51
|
Kearns PKA, Casey HA, Leach JP. Hypothesis: Multiple sclerosis is caused by three-hits, strictly in order, in genetically susceptible persons. Mult Scler Relat Disord 2018; 24:157-174. [PMID: 30015080 DOI: 10.1016/j.msard.2018.06.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/25/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022]
Abstract
Multiple Sclerosis is a chronic, progressive and debilitating neurological disease which, despite extensive study for over 100 years, remains of enigmatic aetiology. Drawn from the epidemiological evidence, there exists a consensus that there are environmental (possibly infectious) factors that contribute to disease pathogenesis that have not yet been fully elucidated. Here we propose a three-tiered hypothesis: 1) a clinic-epidemiological model of multiple sclerosis as a rare late complication of two sequential infections (with the temporal sequence of infections being important); 2) a proposal that the first event is helminthic infection with Enterobius Vermicularis, and the second is Epstein Barr Virus infection; and 3) a proposal for a testable biological mechanism, involving T-Cell exhaustion for Epstein-Barr Virus protein LMP2A. We believe that this model satisfies some of the as-yet unexplained features of multiple sclerosis epidemiology, is consistent with the clinical and neuropathological features of the disease and is potentially testable by experiment. This model may be generalizable to other autoimmune diseases.
Collapse
|
52
|
Differential requirement for dimerization partner DP between E2F-dependent activation of tumor suppressor and growth-related genes. Sci Rep 2018; 8:8438. [PMID: 29855511 PMCID: PMC5981219 DOI: 10.1038/s41598-018-26860-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 01/31/2018] [Indexed: 01/17/2023] Open
Abstract
The transcription factor E2F plays crucial roles in cell proliferation and tumor suppression by activating growth-related genes and pro-apoptotic tumor suppressor genes, respectively. It is generally accepted that E2F binds to target sequences with its heterodimeric partner DP. Here we show that, while knockdown of DP1 expression inhibited ectopic E2F1- or adenovirus E1a-induced expression of the CDC6 gene and cell proliferation, knockdown of DP1 and DP2 expression did not affect ectopic E2F1- or E1a-induced expression of the tumor suppressor ARF gene, an upstream activator of the tumor suppressor p53, activation of p53 or apoptosis. These observations suggest that growth related and pro-apoptotic E2F targets are regulated by distinct molecular mechanisms and contradict the threshold model, which postulates that E2F activation of pro-apoptotic genes requires a higher total activity of activator E2Fs, above that necessary for E2F-dependent activation of growth-related genes.
Collapse
|
53
|
Mex-3B induces apoptosis by inhibiting miR-92a access to the Bim-3'UTR. Oncogene 2018; 37:5233-5247. [PMID: 29849121 DOI: 10.1038/s41388-018-0336-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 03/12/2018] [Accepted: 04/17/2018] [Indexed: 01/06/2023]
Abstract
Cells respond to a variety of cellular stresses, including DNA damage, by regulating genes whose expression modulates cell cycle arrest, DNA repair, senescence, and/or apoptosis. MicroRNAs (miRNAs) play essential roles in both normal development and disease pathogenesis by destabilizing mRNAs and inhibiting translation. In turn, miRNA biogenesis, turnover, and activity can be regulated by specific RNA-binding proteins. Here we show that Mex-3B, an hnRNP K homology (KH) domain-containing RNA-binding protein, critically modulates DNA stress-induced apoptosis by posttranscriptionally upregulating the pro-apoptotic BH3 (Bcl-2 homology region 3)-only family member Bim. Furthermore, our data indicate that binding of Mex-3B to the 3'-untranslated region (3'UTR) of Bim interferes with the interaction of an Argonaute (Ago)-miR-92a complex with a miR-92a target site present in the Bim RNA. Our results provide novel insights into the posttranscriptional mechanisms that are critical for cellular stress responses.
Collapse
|
54
|
GADD45A and CDKN1A are involved in apoptosis and cell cycle modulatory effects of viscumTT with further inactivation of the STAT3 pathway. Sci Rep 2018; 8:5750. [PMID: 29636527 PMCID: PMC5893628 DOI: 10.1038/s41598-018-24075-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/21/2018] [Indexed: 12/28/2022] Open
Abstract
ViscumTT, a whole mistletoe preparation, has shown synergistic induction of apoptosis in several pediatric tumor entities. High therapeutic potential has previously been observed in Ewing's sarcoma, rhabdomyosarcoma, ALL and AML. In this study, we analyzed modulatory effects on the cell cycle by viscumTT in three osteosarcoma cell lines with various TP53 statuses. ViscumTT treatment induced G1 arrest in TP53 wild-type and null-mutant cells, but S arrest in TP53 mutant cells. Blockage of G1/S transition was accompanied by down-regulation of the key regulators CDK4, CCND1, CDK2, CCNE, CCNA. However, investigations on the transcriptional level revealed secondary TP53 participation. Cell cycle arrest was predominantly mediated by transcriptionally increased expression of GADD45A and CDKN1A and decreased SKP2 levels. Enhanced CDKN1A and GADD45A expression further played a role in viscumTT-induced apoptosis with involvement of stress-induced MAPK8 and inactivation of MAPK1/3. Furthermore, viscumTT inhibited the pro-survival pathway STAT3 by dephosphorylation of the two sites, Tyr705 and Ser727, by down-regulation of total STAT3 and its direct downstream targets BIRC5 and C-MYC. Moreover, tests of the efficacy of viscumTT in vivo showing reduction of tumor volume confirmed the high therapeutic potential as an anti-tumoral agent for osteosarcoma.
Collapse
|
55
|
Chemoprevention by Probiotics During 1,2-Dimethylhydrazine-Induced Colon Carcinogenesis in Rats. Dig Dis Sci 2018; 63:900-909. [PMID: 29427224 DOI: 10.1007/s10620-018-4949-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Probiotics are believed to have properties that lower the risk of colon cancer. However, the mechanisms by which they exert their beneficial effects are relatively unknown. AIM To assess the impact of probiotics in preventing induction of colon carcinogenesis in rats. METHODS The rats were divided into six groups viz., normal control, Lactobacillus plantarum (AdF10)-treated, Lactobacillus rhamnosus GG (LGG)-treated, 1,2-dimethylhydrazine (DMH)-treated, L. plantarum (AdF10) + DMH-treated and L. rhamnosus GG (LGG) + DMH-treated. Both the probiotics were supplemented daily at a dose of 2 × 1010 cells per day. DMH at a dose of 30 mg/kg body weight was administered subcutaneously twice a week for the first 4 weeks and then once every week for a duration of 16 weeks. Glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR), glutathione peroxidase (GPx), glutathione-S-transferase (GST) and catalase as protein expression of genes involved in apoptosis were assessed during DMH-induced colon carcinogenesis in rats. RESULTS DMH treatment decreased the activity of GSH, GPx, GST, SOD and catalase. However, AdF10 and LGG supplementation to DMH-treated rats significantly increased the activity of these enzymes. Further, DMH treatment revealed alterations in the protein expressions of various genes involved in the p53-mediated apoptotic pathway such as p53, p21, Bcl-2, Bax, caspase-9 and caspase-3, which, however, were shifted towards normal control levels upon simultaneous supplementation with probiotics. CONCLUSION The present study suggests that probiotics can provide protection against oxidative stress and apoptotic-related protein disregulation during experimentally induced colon carcinogenesis.
Collapse
|
56
|
Ruiz-Moreno C, Jimenez-Del-Rio M, Sierra-Garcia L, Lopez-Osorio B, Velez-Pardo C. Vitamin E synthetic derivate-TPGS-selectively induces apoptosis in jurkat t cells via oxidative stress signaling pathways: implications for acute lymphoblastic leukemia. Apoptosis 2018; 21:1019-32. [PMID: 27364951 DOI: 10.1007/s10495-016-1266-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) is a water-soluble derivative of natural vitamin E commonly used as a drug delivery agent. Recently, TPGS alone has been reported to induce cell death in lung, breast and prostate cancer. However, the effect of TPGS on cancer cell viability remains unclear. Thus, this study was aimed to evaluate the cytotoxic effect of TPGS on human periphral blood lymphocytes (PBL) and on T cell acute lymphocytic leukemia (ALL) Jurkat clone E6-1 cells and its possible mechanism of action. PBL and Jurkat cells were treated with TPGS (10, 20, 40, 60, and 80 μM), and morphological changes in the cell nucleus, mitochondrial membrane potential (ΔΨm), and intracellular reactive oxygen species levels were determined by immune-fluorescence microscopy and flow cytometry. Cellular apoptosis markers were also evaluated by immunocytochemistry. In this study, TPGS induced apoptotic cell death in Jurkat cells, but not in PBL, in a dose-response manner with increasing nuclear DNA fragmentation, increasing cell cycle arrest, and decreasing ΔΨm. Additionally, TPGS increased dichlorofluorescein fluorescence intensity, indicative of H2O2 production, in a dose-independent fashion. TPGS increased DJ-1 Cys(106)-sulfonate, as a marker of intracellular stress and induced the activation of NF-κB, p53 and c-Jun transcription factors. Additionally, it increased the expression of apoptotic markers Bcl-2 related pro-apoptotic proteins Bax and PUMAand activated caspase-3. The antioxidant N-acetyl-L-cysteine and known pharmacological inhibitors protected the cells from the TPGS induced effects. In conclusion, TPGS selectively induces apoptosis in Jurkat cells through two independent but complementary H2O2-mediated signaling pathways. Our findings support the use of TPGS as a potential treatment for ALL.
Collapse
Affiliation(s)
- Cristian Ruiz-Moreno
- Neuroscience Research Group, Faculty of Medicine, Medical Research Institute, University of Antioquia, Calle 70 N° 52-21, and Calle 62 #52-59, Building 1, Room 412, SIU, P. O. Box 1226, Medellín, Colombia
| | - Marlene Jimenez-Del-Rio
- Neuroscience Research Group, Faculty of Medicine, Medical Research Institute, University of Antioquia, Calle 70 N° 52-21, and Calle 62 #52-59, Building 1, Room 412, SIU, P. O. Box 1226, Medellín, Colombia
| | - Ligia Sierra-Garcia
- Material Science Group, Faculty of Chemistry, University of Antioquia, Calle 70 N° 52-21, and Calle 62 #52-59, Building 1, Room 310, SIU, P. O. Box 1226, Medellin, Colombia
| | - Betty Lopez-Osorio
- Material Science Group, Faculty of Chemistry, University of Antioquia, Calle 70 N° 52-21, and Calle 62 #52-59, Building 1, Room 310, SIU, P. O. Box 1226, Medellin, Colombia
| | - Carlos Velez-Pardo
- Neuroscience Research Group, Faculty of Medicine, Medical Research Institute, University of Antioquia, Calle 70 N° 52-21, and Calle 62 #52-59, Building 1, Room 412, SIU, P. O. Box 1226, Medellín, Colombia.
| |
Collapse
|
57
|
FGF1 induces resistance to chemotherapy in ovarian granulosa tumor cells through regulation of p53 mitochondrial localization. Oncogenesis 2018; 7:18. [PMID: 29467390 PMCID: PMC5833868 DOI: 10.1038/s41389-018-0033-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 01/10/2018] [Indexed: 01/04/2023] Open
Abstract
Ovarian cancer remains associated with a high mortality rate and relapse is too frequently seen after chemotherapeutic treatment of granulosa cell tumors (GCTs) or epithelial ovarian cancers (EOCs). It is thus of major importance to progress in the knowledge of the molecular mechanisms underlying chemoresistance of ovarian tumors. Overexpression of Fibroblast Growth Factor 1 (FGF1) is observed in various cancers, correlates with poor survival and could be responsible for resistance to platinum-based chemotherapy of serous ovarian cancers. How FGF1 promotes escape to chemotherapy remains unknown. In previous studies, we showed that FGF1 inhibits p53 transcriptional activities, leading to increased cell survival of neuronal or fibroblast cell lines. In this study, we show that FGF1 favors survival of COV434 cells upon treatment with etoposide and cisplatin, two common chemotherapeutic molecules used for ovarian cancer. Etoposide and cisplatin induced mitochondrial depolarization, cytochrome c release and caspase activation in COV434 cells. Overexpression of FGF1 counteracts these events and thus allows increased survival of ovarian cells. In this study, FGF1 had little effect on p53 stability and transcriptional activities. Etoposide induced p21 expression as expected, but p21 protein levels were even increased in the presence of FGF1. Using RNA interference, we showed that p21 exerts an anti-apoptotic activity in COV434 cells. However abrogating this activity was not sufficient to restore cell death of FGF1-overexpressing cells. We also show for the first time that p53 mitochondrial pathway is involved in the cell death of COV434 cells. Indeed, p53 accumulates at mitochondria upon etoposide treatment and inhibition of p53 mitochondrial localization using pifithrin-µ inhibits apoptosis of COV434 cells. FGF1 also decreases mitochondrial accumulation of p53 induced by etoposide. This constitutes a novel mechanism of action for FGF1 to promote cell survival in response to chemotherapy.
Collapse
|
58
|
Mochizuki M, Nakahara T. Establishment of xenogeneic serum-free culture methods for handling human dental pulp stem cells using clinically oriented in-vitro and in-vivo conditions. Stem Cell Res Ther 2018; 9:25. [PMID: 29394956 PMCID: PMC5797401 DOI: 10.1186/s13287-017-0761-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/04/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022] Open
Abstract
Background Currently, ex-vivo handling of stem cells, including transport after harvest and therapeutic preparation, is generally done in culture media containing fetal bovine serum (FBS), which promotes cell attachment, proliferation, and differentiation. However, because of safety concerns associated with the use of FBS, including potential transmission of zoonotic agents and transplant rejection because of the incorporation of foreign proteins into the stem cells, there is a need for xenogeneic serum-free culture media for clinical handling of stem cells. Methods Dental pulp stem cells were derived from wisdom teeth donated by eight healthy volunteers and cultured in xenogeneic serum-free culture medium (XFM) or xenogeneic serum-containing culture medium (SCM). Cells were subjected to morphological, proliferation, karyotype, differentiation, marker expression, cryopreservation, and cytotoxic susceptibility analyses in vitro, as well as transplantation in vivo. Results In primary culture, XFM cells showed lower adhesion and slightly different morphology, although the single-cell size was similar to that of SCM cells. XFM cells exhibited typical mesenchymal stem cell (MSC) characteristics in vitro and in vivo, including marker gene/protein expression, trilineage differentiation potential, and hard, osteo-dentin tissue formation. Additionally, XFM cells maintained a normal karyotype in vitro and nontumorigenic potential in vivo; however, XFM cells were more susceptible to H2O2 and ultraviolet cytotoxic stimuli. XFM cells formed a multilayered structure showing excessive cell death/division in contrast to the monolayered structure of SCM cells when reaching overconfluence. Proliferation was disrupted in overconfluent XFM cells, and these cells could not be subcultured. Dimethyl sulfoxide-free cryopreserved XFM cells yielded similar results in all of the experiments. Conclusions This study is the first reporting successful isolation and expansion of an MSC population from donor-derived tissue (dental pulp) under xenogeneic serum-free culture conditions, as well as the application of cryopreservation, using a research strategy based on clinically oriented in-vitro and in-vivo experiments. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0761-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mai Mochizuki
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan
| | - Taka Nakahara
- Department of Developmental and Regenerative Dentistry, School of Life Dentistry at Tokyo, The Nippon Dental University, 1-9-20 Fujimi, Chiyoda-ku, Tokyo, 102-8159, Japan.
| |
Collapse
|
59
|
Park S, Oh SS, Lee KW, Lee YK, Kim NY, Kim JH, Yoo J, Kim KD. NDRG2 contributes to cisplatin sensitivity through modulation of BAK-to-Mcl-1 ratio. Cell Death Dis 2018; 9:30. [PMID: 29348517 PMCID: PMC5833685 DOI: 10.1038/s41419-017-0184-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023]
Abstract
The downregulation of N-Myc downstream-regulated gene 2 (NDRG2) is known to be associated with the progression and poor prognosis of several cancers. Sensitivity to anti-cancer may be associated with a good prognosis in cancer patients, and NDRG2, which is induced by p53, sensitizes the cells to chemotherapy. However, the unique function of NDRG2 as an inducer of apoptosis under chemotreatment has not been sufficiently studied. In this study, we investigated the role of NDRG2 in chemo-sensitivity, focusing on cisplatin in U937 histiocytic lymphoma, which has the loss-of-functional mutation in p53. NDRG2 promoted the sensitivity to cisplatin through the modulation of the BAK-to-Mcl-1 ratio. The degradation of Mcl-1 and increase in BAK were mediated by JNK activation and the eIF2α/p-eIF2α pathway, respectively, which depended on PKR activation in NDRG2-overexpressed U937 (U937-NDRG2) cells. NOX5 was highly expressed in U937-NDRG2 cells and contributed to ROS production after cisplatin treatment. ROS scavenging or NOX5-knockdown successfully inhibited the sensitivity of U937-NDRG2 cells to cisplatin. Taken together, these findings indicate that NDRG2 contributed to the increased sensitivity to ciplatin through the modulation of Bak-to-Mcl-1 ratio regulated by NOX5-ROS-PKR pathway; therefore, we suggest that NDRG2 may be a molecular target for improving the efficacy of drug treatment in cancer patients.
Collapse
Affiliation(s)
- Soojong Park
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Sang-Seok Oh
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Ki Won Lee
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Yeon-Kyeong Lee
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Nae Yu Kim
- Department of Internal Medicine, Eulji University School of Medicine, Daejeon, 35233, Republic of Korea
| | - Joo Heon Kim
- Department of Pathology, Eulji University School of Medicine, Daejeon, 35233, Republic of Korea
| | - Jiyun Yoo
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Kwang Dong Kim
- Division of Applied Life Science (BK21 Plus), Gyeongsang National University, Jinju, 52828, Republic of Korea. .,Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea. .,PMBBRC, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
60
|
Scheffold A, Jebaraj BMC, Stilgenbauer S. Venetoclax: Targeting BCL2 in Hematological Cancers. Recent Results Cancer Res 2018; 212:215-242. [PMID: 30069633 DOI: 10.1007/978-3-319-91439-8_11] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Over the last years, targeted anti-cancer therapy with small-molecule inhibitors and antibodies moved to the forefront as a strategy to treat hematological cancers. These novel agents showed outstanding effects in treatment of patients, often irrespective of their underlying genetic features. However, evolution and selection of subclones with continuous treatment leads to disease relapse and resistance toward these novel drugs. Venetoclax (ABT-199) is a novel, orally bioavailable small-molecule inhibitor for selective targeting of B-cell lymphoma 2 (BCL2). Venetoclax is in clinical development and shows high efficacy and safety in particular in the treatment of chronic lymphocytic leukemia (CLL), but preliminarily also in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). The most important and impressive outcomes of venetoclax treatment include a rapid induction of apoptosis and drastic reduction of the tumor bulk within a few hours after administration. Venetoclax was approved by the FDA and EMA in 2016 for patients with previously treated CLL with del(17p13) and patients failing B cell receptor signaling inhibitors (EMA only), on the basis of a single-arm phase II trial demonstrating a tremendous response rate of 79% with complete remission in 20% of cases and an estimated 1-year progression-free survival of 72%. This review focuses on the mode of action, the preclinical models, and outcomes from various clinical trials with venetoclax in different hematologic cancers as well as future development.
Collapse
Affiliation(s)
- Annika Scheffold
- Department of Internal Medicine III, Ulm University, Albert Einstein Allee 23, 89081, Ulm, Germany
| | | | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University, Albert Einstein Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
61
|
Song J, Zhao X, Feng Y, Xu S, Zhang Y, Wei L. Involvement of proapoptotic genes in autophagic cell death induced by irradiation. Cell Death Discov 2017; 3:17068. [PMID: 31098300 PMCID: PMC6515468 DOI: 10.1038/cddiscovery.2017.68] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 08/03/2017] [Accepted: 08/18/2017] [Indexed: 11/21/2022] Open
Abstract
Irradiation is widely used in anticancer therapy; however, the efficiency is limited. Most cancer cells have mutations in apoptosis that they can easily escape the apoptosis induced by irradiation. Autophagy has been known as type II programmed cell death that can be activated by irradiation, especially when apoptosis is blocked, but the underlying molecular mechanism is largely unknown. We demonstrated that proapoptotic genes PUMA and Bid are involved in the regulation of autophagic cell death. When wild-type (WT), Bax-/- and PUMA-/- HCT116 cells were exposed to irradiation, we found that, compared with WT, Bax-/- cells showed significantly decreased cell death because of Bax deficiency; however, surprisingly PUMA-/- cells showed significant increase in cell death although the proapoptotic gene PUMA was knocked out. By analyzing apoptosis via Annexin V-fluorescein isothiocyanate (FITC) assay with flow cytometry, both Bax-/- and PUMA-/- cells showed less apoptosis than WT, suggesting the existence of another type of cell death in PUMA-/- cells. Autophagy was then examined in three cell lines by counting the percentage of cells with punctate GFP-LC3. Although all three cell lines showed significantly increased autophagy activity after irradiation, that of PUMA-/- cells was much higher than the other two cell lines, which suggests that PUMA-/- cells may die through autophagy. This was then confirmed by the decreased cell death in PUMA-/- cells when autophagy was blocked by 3-MA. In addition, we also tested the responses of WT and Bid-/- MEFs to irradiation. Bid-/- MEFs but not WT died through autophagy after irradiation. These results imply the involvement of apoptosis-associated genes such as PUMA and Bid in autophagic cell death, which contributes to identifying the molecular mechanism by which autophagy drives cells to death.
Collapse
Affiliation(s)
- Jianrui Song
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xue Zhao
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China
- Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200217, China
| | - Yi Feng
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Sheng Xu
- Department of Radiotherapy, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China
| | - Yanling Zhang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu 215123, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai 200438, China
| |
Collapse
|
62
|
Contrasting effects of an Mdm2 functional polymorphism on tumor phenotypes. Oncogene 2017; 37:332-340. [PMID: 28925402 DOI: 10.1038/onc.2017.344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/04/2017] [Accepted: 08/08/2017] [Indexed: 12/18/2022]
Abstract
MDM2, an E3 ubiquitin ligase, is a potent inhibitor of the p53 tumor suppressor and is elevated in many human cancers that retain wild-type p53. MDM2 SNP309G is a functional polymorphism that results in elevated levels of MDM2 (due to enhanced SP1 binding to the MDM2 promoter) thus decreasing p53 activity. Mdm2SNP309G/G mice are more prone to spontaneous tumor formation than Mdm2SNP309T/T mice, providing direct evidence for the impact of this SNP in tumor development. We asked whether environmental factors impact SNP309G function and show that SNP309G cooperates with ionizing radiation to exacerbate tumor development. Surprisingly, ultraviolet B light or Benzo(a)pyrene exposure of skin shows that SNP309G allele actually protects against squamous cell carcinoma susceptibility. These contrasting differences led us to interrogate the mechanism by which Mdm2 SNP309 regulates tumor susceptibility in a tissue-specific manner. Although basal Mdm2 levels were significantly higher in most tissues in Mdm2SNP309G/G mice compared with Mdm2SNP309T/T mice, they were significantly lower in Mdm2SNP309G/G keratinocytes, the cell-type susceptible to squamous cell carcinoma. The assessment of potential transcriptional regulators in ENCODE ChIP-seq database identified transcriptional repressor E2F6 as a possible negative regulator of MDM2 expression. Our data show that E2F6 suppresses Mdm2 expression in cells harboring the SNP309G allele but not the SNP309T allele. Thus, Mdm2 SNP309G exhibits tissue-specific regulation and differentially impacts cancer risk.
Collapse
|
63
|
RSK2 is a new Pim2 target with pro-survival functions in FLT3-ITD-positive acute myeloid leukemia. Leukemia 2017; 32:597-605. [PMID: 28914261 DOI: 10.1038/leu.2017.284] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/04/2017] [Accepted: 09/05/2017] [Indexed: 12/25/2022]
Abstract
Acute myeloid leukemia (AML) with the FLT3 internal tandem duplication (FLT3-ITD AML) accounts for 20-30% of AML cases. This subtype usually responds poorly to conventional therapies, and might become resistant to FLT3 tyrosine kinase inhibitors (TKIs) due to molecular bypass mechanisms. New therapeutic strategies focusing on resistance mechanisms are therefore urgently needed. Pim kinases are FLT3-ITD oncogenic targets that have been implicated in FLT3 TKI resistance. However, their precise biological function downstream of FLT3-ITD requires further investigation. We performed high-throughput transcriptomic and proteomic analyses in Pim2-depleted FLT3-ITD AML cells and found that Pim2 predominantly controlled apoptosis through Bax expression and mitochondria disruption. We identified ribosomal protein S6 kinase A3 (RSK2), a 90 kDa serine/threonine kinase involved in the mitogen-activated protein kinase cascade encoded by the RPS6KA3 gene, as a novel Pim2 target. Ectopic expression of an RPS6KA3 allele rescued the viability of Pim2-depleted cells, supporting the involvement of RSK2 in AML cell survival downstream of Pim2. Finally, we showed that RPS6KA3 knockdown reduced the propagation of human AML cells in vivo in mice. Our results point to RSK2 as a novel Pim2 target with translational therapeutic potential in FLT3-ITD AML.
Collapse
|
64
|
Lytic EBV infection investigated by detection of Soluble Epstein-Barr virus ZEBRA in the serum of patients with PTLD. Sci Rep 2017; 7:10479. [PMID: 28874674 PMCID: PMC5585268 DOI: 10.1038/s41598-017-09798-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022] Open
Abstract
The ZEBRA protein (encoded by the BZLF1 gene), is the major transcription factor of EBV, expressed upon EBV lytic cycle activation. Several studies highlighted the critical role of EBV lytic infection as a risk factor for lymphoproliferative disorders like post-transplant lymphoproliferative disease (PTLD). Here, we use an antigen-capture ELISA assay specifically designed to detecting the circulating soluble ZEBRA (sZEBRA) in serum samples (threshold value determined at 40ng/mL). We retrospectively investigated a population of 66 transplanted patients comprising 35 PTLD. All the samples from a control population (30 EBV-seronegative subjects and 25 immunocompetent individuals with EBV serological reactivation), classified as sZEBRA < 40ng/mL were assigned as negative. At PTLD diagnosis, EBV genome (quantified by qPCR with EBV DNA>200 copies/mL) and sZEBRA were detectable in 51% and 60% of cases, respectively. In the patients who developed a pathologically-confirmed PTLD, the mean sZEBRA value in cases, was 399 ng/mL +/− 141 versus 53ng/mL +/− 7 in patients who did not (p < 0,001). This is the first report relating to the detection of the circulating ZEBRA in serum specimens, as well as the first analysis dealing with the lytic cycle of EBV in PTLD patients with this new biomarker.
Collapse
|
65
|
RCC2 is a novel p53 target in suppressing metastasis. Oncogene 2017; 37:8-17. [PMID: 28869598 PMCID: PMC5759027 DOI: 10.1038/onc.2017.306] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 07/14/2017] [Accepted: 07/20/2017] [Indexed: 12/17/2022]
Abstract
RCC2 (also known as TD60) is a highly conserved protein involved in prognosis in colorectal cancer. However, its relationship with tumor development is less understood. Here we demonstrate a signaling pathway defining regulation of RCC2 and its functions in tumor progression. We report that p53 is a transcriptional regulator of RCC2 that acts through its binding to a palindromic motif in the RCC2 promoter. RCC2 physically interacts and deactivates a small GTPase Rac1 that is known to be involved in metastasis. We solved a high-resolution crystal structure of RCC2 and revealed one RCC1-like domain with a unique β-hairpin that is requisite for RCC2 interaction with Rac1. p53 or RCC2 deficiency leads to activation of Rac1 and deterioration of extracellular matrix sensing (haptotaxis) of surface-bound gradients. Ectopic expression of RCC2 restores directional migration in p53-null cells. Our results demonstrate that p53 and RCC2 signaling is important for regulation of cell migration and suppression of metastasis. We propose that the p53/RCC2/Rac1 axis is a potential target for cancer therapy.
Collapse
|
66
|
Synergistic activity and heterogeneous acquired resistance of combined MDM2 and MEK inhibition in KRAS mutant cancers. Oncogene 2017; 36:6581-6591. [PMID: 28783173 PMCID: PMC5700857 DOI: 10.1038/onc.2017.258] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 05/03/2017] [Accepted: 06/23/2017] [Indexed: 01/10/2023]
Abstract
There are currently no effective targeted therapies for KRAS mutant cancers. Therapeutic strategies that combine MEK inhibitors with agents that target apoptotic pathways may be a promising therapeutic approach. We investigated combining MEK and MDM2 inhibitors as a potential treatment strategy for KRAS mutant non-small cell lung cancers and colorectal carcinomas that harbor wild-type TP53. The combination of pimasertib (MEK inhibitor) + SAR405838 (MDM2 inhibitor) was synergistic and induced the expression of PUMA and BIM, led to apoptosis and growth inhibition in vitro, and tumor regression in vivo. Acquired resistance to the combination commonly resulted from the acquisition of TP53 mutations, conferring complete resistance to MDM2 inhibition. In contrast, resistant clones exhibited marked variability in sensitivity to MEK inhibition, which significantly impacted sensitivity to subsequent treatment with alternative MEK inhibitor-based combination therapies. These results highlight both the potential promise and limitations of combining MEK and MDM2 inhibitors for treatment of KRAS mutant NSCLC and CRC.
Collapse
|
67
|
Sun Q, Wang J, Li Y, Zhuang J, Zhang Q, Sun X, Sun D. Synthesis and evaluation of cytotoxic activities of artemisinin derivatives. Chem Biol Drug Des 2017; 90:1019-1028. [DOI: 10.1111/cbdd.13016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/15/2017] [Accepted: 04/23/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Qian Sun
- Marine College; Shandong University at Weihai; Weihai China
| | - Jin Wang
- Marine College; Shandong University at Weihai; Weihai China
| | - Yao Li
- Marine College; Shandong University at Weihai; Weihai China
| | | | - Qian Zhang
- Marine College; Shandong University at Weihai; Weihai China
| | - Xiao Sun
- Marine College; Shandong University at Weihai; Weihai China
| | - Dequn Sun
- Marine College; Shandong University at Weihai; Weihai China
| |
Collapse
|
68
|
Madapura HS, Salamon D, Wiman KG, Lain S, Klein E, Nagy N. cMyc-p53 feedback mechanism regulates the dynamics of T lymphocytes in the immune response. Cell Cycle 2017; 15:1267-75. [PMID: 26985633 DOI: 10.1080/15384101.2016.1160975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Activation and proliferation of T cells are tightly regulated during the immune response. We show here that kinetics of proliferation of PHA activated T cells follows the expression of cMyc. Expression of p53 is also elevated and remains high several days after activation. To investigate the role of p53 in activated T cells, its expression was further elevated with nultin-3 treatment, a small molecule that dissociates the E3 ubiquitin protein ligase MDM2 from p53. Concomitantly, cMyc expression and proliferation decreased. At the other end of the cMyc-p53 axis, inhibition of cMyc with 10058-F4 led to down regulation of p53, likely through the lower level of cMyc induced p14ARF, which is also known to dissociate the p53-MDM2 complex. Both compounds induced cell cycle arrest and apoptosis. We conclude that the feedback regulation between cMyc and p53 is important for the T cell homeostasis. We also show that the two compounds modulating p53 and cMyc levels inhibited proliferation without abolishing the cytotoxic function, thus demonstrating the dichotomy between proliferation and cytotoxicity in activated T cells.
Collapse
Affiliation(s)
- Harsha S Madapura
- a Department of Microbiology , Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden.,b Department of Oncology-Pathology , Cancer Center Karolinska (CCK), Karolinska Institutet , Stockholm , Sweden
| | - Daniel Salamon
- a Department of Microbiology , Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Klas G Wiman
- b Department of Oncology-Pathology , Cancer Center Karolinska (CCK), Karolinska Institutet , Stockholm , Sweden
| | - Sonia Lain
- a Department of Microbiology , Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Eva Klein
- a Department of Microbiology , Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| | - Noémi Nagy
- a Department of Microbiology , Tumor and Cell Biology, Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
69
|
p53-mediated suppression of BiP triggers BIK-induced apoptosis during prolonged endoplasmic reticulum stress. Cell Death Differ 2017. [PMID: 28622297 PMCID: PMC5596431 DOI: 10.1038/cdd.2017.96] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Physiological and pathological conditions that affect the folding capacity of the endoplasmic reticulum (ER) provoke ER stress and trigger the unfolded protein response (UPR). The UPR aims to either restore the balance between newly synthesized and misfolded proteins or if the damage is severe, to trigger cell death. However, the molecular events underlying the switch between repair and cell death are not well understood. The ER-resident chaperone BiP governs the UPR by sensing misfolded proteins and thereby releasing and activating the three mediators of the UPR: PERK, IRE1 and ATF6. PERK promotes G2 cell cycle arrest and cellular repair by inducing the alternative translated p53 isoform p53ΔN40 (p53/47), which activates 14-3-3σ via suppression of p21CDKN1A. Here we show that prolonged ER stress promotes apoptosis via a p53-dependent inhibition of BiP expression. This leads to the release of the pro-apoptotic BH3-only BIK from BiP and activation of apoptosis. Suppression of bip mRNA translation is mediated via the specific binding of p53 to the first 346-nt of the bip mRNA and via a p53 trans-suppression domain located within the first seven N-terminal amino acids of p53ΔN40. This work shows how p53 targets BiP to promote apoptosis during severe ER stress and further illustrates how regulation of mRNA translation has a key role in p53-mediated regulation of gene expression during the UPR.
Collapse
|
70
|
Ju J, Lee GY, Kim YS, Chang HK, Do MS, Park KY. Bamboo Salt Suppresses Colon Carcinogenesis in C57BL/6 Mice with Chemically Induced Colitis. J Med Food 2017; 19:1015-1022. [PMID: 27845862 DOI: 10.1089/jmf.2016.3798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The aim of our experiment was to evaluate the anticancer effect of bamboo salt (BS) on C57BL/6 mice in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon cancer model. BS, solar salt, and purified salt were evaluated for their protective effects during AOM/DSS-induced colon carcinogenesis in C57BL/6 mice. BS, especially after baking for nine separate intervals (BS9x), suppressed colon carcinogenesis in the mice. BS9x decreased colon length shortening, weight-to-length ratios, and tumor counts. Pathological evidence from histological evaluation by hematoxylin and eosin staining also revealed suppression of tumorigenesis. BS9x lowered serum levels of proinflammatory cytokines (TNF-α, IL-6, and IL-1β) to close to those of the Normal group. Additionally, BS9x suppressed colon mRNA expression of proinflammatory factors and significantly regulated mRNA levels of the apoptosis-related factors, Bax and Bcl-2, and the cell cycle-related genes, p21 and p53. Additionally, immunohistochemistry showed that BS promoted p21 expression in the colon. Taken together, the results indicate that BS exhibited anticancer efficacy by modulating apoptosis- and inflammation-related gene expression during colon carcinogenesis in mice, and repetition in baking cycles of BS enhanced its anticancer functionality.
Collapse
Affiliation(s)
- Jaehyun Ju
- 1 Department of Food Science and Nutrition, Pusan National University , Busan, Korea.,2 Department of Food Science and Biotechnology, Cha University , Seongnam, Korea
| | - Ga-Young Lee
- 1 Department of Food Science and Nutrition, Pusan National University , Busan, Korea
| | | | - Hee Kyung Chang
- 4 Department of Pathology, College of Medicine, Kosin University , Busan, Korea
| | - Myoung-Sool Do
- 5 School of Life Science, Handong Global University , Pohang, Korea
| | - Kun-Young Park
- 1 Department of Food Science and Nutrition, Pusan National University , Busan, Korea.,2 Department of Food Science and Biotechnology, Cha University , Seongnam, Korea.,6 Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education , Chongqing, China
| |
Collapse
|
71
|
Modeling the response of a tumor-suppressive network to mitogenic and oncogenic signals. Proc Natl Acad Sci U S A 2017; 114:5337-5342. [PMID: 28484034 DOI: 10.1073/pnas.1702412114] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Intrinsic tumor-suppressive mechanisms protect normal cells against aberrant proliferation. Although cellular signaling pathways engaged in tumor repression have been largely identified, how they are orchestrated to fulfill their function still remains elusive. Here, we built a tumor-suppressive network model composed of three modules responsible for the regulation of cell proliferation, activation of p53, and induction of apoptosis. Numerical simulations show a rich repertoire of network dynamics when normal cells are subject to serum stimulation and adenovirus E1A overexpression. We showed that oncogenic signaling induces ARF and that ARF further promotes p53 activation to inhibit proliferation. Mitogenic signaling activates E2F activators and promotes Akt activation. p53 and E2F1 cooperate to induce apoptosis, whereas Akt phosphorylates p21 to repress caspase activation. These prosurvival and proapoptotic signals compete to dictate the cell fate of proliferation, cell-cycle arrest, or apoptosis. The cellular outcome is also impacted by the kinetic mode (ultrasensitivity or bistability) of p53. When cells are exposed to serum deprivation and recovery under fixed E1A, the shortest starvation time required for apoptosis induction depends on the terminal serum concentration, which was interpreted in terms of the dynamics of caspase-3 activation and cytochrome c release. We discovered that caspase-3 can be maintained active at high serum concentrations and that E1A overexpression sensitizes serum-starved cells to apoptosis. This work elucidates the roles of tumor repressors and prosurvival factors in tumor repression based on a dynamic network analysis and provides a framework for quantitatively exploring tumor-suppressive mechanisms.
Collapse
|
72
|
Deimling SJ, Olsen JB, Tropepe V. The expanding role of the Ehmt2/G9a complex in neurodevelopment. NEUROGENESIS 2017; 4:e1316888. [PMID: 28596979 DOI: 10.1080/23262133.2017.1316888] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/22/2017] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
Abstract
Epigenetic regulators play a crucial role in neurodevelopment. One such epigenetic complex, Ehmt1/2 (G9a/GLP), is essential for repressing gene transcription by methylating H3K9 in a highly tissue- and temporal-specific manner. Recently, data has emerged suggesting that this complex plays additional roles in regulating the activity of numerous other non-histone proteins. While much is known about the downstream effects of Ehmt1/2 function, evidence is only beginning to come to light suggesting the control of Ehmt1/2 function may be, at least in part, due to context-dependent binding partners. Here we review emerging roles for the Ehmt1/2 complex suggesting that it may play a much larger role than previously recognized, and discuss binding partners that we and others have recently characterized which act to coordinate its activity during early neurodevelopment.
Collapse
Affiliation(s)
- Steven J Deimling
- Department of Cell & Systems Biology, University of Toronto, Toronto, Canada
| | - Jonathan B Olsen
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Vincent Tropepe
- Department of Cell & Systems Biology, University of Toronto, Toronto, Canada.,Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Canada; Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Canada
| |
Collapse
|
73
|
Δ133p53 represses p53-inducible senescence genes and enhances the generation of human induced pluripotent stem cells. Cell Death Differ 2017; 24:1017-1028. [PMID: 28362428 DOI: 10.1038/cdd.2017.48] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022] Open
Abstract
p53 functions to induce cellular senescence, which is incompatible with self-renewal of pluripotent stem cells such as induced pluripotent stem cells (iPSC) and embryonic stem cells (ESC). However, p53 also has essential roles in these cells through DNA damage repair for maintaining genomic integrity and high sensitivity to apoptosis for eliminating severely damaged cells. We hypothesized that Δ133p53, a physiological inhibitory p53 isoform, is involved in the balanced regulation of self-renewing capacity, DNA damage repair and apoptosis. We examined 12 lines of human iPSC and their original fibroblasts, as well as three ESC lines, for endogenous protein levels of Δ133p53 and full-length p53 (FL-p53), and mRNA levels of various p53 target genes. While FL-p53 levels in iPSC and ESC widely ranged from below to above those in the fibroblasts, all iPSC and ESC lines expressed elevated levels of Δ133p53. The p53-inducible genes that mediate cellular senescence (p21WAF1, miR-34a, PAI-1 and IGFBP7), but not those for apoptosis (BAX and PUMA) and DNA damage repair (p53R2), were downregulated in iPSC and ESC. Consistent with these endogenous expression profiles, overexpression of Δ133p53 in human fibroblasts preferentially repressed the p53-inducible senescence mediators and significantly enhanced their reprogramming to iPSC. The iPSC lines derived from Δ133p53-overexpressing fibroblasts formed well-differentiated, benign teratomas in immunodeficient mice and had fewer numbers of somatic mutations than an iPSC derived from p53-knocked-down fibroblasts, suggesting that Δ133p53 overexpression is non- or less oncogenic and mutagenic than total inhibition of p53 activities. Overexpressed Δ133p53 prevented FL-p53 from binding to the regulatory regions of p21WAF1 and miR-34a promoters, providing a mechanistic basis for its dominant-negative inhibition of a subset of p53 target genes. This study supports the hypothesis that upregulation of Δ133p53 is an endogenous mechanism that facilitates human somatic cells to become self-renewing pluripotent stem cells with maintained apoptotic and DNA repair activities.
Collapse
|
74
|
The mechanism of Jurkat cells apoptosis induced by Aggregatibacter actinomycetemcomitans cytolethal distending toxin. Apoptosis 2017; 22:841-851. [DOI: 10.1007/s10495-017-1357-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
75
|
Zhang Q, An R, Tian X, Yang M, Li M, Lou J, Xu L, Dong Z. β-Caryophyllene Pretreatment Alleviates Focal Cerebral Ischemia-Reperfusion Injury by Activating PI3K/Akt Signaling Pathway. Neurochem Res 2017; 42:1459-1469. [PMID: 28236211 DOI: 10.1007/s11064-017-2202-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/28/2017] [Accepted: 02/09/2017] [Indexed: 12/29/2022]
Abstract
β-Caryophyllene (BCP) has been reported to be protective against focal cerebral ischemia-reperfusion (I/R) injury by its anti-oxidative and anti-inflammatory features. Recent study demonstrates that the BCP exhibits potential neuroprotection against I/R injury induced apoptosis, however, the mechanism remains unknown. Therefore, we investigate the underlying anti-apoptotic mechanism of BCP pretreatment in I/R injury. Sprague-Dawley rats (pretreated with BCP suspensions or solvent orally for 7 days) were subjected to transient Middle Cerebral Artery Occlusion (MCAO) for 90 min, followed by 24 h reperfusion. Results showed that BCP pretreatment improved the neurologic deficit score, lowered the infarct volume and decreased number of apoptotic cells in the hippocampus. Moreover, in western blot and RT-qPCR detections, BCP pretreatment down-regulated the expressions of Bax and p53, up-regulated the expression of Bcl-2, and enhanced the phosphorylation of Akt on Ser473. Blockage of PI3K activity by wortmannin not only abolished the BCP-induced decreases in infarct volume and neurologic deficit score, but also dramatically abrogated the enhancement of AKt phosphorylation. Our results suggested that BCP pre-treatment protects against I/R injury partly by suppressing apoptosis via PI3K/AKt signaling pathway activation.
Collapse
Affiliation(s)
- Qian Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Ruidi An
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Xiaocui Tian
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Mei Yang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Minghang Li
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Jie Lou
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China
| | - Lu Xu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China.
| | - Zhi Dong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
76
|
Poorebrahim M, Salarian A, Najafi S, Abazari MF, Aleagha MN, Dadras MN, Jazayeri SM, Ataei A, Poortahmasebi V. Regulatory network analysis of Epstein-Barr virus identifies functional modules and hub genes involved in infectious mononucleosis. Arch Virol 2017; 162:1299-1309. [PMID: 28155194 DOI: 10.1007/s00705-017-3242-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 01/24/2017] [Indexed: 12/14/2022]
Abstract
Epstein-Barr virus (EBV) is the most common cause of infectious mononucleosis (IM) and establishes lifetime infection associated with a variety of cancers and autoimmune diseases. The aim of this study was to develop an integrative gene regulatory network (GRN) approach and overlying gene expression data to identify the representative subnetworks for IM and EBV latent infection (LI). After identifying differentially expressed genes (DEGs) in both IM and LI gene expression profiles, functional annotations were applied using gene ontology (GO) and BiNGO tools, and construction of GRNs, topological analysis and identification of modules were carried out using several plugins of Cytoscape. In parallel, a human-EBV GRN was generated using the Hu-Vir database for further analyses. Our analysis revealed that the majority of DEGs in both IM and LI were involved in cell-cycle and DNA repair processes. However, these genes showed a significant negative correlation in the IM and LI states. Furthermore, cyclin-dependent kinase 2 (CDK2) - a hub gene with the highest centrality score - appeared to be the key player in cell cycle regulation in IM disease. The most significant functional modules in the IM and LI states were involved in the regulation of the cell cycle and apoptosis, respectively. Human-EBV network analysis revealed several direct targets of EBV proteins during IM disease. Our study provides an important first report on the response to IM/LI EBV infection in humans. An important aspect of our data was the upregulation of genes associated with cell cycle progression and proliferation.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Salarian
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Najafi
- Department of Microbiology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Foad Abazari
- Department of Genetics, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Maryam Nouri Aleagha
- Department of Genetics, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Mohammad Nasr Dadras
- Center for Disease Control, Ministry of Health and Medical Education (MOHME), Tehran, Iran
| | - Seyed Mohammad Jazayeri
- Hepatitis B Molecular Laboratory, Department of Virology, School of Public Health, Tehran University of Medical Sciences, PO Box 15155-6446, Tehran, Iran
| | - Atousa Ataei
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Vahdat Poortahmasebi
- Hepatitis B Molecular Laboratory, Department of Virology, School of Public Health, Tehran University of Medical Sciences, PO Box 15155-6446, Tehran, Iran.
| |
Collapse
|
77
|
Lieberman HB, Panigrahi SK, Hopkins KM, Wang L, Broustas CG. p53 and RAD9, the DNA Damage Response, and Regulation of Transcription Networks. Radiat Res 2017; 187:424-432. [PMID: 28140789 DOI: 10.1667/rr003cc.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The way cells respond to DNA damage is important since inefficient repair or misrepair of lesions can have deleterious consequences, including mutation, genomic instability, neurodegenerative disorders, premature aging, cancer or death. Whether damage occurs spontaneously as a byproduct of normal metabolic processes, or after exposure to exogenous agents, cells muster a coordinated, complex DNA damage response (DDR) to mitigate potential harmful effects. A variety of activities are involved to promote cell survival, and include DNA repair, DNA damage tolerance, as well as transient cell cycle arrest to provide time for repair before entry into critical cell cycle phases, an event that could be lethal if traversal occurs while damage is present. When such damage is prolonged or not repairable, senescence, apoptosis or autophagy is induced. One major level of DDR regulation occurs via the orchestrated transcriptional control of select sets of genes encoding proteins that mediate the response. p53 is a transcription factor that transactivates specific DDR downstream genes through binding DNA consensus sequences usually in or near target gene promoter regions. The profile of p53-regulated genes activated at any given time varies, and is dependent upon type of DNA damage or stress experienced, exact composition of the consensus DNA binding sequence, presence of other DNA binding proteins, as well as cell context. RAD9 is another protein critical for the response of cells to DNA damage, and can also selectively regulate gene transcription. The limited studies addressing the role of RAD9 in transcription regulation indicate that the protein transactivates at least one of its target genes, p21/waf1/cip1, by binding to DNA sequences demonstrated to be a p53 response element. NEIL1 is also regulated by RAD9 through a similar DNA sequence, though not yet directly verified as a bonafide p53 response element. These findings suggest a novel pathway whereby p53 and RAD9 control the DDR through a shared mechanism involving an overlapping network of downstream target genes. Details and unresolved questions about how these proteins coordinate or compete to execute the DDR through transcriptional reprogramming, as well as biological implications, are discussed.
Collapse
Affiliation(s)
- Howard B Lieberman
- a Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032; and.,b Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| | - Sunil K Panigrahi
- a Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032; and
| | - Kevin M Hopkins
- a Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032; and
| | - Li Wang
- a Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032; and
| | - Constantinos G Broustas
- a Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, New York 10032; and
| |
Collapse
|
78
|
Meng X, Tan J, Li M, Song S, Miao Y, Zhang Q. Sirt1: Role Under the Condition of Ischemia/Hypoxia. Cell Mol Neurobiol 2017; 37:17-28. [PMID: 26971525 PMCID: PMC11482061 DOI: 10.1007/s10571-016-0355-2] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022]
Abstract
Silent information regulator factor 2-related enzyme 1 (sirtuin 1, Sirt1) is a nicotinamide adenine dinucleotide-dependent deacetylase, which can deacetylate histone and non-histone proteins and other transcription factors, and is involved in the regulation of many physiological functions, including cell senescence, gene transcription, energy balance, and oxidative stress. Ischemia/hypoxia injury remains an unresolved and complicated situation in the diseases of ischemia stroke, heart failure, and coronary heart disease, especially among the old folks. Studies have demonstrated that aging could enhance the vulnerability of brain, heart, lung, liver, and kidney to ischemia/hypoxia injury and the susceptibility in old folks to ischemia/hypoxia injury might be associated with Sirt1. In this review, we mainly summarize the role of Sirt1 in modulating pathways against energy depletion and its involvement in oxidative stress, apoptosis, and inflammation under the condition of ischemia/hypoxia.
Collapse
Affiliation(s)
- Xiaofei Meng
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Mengmeng Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | - Shuling Song
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, China.
| |
Collapse
|
79
|
|
80
|
Fink S, Tsai MH, Schnitzler P, Zeier M, Dreger P, Wuchter P, Bulut OC, Behrends U, Delecluse HJ. The Epstein-Barr virus DNA load in the peripheral blood of transplant recipients does not accurately reflect the burden of infected cells. Transpl Int 2016; 30:57-67. [PMID: 27717030 DOI: 10.1111/tri.12871] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 08/06/2016] [Accepted: 09/30/2016] [Indexed: 12/27/2022]
Abstract
Transplant recipients frequently exhibit an increased Epstein-Barr virus (EBV) load in the peripheral blood. Here, we quantitated the EBV-infected cells in the peripheral blood of these patients and defined the mode of viral infection, latent or lytic. These data indicated that there is no strong correlation between the number of infected cells and the EBV load (EBVL). This can be explained by a highly variable number of EBV copies per infected cell and by lytic replication in some cells. The plasma of these patients did not contain any free infectious viruses, but contained nevertheless EBV DNA, sometimes in large amounts, that probably originates from cell debris and contributed to the total EBVL. Some of the investigated samples carried a highly variable number of infected cells in active latency, characterized by an expression of the Epstein-Barr nuclear antigens (EBNA2) protein. However, a third of the samples expressed neither EBNA2 nor lytic proteins. Patients with an increased EBVL represent a heterogeneous group of patients whose infection cannot be characterized by this method alone. Precise characterization of the origin of an increased EBVL, in particular, in terms of the number of EBV-infected cells, requires additional investigations including the number of EBV-encoded small RNA-positive cells.
Collapse
Affiliation(s)
- Susanne Fink
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unit U1074, Heidelberg, Germany.,German center for infection research (DZIF), Heidelberg, Germany.,Nierenzentrum Heidelberg, Heidelberg, Germany
| | - Ming-Han Tsai
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unit U1074, Heidelberg, Germany.,German center for infection research (DZIF), Heidelberg, Germany
| | - Paul Schnitzler
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Peter Dreger
- Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Patrick Wuchter
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Olcay C Bulut
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Uta Behrends
- Children's Hospital Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Henri-Jacques Delecluse
- German Cancer Research Centre (DKFZ) Unit F100, Heidelberg, Germany.,Institut National de la Santé et de la Recherche Médicale (INSERM) Unit U1074, Heidelberg, Germany.,German center for infection research (DZIF), Heidelberg, Germany
| |
Collapse
|
81
|
A possible link between the Epstein-Barr virus infection and autoimmune thyroid disorders. Cent Eur J Immunol 2016; 41:297-301. [PMID: 27833448 PMCID: PMC5099387 DOI: 10.5114/ceji.2016.63130] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 06/21/2016] [Indexed: 11/17/2022] Open
Abstract
The Epstein-Barr virus (EBV), also known as human herpesvirus 4, is a member of the Herpesviridae virus family. EBV infection can cause infectious mononucleosis (IM) in the lytic phase of EBV's life cycle. Past EBV infection is associated with lymphomas, and may also result in certain allergic and autoimmune diseases. Although potential mechanisms of autoimmune diseases have not been clearly elucidated, both genetic and environmental factors, such as infectious agents, are considered to be responsible for their development. In addition, EBV modifies the host immune response. The worldwide prevalence of autoimmune diseases shows how common this pathogen is. Normally, the virus stays in the body and remains dormant throughout life. However, this is not always the case, and a serious EBV-related illness may develop later in life. This explains the chronic course of autoimmune diseases that is often accompanied by exacerbations of symptoms. Based on the present studies, EBV infection can cause autoimmune diseases, such as systemic lupus erythematosus (SLE), multiple sclerosis (MS), rheumatoid arthritis (RA), Sjögren's syndrome, and autoimmune hepatitis. The EBV has also been reported in patients with autoimmune thyroid disorders. Although EBV is not the only agent responsible for the development of autoimmune thyroid diseases, it can be considered a contributory factor.
Collapse
|
82
|
Thyroid hormones improve cardiac function and decrease expression of pro-apoptotic proteins in the heart of rats 14 days after infarction. Apoptosis 2016; 21:184-94. [PMID: 26659365 DOI: 10.1007/s10495-015-1204-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Apoptosis is a key process associated with pathological cardiac remodelling in early-phase post-myocardial infarction. In this context, several studies have demonstrated an anti-apoptotic effect of thyroid hormones (TH). The aim of this study was to evaluate the effects of TH on the expression of proteins associated with the apoptotic process 14 days after infarction. Male Wistar rats (300-350 g) (n = 8/group) were divided into four groups: Sham-operated (SHAM), infarcted (AMI), sham-operated + TH (SHAMT) and infarcted + TH (AMIT). For 12 days, the animals received T3 and T4 [2 and 8 µg/(100 g day)] by gavage. After this, the rats were submitted to haemodynamic and echocardiographic analysis, and then were sacrificed and the heart tissue was collected for molecular analysis. Statistical analyses included two-way ANOVA with Student-Newman-Keuls post test. Ethics Committee number: 23262. TH administration prevented the loss of ventricular wall thickness and improved cardiac function in the infarcted rats 14 days after the injury. AMI rats presented an increase in the pro-apoptotic proteins p53 and JNK. The hormonal treatment prevented this increase in AMIT rats. In addition, TH administration decreased the Bax:Bcl-2 ratio in the infarcted rats. TH administration improved cardiac functional parameters, and decreased the expression of pro-apoptotic proteins 14 days after myocardial infarction.
Collapse
|
83
|
Zhang W, Han D, Wan P, Pan P, Cao Y, Liu Y, Wu K, Wu J. ERK/c-Jun Recruits Tet1 to Induce Zta Expression and Epstein-Barr Virus Reactivation through DNA Demethylation. Sci Rep 2016; 6:34543. [PMID: 27708396 PMCID: PMC5052586 DOI: 10.1038/srep34543] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/15/2016] [Indexed: 12/13/2022] Open
Abstract
DNA demethylation plays an essential role in the reactivation of Epstein-Barr virus (EBV) from latency infection. However, it is unclear how epigenetic modification is initiated in responding to stimuli. Here, we demonstrate that ERK/c-Jun signaling is involved in DNA demethylation of EBV immediate early (IE) gene Zta in response to 12-O-Tetradecanoylphorbol-13-acetate (TPA) stimulation. Remarkably, Ser73 phosphorylation of c-Jun facilitates Zta promoter demethylation and EBV reactivation, whereas knockdown of c-Jun attenuates Zta demethylation and viral reactivation. More importantly, we reveal for the first time that c-Jun interacts with DNA dioxygenase Tet1 and facilitates Tet1 to bind to Zta promoter. The binding of c-Jun and Tet1 to Zta enhances promoter demethylation, resulting in the activation of Zta, the stimulation of BHRF1 (a lytic early gene) and gp350/220 (a lytic late gene), and ultimately the reactivation of EBV. Knockdown of Tet1 attenuates TPA-induced Zta demethylation and EBV reactivation. Thus, TPA activates ERK/c-Jun signaling, which subsequently facilitates Tet1 to bind to Zta promoter, leading to DNA demethylation, gene expression, and EBV reactivation. This study reveals important roles of ERK/c-Jun signaling and Tet1 dioxygenase in epigenetic modification, and provides new insights into the mechanism underlying the regulation of virus latent and lytic infection.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Dongjie Han
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Pin Wan
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Pan Pan
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yanhua Cao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yingle Liu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kailang Wu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jianguo Wu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
84
|
Affiliation(s)
- Gregory F. Erickson
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
85
|
Baudot AD, Crighton D, O'Prey J, Somers J, Sierra Gonzalez P, Ryan KM. p53 directly regulates the glycosidase FUCA1 to promote chemotherapy-induced cell death. Cell Cycle 2016; 15:2299-308. [PMID: 27315169 PMCID: PMC5004703 DOI: 10.1080/15384101.2016.1191714] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/11/2016] [Accepted: 05/14/2016] [Indexed: 12/16/2022] Open
Abstract
p53 is a central factor in tumor suppression as exemplified by its frequent loss in human cancer. p53 exerts its tumor suppressive effects in multiple ways, but the ability to invoke the eradication of damaged cells by programmed cell death is considered a key factor. The ways in which p53 promotes cell death can involve direct activation or engagement of the cell death machinery, or can be via indirect mechanisms, for example though regulation of ER stress and autophagy. We present here another level of control in p53-mediated tumor suppression by showing that p53 activates the glycosidase, FUCA1, a modulator of N-linked glycosylation. We show that p53 transcriptionally activates FUCA1 and that p53 modulates fucosidase activity via FUCA1 up-regulation. Importantly, we also report that chemotherapeutic drugs induce FUCA1 and fucosidase activity in a p53-dependent manner. In this context, while we found that over-expression of FUCA1 does not induce cell death, RNAi-mediated knockdown of endogenous FUCA1 significantly attenuates p53-dependent, chemotherapy-induced apoptotic death. In summary, these findings add an additional component to p53s tumor suppressive response and highlight another mechanism by which the tumor suppressor controls programmed cell death that could potentially be exploited for cancer therapy.
Collapse
Affiliation(s)
- Alice D. Baudot
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | - Diane Crighton
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | - Jim O'Prey
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | - Joanna Somers
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| | | | - Kevin M. Ryan
- Cancer Research UK Beatson Institute, Garscube Estate, Glasgow, Scotland, UK
| |
Collapse
|
86
|
López I, Tournillon AS, Nylander K, Fåhraeus R. p53-mediated control of gene expression via mRNA translation during Endoplasmic Reticulum stress. Cell Cycle 2016; 14:3373-8. [PMID: 26397130 PMCID: PMC4825612 DOI: 10.1080/15384101.2015.1090066] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
p53 is activated by different stress and damage pathways and regulates cell biological responses including cell cycle arrest, repair pathways, apoptosis and senescence. Following DNA damage, the levels of p53 increase and via binding to target gene promoters, p53 induces expression of multiple genes including p21CDKN1A and mdm2. The effects of p53 on gene expression during the DNA damage response are well mimicked by overexpressing p53 under normal conditions. However, stress to the Endoplasmic Reticulum (ER) and the consequent Unfolded Protein Response (UPR) leads to the induction of the p53/47 isoform that lacks the first 40 aa of p53 and to an active suppression of p21CDKN1A transcription and mRNA translation. We now show that during ER stress p53 also suppresses MDM2 protein levels via a similar mechanism. These observations not only raise questions about the physiological role of MDM2 during ER stress but it also reveals a new facet of p53 as a repressor toward 2 of its major target genes during the UPR. As suppression of p21CDKN1A and MDM2 protein synthesis is mediated via their coding sequences, it raises the possibility that p53 controls mRNA translation via a common mechanism that might play an important role in how p53 regulates gene expression during the UPR, as compared to the transcription-dependent gene regulation taking place during the DNA damage response.
Collapse
Affiliation(s)
- Ignacio López
- a Équipe Labellisée Ligue Contre le Cancer; Université Paris 7; INSERM UMR 1162 "Génomique fonctionnelle des tumeurs solides" ; Paris , France
| | - Anne-Sophie Tournillon
- a Équipe Labellisée Ligue Contre le Cancer; Université Paris 7; INSERM UMR 1162 "Génomique fonctionnelle des tumeurs solides" ; Paris , France
| | - Karin Nylander
- b Department of Medical Biosciences ; Umeå University ; Umeå , Sweden
| | - Robin Fåhraeus
- a Équipe Labellisée Ligue Contre le Cancer; Université Paris 7; INSERM UMR 1162 "Génomique fonctionnelle des tumeurs solides" ; Paris , France ,b Department of Medical Biosciences ; Umeå University ; Umeå , Sweden.,c RECAMO; Masaryk Memorial Cancer Institute ; Brno , Czech Republic
| |
Collapse
|
87
|
Positive feedback regulation of p53 transactivity by DNA damage-induced ISG15 modification. Nat Commun 2016; 7:12513. [PMID: 27545325 PMCID: PMC4996943 DOI: 10.1038/ncomms12513] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/11/2016] [Indexed: 12/20/2022] Open
Abstract
p53 plays a pivotal role in tumour suppression under stresses, such as DNA damage. ISG15 has been implicated in the control of tumorigenesis. Intriguingly, the expression of ISG15, UBE1L and UBCH8 is induced by DNA-damaging agents, such as ultraviolet and doxorubicin, which are known to induce p53. Here, we show that the genes encoding ISG15, UBE1L, UBCH8 and EFP, have the p53-responsive elements and their expression is induced in a p53-dependent fashion under DNA damage conditions. Furthermore, DNA damage induces ISG15 conjugation to p53 and this modification markedly enhances the binding of p53 to the promoters of its target genes (for example, CDKN1 and BAX) as well as of its own gene by promoting phosphorylation and acetylation, leading to suppression of cell growth and tumorigenesis. These findings establish a novel feedback circuit between p53 and ISG15-conjugating system for positive regulation of the tumour suppressive function of p53 under DNA damage conditions. The ‘genome guardian' p53 has a well-established role in suppressing tumour development after DNA damage. Here the authors show that expression of the ubiquitin-like protein ISG15 is regulated by p53 which in turn is modified by ISG15 to enhance binding to target gene promoters.
Collapse
|
88
|
Cieniewicz B, Santana AL, Minkah N, Krug LT. Interplay of Murine Gammaherpesvirus 68 with NF-kappaB Signaling of the Host. Front Microbiol 2016; 7:1202. [PMID: 27582728 PMCID: PMC4987367 DOI: 10.3389/fmicb.2016.01202] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/19/2016] [Indexed: 11/13/2022] Open
Abstract
Herpesviruses establish a chronic infection in the host characterized by intervals of lytic replication, quiescent latency, and reactivation from latency. Murine gammaherpesvirus 68 (MHV68) naturally infects small rodents and has genetic and biologic parallels with the human gammaherpesviruses (gHVs), Kaposi's sarcoma-associated herpesvirus and Epstein-Barr virus. The murine gammaherpesvirus model pathogen system provides a platform to apply cutting-edge approaches to dissect the interplay of gammaherpesvirus and host determinants that enable colonization of the host, and that shape the latent or lytic fate of an infected cell. This knowledge is critical for the development of novel therapeutic interventions against the oncogenic gHVs. The nuclear factor kappa B (NF-κB) signaling pathway is well-known for its role in the promotion of inflammation and many aspects of B cell biology. Here, we review key aspects of the virus lifecycle in the host, with an emphasis on the route that the virus takes to gain access to the B cell latency reservoir. We highlight how the murine gammaherpesvirus requires components of the NF-κB signaling pathway to promote replication, latency establishment, and maintenance of latency. These studies emphasize the complexity of gammaherpesvirus interactions with NF-κB signaling components that direct innate and adaptive immune responses of the host. Importantly, multiple facets of NF-κB signaling have been identified that might be targeted to reduce the burden of gammaherpesvirus-associated diseases.
Collapse
Affiliation(s)
- Brandon Cieniewicz
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Alexis L Santana
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Nana Minkah
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| | - Laurie T Krug
- Department of Molecular Genetics and Microbiology, Stony Brook University, Stony Brook NY, USA
| |
Collapse
|
89
|
Jonak K, Kurpas M, Szoltysek K, Janus P, Abramowicz A, Puszynski K. A novel mathematical model of ATM/p53/NF- κB pathways points to the importance of the DDR switch-off mechanisms. BMC SYSTEMS BIOLOGY 2016; 10:75. [PMID: 27526774 PMCID: PMC4986247 DOI: 10.1186/s12918-016-0293-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 06/27/2016] [Indexed: 12/20/2022]
Abstract
Background Ataxia telangiectasia mutated (ATM) is a detector of double-strand breaks (DSBs) and a crucial component of the DNA damage response (DDR) along with p53 and NF- κB transcription factors and Wip1 phosphatase. Despite the recent advances in studying the DDR, the mechanisms of cell fate determination after DNA damage induction is still poorly understood. Results To investigate the importance of various DDR elements with particular emphasis on Wip1, we developed a novel mathematical model of ATM/p53/NF- κB pathways. Our results from in silico and in vitro experiments performed on U2-OS cells with Wip1 silenced to 25 % (Wip1-RNAi) revealed a strong dependence of cellular response to DNA damages on this phosphatase. Notably, Wip1-RNAi cells exhibited lower resistance to ionizing radiation (IR) resulting in smaller clonogenicity and higher apoptotic fraction. Conclusions In this article, we demonstrated that Wip1 plays a role as a gatekeeper of apoptosis and influences the pro-survival behaviour of cells – the level of Wip1 increases to block the apoptotic decision when DNA repair is successful. Moreover, we were able to verify the dynamics of proteins and transcripts, apoptotic fractions and cells viability obtained from stochastic simulations using in vitro approaches. Taken together, we demonstrated that the model can be successfully used in prediction of cellular behaviour after exposure to IR. Thus, our studies may provide further insights into key elements involved in the underlying mechanisms of the DDR. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0293-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katarzyna Jonak
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka, Gliwice, 16, 44-100, Poland
| | - Monika Kurpas
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka, Gliwice, 16, 44-100, Poland
| | - Katarzyna Szoltysek
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wybrzeze Armii Krajowej, Gliwice, 15, 44-400, Poland
| | - Patryk Janus
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wybrzeze Armii Krajowej, Gliwice, 15, 44-400, Poland
| | - Agata Abramowicz
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Wybrzeze Armii Krajowej, Gliwice, 15, 44-400, Poland
| | - Krzysztof Puszynski
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka, Gliwice, 16, 44-100, Poland.
| |
Collapse
|
90
|
Rewiring of the apoptotic TGF-β-SMAD/NFκB pathway through an oncogenic function of p27 in human papillary thyroid cancer. Oncogene 2016; 36:652-666. [PMID: 27452523 DOI: 10.1038/onc.2016.233] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/29/2016] [Accepted: 05/24/2016] [Indexed: 12/25/2022]
Abstract
Papillary thyroid carcinoma (PTC), the most frequent thyroid cancer, is characterized by low proliferation but no apoptosis, presenting frequent lymph-node metastasis. Papillary thyroid carcinoma overexpress transforming growth factor-beta (TGF-β). In human cells, TGF-β has two opposing actions: antitumoral through pro-apoptotic and cytostatic activities, and pro-tumoral promoting growth and metastasis. The switch converting TGF-β from a tumor-suppressor to tumor-promoter has not been identified. In the current study, we have quantified a parallel upregulation of TGF-β and nuclear p27, a CDK2 inhibitor, in samples from PTC. We established primary cultures from follicular epithelium in human homeostatic conditions (h7H medium). TGF-β-dependent cytostasis occurred in normal and cancer cells through p15/CDKN2B induction. However, TGF-β induced apoptosis in normal and benign but not in carcinoma cultures. In normal thyroid cells, TGF-β/SMAD repressed the p27/CDKN1B gene, activating CDK2-dependent SMAD3 phosphorylation to induce p50 NFκB-dependent BAX upregulation and apoptosis. In thyroid cancer cells, oncogene activation prevented TGF-β/SMAD-dependent p27 repression, and CDK2/SMAD3 phosphorylation, leading to p65 NFκB upregulation which repressed BAX, induced cyclin D1 and promoted TGF-β-dependent growth. In PTC samples from patients, upregulation of TGF-β, p27, p65 and cyclin D1 mRNA were significantly correlated, while the expression of the isoform BAX-β, exclusively transcribed in apoptotic cells, was negatively correlated. Additionally, combined ERK and p65 NFκB inhibitors reduced p27 expression and potentiated apoptosis in thyroid cancer cells while not affecting survival in normal thyroid cells. Our results therefore suggest that the oncoprotein p27 reorganizes the effects of TGF-β in thyroid cancer, explaining the slow proliferation but lack of apoptosis and metastatic behavior of PTC.
Collapse
|
91
|
Senichkin VV, Kopeina GS, Zamaraev AV, Lavrik IN, Zhivotovsky BD. Nutrient restriction in combinatory therapy of tumors. Mol Biol 2016. [DOI: 10.1134/s0026893316030109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
92
|
Kapur A, Felder M, Fass L, Kaur J, Czarnecki A, Rathi K, Zeng S, Osowski KK, Howell C, Xiong MP, Whelan RJ, Patankar MS. Modulation of oxidative stress and subsequent induction of apoptosis and endoplasmic reticulum stress allows citral to decrease cancer cell proliferation. Sci Rep 2016; 6:27530. [PMID: 27270209 PMCID: PMC4897611 DOI: 10.1038/srep27530] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/13/2016] [Indexed: 12/17/2022] Open
Abstract
The monoterpenoid, citral, when delivered through PEG-b-PCL nanoparticles inhibits in vivo growth of 4T1 breast tumors. Here, we show that citral inhibits proliferation of multiple human cancer cell lines. In p53 expressing ECC-1 and OVCAR-3 but not in p53-deficient SKOV-3 cells, citral induces G1/S cell cycle arrest and apoptosis as determined by Annexin V staining and increased cleaved caspase3 and Bax and decreased Bcl-2. In SKOV-3 cells, citral induces the ER stress markers CHOP, GADD45, EDEM, ATF4, Hsp90, ATG5, and phospho-eIF2α. The molecular chaperone 4-phenylbutyric acid attenuates citral activity in SKOV-3 but not in ECC-1 and OVCAR-3 cells. In p53-expressing cells, citral increases phosphorylation of serine-15 of p53. Activation of p53 increases Bax, PUMA, and NOXA expression. Inhibition of p53 by pifithrin-α, attenuates citral-mediated apoptosis. Citral increases intracellular oxygen radicals and this leads to activation of p53. Inhibition of glutathione synthesis by L-buthionine sulfoxamine increases potency of citral. Pretreatment with N-acetylcysteine decreases phosphorylation of p53 in citral-treated ECC-1 and OVCAR-3. These results define a p53-dependent, and in the absence of p53, ER stress-dependent mode of action of citral. This study indicates that citral in PEG-b-PCL nanoparticle formulation should be considered for treatment of breast and other tumors.
Collapse
Affiliation(s)
- Arvinder Kapur
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| | - Mildred Felder
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| | - Lucas Fass
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| | - Justanjot Kaur
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| | - Austin Czarnecki
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| | - Kavya Rathi
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| | - San Zeng
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705-2222, USA
| | | | - Colin Howell
- Department of Chemistry and Biochemistry, Oberlin College, Oberlin, OH 44704, USA
| | - May P Xiong
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705-2222, USA
| | - Rebecca J Whelan
- Department of Chemistry and Biochemistry, Oberlin College, Oberlin, OH 44704, USA
| | - Manish S Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI-53792-6188, USA
| |
Collapse
|
93
|
Fukuda K, Uehara Y, Nakata E, Inoue M, Shimazu K, Yoshida T, Kanda H, Nanjo H, Hosoi Y, Yamakoshi H, Iwabuchi Y, Shibata H. A diarylpentanoid curcumin analog exhibits improved radioprotective potential in the intestinal mucosa. Int J Radiat Biol 2016; 92:388-94. [PMID: 27043482 DOI: 10.3109/09553002.2016.1164910] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To best enhance the effects of radiotherapy, it is important to minimize adverse events, including free radical-induced intestinal cell damage. Given the threat of nuclear power plant accidents or nuclear terrorism, there is an urgent need for radioprotectants to counteract the radiation-induced toxicity and/or injuries. Curcumin exhibits protective effects against gamma irradiation; however, its in vivo efficacy is decreased due to the low bioavailability. We examined the radioprotective effect of a newly synthesized curcumin analog, GO-Y031, on 11-Gy X-ray-induced intestinal mucosal damage in mice. MATERIALS AND METHODS The radioprotection experiments were conducted by using C57BL/6J or Jcl:ICR mice. Molecules related to radiation damage, including p53, Bax, Bcl-2, cleaved caspase-3, and reactive carbonyl species (RCS), were investigated immunohistochemically. RESULTS GO-Y031 protected against crypt hypoplasia relative to a mock treatment at 0.5% (weight/weight); the number of crypts were 11.00 ± 2.00/circumference (mm) in treated versus 6.86 ± 0.99/mm in mock-treated C57BL/6 mice (p = 0.0079). GO-Y031 also reduced the levels of RCS, p53, and cleaved caspase-3 accumulation in the irradiated intestinal cells. CONCLUSIONS GO-Y031 suppresses the accumulation of RCS and apoptosis-related molecules in irradiated cells. This compound may be a good primary radioprotective compound.
Collapse
Affiliation(s)
- Koji Fukuda
- a Department of Clinical Oncology , Graduate School of Medicine, Akita University , Akita , Japan
| | - Yoshihiko Uehara
- b Laboratory for Radiation Biology , Tohoku University , Sendai , Japan
| | - Eiko Nakata
- c Department of Radiation Oncology , Graduate School of Medicine, Tohoku University , Sendai , Japan
| | - Masahiro Inoue
- a Department of Clinical Oncology , Graduate School of Medicine, Akita University , Akita , Japan
| | - Kazuhiro Shimazu
- a Department of Clinical Oncology , Graduate School of Medicine, Akita University , Akita , Japan
| | - Taichi Yoshida
- a Department of Clinical Oncology , Graduate School of Medicine, Akita University , Akita , Japan
| | - Hiroaki Kanda
- d Division of Pathology , Cancer Institute, Japanese Foundation for Cancer Research , Tokyo , Japan
| | - Hiroshi Nanjo
- e Department of Clinical Pathology , Graduate School of Medicine, Akita University , Akita , Japan
| | - Yoshio Hosoi
- b Laboratory for Radiation Biology , Tohoku University , Sendai , Japan
| | - Hiroyuki Yamakoshi
- f Department of Organic Chemistry , Graduate School of Pharmaceutical, Tohoku University , Sendai , Japan
| | - Yoshiharu Iwabuchi
- f Department of Organic Chemistry , Graduate School of Pharmaceutical, Tohoku University , Sendai , Japan
| | - Hiroyuki Shibata
- a Department of Clinical Oncology , Graduate School of Medicine, Akita University , Akita , Japan
| |
Collapse
|
94
|
Liao JM, Cao B, Deng J, Zhou X, Strong M, Zeng S, Xiong J, Flemington E, Lu H. TFIIS.h, a new target of p53, regulates transcription efficiency of pro-apoptotic bax gene. Sci Rep 2016; 6:23542. [PMID: 27005522 PMCID: PMC4804275 DOI: 10.1038/srep23542] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/03/2016] [Indexed: 11/09/2022] Open
Abstract
Tumor suppressor p53 transcriptionally regulates hundreds of genes involved in various cellular functions. However, the detailed mechanisms underlying the selection of p53 targets in response to different stresses are still elusive. Here, we identify TFIIS.h, a transcription elongation factor, as a new transcriptional target of p53, and also show that it can enhance the efficiency of transcription elongation of apoptosis-associated bax gene, but not cell cycle-associated p21 (CDKN1A) gene. TFIIS.h is revealed as a p53 target through microarray analysis of RNAs extracted from cells treated with or without inauhzin (INZ), a p53 activator, and further confirmed by RT-q-PCR, western blot, luciferase reporter, and ChIP assays. Interestingly, knocking down TFIIS.h impairs, but overexpressing TFIIS.h promotes, induction of bax, but not other p53 targets including p21, by p53 activation. In addition, overexpression of TFIIS.h induces cell death in a bax- dependent fashion. These findings reveal a mechanism by which p53 utilizes TFIIS.h to selectively promote the transcriptional elongation of the bax gene, upsurging cell death in response to severe DNA damage.
Collapse
Affiliation(s)
- Jun-Ming Liao
- Department of Biochemistry &Molecular Biology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| | - Bo Cao
- Department of Biochemistry &Molecular Biology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| | - Jun Deng
- Department of Biochemistry &Molecular Biology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA.,Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Xiang Zhou
- Department of Biochemistry &Molecular Biology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| | - Michael Strong
- Department of Pathology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| | - Shelya Zeng
- Department of Biochemistry &Molecular Biology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Erik Flemington
- Department of Pathology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| | - Hua Lu
- Department of Biochemistry &Molecular Biology and Cancer Center, Tulane University School of Medicine, 1430, Louisiana, LA 70112, USA
| |
Collapse
|
95
|
Li W, Jiang P, Sun X, Xu S, Ma X, Zhan R. Suppressing H19 Modulates Tumorigenicity and Stemness in U251 and U87MG Glioma Cells. Cell Mol Neurobiol 2016; 36:1219-1227. [PMID: 26983719 PMCID: PMC5047947 DOI: 10.1007/s10571-015-0320-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/18/2015] [Indexed: 01/01/2023]
Abstract
Glioblastoma multiforme (GBM) is a type of malignant carcinoma found in the brain. Its high frequency of occurrence and poor survival rate have garnered much research attention in recent years. Long non-coding RNAs (lncRNAs) are known to be related to the formation and progression of several cancer types by both promoting and suppressing tumor transformation. H19 is one such lncRNA and has been shown to be upregulated in a few types of cancer. In this study, we discovered that the expression of H19 increased in GBM cell lines. H19 knocked down GBM cells also displayed decreased cellular proliferation and a higher apoptosis rate when induced by temozolomide. Interestingly, the GBM cell lines U87MG and U251 were found to express cancer stem cell markers CD133, NANOG, Oct4 and Sox2. Expression of these markers was downregulated in H19-deficient cells. Collectively, these data suggest a role for H19 in contributing to GBM malignancy and the maintenance of its stem cell properties.
Collapse
Affiliation(s)
- Weiguo Li
- Neurosurgery Department, Qilu Hospital, Shandong University, 107 Wenhua West Road, Lixia District, Jinan, 250012, Shandong, China.
| | | | | | - Shujun Xu
- Neurosurgery Department, Qilu Hospital, Shandong University, 107 Wenhua West Road, Lixia District, Jinan, 250012, Shandong, China
| | - Xiangyu Ma
- Neurosurgery Department, Qilu Hospital, Shandong University, 107 Wenhua West Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rucai Zhan
- Neurosurgery Department of No. 3 Hospital of Jinan, Jinan, China
| |
Collapse
|
96
|
Wang Y, Chen Y, Yan Y, Li X, Chen G, He N, Shen S, Chen G, Zhang C, Liao W, Liao Y, Bin J. Loss of CEACAM1, a Tumor-Associated Factor, Attenuates Post-infarction Cardiac Remodeling by Inhibiting Apoptosis. Sci Rep 2016; 6:21972. [PMID: 26911181 PMCID: PMC4766464 DOI: 10.1038/srep21972] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/29/2016] [Indexed: 12/30/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule1 (CEACAM1) is a tumor-associated factor that is known to be involved in apoptosis, but the role of CEACAM1 in cardiovascular disease is unclear. We aims to investigate whether CEACAM1 influences cardiac remodeling in mice with myocardial infarction (MI) and hypoxia-induced cardiomyocyte injury. Both serum in patients and myocardial CEACAM1 levels in mice were significantly increased in response to MI, while levels were elevated in neonatal rat cardiomyocytes (NRCs) exposed to hypoxia. Eight weeks after MI, a lower mortality rate, improved cardiac function, and less cardiac remodeling in CEACAM1 knock-out (KO) mice than in their wild-type (WT) littermates were observed. Moreover, myocardial expression of mitochondrial Bax, cytosolic cytochrome C, and cleaved caspase-3 was significantly lower in CEACAM1 KO mice than in WT mice. In cultured NRCs exposed to hypoxia, recombinant human CEACAM1 (rhCEACAM1) reduced mitochondrial membrane potential, upregulated mitochondrial Bax, increased cytosolic cytochrome C and cleaved caspase-3, and consequently increased apoptosis. RhCEACAM1 also increased the levels of GRP78 and CHOP in NRCs with hypoxia. All of these effects were abolished by silencing CEACAM1. Our study indicates that CEACAM1 exacerbates hypoxic cardiomyocyte injury and post-infarction cardiac remodeling by enhancing cardiomyocyte mitochondrial dysfunction and endoplasmic reticulum stress-induced apoptosis.
Collapse
Affiliation(s)
- Yan Wang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanmei Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yi Yan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinzhong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guojun Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Nvqin He
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuxin Shen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Cardiology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, 450003, China
| | - Gangbin Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chuanxi Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
97
|
Wolfsperger F, Hogh-Binder SA, Schittenhelm J, Psaras T, Ritter V, Bornes L, Huber SM, Jendrossek V, Rudner J. Deubiquitylating enzyme USP9x regulates radiosensitivity in glioblastoma cells by Mcl-1-dependent and -independent mechanisms. Cell Death Dis 2016; 7:e2039. [PMID: 26775694 PMCID: PMC4816183 DOI: 10.1038/cddis.2015.405] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 11/13/2015] [Accepted: 12/11/2015] [Indexed: 12/24/2022]
Abstract
Glioblastoma is a very aggressive form of brain tumor with limited therapeutic options. Usually, glioblastoma is treated with ionizing radiation (IR) and chemotherapy after surgical removal. However, radiotherapy is frequently unsuccessful, among others owing to resistance mechanisms the tumor cells have developed. Antiapoptotic B-cell leukemia (Bcl)-2 family members can contribute to radioresistance by interfering with apoptosis induction in response to IR. Bcl-2 and the closely related Bcl-xL and Mcl-1 are often overexpressed in glioblastoma cells. In contrast to Bcl-2 and Bcl-xL, Mcl-1 is a short-lived protein whose stability is closely regulated by ubiquitylation-dependent proteasomal degradation. Although ubiquitin ligases facilitate degradation, the deubiquitylating enzyme ubiquitin-specific protease 9x (USP9x) interferes with degradation by removing polyubiquitin chains from Mcl-1, thereby stabilizing this protein. Thus, an inability to downregulate Mcl-1 by enhanced USP9x activity might contribute to radioresistance. Here we analyzed the impact of USP9x on Mcl-1 levels and radiosensitivity in glioblastoma cells. Correlating Mcl-1 and USP9x expressions were significantly higher in human glioblastoma than in astrocytoma. Downregulation of Mcl-1 correlated with apoptosis induction in established glioblastoma cell lines. Although Mcl-1 knockdown by siRNA increased apoptosis induction after irradiation in all glioblastoma cell lines, USP9x knockdown significantly improved radiation-induced apoptosis in one of four cell lines and slightly increased apoptosis in another cell line. In the latter two cell lines, USP9x knockdown also increased radiation-induced clonogenic death. The massive downregulation of Mcl-1 and apoptosis induction in A172 cells transfected with USP9x siRNA shows that the deubiquitinase regulates cell survival by regulating Mcl-1 levels. In contrast, USP9x regulated radiosensitivity in Ln229 cells without affecting Mcl-1 levels. We conclude that USP9x can control survival and radiosensitivity in glioblastoma cells by Mcl-1-dependent and Mcl-1-independent mechanisms.
Collapse
Affiliation(s)
- F Wolfsperger
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - S A Hogh-Binder
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - J Schittenhelm
- Department of Neuropathology, Institute of Pathology and Neuropathology, University of Tuebingen, Tuebingen, Germany
| | - T Psaras
- Department of Neurosurgery, University of Tuebingen, Tuebingen, Germany
| | - V Ritter
- Institute for Cell Biology, University Hospital Essen, Essen, Germany
| | - L Bornes
- Institute for Cell Biology, University Hospital Essen, Essen, Germany
| | - S M Huber
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - V Jendrossek
- Institute for Cell Biology, University Hospital Essen, Essen, Germany
| | - J Rudner
- Institute for Cell Biology, University Hospital Essen, Essen, Germany
| |
Collapse
|
98
|
Chakraborty D, Jain CK, Maity A, Ghosh S, Choudhury SR, Jha T, Majumder HK, Mondal NB. Chenopodium album metabolites act as dual topoisomerase inhibitors and induce apoptosis in the MCF7 cell line. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00502g] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Desgalactotigonin and oleanolic acid 3-O-β-d-glucuronide were isolated from Chenopodium album and were evaluated for cytotoxic activity against various cancer cell lines.
Collapse
Affiliation(s)
- Debanjana Chakraborty
- Department of Chemistry
- Indian Institute of Chemical Biology
- Council of Scientific and Industrial Research
- Kolkata 700032
- India
| | - Chetan Kumar Jain
- Molecular Parasitology Laboratory
- Indian Institute of Chemical Biology
- Council of Scientific and Industrial Research
- Kolkata 700032
- India
| | - Arindam Maity
- Department of Chemistry
- Indian Institute of Chemical Biology
- Council of Scientific and Industrial Research
- Kolkata 700032
- India
| | - Shekhar Ghosh
- Department of Chemistry
- Indian Institute of Chemical Biology
- Council of Scientific and Industrial Research
- Kolkata 700032
- India
| | - Susanta Roy Choudhury
- Division of Research, Saroj Gupta Cancer Centre & Research Institute
- Kolkata - 700 063
- India
| | - Tarun Jha
- Department of Pharmaceutical Technology
- Division of Medicinal and Pharmaceutical Chemistry
- Kolkata-700 032
- India
| | - Hemanta K. Majumder
- Molecular Parasitology Laboratory
- Indian Institute of Chemical Biology
- Council of Scientific and Industrial Research
- Kolkata 700032
- India
| | - Nirup B. Mondal
- Department of Chemistry
- Indian Institute of Chemical Biology
- Council of Scientific and Industrial Research
- Kolkata 700032
- India
| |
Collapse
|
99
|
Clewell RA, McMullen PD, Adeleye Y, Carmichael PL, Andersen ME. Pathway Based Toxicology and Fit-for-Purpose Assays. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 856:205-230. [DOI: 10.1007/978-3-319-33826-2_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
100
|
Zhang M, Zhu X, Zhang Y, Cai Y, Chen J, Sivaprakasam S, Gurav A, Pi W, Makala L, Wu J, Pace B, Tuan-Lo D, Ganapathy V, Singh N, Li H. RCAD/Ufl1, a Ufm1 E3 ligase, is essential for hematopoietic stem cell function and murine hematopoiesis. Cell Death Differ 2015; 22:1922-34. [PMID: 25952549 PMCID: PMC4816109 DOI: 10.1038/cdd.2015.51] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/22/2015] [Accepted: 03/24/2015] [Indexed: 12/18/2022] Open
Abstract
The Ufm1 conjugation system is a novel ubiquitin-like modification system, consisting of Ufm1, Uba5 (E1), Ufc1 (E2) and poorly characterized E3 ligase(s). RCAD/Ufl1 (also known as KIAA0776, NLBP and Maxer) was reported to function as a Ufm1 E3 ligase in ufmylation (Ufm1-mediated conjugation) of DDRGK1 and ASC1 proteins. It has also been implicated in estrogen receptor signaling, unfolded protein response (UPR) and neurodegeneration, yet its physiological function remains completely unknown. In this study, we report that RCAD/Ufl1 is essential for embryonic development, hematopoietic stem cell (HSC) survival and erythroid differentiation. Both germ-line and somatic deletion of RCAD/Ufl1 impaired hematopoietic development, resulting in severe anemia, cytopenia and ultimately animal death. Depletion of RCAD/Ufl1 caused elevated endoplasmic reticulum stress and evoked UPR in bone marrow cells. In addition, loss of RCAD/Ufl1 blocked autophagic degradation, increased mitochondrial mass and reactive oxygen species, and led to DNA damage response, p53 activation and enhanced cell death of HSCs. Collectively, our study provides the first genetic evidence for the indispensable role of RCAD/Ufl1 in murine hematopoiesis and development. The finding of RCAD/Ufl1 as a key regulator of cellular stress response sheds a light into the role of a novel protein network including RCAD/Ufl1 and its associated proteins in regulating cellular homeostasis.
Collapse
Affiliation(s)
- M Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - X Zhu
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Y Zhang
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Y Cai
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
- Department of Biology, College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - J Chen
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - S Sivaprakasam
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - A Gurav
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - W Pi
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - L Makala
- Department of Pediatrics, Georgia Regents University, Augusta, GA, USA
| | - J Wu
- Department of Periodontics, College of Dentistry, Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - B Pace
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
- Department of Pediatrics, Georgia Regents University, Augusta, GA, USA
| | - D Tuan-Lo
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - V Ganapathy
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - N Singh
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - H Li
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|