patrik Ernfors
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologias Biomedicas, 38320 Tenerife, Spain
| | - Neus Agell
- Departament de Biologia Cellular, Immunologia i Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Facultat de Medicina, Universitat de Barcelona, C/ Casanova 143, 08036 Barcelona, Spain
| |
Collapse
|
1007
|
Manojlovic-Gacic E, Skender-Gazibara M, Popovic V, Soldatovic I, Boricic N, Raicevic S, Pekic S, Doknic M, Miljic D, Alafuzoff I, Pontén F, Casar-Borota O. Oncogene-Induced Senescence in Pituitary Adenomas--an Immunohistochemical Study. Endocr Pathol 2016; 27:1-11. [PMID: 26573928 DOI: 10.1007/s12022-015-9405-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Oncogene-induced senescence (OIS) serves as an initial barrier to cancer development, being proposed as a possible explanation for the usually benign behavior of the pituitary adenomas. We aimed to explore the immunohistochemical expression of the OIS markers, senescence-associated lysosomal β-galactosidase (SA-β-GAL), p16, and p21 in different types of 345 pituitary adenomas and compared it with the expression in the normal pituitary and in the specimens from the repeated surgeries. SA-β-GAL was overexpressed in the pituitary adenomas, compared to the normal pituitaries. Growth hormone (GH) producing adenomas showed the strongest SA-β-GAL, with densely granulated (DG)-GH adenomas more reactive than the sparsely granulated (SG). Nuclear p21 was decreased in the adenomas, except for the SG-GH adenomas that had higher p21 than the normal pituitaries and the other adenomas. p16 was significantly lower in the adenomas, without type-related differences. SA-β-GAL was slightly lower and p16 slightly higher in the recurrences. Our findings indicate alterations of the senescence program in the different types of pituitary adenomas. Activation of senescence in the pituitary adenomas presents one possible explanation for their usually benign behavior, at least in the GH adenomas that show a synchronous increase of two OIS markers. However, subdivision into GH adenoma subtypes reveals differences that reflect complex regulatory mechanisms influenced by the interplay between the granularity pattern and the hormonal factors, with possible impact on the different clinical behavior of the SG- and DG-GH adenoma subtypes. p16 seems to have a more prominent role in the pituitary tumorigenesis than in the senescence. Recurrent growth in a subset of the pituitary adenomas is not associated with consistent changes in the senescence pattern.
Collapse
Affiliation(s)
- Emilija Manojlovic-Gacic
- Institute of Pathology, Medical Faculty, University of Belgrade, Dr Subotica 1, Belgrade, Serbia.
| | - Milica Skender-Gazibara
- Institute of Pathology, Medical Faculty, University of Belgrade, Dr Subotica 1, Belgrade, Serbia
| | - Vera Popovic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Ivan Soldatovic
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Novica Boricic
- Institute of Pathology, Medical Faculty, University of Belgrade, Dr Subotica 1, Belgrade, Serbia
| | - Savo Raicevic
- Neurosurgery Clinic, Clinical Center of Serbia, Koste Todorovica 4, Belgrade, Serbia
| | - Sandra Pekic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Mirjana Doknic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Dragana Miljic
- Clinic of Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia, Dr Subotica 13, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Dr Subotica 8, Belgrade, Serbia
| | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
- Department of Clinical Pathology and Cytology, Uppsala University Hospital, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
| | - Olivera Casar-Borota
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
- Department of Clinical Pathology and Cytology, Uppsala University Hospital, Rudbeck Laboratory, Dag Hammarskjölds väg 20, Uppsala, Sweden
| |
Collapse
|
1008
|
Khor ES, Noor SM, Wong PF. Expression of zTOR-associated microRNAs in zebrafish embryo treated with rapamycin. Life Sci 2016; 150:67-75. [PMID: 26916825 DOI: 10.1016/j.lfs.2016.02.076] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/17/2016] [Accepted: 02/21/2016] [Indexed: 01/24/2023]
Abstract
AIMS MicroRNAs (miRNAs) are vital in modulating lifespan and various biological processes including vascular function. The pivotal roles of mammalian target of rapamycin (mTOR) in regulating senescence and angiogenesis have been extensively described. However, the roles of its orthologue, zebrafish target of rapamycin (zTOR) in senescence and angiogenesis remain to be unravelled. In the present study, we aimed to investigate the role of zTOR and identify miRNAs associated with senescence and angiogenesis. MAIN METHODS Zebrafish embryos were treated with rapamycin and the inhibition of zTOR and its downstream proteins were validated by immunoblotting. Following the treatment, melanocyte density was quantitated, and senescence and angiogenic responses were determined by senescence-associated beta-galactosidase (SA-β-gal) and endogenous alkaline phosphatase (ALP) staining, respectively. Relative expression of microRNAs were determined by quantitative RT-PCR. KEY FINDINGS Rapamycin (400 nM) suppressed zTOR pathway by down-regulating the phosphorylation of zTOR-associated proteins such as P70S6K and S6K at both 4h post-fertilisation (hpf) and 8hpf while 4E-BP1 was only down-regulated at 8hpf when compared to their respective vehicle controls. Treatment with rapamycin also resulted in significant suppression of melanocyte development and senescence-associated beta-galactosidase (SA-β-gal) activity, and perturbed the development of intersegmental vessels (ISVs) of zebrafish embryos. In addition, the expressions of dre-miR-9-5p and -3p, dre-miR-25-3p and dre-miR-124-3p were significantly up-regulated in embryos treated with rapamycin from 4hpf. SIGNIFICANCE Our findings suggest the involvement of zTOR in embryonic senescence and angiogenesis which could be potentially mediated by selected miRNAs.
Collapse
Affiliation(s)
- Eng-Soon Khor
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Suzita Mohd Noor
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
1009
|
Pan H, Guan D, Liu X, Li J, Wang L, Wu J, Zhou J, Zhang W, Ren R, Zhang W, Li Y, Yang J, Hao Y, Yuan T, Yuan G, Wang H, Ju Z, Mao Z, Li J, Qu J, Tang F, Liu GH. SIRT6 safeguards human mesenchymal stem cells from oxidative stress by coactivating NRF2. Cell Res 2016; 26:190-205. [PMID: 26768768 PMCID: PMC4746611 DOI: 10.1038/cr.2016.4] [Citation(s) in RCA: 259] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/15/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023] Open
Abstract
SIRT6 belongs to the mammalian homologs of Sir2 histone NAD(+)-dependent deacylase family. In rodents, SIRT6 deficiency leads to aging-associated degeneration of mesodermal tissues. It remains unknown whether human SIRT6 has a direct role in maintaining the homeostasis of mesodermal tissues. To this end, we generated SIRT6 knockout human mesenchymal stem cells (hMSCs) by targeted gene editing. SIRT6-deficient hMSCs exhibited accelerated functional decay, a feature distinct from typical premature cellular senescence. Rather than compromised chromosomal stability, SIRT6-null hMSCs were predominately characterized by dysregulated redox metabolism and increased sensitivity to the oxidative stress. In addition, we found SIRT6 in a protein complex with both nuclear factor erythroid 2-related factor 2 (NRF2) and RNA polymerase II, which was required for the transactivation of NRF2-regulated antioxidant genes, including heme oxygenase 1 (HO-1). Overexpression of HO-1 in SIRT6-null hMSCs rescued premature cellular attrition. Our study uncovers a novel function of SIRT6 in maintaining hMSC homeostasis by serving as a NRF2 coactivator, which represents a new layer of regulation of oxidative stress-associated stem cell decay.
Collapse
Affiliation(s)
- Huize Pan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Guan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaomeng Liu
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Jingyi Li
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Lixia Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Junzhi Zhou
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Weizhou Zhang
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ruotong Ren
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
| | - Weiqi Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
| | - Ying Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiping Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Hao
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Tingting Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Guohong Yuan
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hu Wang
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 310036, China
| | - Zhenyu Ju
- Institute of Aging Research, Leibniz Link Partner Group on Stem Cell Aging, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 310036, China
| | - Zhiyong Mao
- School of life sciences and technology, Tongji University, Shanghai 200092, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing 100730, China
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Center for Molecular and Translational Medicine, CMTM, Beijing 100101, China
| | - Guang-Hui Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- FSU-CAS Innovation Institute, Foshan University, Foshan, Guangdong 528000, China
- Center for Molecular and Translational Medicine, CMTM, Beijing 100101, China
- Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
1010
|
Martínez I, García-Carpizo V, Guijarro T, García-Gomez A, Navarro D, Aranda A, Zambrano A. Induction of DNA double-strand breaks and cellular senescence by human respiratory syncytial virus. Virulence 2016; 7:427-42. [PMID: 26809688 DOI: 10.1080/21505594.2016.1144001] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human respiratory syncytial virus (HRSV) accounts for the majority of lower respiratory tract infections during infancy and childhood and is associated with significant morbidity and mortality. HRSV provokes a proliferation arrest and characteristic syncytia in cellular systems such as immortalized epithelial cells. We show here that HRSV induces the expression of DNA damage markers and proliferation arrest such as P-TP53, P-ATM, CDKN1A and γH2AFX in cultured cells secondary to the production of mitochondrial reactive oxygen species (ROS). The DNA damage foci contained γH2AFX and TP53BP1, indicative of double-strand breaks (DSBs) and could be reversed by antioxidant treatments such as N-Acetylcysteine (NAC) or reduced glutathione ethyl ester (GSHee). The damage observed is associated with the accumulation of senescent cells, displaying a canonical senescent phenotype in both mononuclear cells and syncytia. In addition, we show signs of DNA damage and aging such as γH2AFX and CDKN2A expression in the respiratory epithelia of infected mice long after viral clearance. Altogether, these results show that HRSV triggers a DNA damage-mediated cellular senescence program probably mediated by oxidative stress. The results also suggest that this program might contribute to the physiopathology of the infection, tissue remodeling and aging, and might be associated to long-term consequences of HRSV infections.
Collapse
Affiliation(s)
- Isidoro Martínez
- a Department of Molecular Pathology , Spanish National Center for Microbiology, Institute of Health Carlos III , Madrid , Spain
| | - Verónica García-Carpizo
- b Department of Endocrine Physiopathology and Nervous System , Institute for Biomedical Research (IIBM), CSIC-UAM , Madrid , Spain
| | - Trinidad Guijarro
- a Department of Molecular Pathology , Spanish National Center for Microbiology, Institute of Health Carlos III , Madrid , Spain
| | - Ana García-Gomez
- a Department of Molecular Pathology , Spanish National Center for Microbiology, Institute of Health Carlos III , Madrid , Spain
| | - Diego Navarro
- b Department of Endocrine Physiopathology and Nervous System , Institute for Biomedical Research (IIBM), CSIC-UAM , Madrid , Spain
| | - Ana Aranda
- b Department of Endocrine Physiopathology and Nervous System , Institute for Biomedical Research (IIBM), CSIC-UAM , Madrid , Spain
| | - Alberto Zambrano
- a Department of Molecular Pathology , Spanish National Center for Microbiology, Institute of Health Carlos III , Madrid , Spain
| |
Collapse
|
1011
|
Kümper S, Mardakheh FK, McCarthy A, Yeo M, Stamp GW, Paul A, Worboys J, Sadok A, Jørgensen C, Guichard S, Marshall CJ. Rho-associated kinase (ROCK) function is essential for cell cycle progression, senescence and tumorigenesis. eLife 2016; 5:e12994. [PMID: 26765561 PMCID: PMC4798951 DOI: 10.7554/elife.12203] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/13/2016] [Indexed: 12/12/2022] Open
Abstract
Rho-associated kinases 1 and 2 (ROCK1/2) are Rho-GTPase effectors that control key aspects of the actin cytoskeleton, but their role in proliferation and cancer initiation or progression is not known. Here, we provide evidence that ROCK1 and ROCK2 act redundantly to maintain actomyosin contractility and cell proliferation and that their loss leads to cell-cycle arrest and cellular senescence. This phenotype arises from down-regulation of the essential cell-cycle proteins CyclinA, CKS1 and CDK1. Accordingly, while the loss of either Rock1 or Rock2 had no negative impact on tumorigenesis in mouse models of non-small cell lung cancer and melanoma, loss of both blocked tumor formation, as no tumors arise in which both Rock1 and Rock2 have been genetically deleted. Our results reveal an indispensable role for ROCK, yet redundant role for isoforms 1 and 2, in cell cycle progression and tumorigenesis, possibly through the maintenance of cellular contractility.
Collapse
Affiliation(s)
- Sandra Kümper
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Faraz K Mardakheh
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Afshan McCarthy
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Maggie Yeo
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Gordon W Stamp
- Experimental Pathology Laboratory, Cancer Research UK London Research Institute, London, United Kingdom
| | - Angela Paul
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Jonathan Worboys
- Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Amine Sadok
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | - Claus Jørgensen
- Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Sabrina Guichard
- Division of Cancer Biology, Institute of Cancer Research, London, United Kingdom
| | | |
Collapse
|
1012
|
Bai G, Smolka MB, Schimenti JC. Chronic DNA Replication Stress Reduces Replicative Lifespan of Cells by TRP53-Dependent, microRNA-Assisted MCM2-7 Downregulation. PLoS Genet 2016; 12:e1005787. [PMID: 26765334 PMCID: PMC4713100 DOI: 10.1371/journal.pgen.1005787] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/15/2015] [Indexed: 11/18/2022] Open
Abstract
Circumstances that compromise efficient DNA replication, such as disruptions to replication fork progression, cause a state known as DNA replication stress (RS). Whereas normally proliferating cells experience low levels of RS, excessive RS from intrinsic or extrinsic sources can trigger cell cycle arrest and senescence. Here, we report that a key driver of RS-induced senescence is active downregulation of the Minichromosome Maintenance 2–7 (MCM2-7) factors that are essential for replication origin licensing and which constitute the replicative helicase core. Proliferating cells produce high levels of MCM2-7 that enable formation of dormant origins that can be activated in response to acute, experimentally-induced RS. However, little is known about how physiological RS levels impact MCM2-7 regulation. We found that chronic exposure of primary mouse embryonic fibroblasts (MEFs) to either genetically-encoded or environmentally-induced RS triggered gradual MCM2-7 repression, followed by inhibition of replication and senescence that could be accelerated by MCM hemizygosity. The MCM2-7 reduction in response to RS is TRP53-dependent, and involves a group of Trp53-dependent miRNAs, including the miR-34 family, that repress MCM expression in replication-stressed cells before they undergo terminal cell cycle arrest. miR-34 ablation partially rescued MCM2-7 downregulation and genomic instability in mice with endogenous RS. Together, these data demonstrate that active MCM2-7 repression is a physiologically important mechanism for RS-induced cell cycle arrest and genome maintenance on an organismal level. Duplication of the genome by DNA replication is essential for cell proliferation. DNA replication is initiated from many sites (“origins”) along chromosomes that are bound by replication licensing proteins, including MCM2-7. They are also core components of the replication helicase complex that unwinds double stranded DNA to expose single stranded DNA that is the template for DNA polymerase. Eukaryotic DNA replication machinery faces many challenges to duplicate the complex and massive genome. Circumstances that inhibit progression of the replication machinery cause “replication stress” (RS). Cells can counteract RS by utilizing “dormant” or “backup” origins. Abundant MCM2-7 expression sufficiently licenses dormant origins, but reducing MCMs compromises cellular responses to RS. We show that MCM2-7 expression is downregulated in cells experiencing chronic RS, and this depends on the TRP53 tumor suppressor and microRNAs it regulates. Extended RS eventually reduces MCMs to a point that terminal cell cycle arrest occurs. We propose that this mechanism is a crucial protection against neoplasia.
Collapse
Affiliation(s)
- Gongshi Bai
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Marcus B. Smolka
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- Weill Institute for Cellular and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Center for Vertebrate Genomics, Cornell University, Ithaca, New York, United States of America
| | - John C. Schimenti
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- Center for Vertebrate Genomics, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
1013
|
Marmary Y, Adar R, Gaska S, Wygoda A, Maly A, Cohen J, Eliashar R, Mizrachi L, Orfaig-Geva C, Baum BJ, Rose-John S, Galun E, Axelrod JH. Radiation-Induced Loss of Salivary Gland Function Is Driven by Cellular Senescence and Prevented by IL6 Modulation. Cancer Res 2016; 76:1170-80. [PMID: 26759233 DOI: 10.1158/0008-5472.can-15-1671] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/19/2015] [Indexed: 11/16/2022]
Abstract
Head and neck cancer patients treated by radiation commonly suffer from a devastating side effect known as dry-mouth syndrome, which results from the irreversible loss of salivary gland function via mechanisms that are not completely understood. In this study, we used a mouse model of radiation-induced salivary hypofunction to investigate the outcomes of DNA damage in the head and neck region. We demonstrate that the loss of salivary function was closely accompanied by cellular senescence, as evidenced by a persistent DNA damage response (γH2AX and 53BP1) and the expression of senescence-associated markers (SA-βgal, p19ARF, and DcR2) and secretory phenotype (SASP) factors (PAI-1 and IL6). Notably, profound apoptosis or necrosis was not observed in irradiated regions. Signs of cellular senescence were also apparent in irradiated salivary glands surgically resected from human patients who underwent radiotherapy. Importantly, using IL6 knockout mice, we found that sustained expression of IL6 in the salivary gland long after initiation of radiation-induced DNA damage was required for both senescence and hypofunction. Additionally, we demonstrate that IL6 pretreatment prevented both senescence and salivary gland hypofunction via a mechanism involving enhanced DNA damage repair. Collectively, these results indicate that cellular senescence is a fundamental mechanism driving radiation-induced damage in the salivary gland and suggest that IL6 pretreatment may represent a promising therapeutic strategy to preserve salivary gland function in head and neck cancer patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Yitzhak Marmary
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Revital Adar
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Svetlana Gaska
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Annette Wygoda
- Department of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Alexander Maly
- Department of Pathology, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Jonathan Cohen
- Department of Otolaryngology/Head and Neck Surgery, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Ron Eliashar
- Department of Otolaryngology/Head and Neck Surgery, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Lina Mizrachi
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Carmit Orfaig-Geva
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Bruce J Baum
- Molecular Physiology and Therapeutics Branch National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Eithan Galun
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Jonathan H Axelrod
- Goldyne-Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel.
| |
Collapse
|
1014
|
Klein D, Schmetter A, Imsak R, Wirsdörfer F, Unger K, Jastrow H, Stuschke M, Jendrossek V. Therapy with Multipotent Mesenchymal Stromal Cells Protects Lungs from Radiation-Induced Injury and Reduces the Risk of Lung Metastasis. Antioxid Redox Signal 2016; 24:53-69. [PMID: 26066676 DOI: 10.1089/ars.2014.6183] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS Previous thorax irradiation promotes metastatic spread of tumor cells to the lung. We hypothesized that vascular damage facilitates lung metastasis after thorax irradiation and that therapeutically applied multipotent mesenchymal stromal cells (MSCs) with reported repair activity may prevent these adverse effects of ionizing radiation by protecting lung endothelia from radiation-induced damage. RESULTS Previous whole-thorax irradiation (WTI) with 15 Gy significantly enhanced seeding and metastatic growth of tumor cells in the lung. WTI was further associated with endothelial cell damage, senescence of lung epithelial cells, and upregulation of invasion- and inflammation-promoting soluble factors, for example, endothelial matrix metalloproteinase 2 (Mmp2), its activator Mmp14, the cofactor tissue inhibitor of metalloproteinases 2 (Timp2), chemokine (C-C motif) ligand 2 (Ccl2), and urokinase-type plasminogen activator (Plau/uPA), and recruitment of CD11b+CD11c- myelomonocytic cells. Inhibition of Mmp2 counteracted radiation-induced vascular dysfunction without preventing increased metastasis. In contrast, therapy with bone marrow or aorta-derived MSCs within 2 weeks postirradiation antagonized radiation-induced damage to resident cells as well as the resulting secretome changes and abrogated the metastasis-promoting effects of WTI. INNOVATION Therapy with MSCs protects lungs from radiation-induced injury and reduces the risk of lung metastasis. MSC-mediated inhibition of Mmp2 mediates their protective effects at the vasculature. Furthermore, local and systemic effects such as inhibition of radiation-induced senescence of bronchial epithelial cells and associated secretion of immunomodulatory factors may participate in the inhibitory effect of MSCs on lung metastasis. CONCLUSION MSC therapy is a promising strategy to prevent radiation-induced lung injury and the resulting increased risk of metastasis.
Collapse
Affiliation(s)
- Diana Klein
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Alexandra Schmetter
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Roze Imsak
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Florian Wirsdörfer
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Kristian Unger
- 2 Research Unit Radiation Cytogenetics, Helmholtz-Zentrum München, German Research Center for Environmental Health , Neuherberg, Germany and Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer', Helmholtz-Zentrum München, Neuherberg, Germany
| | - Holger Jastrow
- 3 Institute of Anatomy, University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Martin Stuschke
- 4 Department of Radiotherapy, University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Verena Jendrossek
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| |
Collapse
|
1015
|
Vynnytska-Myronovska BO, Kurlishchuk Y, Chen O, Bobak Y, Dittfeld C, Hüther M, Kunz-Schughart LA, Stasyk OV. Arginine starvation in colorectal carcinoma cells: Sensing, impact on translation control and cell cycle distribution. Exp Cell Res 2016; 341:67-74. [PMID: 26751966 DOI: 10.1016/j.yexcr.2016.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/04/2015] [Accepted: 01/01/2016] [Indexed: 11/15/2022]
Abstract
Tumor cells rely on a continued exogenous nutrient supply in order to maintain a high proliferative activity. Although a strong dependence of some tumor types on exogenous arginine sources has been reported, the mechanisms of arginine sensing by tumor cells and the impact of changes in arginine availability on translation and cell cycle regulation are not fully understood. The results presented herein state that human colorectal carcinoma cells rapidly exhaust the internal arginine sources in the absence of exogenous arginine and repress global translation by activation of the GCN2-mediated pathway and inhibition of mTOR signaling. Tumor suppressor protein p53 activation and G1/G0 cell cycle arrest support cell survival upon prolonged arginine starvation. Cells with the mutant or deleted TP53 fail to stop cell cycle progression at defined cell cycle checkpoints which appears to be associated with reduced recovery after durable metabolic stress triggered by arginine withdrawal.
Collapse
Affiliation(s)
- Bozhena O Vynnytska-Myronovska
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov str., 14/16, Lviv 79005, Ukraine; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Fetscherstr. 74, 01307 Dresden, Germany
| | - Yuliya Kurlishchuk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov str., 14/16, Lviv 79005, Ukraine; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Fetscherstr. 74, 01307 Dresden, Germany
| | - Oleh Chen
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov str., 14/16, Lviv 79005, Ukraine; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Fetscherstr. 74, 01307 Dresden, Germany
| | - Yaroslav Bobak
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov str., 14/16, Lviv 79005, Ukraine
| | - Claudia Dittfeld
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Fetscherstr. 74, 01307 Dresden, Germany
| | - Melanie Hüther
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Fetscherstr. 74, 01307 Dresden, Germany
| | - Leoni A Kunz-Schughart
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Fetscherstr. 74, 01307 Dresden, Germany; Department of Oncology, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Oleh V Stasyk
- Department of Cell Signaling, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov str., 14/16, Lviv 79005, Ukraine.
| |
Collapse
|
1016
|
Zhang C, Wang D, Zhang L, Guo JF, Ren AM. Theoretical investigation and design of two-photon fluorescent probes for visualizing β-galactosidase activity in living cells. RSC Adv 2016. [DOI: 10.1039/c6ra11712k] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The two-photon fluorescent probes show dual signal for β-gal bio-imaging.
Collapse
Affiliation(s)
- Chun Zhang
- Institute of Theoretical Chemistry
- Jilin University
- Changchun 130023
- China
| | - Dan Wang
- Institute of Theoretical Chemistry
- Jilin University
- Changchun 130023
- China
| | - Li Zhang
- Institute of Theoretical Chemistry
- Jilin University
- Changchun 130023
- China
| | - Jing-Fu Guo
- School of Physics
- Northeast Normal University
- Changchun 130021
- China
| | - Ai-Min Ren
- Institute of Theoretical Chemistry
- Jilin University
- Changchun 130023
- China
| |
Collapse
|
1017
|
Bunel V, Antoine MH, Stévigny C, Nortier J, Duez P. New in vitro insights on a cell death pathway induced by magnolol and honokiol in aristolochic acid tubulotoxicity. Food Chem Toxicol 2016; 87:77-87. [DOI: 10.1016/j.fct.2015.11.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022]
|
1018
|
Khongkow P, Middleton AW, Wong JPM, Kandola NK, Kongsema M, de Moraes GN, Gomes AR, Lam EWF. In Vitro Methods for Studying the Mechanisms of Resistance to DNA-Damaging Therapeutic Drugs. Methods Mol Biol 2016; 1395:39-53. [PMID: 26910067 DOI: 10.1007/978-1-4939-3347-1_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Most commonly used anticancer drugs exert their effects mainly by causing DNA damage. The enhancement in DNA damage response (DDR) is considered a key mechanism that enables cancer cells to survive through eliminating the damaged DNA lesions and thereby developing resistance to DNA-damaging agents. This chapter describes the four experimental approaches for studying DDR and genotoxic drug resistance, including the use of γ-H2AX and comet assays to monitor DNA damage and repair capacity as well as the use of clonogenic and β-galactosidase staining assays to assess long-term cell fate after DNA-damaging treatment. Finally, we also present examples of these methods currently used in our laboratory for studying the role of FOXM1 in DNA damage-induced senescence and epirubicin resistance.
Collapse
Affiliation(s)
- Pasarat Khongkow
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Anna W Middleton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Jocelyn P-M Wong
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Navrohit K Kandola
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mesayamas Kongsema
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Gabriela Nestal de Moraes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Ana R Gomes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
1019
|
Platinum(II) phenanthroimidazole G-quadruplex ligand induces selective telomere shortening in A549 cancer cells. Biochimie 2015; 121:287-97. [PMID: 26724375 DOI: 10.1016/j.biochi.2015.12.015] [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: 07/28/2015] [Accepted: 12/23/2015] [Indexed: 11/21/2022]
Abstract
Telomere maintenance, achieved by the binding of protective shelterin capping proteins to telomeres and by either telomerase or a recombination-based alternative lengthening of telomere (ALT) mechanism, is critical for cell proliferation and survival. Extensive telomere shortening or loss of telomere integrity activates DNA damage checkpoints, leading to cell senescence or death. Although telomerase upregulation is an attractive target for anti-cancer therapy, the lag associated with telomere shortening and the potential activation of ALT pose a challenge. An alternative approach is to modify telomere interactions with binding proteins (telomere uncapping). G-quadruplex ligands stabilize structures generated from single-stranded G-rich 3'-telomere end (G-quadruplex) folding, which in principle, cannot be elongated by telomerase, thus leading to telomere shortening. Ligands can also mediate rapid anti-proliferative effects by telomere uncapping. We previously reported that the G-quadruplex ligand, phenylphenanthroimidazole ethylenediamine platinum(II) (PIP), inhibits telomerase activity in vitro[47]. In the current study, a long-term seeding assay showed that PIP significantly inhibited the seeding capacity of A549 lung cancer cells and to a lesser extent primary MRC5 fibroblast cells. Importantly, treatment with PIP caused a significant dose- and time-dependent decrease in average telomere length of A549 but not MRC5 cells. Moreover, cell cycle analysis revealed a significant increase in G1 arrest upon treatment of A549 cells, but not MRC5 cells. Both apoptosis and cellular senescence may contribute to the anti-proliferative effects of PIP. Our studies validate the development of novel and specific therapeutic ligands targeting telomeric G-quadruplex structures in cancer cells.
Collapse
|
1020
|
Ding Y, Chen J, Okon IS, Zou MH, Song P. Absence of AMPKα2 accelerates cellular senescence via p16 induction in mouse embryonic fibroblasts. Int J Biochem Cell Biol 2015; 71:72-80. [PMID: 26718972 DOI: 10.1016/j.biocel.2015.12.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/30/2015] [Accepted: 12/18/2015] [Indexed: 01/22/2023]
Abstract
Emerging evidence suggests that activation of adenosine monophosphate-activated protein kinase (AMPK), an energy gauge and redox sensor, delays aging process. However, the molecular mechanisms by which AMPKα isoform regulates cellular senescence remain largely unknown. The aim of this study was to determine if AMPKα deletion contributes to the accelerated cell senescence by inducing p16(INK4A) (p16) expression thereby arresting cell cycle. The markers of cellular senescence, cell cycle proteins, and reactive oxygen species (ROS) were monitored in cultured mouse embryonic fibroblasts (MEFs) isolated from wild type (WT, C57BL/6J), AMPKα1, or AMPKα2 homozygous deficient (AMPKα1(-/-), AMPKα2(-/-)) mice by Western blot and cellular immunofluorescence staining, as well as immunohistochemistry (IHC) in skin tissue of young and aged mice. Deletion of AMPKα2, the minor isoform of AMPKα, but not AMPKα1 in high-passaged MEFs led to spontaneous cell senescence demonstrated by accumulation of senescence-associated-β-galactosidase (SA-β-gal) staining and foci formation of heterochromatin protein 1 homolog gamma (HP1γ). It was shown here that AMPKα2 deletion upregulates cyclin-dependent kinase (CDK) inhibitor, p16, which arrests cell cycle. Furthermore, AMPKα2 null cells exhibited elevated ROS production. Interestingly, knockdown of HMG box-containing protein 1 (HBP1) partially blocked the cellular senescence of AMPKα2-deleted MEFs via the reduction of p16. Finally, dermal cells senescence, including fibroblasts senescence evidenced by the staining of p16, HBP1, and Ki-67, in the skin of aged AMPKα2(-/-) mice was enhanced when compared with that in wild type mice. Taken together, our results suggest that AMPKα2 isoform plays a fundamental role in anti-oxidant stress and anti-senescence.
Collapse
Affiliation(s)
- Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Jie Chen
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Imoh Sunday Okon
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
1021
|
Clearance of senescent cells by ABT263 rejuvenates aged hematopoietic stem cells in mice. Nat Med 2015; 22:78-83. [PMID: 26657143 DOI: 10.1038/nm.4010] [Citation(s) in RCA: 1298] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/16/2015] [Indexed: 12/15/2022]
Abstract
Senescent cells (SCs) accumulate with age and after genotoxic stress, such as total-body irradiation (TBI). Clearance of SCs in a progeroid mouse model using a transgenic approach delays several age-associated disorders, suggesting that SCs play a causative role in certain age-related pathologies. Thus, a 'senolytic' pharmacological agent that can selectively kill SCs holds promise for rejuvenating tissue stem cells and extending health span. To test this idea, we screened a collection of compounds and identified ABT263 (a specific inhibitor of the anti-apoptotic proteins BCL-2 and BCL-xL) as a potent senolytic drug. We show that ABT263 selectively kills SCs in culture in a cell type- and species-independent manner by inducing apoptosis. Oral administration of ABT263 to either sublethally irradiated or normally aged mice effectively depleted SCs, including senescent bone marrow hematopoietic stem cells (HSCs) and senescent muscle stem cells (MuSCs). Notably, this depletion mitigated TBI-induced premature aging of the hematopoietic system and rejuvenated the aged HSCs and MuSCs in normally aged mice. Our results demonstrate that selective clearance of SCs by a pharmacological agent is beneficial in part through its rejuvenation of aged tissue stem cells. Thus, senolytic drugs may represent a new class of radiation mitigators and anti-aging agents.
Collapse
|
1022
|
Perrigue PM, Najbauer J, Jozwiak AA, Barciszewski J, Aboody KS, Barish ME. Planarians as a model of aging to study the interaction between stem cells and senescent cells in vivo. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2015; 5:30052. [PMID: 26654402 PMCID: PMC4696462 DOI: 10.3402/pba.v5.30052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/06/2015] [Accepted: 11/06/2015] [Indexed: 02/07/2023]
Abstract
The depletion of stem cell pools and the accumulation of senescent cells in animal tissues are linked to aging. Planarians are invertebrate flatworms and are unusual in that their stem cells, called neoblasts, are constantly replacing old and dying cells. By eliminating neoblasts in worms via irradiation, the biological principles of aging are exposed in the absence of wound healing and regeneration, making planaria a powerful tool for aging research.
Collapse
Affiliation(s)
- Patrick M Perrigue
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland;
| | - Joseph Najbauer
- Department of Immunology and Biotechnology, University of Pécs, Pécs, Hungary
| | - Agnieszka A Jozwiak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Jan Barciszewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Karen S Aboody
- Department of Neurosciences, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA.,Division of Neurosurgery, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| | - Michael E Barish
- Department of Neurosciences, City of Hope National Medical Center and Beckman Research Institute, Duarte, CA, USA
| |
Collapse
|
1023
|
Sharma K, Goehe RW, Di X, Hicks MA, Torti SV, Torti FM, Harada H, Gewirtz DA. A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB 1089. Autophagy 2015; 10:2346-61. [PMID: 25629933 DOI: 10.4161/15548627.2014.993283] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The standard of care for unresectable lung cancer is chemoradiation. However, therapeutic options are limited and patients are rarely cured. We have previously shown that vitamin D and vitamin D analogs such as EB 1089 can enhance the response to radiation in breast cancer through the promotion of a cytotoxic form of autophagy. In A549 and H460 non-small cell lung cancer (NSCLC) cells, 1,25-D3 (the hormonally active form of vitamin D) and EB 1089 prolonged the growth arrest induced by radiation alone and suppressed proliferative recovery, which translated to a significant reduction in clonogenic survival. In H838 or H358 NSCLC cells, which lack VDR/vitamin D receptor or functional TP53, respectively, 1,25-D3 failed to modify the extent of radiation-induced growth arrest or suppress proliferative recovery post-irradiation. Sensitization to radiation in H1299 NSCLC cells was evident only when TP53 was induced in otherwise tp53-null H1299 NSCLC cells. Sensitization was not associated with increased DNA damage, decreased DNA repair or an increase in apoptosis, necrosis, or senescence. Instead sensitization appeared to be a consequence of the conversion of the cytoprotective autophagy induced by radiation alone to a novel cytostatic form of autophagy by the combination of 1,25-D3 or EB 1089 with radiation. While both pharmacological and genetic suppression of autophagy or inhibition of AMPK phosphorylation sensitized the NSCLC cells to radiation alone, inhibition of the cytostatic autophagy induced by the combination treatment reversed sensitization. Evidence for selectivity was provided by lack of radiosensitization in normal human bronchial cells and cardiomyocytes. Taken together, these studies have identified a unique cytostatic function of autophagy that appears to be mediated by VDR, TP53, and possibly AMPK in the promotion of an enhanced response to radiation by 1,25-D3 and EB 1089 in NSCLC.
Collapse
Key Words
- ACTB, actin, β
- AMPK, AMP activated protein kinase
- ANXA5, annexin A5
- ATG5, autophagy related 5
- AVO, acidic vesicular organelles
- BECN1, Beclin 1, autophagy-related
- Baf, bafilomycin A1
- FACS, fluorescence activating cell sorting
- GFP, green fluorescent protein; H2AFX/H2AX, H2A histone family, member X
- GLB, galactosidase
- MAP1LC3/LC3, microtubule-associated protein 1 light chain 3
- NSCLC
- NSCLC, non-small cell lung cancer
- PI, propidium iodide
- SQSTM1, sequestosome 1
- TP53, tumor protein p53
- VDR, vitamin D (1, 25-dihydroxyvitamin D3) receptor
- WT, wild-type; LC3 and LC3-II acronyms have been used to indicate the LC3B and LC3BII isoforms respectively
- autophagy
- cytoprotective
- cytostatic
- radiation
- β
Collapse
Affiliation(s)
- Khushboo Sharma
- a Department of Pharmacology and Pharmacology; Massey Cancer Center ; Virginia Commonwealth University ; Richmond , VA USA
| | | | | | | | | | | | | | | |
Collapse
|
1024
|
Chang TC, Tang W, Koh WJH, Rettie AJE, Emond MJ, Monnat RJ, Folch A. Microwell arrays reveal cellular heterogeneity during the clonal expansion of transformed human cells. TECHNOLOGY 2015; 3:163-171. [PMID: 27158641 PMCID: PMC4854201 DOI: 10.1142/s2339547815200046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
We developed micromolded microwell arrays to study the proliferation and senescence of single cells. Microwell arrays were designed to be compatible with conventional cell culture protocols to simplify cell loading, cell culture, and imaging. We demonstrated the utility of these arrays by measuring the proliferation and senescence of isogenic cells which expressed or had been depleted of the human Werner syndrome protein. Our results allowed us to reveal cell-to-cell heterogeneity in proliferation in WRN+ and WRN-depleted fibroblasts during clonal growth.
Collapse
Affiliation(s)
- Tim C Chang
- Departments of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Weiliang Tang
- Departments of Pathology, University of Washington, Seattle, WA 98195, USA
| | - William Jen Hoe Koh
- Departments of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | | | - Mary J Emond
- Departments of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Raymond J Monnat
- Departments of Pathology, University of Washington, Seattle, WA 98195, USA; Departments of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Albert Folch
- Departments of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
1025
|
Zhou Y, Ino K, Shiku H, Matsue T. Evaluation of senescence in individual MCF-7 spheroids based on electrochemical measurement of senescence-associated β-galactosidase activity. Electrochim Acta 2015. [DOI: 10.1016/j.electacta.2015.10.115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
1026
|
Capasso S, Alessio N, Squillaro T, Di Bernardo G, Melone MA, Cipollaro M, Peluso G, Galderisi U. Changes in autophagy, proteasome activity and metabolism to determine a specific signature for acute and chronic senescent mesenchymal stromal cells. Oncotarget 2015; 6:39457-39468. [PMID: 26540573 PMCID: PMC4741838 DOI: 10.18632/oncotarget.6277] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/02/2015] [Indexed: 12/11/2022] Open
Abstract
A sharp definition of what a senescent cell is still lacking since we do not have in depth understanding of mechanisms that induce cellular senescence. In addition, senescent cells are heterogeneous, in that not all of them express the same genes and present the same phenotype. To further clarify the classification of senescent cells, hints may be derived by the study of cellular metabolism, autophagy and proteasome activity. In this scenario, we decided to study these biological features in senescence of Mesenchymal Stromal Cells (MSC). These cells contain a subpopulation of stem cells that are able to differentiate in mesodermal derivatives (adipocytes, chondrocytes, osteocytes). In addition, they can also contribute to the homeostatic maintenance of many organs, hence, their senescence could be very deleterious for human body functions. We induced MSC senescence by oxidative stress, doxorubicin treatment, X-ray irradiation and replicative exhaustion. The first three are considered inducers of acute senescence while extensive proliferation triggers replicative senescence also named as chronic senescence. In all conditions, but replicative and high IR dose senescence, we detected a reduction of the autophagic flux, while proteasome activity was impaired in peroxide-treated and irradiated cells. Differences were observed also in metabolic status. In general, all senescent cells evidenced metabolic inflexibility and prefer to use glucose as energy fuel. Irradiated cells with low dose of X-ray and replicative senescent cells show a residual capacity to use fatty acids and glutamine as alternative fuels, respectively. Our study may be useful to discriminate among different senescent phenotypes.
Collapse
Affiliation(s)
- Stefania Capasso
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Tiziana Squillaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
| | - Mariarosa A. Melone
- Institute of Bioscience and Bioresources, CNR, Naples, Italy
- Department of Clinical and Experimental Medicine and Surgery, Division of Neurology, Second University of Naples, Naples, Italy
| | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
| | | | - Umberto Galderisi
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, PA, USA
- Department of Experimental Medicine, Biotechnology and Molecular Biology Section, Second University of Naples, Naples, Italy
- Institute of Bioscience and Bioresources, CNR, Naples, Italy
| |
Collapse
|
1027
|
Tominaga T, Dutta RK, Joladarashi D, Doi T, Reddy JK, Kanwar YS. Transcriptional and Translational Modulation of myo-Inositol Oxygenase (Miox) by Fatty Acids: IMPLICATIONS IN RENAL TUBULAR INJURY INDUCED IN OBESITY AND DIABETES. J Biol Chem 2015; 291:1348-67. [PMID: 26578517 DOI: 10.1074/jbc.m115.698191] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Indexed: 11/06/2022] Open
Abstract
The kidney is one of the target organs for various metabolic diseases, including diabetes, metabolic syndrome, and obesity. Most of the metabolic studies underscore glomerular pathobiology, although the tubulo-interstitial compartment has been underemphasized. This study highlights mechanisms concerning the pathobiology of tubular injury in the context of myo-inositol oxygenase (Miox), a tubular enzyme. The kidneys of mice fed a high fat diet (HFD) had increased Miox expression and activity, and the latter was related to phosphorylation of serine/threonine residues. Also, expression of sterol regulatory element-binding protein1 (Srebp1) and markers of cellular/nuclear damage was increased along with accentuated apoptosis and loss of tubular brush border. Similar results were observed in cells treated with palmitate/BSA. Multiple sterol-response elements and E-box motifs were found in the miox promoter, and its activity was modulated by palmitate/BSA. Electrophoretic mobility and ChIP assays confirmed binding of Srebp to consensus sequences of the miox promoter. Exposure of palmitate/BSA-treated cells to rapamycin normalized Miox expression and prevented Srebp1 nuclear translocation. In addition, rapamycin treatment reduced p53 expression and apoptosis. Like rapamycin, srebp siRNA reduced Miox expression. Increased expression of Miox was associated with the generation of reactive oxygen species (ROS) in kidney tubules of mice fed an HFD and cell exposed to palmitate/BSA. Both miox and srebp1 siRNAs reduced generation of ROS. Collectively, these findings suggest that HFD or fatty acids modulate transcriptional, translational, and post-translational regulation of Miox expression/activity and underscore Miox being a novel target of the transcription factor Srebp1. Conceivably, activation of the mTORC1/Srebp1/Miox pathway leads to the generation of ROS culminating into tubulo-interstitial injury in states of obesity.
Collapse
Affiliation(s)
- Tatsuya Tominaga
- From the Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 and
| | - Rajesh K Dutta
- From the Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 and
| | - Darukeshwara Joladarashi
- From the Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 and
| | - Toshio Doi
- the Department of Nephrology, University of Tokushima, Tokushima, Japan
| | - Janardan K Reddy
- From the Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 and
| | - Yashpal S Kanwar
- From the Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611 and
| |
Collapse
|
1028
|
Lompardía SL, Díaz M, Papademetrio DL, Mascaró M, Pibuel M, Álvarez E, Hajos SE. Hyaluronan oligomers sensitize chronic myeloid leukemia cell lines to the effect of Imatinib. Glycobiology 2015; 26:343-52. [DOI: 10.1093/glycob/cwv107] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
|
1029
|
Molecular Characterization of Growth Hormone-producing Tumors in the GC Rat Model of Acromegaly. Sci Rep 2015; 5:16298. [PMID: 26549306 PMCID: PMC4637865 DOI: 10.1038/srep16298] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/08/2015] [Indexed: 12/14/2022] Open
Abstract
Acromegaly is a disorder resulting from excessive production of growth hormone (GH) and consequent increase of insulin-like growth factor 1 (IGF-I), most frequently caused by pituitary adenomas. Elevated GH and IGF-I levels results in wide range of somatic, cardiovascular, endocrine, metabolic, and gastrointestinal morbidities. Subcutaneous implantation of the GH-secreting GC cell line in rats leads to the formation of tumors. GC tumor-bearing rats develop characteristics that resemble human acromegaly including gigantism and visceromegaly. However, GC tumors remain poorly characterized at a molecular level. In the present work, we report a detailed histological and molecular characterization of GC tumors using immunohistochemistry, molecular biology and imaging techniques. GC tumors display histopathological and molecular features of human GH-producing tumors, including hormone production, cell architecture, senescence activation and alterations in cell cycle gene expression. Furthermore, GC tumors cells displayed sensitivity to somatostatin analogues, drugs that are currently used in the treatment of human GH-producing adenomas, thus supporting the GC tumor model as a translational tool to evaluate therapeutic agents. The information obtained would help to maximize the usefulness of the GC rat model for research and preclinical studies in GH-secreting tumors.
Collapse
|
1030
|
Bartosova M, Rudolf A, Pichl S, Schmidt K, Okun JG, Straub BK, Rutkowski R, Witowski J, Schmitt CP. Increased storage and secretion of phosphatidylcholines by senescent human peritoneal mesothelial cells. Clin Exp Nephrol 2015; 20:544-551. [PMID: 26527208 DOI: 10.1007/s10157-015-1192-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/24/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND/AIMS Human peritoneal mesothelial cells (HPMC) secrete phosphatidylcholines (PC) which form a lipid bilayer lining the peritoneum. They prevent frictions and adhesions and act as a barrier to the transport of water-soluble solutes while permitting water flux. PC may play an essential role in peritoneal integrity and function, the role of PD induced HPMC senescence on PC homeostasis, however, is unknown. METHODS HPMC cell lines were isolated from four non-uremic patients. Expression of the three PC synthesis genes (rt-PCR), and cellular storage and secretion of PC (ESI-mass-spectrometry) were analyzed in young and senescent HPMC (>Hayflick-limit). RESULTS Senescent cells displayed significantly altered morphology; flow cytometry demonstrated extensive staining for senescence-associated beta galactosidase. Nine different PC were detected in HPMC with palmitoyl-myristoyl phosphatidylcholine (PMPC) being most abundant. In senescent HPMC mRNA expression of the three key PC synthesis genes was 1.5-, 2.4- and 6-fold increased as compared to young HPMC, with the latter, phosphatidylcholine cytidylyltransferase, being rate limiting. Intracellular storage of the nine PC was 75-450 % higher in senescent vs. young HPMC, PC secretion rates were 100-300 % higher. Intracellular PC concentrations were not correlated with the PC secretion rates. Electron microscopy demonstrated lamellar bodies, the primary storage site of PC, in senescent but not in young cells. CONCLUSION Senescent HPMC store and secrete substantially more PC than young cells. Our findings indicate a novel protective mechanism, which should counteract peritoneal damage induced by chronic exposure to PD fluids.
Collapse
Affiliation(s)
- Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | | | - Sebastian Pichl
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Kathrin Schmidt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Jürgen G Okun
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Beate K Straub
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Claus P Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| |
Collapse
|
1031
|
Dezutter-Dambuyant C, Durand I, Alberti L, Bendriss-Vermare N, Valladeau-Guilemond J, Duc A, Magron A, Morel AP, Sisirak V, Rodriguez C, Cox D, Olive D, Caux C. A novel regulation of PD-1 ligands on mesenchymal stromal cells through MMP-mediated proteolytic cleavage. Oncoimmunology 2015; 5:e1091146. [PMID: 27141350 PMCID: PMC4839348 DOI: 10.1080/2162402x.2015.1091146] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022] Open
Abstract
Whether fibroblasts regulate immune response is a crucial issue in the modulation of inflammatory responses. Herein, we demonstrate that foreskin fibroblasts (FFs) potently inhibit CD3+ T cell proliferation through a mechanism involving early apoptosis of activated T cells. Using blocking antibodies, we demonstrate that the inhibition of T cell proliferation occurs through cell-to-cell interactions implicating PD-1 receptor expressed on T cells and its ligands, PD-L1 and PD-L2, on fibroblasts. Dual PD-1 ligand neutralization is required to abrogate (i) binding of the PD-1-Fc fusion protein, (ii) early apoptosis of T cells, and (iii) inhibition of T cell proliferation. Of utmost importance, we provide the first evidence that PD-1 ligand expression is regulated through proteolytic cleavage by endogenous matrix metalloproteinases (MMPs) without transcriptional alteration during culture-time. Using (i) different purified enzymatic activities, (ii) MMP-specific inhibitors, and (iii) recombinant human MMP-9 and MMP-13, we demonstrated that in contrast to CD80/CD86, PD-L1 was selectively cleaved by MMP-13, while PD-L2 was sensitive to broader MMP activities. Their cleavage by exogenous MMP-9 and MMP-13 with loss of PD-1 binding domain resulted in the reversion of apoptotic signals on mitogen-activated CD3+ T cells. We suggest that MMP-dependent cleavage of PD-1 ligands on fibroblasts may limit their immunosuppressive capacity and thus contribute to the exacerbation of inflammation in tissues. In contrast, carcinoma-associated fibroblasts appear PD-1 ligand-depleted through MMP activity that may impair physical deletion of exhausted defective memory T cells through apoptosis and facilitate their regulatory functions. These observations should be considered when using the powerful PD-1/PD-L1 blocking immunotherapies.
Collapse
Affiliation(s)
- Colette Dezutter-Dambuyant
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Isabelle Durand
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Laurent Alberti
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Nathalie Bendriss-Vermare
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jenny Valladeau-Guilemond
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Adeline Duc
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Audrey Magron
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Anne-Pierre Morel
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Vanja Sisirak
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Céline Rodriguez
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - David Cox
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| | - Daniel Olive
- Aix-Marseille Université, Marseille, France, Inserm U1068, Center de Recherche en Cancérologie de Marseille (CRCM), Immunity & Cancer Institut Paoli-Calmettes; Aix-Marseille Université UM 105, CNRS UMR 7258, IBiSA Cancer Immunomonitoring Platform, Marseilles, France
| | - Christophe Caux
- Université de Lyon, Lyon, France; Université Lyon 1, ISPB, Lyon, France; INSERM U1052, Center de Recherche en Cancérologie de Lyon, Lyon, France; CNRS UMR5286, Center de Recherche en Cancérologie de Lyon, Lyon, France
| |
Collapse
|
1032
|
Abstract
Given the irreversible nature of nephron loss, aging of the kidney is of special interest to diagnostic and toxicologic pathologists. There are many similarities among histologic lesions in aged human and canine kidneys, including increased frequency of glomerulosclerosis, interstitial fibrosis, and tubular atrophy. Unfortunately, there are few studies in which renal tissue from aged healthy dogs was adequately examined with advanced diagnostics—namely, transmission electron microscopy and immunofluorescence—so age-associated changes in canine podocytes and glomerular basement membranes are poorly characterized. An age-associated decrease in the glomerular filtration rate in humans and dogs (specifically small breed dogs) has been documented. Although lesions in aged rats and mice differ somewhat from those of aged dogs and humans, the knowledge gained from rodent models is still vital to elucidating the pathogenesis of age-associated renal disease. Many novel molecules implicated in renal aging have been identified through genetically modified rodent models and transcriptomic and proteomic analysis of human kidneys. These molecules represent intriguing therapeutic targets and diagnostic biomarkers. Likewise, influencing critical pathways of cellular aging, such as telomere shortening, cellular senescence, and autophagy, could improve renal function in the elderly.
Collapse
Affiliation(s)
- R. E. Cianciolo
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - S. L. Benali
- Dipartimento di Biomedicina comparata e Alimentazione, Università di Padova, Legnaro, Italy
| | - L. Aresu
- Dipartimento di Biomedicina comparata e Alimentazione, Università di Padova, Legnaro, Italy
| |
Collapse
|
1033
|
Fernández Larrosa PN, Ruíz Grecco M, Mengual Gómez D, Alvarado CV, Panelo LC, Rubio MF, Alonso DF, Gómez DE, Costas MA. RAC3 more than a nuclear receptor coactivator: a key inhibitor of senescence that is downregulated in aging. Cell Death Dis 2015; 6:e1902. [PMID: 26469953 PMCID: PMC4632280 DOI: 10.1038/cddis.2015.218] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 06/24/2015] [Accepted: 07/01/2015] [Indexed: 11/10/2022]
Abstract
Receptor-associated coactivator 3 (RAC3) is a nuclear receptor coactivator usually overexpressed in tumors that exerts oncogenic functions in the cytoplasm and the nucleus. Although as part of its oncogenic actions it was previously identified as an inhibitor of apoptosis and autophagy, its expression is required in order to preserve the pluripotency and embryonic stem cell self-renewal. In this work we investigated its role in cellular senescence. We found that RAC3 overexpression in the nontumoral HEK293 cells inhibits the premature senescence induced by hydrogen peroxide or rapamycin. The mechanism involves not only the inhibition of autophagy early induced by these stimuli in the pathway to senescence, but also the increase in levels and nuclear localization of both the cell cycle suppressors p53/p21 and the longevity promoters FOXO1A, FOXO3A and SIRT1. Furthermore, we found that RAC3 overexpression is required in order to maintain the telomerase activity. In tumoral HeLa cells its activity was inhibited by depletion of RAC3 inducing replicative senescence. Moreover, we demonstrated that in vivo, levels of RAC3 are downregulated in the liver from aged as compared with young rats, whereas the levels of p21 are increased, correlating with the expected senescent cell contents in aged tissues. A similar downregulation of RAC3 was observed in the premature and replicative senescence of human fetal WI-38 cells and premature senescence of hepatocyte HepG2 cell line. Taken together, all these results demonstrate that RAC3 is an inhibitor of senescence whose downregulation in aged individuals could be probably a tumor suppressor mechanism, avoiding the clonal expansion of risky old cells having damaged DNA.
Collapse
Affiliation(s)
- P N Fernández Larrosa
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - M Ruíz Grecco
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - D Mengual Gómez
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - C V Alvarado
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - L C Panelo
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - M F Rubio
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| | - D F Alonso
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - D E Gómez
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, R. Sáenz Peña 352, Bernal, Buenos Aires B1876BXD Argentina
| | - M A Costas
- Laboratorio de Biología Molecular y Apoptosis, Instituto de Investigaciones Médicas Alfredo Lanari, IDIM-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Combatientes de Malvinas 3150, Buenos Aires C1427ARO, Argentina
| |
Collapse
|
1034
|
Nozaki T, Nikai S, Okabe R, Nagahama K, Eto N. A novel in vitro model of sarcopenia using BubR1 hypomorphic C2C12 myoblasts. Cytotechnology 2015; 68:1705-15. [PMID: 26464273 DOI: 10.1007/s10616-015-9920-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/01/2015] [Indexed: 12/23/2022] Open
Abstract
Sarcopenia is the age-related loss of skeletal muscle mass and function with adverse outcomes that include physical disability, poor quality of life, and death. The detailed molecular mechanisms remain unknown. An in vitro muscle atrophy model is needed to enable mechanistic studies. To create such a model, we employed BubR1 insufficiency which causes premature ageing in mice. Using C2C12 cells, a recognized in vitro model of the skeletal muscle cell, we obtained the BubR1 hypomorphic C2C12 (C2C12BKD) cells by using shRNA. The resulting C2C12BKD cells displayed several characteristics of the sarcopenic muscle cell. In C2C12BKD cells, formation of myotubes, assessed by analysis of fusion index, was markedly reduced as was the expression of myogenin and MyoD, two marker genes for myogenesis. Moreover, the cells showed increased expression of the muscle-specific ubiquitin ligases Atrogin-1 and MuRF-1, indicating increased protein degradation through the ubiquitin-proteasome dependent proteolytic pathway. These results suggest that C2C12BKD cells are potentially useful as a novel in vitro model of sarcopenia.
Collapse
Affiliation(s)
- Takateru Nozaki
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, 889-2192, Japan
| | - Shiori Nikai
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, 889-2192, Japan
| | - Ryo Okabe
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, 889-2192, Japan
| | - Kiyoko Nagahama
- Interdisciplinary Graduate School of Agriculture and Engineering, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, 889-2192, Japan
| | - Nozomu Eto
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, 889-2192, Japan.
- Interdisciplinary Graduate School of Agriculture and Engineering, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki, 889-2192, Japan.
| |
Collapse
|
1035
|
Wild-Type Hras Suppresses the Earliest Stages of Tumorigenesis in a Genetically Engineered Mouse Model of Pancreatic Cancer. PLoS One 2015; 10:e0140253. [PMID: 26452271 PMCID: PMC4599940 DOI: 10.1371/journal.pone.0140253] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/23/2015] [Indexed: 01/05/2023] Open
Abstract
Oncogenic, activating mutations in KRAS initiate pancreatic cancer. There are, however, two other Ras family members, Nras and Hras, which can be activated in the presence of oncogenic Kras. The role of these wild-type Ras proteins in cancer remains unclear, as their disruption has been shown to enhance or inhibit tumorigenesis depending upon the context. As pancreatic cancer is critically dependent upon Ras signaling, we tested and now report that loss of Hras increases tumor load and reduces survival in an oncogenic Kras-driven pancreatic adenocarcinoma mouse model. These effects were traced to the earliest stages of pancreatic cancer, suggesting that wild-type Hras may suppress tumor initiation. In normal cells, activated Ras can suppress proliferation through p53-dependent mechanisms. We find that the tumor suppressive effects of Hras are nullified in a homozygous mutant p53 background. As such, loss of wild-type Hras fosters the earliest stages of pancreatic cancer in a p53-dependent manner.
Collapse
|
1036
|
Mazur PK, Herner A, Mello SS, Wirth M, Hausmann S, Sánchez-Rivera FJ, Lofgren SM, Kuschma T, Hahn SA, Vangala D, Trajkovic-Arsic M, Gupta A, Heid I, Noël PB, Braren R, Erkan M, Kleeff J, Sipos B, Sayles LC, Heikenwalder M, Heßmann E, Ellenrieder V, Esposito I, Jacks T, Bradner JE, Khatri P, Sweet-Cordero EA, Attardi LD, Schmid RM, Schneider G, Sage J, Siveke JT. Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat Med 2015; 21:1163-71. [PMID: 26390243 PMCID: PMC4959788 DOI: 10.1038/nm.3952] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/26/2015] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers and shows resistance to any therapeutic strategy used. Here we tested small-molecule inhibitors targeting chromatin regulators as possible therapeutic agents in PDAC. We show that JQ1, an inhibitor of the bromodomain and extraterminal (BET) family of proteins, suppresses PDAC development in mice by inhibiting both MYC activity and inflammatory signals. The histone deacetylase (HDAC) inhibitor SAHA synergizes with JQ1 to augment cell death and more potently suppress advanced PDAC. Finally, using a CRISPR-Cas9-based method for gene editing directly in the mouse adult pancreas, we show that de-repression of p57 (also known as KIP2 or CDKN1C) upon combined BET and HDAC inhibition is required for the induction of combination therapy-induced cell death in PDAC. SAHA is approved for human use, and molecules similar to JQ1 are being tested in clinical trials. Thus, these studies identify a promising epigenetic-based therapeutic strategy that may be rapidly implemented in fatal human tumors.
Collapse
Affiliation(s)
- Pawel K Mazur
- Department of Pediatrics, Stanford University School of Medicine, California, USA
- Department of Genetics, Stanford University School of Medicine, California, USA
| | - Alexander Herner
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stephano S Mello
- Department of Genetics, Stanford University School of Medicine, California, USA
- Department of Radiation Oncology, Stanford University School of Medicine, California, USA
| | - Matthias Wirth
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Simone Hausmann
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Francisco J Sánchez-Rivera
- David H. Koch Institute for Integrative Cancer Research, Department of Biology, and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Shane M Lofgren
- Department of Medicine, Stanford University School of Medicine, California, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, California, USA
| | - Timo Kuschma
- Department of Pediatrics, Stanford University School of Medicine, California, USA
- Department of Genetics, Stanford University School of Medicine, California, USA
| | - Stephan A Hahn
- Department of Molecular Gastrointestinal Oncology, Ruhr-University Bochum, Bochum, Germany
| | - Deepak Vangala
- Ruhr-University Bochum, Medical Clinic, Knappschaftskrankenhaus, Bochum, Germany
| | - Marija Trajkovic-Arsic
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Aayush Gupta
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Irina Heid
- Institute of Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Peter B Noël
- Institute of Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Rickmer Braren
- Institute of Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Mert Erkan
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jörg Kleeff
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Bence Sipos
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | - Leanne C Sayles
- Department of Pediatrics, Stanford University School of Medicine, California, USA
| | - Mathias Heikenwalder
- Institute of Virology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research center (DKFZ) Heidelberg, Germany
| | - Elisabeth Heßmann
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Irene Esposito
- Institute of Pathology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Department of Biology, and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - James E Bradner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Purvesh Khatri
- Department of Medicine, Stanford University School of Medicine, California, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, California, USA
| | | | - Laura D Attardi
- Department of Genetics, Stanford University School of Medicine, California, USA
- Department of Radiation Oncology, Stanford University School of Medicine, California, USA
| | - Roland M Schmid
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guenter Schneider
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Julien Sage
- Department of Pediatrics, Stanford University School of Medicine, California, USA
- Department of Genetics, Stanford University School of Medicine, California, USA
| | - Jens T Siveke
- Second Department of Internal Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
1037
|
Jung YR, Kim EJ, Choi HJ, Park JJ, Kim HS, Lee YJ, Park MJ, Lee M. Aspirin Targets SIRT1 and AMPK to Induce Senescence of Colorectal Carcinoma Cells. Mol Pharmacol 2015; 88:708-19. [PMID: 26219912 DOI: 10.1124/mol.115.098616] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/27/2015] [Indexed: 12/11/2022] Open
Abstract
Cancer therapies attempt to destroy the entire tumor, but this tends to require toxic compounds and high doses of radiation. Recently, considerable attention has focused on therapy-induced senescence (TIS), which can be induced in cancer cells by low doses of therapeutic drugs or radiation and provides a barrier to tumor development. However, the molecular mechanisms governing TIS remain elusive. Special attention has been paid to the potential chemopreventive effect of aspirin against human colorectal cancer. In this study, we investigated the effects of aspirin on TIS of human colorectal carcinoma (CRC) cells and show that it occurs via sirtuin 1 (SIRT1) and AMP-activated protein kinase (AMPK), two key regulators of cellular metabolism. Aspirin increased the senescence of CRC cells, increased the protein levels of SIRT1, phospho-AMPK (T172), and phospho-acetyl CoA carboxylase (S79), and reduced the cellular level of ATP. Small-interfering RNA-mediated downregulation or pharmacological inhibition of SIRT1 or AMPK significantly attenuated the aspirin-induced cellular senescence in CRC cells. In contrast, treatment with a SIRT1 agonist or an AMP analog induced cellular senescence. Remarkably, SIRT1 knockdown abrogated the aspirin-induced activation of AMPK, and vice versa. During the progression of aspirin-induced cellular senescence in CRC cells, SIRT1 showed increased deacetylase activity at a relatively early time point but was characterized by decreased activity with increased cytoplasmic localization at a later time point. Collectively, these novel findings suggest that aspirin could provide anticancer effects by inducing senescence in human CRC cells through the reciprocal regulation of SIRT1-AMPK pathways.
Collapse
Affiliation(s)
- Yu Ri Jung
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Eun Ju Kim
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hyeong Jwa Choi
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Jung-Jin Park
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hak-Su Kim
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Yoon-Jin Lee
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Myung-Jin Park
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Minyoung Lee
- Division of Radiation Effect (Y.R.J., E.J.K., H.J.C., Y.-J.L., M.L.), Division of Radiation Cancer Research (M.-J.P.), Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea; Department of Biochemistry and Medical Research Center (J.-J.P.), College of Medicine, Chungbuk National University, Cheongju, Republic of Korea; and Department of Pharmacology and Hypoxia-Related Diseases Research Center (H.-S.K.), College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
1038
|
Purple sweet potato color inhibits endothelial premature senescence by blocking the NLRP3 inflammasome. J Nutr Biochem 2015; 26:1029-40. [DOI: 10.1016/j.jnutbio.2015.04.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 04/09/2015] [Accepted: 04/16/2015] [Indexed: 12/31/2022]
|
1039
|
Stickel SA, Gomes NP, Frederick B, Raben D, Su TT. Bouvardin is a Radiation Modulator with a Novel Mechanism of Action. Radiat Res 2015; 184:392-403. [PMID: 26414509 DOI: 10.1667/rr14068.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Protein synthesis is essential for growth, proliferation and survival of cells. Translation factors are overexpressed in many cancers and in preclinical models, their experimental inhibition has been shown to inhibit cancer growth. Differential regulation of translation also occurs upon exposure to cancer-relevant stressors such as hypoxia and ionizing radiation. The failure to regulate translation has been shown to interfere with recovery after genotoxic stress. These findings suggest that modulation of translation, alone or in conjunction with genotoxins, may be therapeutic in oncology. Yet, only two drugs that directly inhibit translation are FDA-approved for oncology therapies used today. We have previously identified the protein synthesis inhibitor, bouvardin in a screen for small molecule enhancers of ionizing radiation in Drosophila melanogaster . Bouvardin was independently identified in a screen for selective inhibitors of engineered human breast cancer stem cells. Here we report the effect of bouvardin treatment in preclinical models of head and neck cancer (HNC) and glioma, two cancer types for which radiation therapy is the most common treatment. Our data show that bouvardin treatment blocked translation elongation on human ribosomes and suggest that it did so by blocking the dissociation of elongation factor 2 from the ribosome. Bouvardin and radiation enhanced the induction of clonogenic death in HNC and glioma cells, although by different mechanisms. Bouvardin treatment enhanced the radiation-induced antitumor effects in HNC tumor xenografts in mice. These data suggest that inhibition of translation elongation, particularly in combination with radiation treatment, may be a promising treatment option for cancer.
Collapse
Affiliation(s)
- Stefanie A Stickel
- a Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado
| | - Nathan P Gomes
- a Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado;,d SuviCa, Inc., Boulder, Colorado
| | - Barbara Frederick
- a Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado;,c Department of Radiation Oncology, University of Colorado Health Sciences Campus, Aurora, Colorado; and
| | - David Raben
- a Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado;,c Department of Radiation Oncology, University of Colorado Health Sciences Campus, Aurora, Colorado; and
| | - Tin Tin Su
- a Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado;,b University of Colorado, Comprehensive Cancer Center, Aurora, Colorado
| |
Collapse
|
1040
|
Seignez C, Martin A, Rollet CE, Racoeur C, Scagliarini A, Jeannin JF, Bettaieb A, Paul C. Senescence of tumor cells induced by oxaliplatin increases the efficiency of a lipid A immunotherapy via the recruitment of neutrophils. Oncotarget 2015; 5:11442-51. [PMID: 25347345 PMCID: PMC4294335 DOI: 10.18632/oncotarget.2556] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/30/2014] [Indexed: 01/01/2023] Open
Abstract
Management of advanced colorectal cancer is challenging due to the lack of efficient therapy. The lipid A, OM-174 exhibited antitumor activity in colorectal cancer. We explored the anticancer efficacy of this compound in rats bearing large colorectal tumors in combination with the platinum derivative drugs oxaliplatin and cisplatin. While each drug used alone exhibited partial antitumor activity, sequential treatment with oxaliplatin or cisplatin for one week followed by lipid A injections induced a great regression of colorectal tumors, with more than 95% of rats cured from their tumors. This potent antitumor efficacy of the combined treatments was correlated to the sequential induction of cellular senescence by oxaliplatin, and of apoptosis, mainly triggered by the lipid A. Moreover, a recruitment of tumor-associated neutrophils with N1 phenotype as attested by the expression of inducible nitric oxide synthase was observed with combination of oxaliplatin and lipid A. Neutrophil recruitment within tumor microenvironment was due to oxaliplatin and lipid A-dependent release of neutrophil specific chemoattractants such as cxcl1 and 2. However the N1 phenotype is only dependent of lipid A treatment. These results suggest that the combination of chemotherapy with an immunotherapy is a promising approach to treat patients with advanced colorectal cancer.
Collapse
Affiliation(s)
- Cédric Seignez
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Amandine Martin
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Claire-Emmanuelle Rollet
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Cindy Racoeur
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Alessandra Scagliarini
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Jean-François Jeannin
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Ali Bettaieb
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| | - Catherine Paul
- EPHE Cancer Immunotherapy Laboratory, EA7269 EPHE-University of burgundy, Dijon, F-21000, France
| |
Collapse
|
1041
|
MOF maintains transcriptional programs regulating cellular stress response. Oncogene 2015; 35:2698-710. [PMID: 26387537 PMCID: PMC4893634 DOI: 10.1038/onc.2015.335] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Revised: 07/09/2015] [Accepted: 08/04/2015] [Indexed: 12/14/2022]
Abstract
MOF (MYST1, KAT8) is the major H4K16 lysine acetyltransferase (KAT) in Drosophila and mammals and is essential for embryonic development. However, little is known regarding the role of MOF in specific cell lineages. Here we analyze the differential role of MOF in proliferating and terminally differentiated tissues at steady state and under stress conditions. In proliferating cells, MOF directly binds and maintains the expression of genes required for cell cycle progression. In contrast, MOF is dispensable for terminally differentiated, postmitotic glomerular podocytes under physiological conditions. However, in response to injury, MOF is absolutely critical for podocyte maintenance in vivo. Consistently, we detect defective nuclear, endoplasmic reticulum and Golgi structures, as well as presence of multivesicular bodies in vivo in podocytes lacking Mof following injury. Undertaking genome-wide expression analysis of podocytes, we uncover several MOF-regulated pathways required for stress response. We find that MOF, along with the members of the non-specific lethal but not the male-specific lethal complex, directly binds to genes encoding the lysosome, endocytosis and vacuole pathways, which are known regulators of podocyte maintenance. Thus, our work identifies MOF as a key regulator of cellular stress response in glomerular podocytes.
Collapse
|
1042
|
|
1043
|
Dasgupta T, Olow AK, Yang X, Hashizume R, Nicolaides TP, Tom M, Aoki Y, Berger MS, Weiss WA, Stalpers LJA, Prados M, James CD, Mueller S, Haas-Kogan DA. Survival advantage combining a BRAF inhibitor and radiation in BRAF V600E-mutant glioma. J Neurooncol 2015; 126:385-93. [PMID: 26384810 DOI: 10.1007/s11060-015-1939-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/12/2015] [Indexed: 01/05/2023]
Abstract
Radiation (RT) is critical to the treatment of high-grade gliomas (HGGs) but cures remain elusive. The BRAF mutation V600E is critical to the pathogenesis of 10-20% of pediatric gliomas, and a small proportion of adult HGGs. Here we aim to determine whether PLX4720, a specific BRAF V600E inhibitor, enhances the activity of RT in human HGGs in vitro and in vivo. Patient-derived HGG lines harboring wild-type BRAF or BRAF V600E were assessed in vitro to determine IC50 values, cell cycle arrest, apoptosis and senescence and elucidate mechanisms of combinatorial activity. A BRAF V600E HGG intracranial xenograft mouse model was used to evaluate in vivo combinatorial efficacy of PLX4720+RT. Tumors were harvested for immunohistochemistry to quantify cell cycle arrest and apoptosis. RT+PLX4720 exhibited greater anti-tumor effects than either monotherapy in BRAF V600E but not in BRAF WT lines. In vitro studies showed increased Annexin V and decreased S phase cells in BRAF V600E gliomas treated with PLX4720+RT, but no significant changes in β-galactosidase levels. In vivo, concurrent and sequential PLX4720+RT each significantly prolonged survival compared to monotherapies, in the BRAF V600E HGG model. Immunohistochemistry of in vivo tumors demonstrated that PLX4720+RT decreased Ki-67 and phospho-MAPK, and increased γH2AX and p21 compared to control mice. BRAF V600E inhibition enhances radiation-induced cytotoxicity in BRAF V600E-mutated HGGs, in vitro and in vivo, effects likely mediated by apoptosis and cell cycle, but not senescence. These studies provide the pre-clinical rationale for clinical trials of concurrent radiotherapy and BRAF V600E inhibitors.
Collapse
Affiliation(s)
- Tina Dasgupta
- Valley Radiotherapy Associates, 1500 Rosecrans Avenue, Manhattan Beach, CA, 90266, USA.
| | - Aleksandra K Olow
- Department of Laboratory Medicine, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
- Department of Radiation Oncology, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Xiaodong Yang
- Department of Radiation Oncology, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Rintaro Hashizume
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Theodore P Nicolaides
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Maxwell Tom
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Yasuyuki Aoki
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - William A Weiss
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Lukas J A Stalpers
- Department of Radiotherapy, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - C David James
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sabine Mueller
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, 94143, USA
- Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California, San Francisco (UCSF), San Francisco, CA, 94143, USA
| |
Collapse
|
1044
|
Gonzalez LC, Ghadaouia S, Martinez A, Rodier F. Premature aging/senescence in cancer cells facing therapy: good or bad? Biogerontology 2015; 17:71-87. [DOI: 10.1007/s10522-015-9593-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/22/2015] [Indexed: 01/07/2023]
|
1045
|
Abdelmohsen K, Gorospe M. Noncoding RNA control of cellular senescence. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 6:615-29. [PMID: 26331977 DOI: 10.1002/wrna.1297] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/19/2015] [Accepted: 07/20/2015] [Indexed: 12/23/2022]
Abstract
Senescent cells accumulate in normal tissues with advancing age and arise by long-term culture of primary cells. Senescence develops following exposure to a range of stress-causing agents and broadly influences the physiology and pathology of tissues, organs, and systems in the body. While many proteins are known to control senescence, numerous noncoding (nc)RNAs are also found to promote or repress the senescent phenotype. Here, we review the regulatory ncRNAs (primarily microRNAs and lncRNAs) identified to-date as key modulators of senescence. We highlight the major senescent pathways (p53/p21 and pRB/p16), as well as the senescence-associated secretory phenotype (SASP) and other senescence-associated events governed by ncRNAs, and discuss the importance of understanding comprehensively the ncRNAs implicated in cell senescence.
Collapse
Affiliation(s)
- Kotb Abdelmohsen
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
1046
|
Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, Raguz S, Acosta JC, Innes AJ, Banito A, Georgilis A, Montoya A, Wolter K, Dharmalingam G, Faull P, Carroll T, Martínez-Barbera JP, Cutillas P, Reisinger F, Heikenwalder M, Miller RA, Withers D, Zender L, Thomas GJ, Gil J. mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat Cell Biol 2015; 17:1205-17. [PMID: 26280535 PMCID: PMC4589897 DOI: 10.1038/ncb3225] [Citation(s) in RCA: 545] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/20/2015] [Indexed: 12/15/2022]
Abstract
Senescent cells secrete a combination of factors collectively known as the senescence-associated secretory phenotype (SASP). The SASP reinforces senescence and activates an immune surveillance response, but it can also show pro-tumorigenic properties and contribute to age-related pathologies. In a drug screen to find new SASP regulators, we uncovered the mTOR inhibitor rapamycin as a potent SASP suppressor. Here we report a mechanism by which mTOR controls the SASP by differentially regulating the translation of the MK2 (also known as MAPKAPK2) kinase through 4EBP1. In turn, MAPKAPK2 phosphorylates the RNA-binding protein ZFP36L1 during senescence, inhibiting its ability to degrade the transcripts of numerous SASP components. Consequently, mTOR inhibition or constitutive activation of ZFP36L1 impairs the non-cell-autonomous effects of senescent cells in both tumour-suppressive and tumour-promoting contexts. Altogether, our results place regulation of the SASP as a key mechanism by which mTOR could influence cancer, age-related diseases and immune responses.
Collapse
Affiliation(s)
- Nicolás Herranz
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Suchira Gallage
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Metabolic Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Massimiliano Mellone
- Cancer Sciences Unit, Cancer Research UK Centre, Somers Building, University of Southampton, Southampton SO16 6YD, UK
| | - Torsten Wuestefeld
- Division of Molecular Oncology of Solid Tumors, Dept. of Internal Medicine I, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Sabrina Klotz
- Division of Molecular Oncology of Solid Tumors, Dept. of Internal Medicine I, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Christopher J. Hanley
- Cancer Sciences Unit, Cancer Research UK Centre, Somers Building, University of Southampton, Southampton SO16 6YD, UK
| | - Selina Raguz
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Juan Carlos Acosta
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Andrew J Innes
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Ana Banito
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Athena Georgilis
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Alex Montoya
- Proteomics Facility; MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Katharina Wolter
- Division of Molecular Oncology of Solid Tumors, Dept. of Internal Medicine I, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Gopuraja Dharmalingam
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Peter Faull
- Proteomics Facility; MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Thomas Carroll
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | | | - Pedro Cutillas
- Proteomics Facility; MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Florian Reisinger
- Institute for Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
| | - Mathias Heikenwalder
- Institute for Virology, Technische Universität München/Helmholtz Zentrum München, Munich, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research (DKFZ), Heidelberg, Germany
| | - Richard A. Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Dominic Withers
- Metabolic Signalling Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Lars Zender
- Division of Molecular Oncology of Solid Tumors, Dept. of Internal Medicine I, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Gareth J. Thomas
- Cancer Sciences Unit, Cancer Research UK Centre, Somers Building, University of Southampton, Southampton SO16 6YD, UK
| | - Jesús Gil
- Cell Proliferation Group, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
- Epigenetics Section, MRC Clinical Sciences Centre, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| |
Collapse
|
1047
|
Acrylamide induces accelerated endothelial aging in a human cell model. Food Chem Toxicol 2015; 83:140-5. [DOI: 10.1016/j.fct.2015.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/28/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022]
|
1048
|
Rhus coriaria induces senescence and autophagic cell death in breast cancer cells through a mechanism involving p38 and ERK1/2 activation. Sci Rep 2015; 5:13013. [PMID: 26263881 PMCID: PMC4532997 DOI: 10.1038/srep13013] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/13/2015] [Indexed: 12/29/2022] Open
Abstract
Here, we investigated the anticancer effect of Rhus coriaria on three breast cancer cell lines. We demonstrated that Rhus coriaria ethanolic extract (RCE) inhibits the proliferation of these cell lines in a time- and concentration-dependent manner. RCE induced senescence and cell cycle arrest at G1 phase. These changes were concomitant with upregulation of p21, downregulation of cyclin D1, p27, PCNA, c-myc, phospho-RB and expression of senescence-associated β-galactosidase activity. No proliferative recovery was detected after RCE removal. Annexin V staining and PARP cleavage analysis revealed a minimal induction of apoptosis in MDA-MB-231 cells. Electron microscopy revealed the presence of autophagic vacuoles in RCE-treated cells. Interestingly, blocking autophagy by 3-methyladenine (3-MA) or chloroquine (CQ) reduced RCE-induced cell death and senescence. RCE was also found to activate p38 and ERK1/2 signaling pathways which coincided with induction of autophagy. Furthermore, we found that while both autophagy inhibitors abolished p38 phosphorylation, only CQ led to significant decrease in pERK1/2. Finally, RCE induced DNA damage and reduced mutant p53, two events that preceded autophagy. Our findings provide strong evidence that R. coriaria possesses strong anti-breast cancer activity through induction of senescence and autophagic cell death, making it a promising alternative or adjunct therapeutic candidate against breast cancer.
Collapse
|
1049
|
Gao Q, Yu GY, Shi JY, Li LH, Zhang WJ, Wang ZC, Yang LX, Duan M, Zhao H, Wang XY, Zhou J, Qiu SJ, Jeong LS, Jia LJ, Fan J. Neddylation pathway is up-regulated in human intrahepatic cholangiocarcinoma and serves as a potential therapeutic target. Oncotarget 2015; 5:7820-32. [PMID: 25229838 PMCID: PMC4202163 DOI: 10.18632/oncotarget.2309] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Therapeutic intervention in neddylation pathway is an emerging area for cancer treatment. Herein, we evaluated the clinical relevance and therapeutic potential of targeting this pathway in intrahepatic cholangiocarcinoma (ICC). Immunohistochemistry of neddylation pathway components in a cohort of 322 cases showed that E1 (NAE1 and UBA3) and E2 (UBC12) enzymes, as well as global NEDD8 conjugation, were upregulated in over 2/3 of human ICC. Notably, NAE1 was identified as an independent prognosticator for postoperative recurrence (P=0.009) and a combination of NEDD8 and NAE1 provided a better power for predicting patient clinical outcomes. In vitro treatment with MLN4924, a small-molecule NEDD8-activating enzyme inhibitor, led to a dose-dependent decrease of viability in both established and primary cholangiocarcinoma cell lines. Additionally, MLN4924 exhibited at least additive effect when combined with cisplatin. By blocking cullins neddylation, MLN4924 inactivated Cullin-Ring ligase (CRL) and caused the accumulation of CRL substrates that triggered cell cycle arrest, senescence or apoptosis. Meanwhile, MLN4924 was well-tolerated and significantly inhibited tumor growth in xenograft model of cholangiocarcinoma. Taken together, our findings indicated that upregulated neddylation pathway was involved in ICC progression and interference in this pathway could be a promising target for ICC therapy.
Collapse
Affiliation(s)
- Qiang Gao
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China. Contributed equally to this work
| | - Guang-Yang Yu
- Cancer Institute, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China. Contributed equally to this work
| | - Jie-Yi Shi
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China. Contributed equally to this work
| | - Li-Hui Li
- Cancer Institute, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China. Contributed equally to this work
| | - Wen-Juan Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Chao Wang
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Liu-Xiao Yang
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Meng Duan
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Hu Zhao
- Department of Laboratory Medicine, Laboratory of Molecular Biology of Huadong Hospital, Fudan University, Shanghai, P. R. China
| | - Xiao-Ying Wang
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China. Institute of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Shuang-Jian Qiu
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Lak Shin Jeong
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Li-Jun Jia
- Cancer Institute, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital and Shanghai Medical School, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China. Institute of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| |
Collapse
|
1050
|
Molinos M, Almeida CR, Gonçalves RM, Barbosa MA. Improvement of Bovine Nucleus Pulposus Cells Isolation Leads to Identification of Three Phenotypically Distinct Cell Subpopulations. Tissue Eng Part A 2015; 21:2216-27. [DOI: 10.1089/ten.tea.2014.0461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Maria Molinos
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Catarina R. Almeida
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Raquel M. Gonçalves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Mário A. Barbosa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|