51
|
MHY440, a Novel Topoisomerase Ι Inhibitor, Induces Cell Cycle Arrest and Apoptosis via a ROS-Dependent DNA Damage Signaling Pathway in AGS Human Gastric Cancer Cells. Molecules 2018; 24:molecules24010096. [PMID: 30597845 PMCID: PMC6337620 DOI: 10.3390/molecules24010096] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/23/2018] [Accepted: 12/24/2018] [Indexed: 12/31/2022] Open
Abstract
We investigated the antitumor activity and action mechanism of MHY440 in AGS human gastric cancer cells. MHY440 inhibited topoisomerase (Topo) Ι activity and was associated with a DNA damage response signaling pathway. It exhibited a stronger anti-proliferative effect on AGS cells relative to Hs27 human foreskin fibroblast cells, and this effect was both time- and concentration-dependent. MHY440 also increased cell arrest in the G2/M phase by decreasing cyclin B1, Cdc2, and Cdc25c, and upregulating p53 and p73. MHY440 induced AGS cell apoptosis through the upregulation of Fas-L, Fas, and Bax as well as the proteolysis of BH3 interacting-domain death agonist and poly(ADP-ribose) polymerase. It also contributed to the loss of mitochondrial membrane potential. The apoptotic cell death induced by MHY440 was inhibited by pretreatment with Z-VAD-FMK, a pan-caspase inhibitor, indicating that apoptosis was caspase-dependent. Moreover, the apoptotic effect of MHY440 was reactive oxygen species (ROS)-dependent, as evidenced by the inhibition of MHY440-induced PARP cleavage and ROS generation via N-acetylcysteine-induced ROS scavenging. Taken together, MHY440 showed anticancer effects by inhibiting Topo I, regulating the cell cycle, inducing apoptosis through caspase activation, and generating ROS, suggesting that MHY440 has considerable potential as a therapeutic agent for human gastric cancer.
Collapse
|
52
|
Fan Y, Sanyal S, Bruzzone R. Breaking Bad: How Viruses Subvert the Cell Cycle. Front Cell Infect Microbiol 2018; 8:396. [PMID: 30510918 PMCID: PMC6252338 DOI: 10.3389/fcimb.2018.00396] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023] Open
Abstract
Interactions between the host and viruses during the course of their co-evolution have not only shaped cellular function and the immune system, but also the counter measures employed by viruses. Relatively small genomes and high replication rates allow viruses to accumulate mutations and continuously present the host with new challenges. It is therefore, no surprise that they either escape detection or modulate host physiology, often by redirecting normal cellular pathways to their own advantage. Viruses utilize a diverse array of strategies and molecular targets to subvert host cellular processes, while evading detection. These include cell-cycle regulation, major histocompatibility complex-restricted antigen presentation, intracellular protein transport, apoptosis, cytokine-mediated signaling, and humoral immune responses. Moreover, viruses routinely manipulate the host cell cycle to create a favorable environment for replication, largely by deregulating cell cycle checkpoints. This review focuses on our current understanding of the molecular aspects of cell cycle regulation that are often targeted by viruses. Further study of their interactions should provide fundamental insights into cell cycle regulation and improve our ability to exploit these viruses.
Collapse
Affiliation(s)
- Ying Fan
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sumana Sanyal
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,LKS Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, LKS Faculty of Medicine, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| |
Collapse
|
53
|
Aasland D, Götzinger L, Hauck L, Berte N, Meyer J, Effenberger M, Schneider S, Reuber EE, Roos WP, Tomicic MT, Kaina B, Christmann M. Temozolomide Induces Senescence and Repression of DNA Repair Pathways in Glioblastoma Cells via Activation of ATR-CHK1, p21, and NF-κB. Cancer Res 2018; 79:99-113. [PMID: 30361254 DOI: 10.1158/0008-5472.can-18-1733] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/07/2018] [Accepted: 10/22/2018] [Indexed: 11/16/2022]
Abstract
The DNA-methylating drug temozolomide, which induces cell death through apoptosis, is used for the treatment of malignant glioma. Here, we investigate the mechanisms underlying the ability of temozolomide to induce senescence in glioblastoma cells. Temozolomide-induced senescence was triggered by the specific DNA lesion O6-methylguanine (O6MeG) and characterized by arrest of cells in the G2-M phase. Inhibitor experiments revealed that temozolomide-induced senescence was initiated by damage recognition through the MRN complex, activation of the ATR/CHK1 axis of the DNA damage response pathway, and mediated by degradation of CDC25c. Temozolomide-induced senescence required functional p53 and was dependent on sustained p21 induction. p53-deficient cells, not expressing p21, failed to induce senescence, but were still able to induce a G2-M arrest. p14 and p16, targets of p53, were silenced in our cell system and did not seem to play a role in temozolomide-induced senescence. In addition to p21, the NF-κB pathway was required for senescence, which was accompanied by induction of the senescence-associated secretory phenotype. Upon temozolomide exposure, we found a strong repression of the mismatch repair proteins MSH2, MSH6, and EXO1 as well as the homologous recombination protein RAD51, which was downregulated by disruption of the E2F1/DP1 complex. Repression of these repair factors was not observed in G2-M arrested p53-deficient cells and, therefore, it seems to represent a specific trait of temozolomide-induced senescence. SIGNIFICANCE: These findings reveal a mechanism by which the anticancer drug temozolomide induces senescence and downregulation of DNA repair pathways in glioma cells.
Collapse
Affiliation(s)
- Dorthe Aasland
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Laura Götzinger
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Laura Hauck
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Nancy Berte
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Jessica Meyer
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | | | - Simon Schneider
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Emelie E Reuber
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Wynand P Roos
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany
| | - Maja T Tomicic
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany.
| | - Bernd Kaina
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany.
| | - Markus Christmann
- Department of Toxicology, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
54
|
Qi D, Hu L, Jiao T, Zhang T, Tong X, Ye X. Phosphatase Cdc25A Negatively Regulates the Antiviral Immune Response by Inhibiting TBK1 Activity. J Virol 2018; 92:e01118-18. [PMID: 30021902 PMCID: PMC6146813 DOI: 10.1128/jvi.01118-18] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 07/08/2018] [Indexed: 01/27/2023] Open
Abstract
The phosphatase Cdc25A plays an important role in cell cycle regulation by dephosphorylating its substrates, such as cyclin-dependent kinases. In this study, we demonstrate that Cdc25A negatively regulates RIG-I-mediated antiviral signaling. We found that ectopic expression of Cdc25A in 293T cells inhibits the activation of beta interferon (IFN-β) induced by Sendai virus and poly(I·C), while knockdown of Cdc25A enhances the transcription of IFN-β stimulated by RNA virus infection. The inhibitory effect of Cdc25A on the antiviral immune response is mainly dependent on its phosphatase activity. Data from a luciferase assay indicated that Cdc25A can inhibit TBK1-mediated activation of IFN-β. Further analysis indicated that Cdc25A can interact with TBK1 and reduce the phosphorylation of TBK1 at S172, which in turn decreases the phosphorylation of its downstream substrate IRF3. Consistently, knockdown of Cdc25A upregulates the phosphorylation of both TBK1-S172 and IRF3 in Sendai virus-infected or TBK1-transfected 293T cells. In addition, we confirmed that Cdc25A can directly dephosphorylate TBK1-S172-p. These results demonstrate that Cdc25A inhibits the antiviral immune response by reducing the active form of TBK1. Using herpes simplex virus 1 (HSV-1) infection, an IFN-β reporter assay, and reverse transcription-quantitative PCR (RT-qPCR), we demonstrated that Cdc25A can also inhibit DNA virus-induced activation of IFN-β. Using a vesicular stomatitis virus (VSV) infection assay, we confirmed that Cdc25A can repress the RIG-I-like receptor (RLR)-mediated antiviral immune response and influence the antiviral status of cells. In conclusion, we demonstrate that Cdc25A negatively regulates the antiviral immune response by inhibiting TBK1 activity.IMPORTANCE The RLR-mediated antiviral immune response is critical for host defense against RNA virus infection. However, the detailed mechanism for balancing the RLR signaling pathway in host cells is not well understood. We found that the phosphatase Cdc25A negatively regulates the RNA virus-induced innate immune response. Our studies indicate that Cdc25A inhibits the RLR signaling pathway via its phosphatase activity. We demonstrated that Cdc25A reduces TBK1 activity and consequently restrains the activation of IFN-β transcription as well as the antiviral status of nearby cells. We showed that Cdc25A can also inhibit DNA virus-induced activation of IFN-β. Taken together, our findings uncover a novel function and mechanism for Cdc25A in regulating antiviral immune signaling. These findings reveal Cdc25A as an important negative regulator of antiviral immunity and demonstrate its role in maintaining host cell homeostasis following viral infection.
Collapse
Affiliation(s)
- Dandan Qi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lei Hu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Tong Jiao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Tinghong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomei Tong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xin Ye
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
55
|
Kim D, Liu Y, Oberly S, Freire R, Smolka MB. ATR-mediated proteome remodeling is a major determinant of homologous recombination capacity in cancer cells. Nucleic Acids Res 2018; 46:8311-8325. [PMID: 30010936 PMCID: PMC6144784 DOI: 10.1093/nar/gky625] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/28/2018] [Indexed: 12/20/2022] Open
Abstract
The ATR kinase is crucial for genome maintenance, but the mechanisms by which ATR controls the DNA repair machinery are not fully understood. Here, we find that long-term chronic inhibition of ATR signaling severely impairs the ability of cells to utilize homologous recombination (HR)-mediated DNA repair. Proteomic analysis shows that chronic ATR inhibition depletes the abundance of key HR factors, suggesting that spontaneous ATR signaling enhances the capacity of cells to use HR-mediated repair by controlling the abundance of the HR machinery. Notably, ATR controls the abundance of HR factors largely via CHK1-dependent transcription, and can also promote stabilization of specific HR proteins. Cancer cells exhibit a strong dependency on ATR signaling for maintaining elevated levels of HR factors, and we propose that increased constitutive ATR signaling caused by augmented replication stress in cancer cells drives the enhanced HR capacity observed in certain tumor types. Overall, these findings define a major pro-HR function for ATR and have important implications for therapy by providing rationale for sensitizing HR-proficient cancer cells to PARP inhibitors.
Collapse
Affiliation(s)
- Dongsung Kim
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Yi Liu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Susannah Oberly
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologias Biomedicas, 38320 Tenerife, Spain
| | - Marcus B Smolka
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- To whom correspondence should be addressed. Tel: +1 607 255 0274; Fax: +1 607 255 5961;
| |
Collapse
|
56
|
Zhang W, Zeng Q, Ban Z, Cao J, Chu T, Lei D, Liu C, Guo W, Zeng X. Effects of let-7c on the proliferation of ovarian carcinoma cells by targeted regulation of CDC25a gene expression. Oncol Lett 2018; 16:5543-5550. [PMID: 30405749 DOI: 10.3892/ol.2018.9327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/03/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs serve a role in the development of ovarian cancer (OC). The present study investigated whether let-7c is able to regulate the proliferation of OC cells by targeting cell division cycle 25A (CDC25a). The reverse transcription-quantitative polymerase chain reaction was performed to detect the expression of let-7c in OC specimens. Let-7c agomir was transfected into OC cells, and the proliferation and apoptosis of OC cells were detected. A dual-luciferase assay and western blotting were performed to analyze whether CDC25a was the target gene of let-7c as well as its interaction site. The results revealed that, in OC tissue, let-7c was downregulated when compared with normal ovarian tissue. A Cell Counting Kit-8 (CCK8) assay, colony formation assay and flow cytometry demonstrated that increased expression of let-7c was able to inhibit the proliferation and increase the apoptosis of OC cells. Western blotting revealed that upregulated let-7c is able to decrease the expression of CDC25a, and a dual-luciferase assay and a recovery assay demonstrated that let-7c was able to regulate the expression of the 3' untranslated region of CDC25a. Therefore, the roles of let-7c in inhibiting the proliferation and promoting the apoptosis of OC cells may be realized through the regulation of the expression of CDC25a. The results of the present study revealed that let-7c may be a novel target in the diagnosis and treatment of OC.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qingru Zeng
- Department of Ultrasound, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhenying Ban
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jing Cao
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Tianjiao Chu
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Dongmei Lei
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chi Liu
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Wentao Guo
- Pathogen Biology Laboratory, The Basic Medical College of Guangdong Medical University, Dongguan, Guangdong 523000, P.R. China
| | - Xianxu Zeng
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
57
|
Qiu L, Zhao C, Wang P, Fan S, Yan L, Xie B, Jiang S, Wang S, Lin H. Genomic structure, expression, and functional characterization of checkpoint kinase 1 from Penaeus monodon. PLoS One 2018; 13:e0198036. [PMID: 29795680 PMCID: PMC5967826 DOI: 10.1371/journal.pone.0198036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/11/2018] [Indexed: 12/14/2022] Open
Abstract
Chk1 is a cell-cycle regulator. Chk1 has been identified in organisms ranging from yeast to humans, but few researchers have studied Chk1 in shrimps. We cloned Chk1 from the black tiger shrimp (Penaeus monodon). The full-length cDNA sequence of PmChk1 had 3,334 base pairs (bp), with an open reading frame of 1,455 bp. The complete genomic sequence of PmChk1 (11,081 bp) contained 10 exons separated by nine introns. qRT-PCR showed that PmChk1 was highly expressed in the ovaries and gills of P. monodon. The lowest PmChk1 expression was noted in stage III of ovarian development in P. monodon. PmChk1 expression decreased significantly after injection of 5-hydroxytryptamine and eyestalk ablation in P. monodon ovaries. RNA interference experiments were undertaken to examine the expression of PmChk1, PmCDC2, and PmCyclin B. PmChk1 knockdown in the ovaries and hepatopancreas by dsRNA-Chk1 was successful. The localization and level of PmChk1 expression in the hepatopancreas was studied using in situ hybridization, which showed that data were in accordance with those of qRT-PCR. The Gonadosomatic Index of P. monodon after dsRNA-Chk1 injection was significantly higher than that after injection of dsRNA-GFP or phosphate-buffered saline. These data suggest that PmChk1 may have important roles in the ovarian maturation of P. monodon.
Collapse
Affiliation(s)
- Lihua Qiu
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
- Key Laboratory of Aquatic Genomics, Ministry of Agriculture, Beijing, China
| | - Chao Zhao
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
| | - Pengfei Wang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
| | - Sigang Fan
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
| | - Lulu Yan
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
| | - Bobo Xie
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
| | - Shigui Jiang
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture, Guangzhou, China
- South China Sea Resource Exploitation and Protection Collaborative Innovation Center, Sun Yat-Sen University, Guangzhou, China
- * E-mail:
| | - Shu Wang
- Chinese Academy of Fishery Sciences, Beijing, China
| | - Heizhao Lin
- Shenzhen Base of South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shenzhen, PR China
| |
Collapse
|
58
|
Tian C, Han Z, Li Y, Wang M, Yang J, Wang X, Zhang Z, Liu J. Synthesis and biological evaluation of 2,6-disubstituted-9H-purine, 2,4-disubstitued-thieno[3,2-d]pyrimidine and -7H-pyrrolo[2,3-d]pyrimidine analogues as novel CHK1 inhibitors. Eur J Med Chem 2018; 151:836-848. [DOI: 10.1016/j.ejmech.2018.03.075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/14/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
|
59
|
Kizhedathu A, Bagul AV, Guha A. Negative regulation of G2-M by ATR (mei-41)/Chk1(Grapes) facilitates tracheoblast growth and tracheal hypertrophy in Drosophila. eLife 2018; 7:29988. [PMID: 29658881 PMCID: PMC5953539 DOI: 10.7554/elife.29988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 04/12/2018] [Indexed: 11/21/2022] Open
Abstract
Imaginal progenitors in Drosophila are known to arrest in G2 during larval stages and proliferate thereafter. Here we investigate the mechanism and implications of G2 arrest in progenitors of the adult thoracic tracheal epithelium (tracheoblasts). We report that tracheoblasts pause in G2 for ~48–56 h and grow in size over this period. Surprisingly, tracheoblasts arrested in G2 express drivers of G2-M like Cdc25/String (Stg). We find that mechanisms that prevent G2-M are also in place in this interval. Tracheoblasts activate Checkpoint Kinase 1/Grapes (Chk1/Grp) in an ATR/mei-41-dependent manner. Loss of ATR/Chk1 led to precocious mitotic entry ~24–32 h earlier. These divisions were apparently normal as there was no evidence of increased DNA damage or cell death. However, induction of precocious mitoses impaired growth of tracheoblasts and the tracheae they comprise. We propose that ATR/Chk1 negatively regulate G2-M in developing tracheoblasts and that G2 arrest facilitates cellular and hypertrophic organ growth. Every organism begins as a single cell. That cell, and all the other cells it generates over time, need to divide at the right time and in the right place to develop into an adult. As they do so, they pass through the stages of the cell cycle. As cells prepare to divide they enter into the first growth phase, G1, ramping up their metabolic activity. They then enter S phase, duplicate their DNA, and subsequently a second growth phase G2. Finally, during the mitotic phase, the chromosome separate and cells undergo cytokinesis to form new cells. Dividing cells can pause at certain stages of the cell cycle to assess whether the conditions are suitable to proceed. The length of the pause depends on the stage of development and the cell type. Signals around the cell provide the cues that it needs to make the decision. The fruit fly Drosophila melanogaster, for example, undergoes metamorphosis during development, meaning it transforms from a larva into an adult. The larva contains small patches of ‘progenitor’ cells that form the adult tissue. These remain paused for various intervals during larval life and restart their cell cycle as the animal develops. A key challenge in biology is to understand how these progenitors pause and what makes them start dividing again. Here, Kizhedathu, Bagul and Guha uncover a new mechanism that pauses the cell cycle in developing animal cells. Progenitors of the respiratory system in the adult fruit fly pause at the G2 stage of the cell cycle during larval life. Some of these progenitors, from a part of the larva called the dorsal trunk, go on to form the structures of the adult respiratory system. By counting the cells and tracking their dynamics with fluorescent labels, Kizhedathu et al. revealed that the progenitor cells pause for between 48 and to 56 hours. Previous research suggested that this pause happens because the cells lack a protein essential for mitosis called Cdc25/String. However, these progenitors were producing Cdc25/String. They stopped dividing because they also made another protein, known as Checkpoint Kinase 1/Grapes (Chk1/Grp). Chk1 is known to add a chemical modification to Cdc25, which dampens its activity and stops the cell cycle from progressing. This is likely what allow the flies to co-ordinate their development and give the cells more time to grow. When Chk1 was experimentally removed, it reactivated the paused cells sooner, resulting in smaller cells and a smaller respiratory organ. This work extends our understanding of stem cell dynamics and growth during development. Previous work has shown that cells that give rise to muscles and the neural tube (the precursor of the central nervous system) also pause their cell cycle in G2. Understanding more about how this happens could open new avenues for research into developmental disease.
Collapse
Affiliation(s)
- Amrutha Kizhedathu
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,SASTRA University, Thanjavur, India
| | - Archit V Bagul
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Arjun Guha
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| |
Collapse
|
60
|
Oo ZY, Stevenson AJ, Proctor M, Daignault SM, Walpole S, Lanagan C, Chen J, Škalamera D, Spoerri L, Ainger SA, Sturm RA, Haass NK, Gabrielli B. Endogenous Replication Stress Marks Melanomas Sensitive to CHEK1 Inhibitors In Vivo. Clin Cancer Res 2018. [PMID: 29535131 DOI: 10.1158/1078-0432.ccr-17-2701] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: Checkpoint kinase 1 inhibitors (CHEK1i) have single-agent activity in vitro and in vivo Here, we have investigated the molecular basis of this activity.Experimental Design: We have assessed a panel of melanoma cell lines for their sensitivity to the CHEK1i GNE-323 and GDC-0575 in vitro and in vivo The effects of these compounds on responses to DNA replication stress were analyzed in the hypersensitive cell lines.Results: A subset of melanoma cell lines is hypersensitive to CHEK1i-induced cell death in vitro, and the drug effectively inhibits tumor growth in vivo In the hypersensitive cell lines, GNE-323 triggers cell death without cells entering mitosis. CHEK1i treatment triggers strong RPA2 hyperphosphorylation and increased DNA damage in only hypersensitive cells. The increased replication stress was associated with a defective S-phase cell-cycle checkpoint. The number and intensity of pRPA2 Ser4/8 foci in untreated tumors appeared to be a marker of elevated replication stress correlated with sensitivity to CHEK1i.Conclusions: CHEK1i have single-agent activity in a subset of melanomas with elevated endogenous replication stress. CHEK1i treatment strongly increased this replication stress and DNA damage, and this correlated with increased cell death. The level of endogenous replication is marked by the pRPA2Ser4/8 foci in the untreated tumors, and may be a useful marker of replication stress in vivoClin Cancer Res; 24(12); 2901-12. ©2018 AACR.
Collapse
Affiliation(s)
- Zay Yar Oo
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Alexander J Stevenson
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Martina Proctor
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Sheena M Daignault
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Sebastian Walpole
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Catherine Lanagan
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - James Chen
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Dubravka Škalamera
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Loredana Spoerri
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Stephen A Ainger
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Richard A Sturm
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Nikolas K Haass
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| | - Brian Gabrielli
- Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia. .,The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland. Australia
| |
Collapse
|
61
|
Kjeldsen E, Nielsen CJF, Roy A, Tesauro C, Jakobsen AK, Stougaard M, Knudsen BR. Characterization of Camptothecin-induced Genomic Changes in the Camptothecin-resistant T-ALL-derived Cell Line CPT-K5. Cancer Genomics Proteomics 2018; 15:91-114. [PMID: 29496689 PMCID: PMC5892604 DOI: 10.21873/cgp.20068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/27/2022] Open
Abstract
Acquisition of resistance to topoisomerase I (TOP1)-targeting camptothecin (CPT) derivatives is a major clinical problem. Little is known about the underlying chromosomal and genomic mechanisms. We characterized the CPT-K5 cell line expressing mutant CPT-resistant TOP1 and its parental T-cell derived acute lymphoblastic leukemia CPT-sensitive RPMI-8402 cell line by karyotyping and molecular genetic methods, including subtractive oligo-based array comparative genomic hybridization (soaCGH) analysis. Karyotyping revealed that CPT-K5 cells had acquired additional structural aberrations and a reduced modal chromosomal number compared to RPMI-8402. soaCGH analysis identified vast copy number alterations and >200 unbalanced DNA breakpoints distributed unevenly across the chromosomal complement in CPT-K5. In addition, the short tandem repeat alleles were found to be highly different between CPT-K5 and its parental cell line. We identified copy number alterations affecting genes important for maintaining genome integrity and reducing CPT-induced DNA damage. We show for the first time that short tandem repeats are targets for TOP1 cleavage, that can be differentially stimulated by CPT.
Collapse
Affiliation(s)
- Eigil Kjeldsen
- Cancer Cytogenetics Section, HemoDiagnostic Laboratory, Aarhus University Hospital, Aarhus, Denmark
| | - Christine J F Nielsen
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| | - Amit Roy
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| | - Cinzia Tesauro
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| | | | - Magnus Stougaard
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitta R Knudsen
- Department of Molecular Biology and Genetics, C.F. Møllers Allé, Aarhus University, Aarhus, Denmark
| |
Collapse
|
62
|
Wayne J, Brooks T, Massey AJ. Inhibition of Chk1 with the small molecule inhibitor V158411 induces DNA damage and cell death in an unperturbed S-phase. Oncotarget 2018; 7:85033-85048. [PMID: 27829224 PMCID: PMC5356717 DOI: 10.18632/oncotarget.13119] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/22/2016] [Indexed: 12/30/2022] Open
Abstract
Chk1 kinase is a critical component of the DNA damage response checkpoint and Chk1 inhibitors are currently under clinical investigation. Chk1 suppresses oncogene-induced replication stress with Chk1 inhibitors demonstrating activity as a monotherapy in numerous cancer types. Understanding the mechanism by which Chk1 inhibitors induce DNA damage and cancer cell death is essential for their future clinical development. Here we characterize the mechanism by which the novel Chk1 inhibitor (V158411) increased DNA damage and cell death in models of human cancer. V158411 induced a time- and concentration-dependent increase in γH2AX-positive nuclei that was restricted to cells actively undergoing DNA synthesis. γH2AX induction was an early event and correlated with activation of the ATR/ATM/DNA-PKcs DNA damage response pathways. The appearance of γH2AX positive nuclei preceded ssDNA appearance and RPA exhaustion. Complete and sustained inhibition of Chk1 kinase was necessary to activate a robust γH2AX induction and growth inhibition. Chk1 inhibitor cytotoxicity correlated with induction of DNA damage with cells undergoing apoptosis, mitotic slippage and DNA damage-induced permanent cell cycle arrest. We identified two distinct classes of Chk1 inhibitors: those that induced a strong increase in γH2AX, pChk1 (S317) and pRPA32 (S4/S8) (including V158411, LY2603618 and ARRY-1A) and those that did not (including MK-8776 and GNE-900). Tumor cell death, induced through increased DNA damage, coupled with abrogation of cell cycle checkpoints makes selective inhibitors of Chk1 a potentially useful therapeutic treatment for multiple human cancers.
Collapse
|
63
|
Davis T, van Niekerk G, Peres J, Prince S, Loos B, Engelbrecht AM. Doxorubicin resistance in breast cancer: A novel role for the human protein AHNAK. Biochem Pharmacol 2018; 148:174-183. [DOI: 10.1016/j.bcp.2018.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/03/2018] [Indexed: 12/24/2022]
|
64
|
Zhang Y, Chang JF, Sun J, Chen L, Yang XM, Tang HY, Jing YY, Kang X, He ZM, Wu JY, Wei HM, Wang DL, Xu RG, Zhu RB, Shen Y, Zeng SY, Wang C, Liu KN, Zhang Y, Mao ZY, Jiang CZ, Sun FL. Histone H3K27 methylation is required for NHEJ and genome stability by modulating the dynamics of FANCD2 on chromatin. J Cell Sci 2018; 131:jcs.215525. [PMID: 29760279 DOI: 10.1242/jcs.215525] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022] Open
Abstract
Dysregulation of homeostatic balance in di- and tri-methyl H3K27 levels or that caused by mis-sense mutations of histone H3 (H3K27M) was reported to be associated with various types of cancers. In this study, we found that reduction in H3K27me2/3 caused by H3.1K27M, a mutation of H3 variants found in DIPG patients, dramatically attenuated the presence of 53BP1 foci and NHEJ repair capability in HDF cells. H3.1K27M cells showed increased rates of genomic insertions/deletions (In/Dels) and copy number variations (CNVs), as well as augmented p53-dependent apoptotic cells. We further showed that both hypo-H3K27me2/3 and H3.1K27M interacted with FANCD2, a central player to orchestrate DNA repair pathway choice. H3.1K27M triggered an accumulation of FANCD2 on chromatin, supporting the interaction between H3.1K27M and FANCD2. Most interestingly, knock-down of FANCD2 in H3.1K27M cells recovered the number of 53BP1 foci, NHEJ efficiency and apoptosis rate. Although these findings in HDF cells may differ from the case of endogenous H3.1K27M mutant regulation in the specific tumor context of DIPG, our results suggest a new model by which H3K27me2/3 facilitates NHEJ and the maintenance of genome stability.
Collapse
Affiliation(s)
- Ye Zhang
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Jian-Feng Chang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Jin Sun
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Lu Chen
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Xiao-Mei Yang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Huan-Yin Tang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Yuan-Ya Jing
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Xuan Kang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Zhi-Min He
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Jun-Yu Wu
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Hui-Min Wei
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Da-Liang Wang
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Rong-Gang Xu
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Rui-Bao Zhu
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| | - Ying Shen
- School of Software Engineering, Tongji University, Shanghai 200092, PR China
| | - Shi-Yang Zeng
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Chen Wang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Kui-Nan Liu
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Yong Zhang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Zhi-Ying Mao
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Ci-Zhong Jiang
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
| | - Fang-Lin Sun
- Research Center for Translational Medicine at East Hospital, School of Life Sciences and Technology, Advanced Institute of Translational Medicine, Tongji University, Shanghai 200092, PR China
- School of Medicine, Tsinghua University, Beijing 100084, PR China
| |
Collapse
|
65
|
Shen T, Zhou H, Shang C, Luo Y, Wu Y, Huang S. Ciclopirox activates ATR-Chk1 signaling pathway leading to Cdc25A protein degradation. Genes Cancer 2018; 9:39-52. [PMID: 29725502 PMCID: PMC5931253 DOI: 10.18632/genesandcancer.166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/11/2018] [Indexed: 02/05/2023] Open
Abstract
Ciclopirox olamine (CPX), an off-patent anti-fungal drug, has been found to inhibit the G1-cyclin dependent kinases partly by increasing the phosphorylation and degradation of Cdc25A. However, little is known about the molecular target(s) of CPX responsible for Cdc25A degradation. Here, we show that CPX induced the degradation of Cdc25A neither by increasing CK1α or decreasing DUB3 expression, nor via activating GSK3β, but through activating Chk1 in rhabdomyosarcoma (Rh30) and breast carcinoma (MDA-MB-231) cells. This is strongly supported by the findings that inhibition of Chk1 with TCS2312 or knockdown of Chk1 profoundly attenuated CPX-induced Cdc25A degradation in the cells. Furthermore, we observed that CPX caused DNA damage, which was independent of reactive oxygen species (ROS) induction, but related to iron chelation. CPX treatment resulted in the activation of ataxia telangiectasia mutated (ATM) and ATM-and RAD3-related (ATR) kinases. Treatment with Ku55933 (a selective ATM inhibitor) failed to prevent CPX-induced Chk1 phosphorylation and Cdc25A degradation. In contrast, knockdown of ATR conferred high resistance to CPX-induced Chk1 phosphorylation and Cdc25A degradation. Therefore, the results suggest that CPX-induced degradation of Cdc25A is attributed to the activation of ATR-Chk1 signaling pathway, a consequence of iron chelation-induced DNA damage.
Collapse
Affiliation(s)
- Tao Shen
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Hongyu Zhou
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Chaowei Shang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- State Key Laboratory of Biotherapy / Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Yang Wu
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- State Key Laboratory of Biotherapy / Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
66
|
Liu H, Luo Q, Cui H, Deng H, Kuang P, Lu Y, Fang J, Zuo Z, Deng J, Li Y, Wang X, Zhao L. Sodium fluoride causes hepatocellular S-phase arrest by activating ATM-p53-p21 and ATR-Chk1-Cdc25A pathways in mice. Oncotarget 2017; 9:4318-4337. [PMID: 29435105 PMCID: PMC5796976 DOI: 10.18632/oncotarget.23093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/14/2017] [Indexed: 12/26/2022] Open
Abstract
In this study, experimental pathology, flow cytometry (FCM), quantitative real-time polymerase chain reaction (qRT-PCR), and western blot (WB) were used to evaluate the effects of sodium fluoride (NaF) on hepatocellular cell cycle progression in mice. A total of 240 ICR mice were divided equally into four groups; the experimental groups received 12, 24, or 48 mg/kg NaF intragastrically for 42 days, while the control group received distilled water. Doses of NaF above 12 mg/kg increased the percentage of cells in S phase (S-phase arrest), reduced percentages of cells in G0/G1 or G2/M phase, and activated the ATM-p53-p21 and ATR-Chk1-Cdc25A pathways. Activation of these pathways was characterized by up-regulation of ATM, p53, p21, ATR, and Chk1 mRNA and protein expression, and down-regulation of Cdc25A, cyclin E, cyclin A, CDK2, CDK4, and proliferating cell nuclear antigen (PCNA) mRNA and protein expression. These results indicate that NaF caused S-phase arrest by activating the ATM-p53-p21 and ATR-Chk1-Cdc25A pathways.
Collapse
Affiliation(s)
- Huan Liu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China
| | - Qin Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| | - Huidan Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China
| | - Ping Kuang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China
| | - Yujiao Lu
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| | - Yinglun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| | - Xun Wang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| | - Ling Zhao
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu, China.,Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu, China
| |
Collapse
|
67
|
Carrassa L, Damia G. DNA damage response inhibitors: Mechanisms and potential applications in cancer therapy. Cancer Treat Rev 2017; 60:139-151. [PMID: 28961555 DOI: 10.1016/j.ctrv.2017.08.013] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/26/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023]
Abstract
Over the last decade the unravelling of the molecular mechanisms of the DNA damage response pathways and of the genomic landscape of human tumors have paved the road to new therapeutic approaches in oncology. It is now clear that tumors harbour defects in different DNA damage response steps, mainly signalling and repair, rendering them more dependent on the remaining pathways. We here focus on the proteins ATM, ATR, CHK1 and WEE1, reviewing their roles in the DNA damage response and as targets in cancer therapy. In the last decade specific inhibitors of these proteins have been designed, and their potential antineoplastic activity has been explored both in monotherapy strategies against tumors with specific defects (synthetic lethality approach) and in combination with radiotherapy or chemotherapeutic or molecular targeted agents. The preclinical and clinical evidence of antitumor activity of these inhibitors emanating from these research efforts will be critically reviewed. Lastly, the potential therapeutic feasibility of combining together such inhibitors with the aim to target particular subsets of tumors will be also discussed.
Collapse
Affiliation(s)
- Laura Carrassa
- Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| | - Giovanna Damia
- Laboratory of Molecular Pharmacology, Department of Oncology, IRCCS - Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| |
Collapse
|
68
|
Ali I, Lone MN, Aboul-Enein HY. Imidazoles as potential anticancer agents. MEDCHEMCOMM 2017; 8:1742-1773. [PMID: 30108886 PMCID: PMC6084102 DOI: 10.1039/c7md00067g] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/21/2017] [Indexed: 12/19/2022]
Abstract
Cancer is a black spot on the face of humanity in this era of science and technology. Presently, several classes of anticancer drugs are available in the market, but issues such as toxicity, low efficacy and solubility have decreased the overall therapeutic indices. Thus, the search for new promising anticancer agents continues, and the battle against cancer is far from over. Imidazole is an aromatic diazole and alkaloid with anticancer properties. There is considerable interest among scientists in developing imidazoles as safe alternatives to anticancer chemotherapy. The present article describes the structural, chemical, and biological features of imidazoles. Several classes of imidazoles as anticancer agents based on their mode of action have been critically discussed. A careful observation has been made into pharmacologically active imidazoles with better or equal therapeutic effects compared to well-known imidazole-based anticancer drugs, which are available on the market. A brief discussion of the toxicities of imidazoles has been made. Finally, the current challenges and future perspectives of imidazole based anticancer drug development are conferred.
Collapse
Affiliation(s)
- Imran Ali
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi-110025 , India . ;
| | - Mohammad Nadeem Lone
- Department of Chemistry , Jamia Millia Islamia (Central University) , New Delhi-110025 , India . ;
| | - Haasan Y Aboul-Enein
- Pharmaceutical and Medicinal Chemistry Department , Pharmaceutical and Drug Industries Research Division , National Research Centre , Dokki , Giza 12622 , Egypt
| |
Collapse
|
69
|
Nakajima T, Wang B, Ono T, Uehara Y, Nakamura S, Ichinohe K, Braga-Tanaka I, Tanaka S, Tanaka K, Nenoi M. Differences in sustained alterations in protein expression between livers of mice exposed to high-dose-rate and low-dose-rate radiation. JOURNAL OF RADIATION RESEARCH 2017; 58:421-429. [PMID: 28201773 PMCID: PMC5570048 DOI: 10.1093/jrr/rrw133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Indexed: 05/13/2023]
Abstract
Molecular mechanisms of radiation dose-rate effects are not well understood. Among many possibilities, long-lasting sustained alterations in protein levels would provide critical information. To evaluate sustained effects after acute and chronic radiation exposure, we analyzed alterations in protein expression in the livers of mice. Acute exposure consisted of a lethal dose of 8 Gy and a sublethal dose of 4 Gy, with analysis conducted 6 days and 3 months after irradiation, respectively. Chronic irradiation consisted of a total dose of 8 Gy delivered over 400 days (20 mGy/day). Analyses following chronic irradiation were done immediately and at 3 months after the end of the exposure. Based on antibody arrays of protein expression following both acute lethal and sublethal dose exposures, common alterations in the expression of two proteins were detected. In the sublethal dose exposure, the expression of additional proteins was altered 3 months after irradiation. Immunohistochemical analysis showed that the increase in one of the two commonly altered proteins, MyD88, was observed around blood vessels in the liver. The alterations in protein expression after chronic radiation exposure were different from those caused by acute radiation exposures. Alterations in the expression of proteins related to inflammation and apoptosis, such as caspase 12, were observed even at 3 months after the end of the chronic radiation exposure. The alterations in protein expression depended on the dose, the dose rate, and the passage of time after irradiation. These changes could be involved in long-term effects of radiation in the liver.
Collapse
Affiliation(s)
- Tetsuo Nakajima
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, 9-1, Anagawa-4-chome, Inage-ku, Chiba-shi 263-8555, Japan
- Corresponding author. National Institute of Radiological Sciences, National Institutes of Quantum and Radiolgical Science and Technology, 9-1, Anagawa-4-chome, Inage-ku, Chiba-shi 263-8555, Japan. Tel: +81-43-206-3086; Fax: +81-43-255-6497;
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, 9-1, Anagawa-4-chome, Inage-ku, Chiba-shi 263-8555, Japan
| | - Tetsuya Ono
- Department of Radiobiology, Institute for Environmental Sciences, 1-7, Ienomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Yoshihiko Uehara
- Department of Cell Biology, Tohoku University School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Shingo Nakamura
- Department of Radiobiology, Institute for Environmental Sciences, 1-7, Ienomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Kazuaki Ichinohe
- Department of Radiobiology, Institute for Environmental Sciences, 1-7, Ienomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Ignacia Braga-Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, 1-7, Ienomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Satoshi Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, 1-7, Ienomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Kimio Tanaka
- Department of Radiobiology, Institute for Environmental Sciences, 1-7, Ienomae, Obuchi, Rokkasho-mura, Kamikita-gun, Aomori 039-3212, Japan
| | - Mitsuru Nenoi
- National Institute of Radiological Sciences, National Institutes of Quantum and Radiological Science and Technology, 9-1, Anagawa-4-chome, Inage-ku, Chiba-shi 263-8555, Japan
| |
Collapse
|
70
|
Wei J, Zhang L, Ren L, Zhang J, Liu J, Duan J, Yu Y, Li Y, Peng C, Zhou X, Sun Z. Endosulfan induces cell dysfunction through cycle arrest resulting from DNA damage and DNA damage response signaling pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 589:97-106. [PMID: 28273598 DOI: 10.1016/j.scitotenv.2017.02.154] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/07/2017] [Accepted: 02/18/2017] [Indexed: 06/06/2023]
Abstract
Our previous study showed that endosulfan increases the risk of cardiovascular disease. To identify toxic mechanism of endosulfan, we conducted an animal study for which 32 male Wistar rats were randomly and equally divided into four groups: Control group (corn oil only) and three treatment groups (1, 5 and 10mgkg-1·d-1). The results showed that exposure to endosulfan resulted in injury of cardiac tissue with impaired mitochondria integrity and elevated 8-OHdG expression in myocardial cells. Moreover, endosulfan increased the expressions of Fas, FasL, Caspase-8, Cleaved Caspase-8, Caspase-3 and Cleaved Caspase-3 in cardiac tissue. In vitro, human umbilical vein endothelial cells (HUVECs) were treated with different concentrations of endosulfan (1, 6 and 12μgmL-1) for 24h. An inhibitor for Ataxia Telangiectasia Mutated Protein (ATM) (Ku-55933, 10μM) was added in 12μgmL-1 group for 2h before exposure to endosulfan. Results showed that endosulfan induced DNA damage and activated DNA damage response signaling pathway (ATM/Chk2 and ATR/Chk1) and consequent cell cycle checkpoint. Furthermore, endosulfan promoted the cell apoptosis through death receptor pathway resulting from oxidative stress. The results provide a new insight for mechanism of endosulfan-induced cardiovascular toxicity which will be helpful in future prevention of cardiovascular diseases induced by endosulfan.
Collapse
Affiliation(s)
- Jialiu Wei
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lianshuang Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Lihua Ren
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jin Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Jianhui Liu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Junchao Duan
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yang Yu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yanbo Li
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Cheng Peng
- National Research Centre for Environmental Toxicology (Entox), Queensland Alliance for Environmental Health Science (QAEHS), The University of Queensland, Coopers Plains, Brisbane, QLD 4108, Australia
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| | - Zhiwei Sun
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, 100069 Beijing, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| |
Collapse
|
71
|
Goss KL, Koppenhafer SL, Harmoney KM, Terry WW, Gordon DJ. Inhibition of CHK1 sensitizes Ewing sarcoma cells to the ribonucleotide reductase inhibitor gemcitabine. Oncotarget 2017; 8:87016-87032. [PMID: 29152060 PMCID: PMC5675612 DOI: 10.18632/oncotarget.18776] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
Ewing sarcoma is a bone and soft tissue sarcoma that occurs in children and young adults. The EWS-FLI1 gene fusion is the driver mutation in most Ewing sarcoma tumors and functions, in part, as an aberrant transcription factor. We recently identified that Ewing sarcoma cells are sensitive to inhibition of ribonucleotide reductase (RNR), which catalyzes the formation of deoxyribonucleotides from ribonucleotides. In this report, we show that Ewing sarcoma cells are sensitive to treatment with clofarabine, which is a nucleoside analogue and allosteric inhibitor of RNR. However, clofarabine is a reversible inhibitor of RNR and we found that the effect of clofarabine is limited when using a short (6-hour) drug treatment. Gemcitabine, on the other hand, is an irreversible inhibitor of the RRM1 subunit of RNR and this drug induces apoptosis in Ewing sarcoma cells when used in both 6-hour and longer drug treatments. Treatment of Ewing sarcoma cells with gemcitabine also results in activation of checkpoint kinase 1 (CHK1), which is a critical mediator of cell survival in the setting of impaired DNA replication. Notably, inhibition of CHK1 function in Ewing sarcoma cells using a small-molecule CHK1 inhibitor, or siRNA knockdown, in combination with gemcitabine results in increased toxicity both in vitro and in vivo in a mouse xenograft experiment. Overall, our results provide insight into Ewing sarcoma biology and identify a candidate therapeutic target, and drug combination, in Ewing sarcoma.
Collapse
Affiliation(s)
- Kelli L Goss
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Stacia L Koppenhafer
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Kathryn M Harmoney
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, Iowa 52242, USA
| | - William W Terry
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, Iowa 52242, USA
| | - David J Gordon
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
72
|
Konyar D, Erdas O, Alpaslan FN, Buyukbingol E. An application of CIFAP for predicting the binding affinity of Chk1 inhibitors derived from 2-aminothiazole-4-carboxamide. J Mol Recognit 2017. [PMID: 28620979 DOI: 10.1002/jmr.2642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Investigation of protein-ligand interactions obtained from experiments has a crucial part in the design of newly discovered and effective drugs. Analyzing the data extracted from known interactions could help scientists to predict the binding affinities of promising ligands before conducting experiments. The objective of this study is to advance the CIFAP (compressed images for affinity prediction) method, which is relevant to a protein-ligand model, identifying 2D electrostatic potential images by separating the binding site of protein-ligand complexes and using the images for predicting the computational affinity information represented by pIC50 values. The CIFAP method has 2 phases, namely, data modeling and prediction. In data modeling phase, the separated 3D structure of the binding pocket with the ligand inside is fitted into an electrostatic potential grid box, which is then compressed through 3 orthogonal directions into three 2D images for each protein-ligand complex. Sequential floating forward selection technique is performed for acquiring prediction patterns from the images. In the prediction phase, support vector regression (SVR) and partial least squares regression are used for testing the quality of the CIFAP method for predicting the binding affinity of 45 CHK1 inhibitors derived from 2-aminothiazole-4-carboxamide. The results show that the CIFAP method using both support vector regression and partial least squares regression is very effective for predicting the binding affinities of CHK1-ligand complexes with low-error values and high correlation. As a future work, the results could be improved by working on the pose of the ligands inside the grid.
Collapse
Affiliation(s)
- Dilan Konyar
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara University, Ankara, Turkey
| | - Ozlem Erdas
- Department of Computer Engineering, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Ferda Nur Alpaslan
- Department of Computer Engineering, Middle East Technical University, Ankara, Turkey
| | - Erdem Buyukbingol
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Ankara University, Ankara, Turkey
| |
Collapse
|
73
|
Borg NA, Dixit VM. Ubiquitin in Cell-Cycle Regulation and Dysregulation in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2017. [DOI: 10.1146/annurev-cancerbio-040716-075607] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Uncontrolled cell proliferation and genomic instability are common features of cancer and can arise from, respectively, the loss of cell-cycle control and defective checkpoints. Ubiquitin-mediated proteolysis, ultimately executed by ubiquitin-ligating enzymes (E3s), plays a key part in cell-cycle regulation and is dominated by two multisubunit E3s, the anaphase-promoting complex (or cyclosome) (APC/C) and SKP1–cullin-1–F-box (SCF) complex. We highlight the role of APC/C and the SCF bound to F-box proteins, FBXW7, SKP2, and β-TrCP, in regulating the abundance of select fundamental proteins, primarily during the cell cycle, that are associated with human cancer. The clinical success of the first proteasome inhibitor, bortezomib, in treating multiple myeloma and mantle-cell lymphoma set the precedent for viewing the ubiquitin–proteasome system as a druggable target for cancer. Given that there are more E3s than kinases, selective, small-molecule E3 inhibitors have the potential of opening up another dimension in the therapeutic armamentarium for the treatment of cancer.
Collapse
Affiliation(s)
- Natalie A. Borg
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Vishva M. Dixit
- Department of Physiological Chemistry, Genentech Inc., South San Francisco, California 94080
| |
Collapse
|
74
|
Pal HC, Katiyar SK. Cryptolepine, a Plant Alkaloid, Inhibits the Growth of Non-Melanoma Skin Cancer Cells through Inhibition of Topoisomerase and Induction of DNA Damage. Molecules 2016; 21:E1758. [PMID: 28009843 PMCID: PMC6273109 DOI: 10.3390/molecules21121758] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/13/2016] [Accepted: 12/17/2016] [Indexed: 01/03/2023] Open
Abstract
Topoisomerases have been shown to have roles in cancer progression. Here, we have examined the effect of cryptolepine, a plant alkaloid, on the growth of human non-melanoma skin cancer cells (NMSCC) and underlying mechanism of action. For this purpose SCC-13 and A431 cell lines were used as an in vitro model. Our study reveals that SCC-13 and A431 cells express higher levels as well as activity of topoisomerase (Topo I and Topo II) compared with normal human epidermal keratinocytes. Treatment of NMSCC with cryptolepine (2.5, 5.0 and 7.5 µM) for 24 h resulted in marked decrease in topoisomerase activity, which was associated with substantial DNA damage as detected by the comet assay. Cryptolepine induced DNA damage resulted in: (i) an increase in the phosphorylation of ATM/ATR, BRCA1, Chk1/Chk2 and γH2AX; (ii) activation of p53 signaling cascade, including enhanced protein expressions of p16 and p21; (iii) downregulation of cyclin-dependent kinases, cyclin D1, cyclin A, cyclin E and proteins involved in cell division (e.g., Cdc25a and Cdc25b) leading to cell cycle arrest at S-phase; and (iv) mitochondrial membrane potential was disrupted and cytochrome c released. These changes in NMSCC by cryptolepine resulted in significant reduction in cell viability, colony formation and increase in apoptotic cell death.
Collapse
Affiliation(s)
- Harish C Pal
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Santosh K Katiyar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA.
| |
Collapse
|
75
|
Ismail NS, Ali GM, Ibrahim DA, Elmetwali AM. Medicinal attributes of pyrazolo[1,5-a]pyrimidine based scaffold derivatives targeting kinases as anticancer agents. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2016. [DOI: 10.1016/j.fjps.2016.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
76
|
Infante JR, Hollebecque A, Postel-Vinay S, Bauer TM, Blackwood EM, Evangelista M, Mahrus S, Peale FV, Lu X, Sahasranaman S, Zhu R, Chen Y, Ding X, Murray ER, Schutzman JL, Lauchle JO, Soria JC, LoRusso PM. Phase I Study of GDC-0425, a Checkpoint Kinase 1 Inhibitor, in Combination with Gemcitabine in Patients with Refractory Solid Tumors. Clin Cancer Res 2016; 23:2423-2432. [PMID: 27815358 DOI: 10.1158/1078-0432.ccr-16-1782] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 11/16/2022]
Abstract
Purpose: Chk1 inhibition potentiates DNA-damaging chemotherapy by overriding cell-cycle arrest and genome repair. This phase I study evaluated the Chk1 inhibitor GDC-0425 given in combination with gemcitabine to patients with advanced solid tumors.Experimental Design: Patients received GDC-0425 alone for a 1-week lead-in followed by 21-day cycles of gemcitabine plus GDC-0425. Gemcitabine was initially administered at 750 mg/m2 (Arm A), then increased to 1,000 mg/m2 (Arm B), on days 1 and 8 in a 3 + 3 + 3 dose escalation to establish maximum tolerated dose (MTD). GDC-0425 was initially administered daily for three consecutive days; however, dosing was abbreviated to a single day on the basis of pharmacokinetics and tolerability. TP53 mutations were evaluated in archival tumor tissue. On-treatment tumor biopsies underwent pharmacodynamic biomarker analyses.Results: Forty patients were treated with GDC-0425. The MTD of GDC-0425 was 60 mg when administered approximately 24 hours after gemcitabine 1,000 mg/m2 Dose-limiting toxicities included thrombocytopenia (n = 5), neutropenia (n = 4), dyspnea, nausea, pyrexia, syncope, and increased alanine aminotransferase (n = 1 each). Common related adverse events were nausea (48%); anemia, neutropenia, vomiting (45% each); fatigue (43%); pyrexia (40%); and thrombocytopenia (35%). The GDC-0425 half-life was approximately 15 hours. There were two confirmed partial responses in patients with triple-negative breast cancer (TP53-mutated) and melanoma (n = 1 each) and one unconfirmed partial response in a patient with cancer of unknown primary origin.Conclusions: Chk1 inhibition with GDC-0425 in combination with gemcitabine was tolerated with manageable bone marrow suppression. The observed preliminary clinical activity warrants further investigation of this chemopotentiation strategy. Clin Cancer Res; 23(10); 2423-32. ©2016 AACR.
Collapse
Affiliation(s)
- Jeffrey R Infante
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee.
| | - Antoine Hollebecque
- Départemement d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Sophie Postel-Vinay
- Départemement d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,INSERM, U981, Villejuif, France
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | | | | | - Sami Mahrus
- Genentech, Inc., South San Francisco, California
| | | | - Xuyang Lu
- Genentech, Inc., South San Francisco, California
| | | | - Rui Zhu
- Genentech, Inc., South San Francisco, California
| | - Yuan Chen
- Genentech, Inc., South San Francisco, California
| | - Xiao Ding
- Genentech, Inc., South San Francisco, California
| | | | | | | | - Jean-Charles Soria
- Départemement d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,INSERM, U981, Villejuif, France
| | | |
Collapse
|
77
|
Visconti R, Della Monica R, Grieco D. Cell cycle checkpoint in cancer: a therapeutically targetable double-edged sword. J Exp Clin Cancer Res 2016; 35:153. [PMID: 27670139 PMCID: PMC5037895 DOI: 10.1186/s13046-016-0433-9] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023] Open
Abstract
Major currently used anticancer therapeutics either directly damage DNA or target and upset basic cell division mechanisms like DNA replication and chromosome segregation. These insults elicit activation of cell cycle checkpoints, safeguard mechanisms that cells implement to correctly complete cell cycle phases, repair damage or eventually commit suicide in case damage is unrepairable. Although cancer cells appear to be advantageously defective in some aspects of checkpoint physiology, recent acquisitions on the biochemical mechanisms of the various checkpoints are offering new therapeutic approaches against cancer. Indeed, chemical manipulation of these mechanisms is providing new therapeutic strategies and tools to increase the killing efficacy of major cancer therapeutics as well as to directly promote cancer cell death. In this review we summarize developing concepts on how targeting cell cycle checkpoints may provide substantial improvement to cancer therapy.
Collapse
Affiliation(s)
| | - Rosa Della Monica
- DMMBM, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via G. Salvatore 486, 80145 Naples, Italy
| | - Domenico Grieco
- DMMBM, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy
- CEINGE Biotecnologie Avanzate, Via G. Salvatore 486, 80145 Naples, Italy
| |
Collapse
|
78
|
Ray A, Blevins C, Wani G, Wani AA. ATR- and ATM-Mediated DNA Damage Response Is Dependent on Excision Repair Assembly during G1 but Not in S Phase of Cell Cycle. PLoS One 2016; 11:e0159344. [PMID: 27442013 PMCID: PMC4956099 DOI: 10.1371/journal.pone.0159344] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/30/2016] [Indexed: 11/18/2022] Open
Abstract
Cell cycle checkpoint is mediated by ATR and ATM kinases, as a prompt early response to a variety of DNA insults, and culminates in a highly orchestrated signal transduction cascade. Previously, we defined the regulatory role of nucleotide excision repair (NER) factors, DDB2 and XPC, in checkpoint and ATR/ATM-dependent repair pathway via ATR and ATM phosphorylation and recruitment to ultraviolet radiation (UVR)-induced damage sites. Here, we have dissected the molecular mechanisms of DDB2- and XPC- mediated regulation of ATR and ATM recruitment and activation upon UVR exposures. We show that the ATR and ATM activation and accumulation to UVR-induced damage not only depends on DDB2 and XPC, but also on the NER protein XPA, suggesting that the assembly of an active NER complex is essential for ATR and ATM recruitment. ATR and ATM localization and H2AX phosphorylation at the lesion sites occur as early as ten minutes in asynchronous as well as G1 arrested cells, showing that repair and checkpoint-mediated by ATR and ATM starts early upon UV irradiation. Moreover, our results demonstrated that ATR and ATM recruitment and H2AX phosphorylation are dependent on NER proteins in G1 phase, but not in S phase. We reasoned that in G1 the UVR-induced ssDNA gaps or processed ssDNA, and the bound NER complex promote ATR and ATM recruitment. In S phase, when the UV lesions result in stalled replication forks with long single-stranded DNA, ATR and ATM recruitment to these sites is regulated by different sets of proteins. Taken together, these results provide evidence that UVR-induced ATR and ATM recruitment and activation differ in G1 and S phases due to the existence of distinct types of DNA lesions, which promote assembly of different proteins involved in the process of DNA repair and checkpoint activation.
Collapse
Affiliation(s)
- Alo Ray
- Department of Radiology, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Chessica Blevins
- Department of Radiology, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Gulzar Wani
- Department of Radiology, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Altaf A Wani
- Department of Radiology, Department of Molecular and Cellular Biochemistry, James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio, 43210, United States of America
| |
Collapse
|
79
|
Wang S, Lei T, Zhang M. The Reversal Effect and Its Mechanisms of Tetramethylpyrazine on Multidrug Resistance in Human Bladder Cancer. PLoS One 2016; 11:e0157759. [PMID: 27391608 PMCID: PMC4938409 DOI: 10.1371/journal.pone.0157759] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 06/03/2016] [Indexed: 01/11/2023] Open
Abstract
Chemotherapy is an important strategy for the treatment of bladder cancer. However, the main problem limiting the success of chemotherapy is the development of multidrug resistance (MDR). To improve the management of bladder cancer, it is an urgent matter to search for strategies to reverse MDR. We chose three kinds of herbal medicines including ginsenoside Rh2, (-)-Epigallocatechin gallate (EGCG) and Tetramethylpyrazine (TMP) to detect their effects on bladder cancer. Reversal effects of these three herbal medicines for drug resistance in adriamycin (ADM)-resistant Pumc-91 cells (Pumc-91/ADM) were assessed by Cell Counting Kit-8 (CCK-8) cell proliferation assay system. The mechanisms of reversal effect for TMP were explored in Pumc-91/ADM and T24/DDP cells. After Pumc-91/ADM and T24/DDP cells were treated with TMP, cell cycle distribution analysis was performed by flow cytometry. The expression of MRP1, GST, BCL-2, LRP and TOPO-II was evaluated using quantitative real-time polymerase chain reaction (qRT-PCR), immunefluorescence assay and western blot. It was observed that TMP was capable of enhancing the cytotoxicity of anticancer agents on Pumc-91/ADM cells in response to ADM, however Rh2 and EGCG were unable to. The reversal effect of TMP was also demonstrated in T24/DDP cells. Moreover, the treatment with TMP in Pumc-91/ADM and T24/DDP cells led to an increased of G1 phase accompanied with a concomitant decrease of cell numbers in S phase. Compared to the control group, an obvious decrease of MRP1, GST, BCL-2 and an increase of TOPO-II were shown in TMP groups with a dose-dependency in mRNA and protein levels. However, there was no difference on LRP expression between TMP groups and the control group. TMP could effectively reverse MDR of Pumc-91/ADM and T24/DDP cells and its mechanisms might be correlated with the alteration of MRP1, GST, BCL-2 and TOPO-II. TMP might be a potential candidate for reversing drug resistance in bladder cancer chemotherapy.
Collapse
Affiliation(s)
- Shanshan Wang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Ting Lei
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
| | - Man Zhang
- Clinical Laboratory Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Urinary Cellular Molecular Diagnostics, Beijing, China
- * E-mail:
| |
Collapse
|
80
|
Ding X, Chen Y, Sahasranaman S, Shi Y, McKnight J, Dean B. A supported liquid extraction LC-MS/MS method for determination of concentrations of GDC-0425, a small molecule Checkpoint kinase 1 inhibitor, in human plasma. Biomed Chromatogr 2016; 30:1984-1991. [DOI: 10.1002/bmc.3775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 01/04/2016] [Accepted: 05/27/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Xiao Ding
- Genentech; Drug Metabolism and Pharmacokinetics; 1 DNA Way South San Francisco CA USA
| | - Yuan Chen
- Genentech; Drug Metabolism and Pharmacokinetics; 1 DNA Way South San Francisco CA USA
| | - Srikumar Sahasranaman
- Genentech; Small Molecule Clinical Pharmacology; 1 DNA Way South San Francisco CA USA
| | - Yifan Shi
- Covance Laboratories; 3301 Kinsman Blvd. Madison WI USA
| | | | - Brian Dean
- Genentech; Drug Metabolism and Pharmacokinetics; 1 DNA Way South San Francisco CA USA
| |
Collapse
|
81
|
Colis LC, Herzon SB. Synergistic potentiation of (-)-lomaiviticin A cytotoxicity by the ATR inhibitor VE-821. Bioorg Med Chem Lett 2016; 26:3122-3126. [PMID: 27177826 PMCID: PMC4899226 DOI: 10.1016/j.bmcl.2016.04.090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 11/23/2022]
Abstract
(-)-Lomaiviticin A (1) is a cytotoxic bacterial metabolite that induces double-strand breaks in DNA. Here we show that the cytotoxicity of (-)-lomaiviticin A (1) is synergistically potentiated in the presence of VE-821 (7), an inhibitor of ataxia telangiectasia and Rad3-related protein (ATR). While 0.5nM 1 or 10μM 7 alone are non-lethal to K562 cells, co-incubation of the two leads to high levels of cell kill (81% and 94% after 24 and 48h, respectively). Mechanistic data indicate that cells treated with 1 and 7 suffer extensive DNA double-strand breaks and apoptosis. These data suggest combinations of 1 and 7 may be a valuable chemotherapeutic strategy.
Collapse
Affiliation(s)
- Laureen C Colis
- Department of Chemistry, Yale University, New Haven, CT 06520, United States
| | - Seth B Herzon
- Department of Chemistry, Yale University, New Haven, CT 06520, United States; Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
82
|
Osborne JD, Matthews TP, McHardy T, Proisy N, Cheung KMJ, Lainchbury M, Brown N, Walton MI, Eve PD, Boxall KJ, Hayes A, Henley AT, Valenti MR, De Haven Brandon AK, Box G, Jamin Y, Robinson SP, Westwood IM, van Montfort RLM, Leonard PM, Lamers MBAC, Reader JC, Aherne GW, Raynaud FI, Eccles SA, Garrett MD, Collins I. Multiparameter Lead Optimization to Give an Oral Checkpoint Kinase 1 (CHK1) Inhibitor Clinical Candidate: (R)-5-((4-((Morpholin-2-ylmethyl)amino)-5-(trifluoromethyl)pyridin-2-yl)amino)pyrazine-2-carbonitrile (CCT245737). J Med Chem 2016; 59:5221-37. [PMID: 27167172 DOI: 10.1021/acs.jmedchem.5b01938] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multiparameter optimization of a series of 5-((4-aminopyridin-2-yl)amino)pyrazine-2-carbonitriles resulted in the identification of a potent and selective oral CHK1 preclinical development candidate with in vivo efficacy as a potentiator of deoxyribonucleic acid (DNA) damaging chemotherapy and as a single agent. Cellular mechanism of action assays were used to give an integrated assessment of compound selectivity during optimization resulting in a highly CHK1 selective adenosine triphosphate (ATP) competitive inhibitor. A single substituent vector directed away from the CHK1 kinase active site was unexpectedly found to drive the selective cellular efficacy of the compounds. Both CHK1 potency and off-target human ether-a-go-go-related gene (hERG) ion channel inhibition were dependent on lipophilicity and basicity in this series. Optimization of CHK1 cellular potency and in vivo pharmacokinetic-pharmacodynamic (PK-PD) properties gave a compound with low predicted doses and exposures in humans which mitigated the residual weak in vitro hERG inhibition.
Collapse
|
83
|
Zhang D, Piao HL, Li YH, Qiu Q, Li DJ, Du MR, Tsang BK. Inhibition of AKT sensitizes chemoresistant ovarian cancer cells to cisplatin by abrogating S and G2/M arrest. Exp Mol Pathol 2016; 100:506-13. [PMID: 27163202 DOI: 10.1016/j.yexmp.2016.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 04/15/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is frequently altered in human malignancies and Akt over-expression and/or activation induces malignant transformation and chemoresistance. However, the role of Akt in the mechanisms of chemoresistance remains elusive. Here we reported that cisplatin treatment of chemosensitive, but not resistant, ovarian cancer cells (OVCAs) markedly increased the cell proportion in sub-G1 phase. Cisplatin however caused a significant accumulation of the resistant cells in S and G2/M phases, which was associated with a rapid and sustained checkpoint kinase 1 (Chk1) activation. In contrast, while cisplatin also elicited a rapid activation of Chk1 in sensitive cells, it markedly decreased total ChK1 and phospho-Chk1 contents over 12 h. Over-expression of dominant negative (DN)-AKT alone increased phospho-Chk1 content, and induced G2/M arrest and apoptosis. However, it inhibited Chk1 activation and G2/M arrest with combination of cisplatin treatment, resulting in p53-independent apoptosis. Furthermore, the responses of the chemoresistant cells to cisplatin were attenuated with forced expression of constitutive active AKT2. Chk1 knock-down also facilitated cisplatin-induced apoptosis in chemoresistant cells. Our studies implicate that, in addition to its cell survival and anti-apoptotic actions, Akt might also play an important role in the regulation of G2-M transition, possibly via up-regulation of Chk1 activity and stability. These data provide strong support for the concept that Akt is important in cell cycle regulation in the control of chemosensitivity in OVCAs and offers an alternate regulatory pathway for the development of rationale therapy for cisplatin-resistant ovarian cancer.
Collapse
Affiliation(s)
- Di Zhang
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Hai-Lan Piao
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Yan-Hong Li
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Qing Qiu
- Department of Obstetrics and Gynecology, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Canada; Department of Cellular and Molecular Medicine, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Canada; Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China
| | - Mei-Rong Du
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, 200011, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, 200011, China; Department of Obstetrics and Gynecology, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Canada.
| | - Benjamin K Tsang
- State Key Laboratory of Quality Research in Chinese Medicine and the Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau; Department of Obstetrics and Gynecology, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Canada; Department of Cellular and Molecular Medicine, and Interdisciplinary School of Health Sciences, University of Ottawa, Ottawa, Canada; Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Canada.
| |
Collapse
|
84
|
Jamatia R, Gupta A, Pal AK. Superparamagnetic Copper Ferrite Nanoparticles Catalyzed One Step Regioselective Synthesis of Dibenzodiazepinones via Ligand and Base Free Ullmann Type Coupling Reaction. ChemistrySelect 2016. [DOI: 10.1002/slct.201500038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ramen Jamatia
- Department of Chemistry; North-Eastern Hill University; Shillong- 793022 Meghalaya INDIA
| | - Ajay Gupta
- Department of Chemistry; North-Eastern Hill University; Shillong- 793022 Meghalaya INDIA
| | - Amarta Kumar Pal
- Department of Chemistry; North-Eastern Hill University; Shillong- 793022 Meghalaya INDIA
| |
Collapse
|
85
|
Preet R, Siddharth S, Satapathy SR, Das S, Nayak A, Das D, Wyatt MD, Kundu CN. Chk1 inhibitor synergizes quinacrine mediated apoptosis in breast cancer cells by compromising the base excision repair cascade. Biochem Pharmacol 2016; 105:23-33. [DOI: 10.1016/j.bcp.2016.01.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/25/2016] [Indexed: 11/26/2022]
|
86
|
Ogitani Y, Aida T, Hagihara K, Yamaguchi J, Ishii C, Harada N, Soma M, Okamoto H, Oitate M, Arakawa S, Hirai T, Atsumi R, Nakada T, Hayakawa I, Abe Y, Agatsuma T. DS-8201a, A Novel HER2-Targeting ADC with a Novel DNA Topoisomerase I Inhibitor, Demonstrates a Promising Antitumor Efficacy with Differentiation from T-DM1. Clin Cancer Res 2016; 22:5097-5108. [DOI: 10.1158/1078-0432.ccr-15-2822] [Citation(s) in RCA: 353] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/20/2016] [Indexed: 12/31/2022]
|
87
|
Pauty J, Côté MF, Rodrigue A, Velic D, Masson JY, Fortin S. Investigation of the DNA damage response to SFOM-0046, a new small-molecule drug inducing DNA double-strand breaks. Sci Rep 2016; 6:23302. [PMID: 27001483 PMCID: PMC4802344 DOI: 10.1038/srep23302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 03/03/2016] [Indexed: 01/25/2023] Open
Abstract
2-Ethylphenyl 4-(3-ethylureido)benzenesulfonate (SFOM-0046) is a novel anticancer agent that arrests cell cycle in S-phase and causes DNA replication stress leading to the phosphorylation of H2AX into γ-H2AX. First, using the M21, HT29, HT-1080 and HeLa cell lines, we confirmed that S-phase cell cycle arrest and γ-H2AX foci induction by SFOM-0046 is a general mechanism occurring in diverse cancer cell lines. In addition to γ-H2AX, SFOM-0046 activates preferentially ATR-Chk1 in M21 and HT29 cells while both ATR-Chk1 and ATM-Chk2 pathways are activated in HCT116 cells. Co-localization of SFOM-0046-induced 53BP1 foci with γ-H2AX foci validates that the DNA damage generated corresponds to double-strand-breaks (DSBs). Consistent with an S-phase arrest, SFOM-0046 treatment induces RAD51 foci formation but not DNA-PKcs foci, confirming that homologous recombination is the major DSB repair pathway targeted by the drug. Furthermore, using isogenic HCT116 p53+/+ and HCT116 p53−/− cells, we showed that p53 plays a key role in the survival mechanism to SFOM-0046. Finally, SFOM-0046 exhibits a dose-dependent antitumor activity on human fibrosarcoma HT-1080 tumours grafted onto chick chorioallantoic membranes without showing embryo toxicity even at high doses. Altogether, our results highlight SFOM-0046 as a very promising drug that induces a replication stress response.
Collapse
Affiliation(s)
- Joris Pauty
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Marie-France Côté
- CHU de Québec Research Center, Oncology Axis, Hôpital Saint-François d'Assise, 10 de l'Espinay, Quebec City, QC, G1L 3L5, Canada
| | - Amélie Rodrigue
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Denis Velic
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Jean-Yves Masson
- Genome Stability Laboratory, CHU de Québec Research Center, Oncology Axis, Hôtel-Dieu-de-Québec, 9 McMahon, Quebec City, QC, G1R 2J6, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Laval University, Quebec City, QC, G1V 0A6, Canada
| | - Sébastien Fortin
- CHU de Québec Research Center, Oncology Axis, Hôpital Saint-François d'Assise, 10 de l'Espinay, Quebec City, QC, G1L 3L5, Canada.,Faculty of Pharmacy, Laval University, Quebec City, QC, G1V 0A6, Canada
| |
Collapse
|
88
|
Choi HK, Ryu H, Son AR, Seo B, Hwang SG, Song JY, Ahn J. The novel anthraquinone derivative IMP1338 induces death of human cancer cells by p53-independent S and G2/M cell cycle arrest. Biomed Pharmacother 2016; 79:308-14. [PMID: 27044842 DOI: 10.1016/j.biopha.2016.02.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 10/22/2022] Open
Abstract
To identify novel small molecules that induce selective cancer cell death, we screened a chemical library containing 1040 compounds in HT29 colon cancer and CCD18-Co normal colon cells, using a phenotypic cell-based viability assay system with the Cell Counting Kit-8 (CCK-8). We discovered a novel anthraquinone derivative, N-(4-[{(9,10-dioxo-9,10-dihydro-1-anthracenyl)sulfonyl}amino]phenyl)-N-methylacetamide (IMP1338), which was cytotoxic against the human colon cancer cells tested. The MTT cell viability assay showed that treatment with IMP1338 selectively inhibited HCT116, HCT116 p53(-/-), HT29, and A549 cancer cell proliferation compared to that of Beas2B normal epithelial cells. To elucidate the cellular mechanism underlying the cytotoxicity of IMP1338, we examined the effect of IMP1338 on the cell cycle distribution and death of cancer cells. IMP1338 treatment significantly arrested the cell cycle at S and G2/M phases by DNA damage and led to apoptotic cell death, which was determined using FACS analysis with Annexin V/PI double staining. Furthermore, IMP1338 increased caspase-3 cleavage in wild-type p53, p53 knockout HCT116, and HT29 cells as determined using immunoblotting. In addition, IMP1338 markedly induced the phosphorylation of histone H2AX and Chk1 in both cell lines while the combination of 5-fluorouracil (5-FU) and radiation inhibited the viability of HCT116, HCT116 p53(-/-), and HT29 cells compared to 5-FU or radiation alone. Our findings indicated that IMP1338 induced p53-independent cell death through S and G2/M phase arrest as well as DNA damage. These results provide a basis for future investigations assessing the promising anticancer properties of IMP1338.
Collapse
Affiliation(s)
- Hyun Kyung Choi
- Department of Medicinal Chemistry, Jungwon University, 85 Munmuro, Goesan 28024, South Korea
| | - Hwani Ryu
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - A-Rang Son
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - Bitna Seo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea
| | - Jie-Young Song
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea.
| | - Jiyeon Ahn
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, 75 Nowonro Nowongu, Seoul 01812, South Korea.
| |
Collapse
|
89
|
Praveen Kumar C, Reddy TS, Mainkar PS, Bansal V, Shukla R, Chandrasekhar S, Hügel HM. Synthesis and biological evaluation of 5,10-dihydro-11 H -dibenzo[ b,e ][1,4]diazepin-11-one structural derivatives as anti-cancer and apoptosis inducing agents. Eur J Med Chem 2016; 108:674-686. [DOI: 10.1016/j.ejmech.2015.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/19/2015] [Accepted: 12/02/2015] [Indexed: 01/31/2023]
|
90
|
Chk1 Activation Protects Rad9A from Degradation as Part of a Positive Feedback Loop during Checkpoint Signalling. PLoS One 2015; 10:e0144434. [PMID: 26658951 PMCID: PMC4676731 DOI: 10.1371/journal.pone.0144434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/18/2015] [Indexed: 11/19/2022] Open
Abstract
Phosphorylation of Rad9A at S387 is critical for establishing a physical interaction with TopBP1, and to downstream activation of Chk1 for checkpoint activation. We have previously demonstrated a phosphorylation of Rad9A that occurs at late time points in cells exposed to genotoxic agents, which is eliminated by either Rad9A overexpression, or conversion of S387 to a non-phosphorylatable analogue. Based on this, we hypothesized that this late Rad9A phosphorylation is part of a feedback loop regulating the checkpoint. Here, we show that Rad9A is hyperphosphorylated and accumulates in cells exposed to bleomycin. Following the removal of bleomycin, Rad9A is polyubiquitinated, and Rad9A protein levels drop, indicating an active degradation process for Rad9A. Chk1 inhibition by UCN-01 or siRNA reduces Rad9A levels in cells synchronized in S-phase or exposed to DNA damage, indicating that Chk1 activation is required for Rad9A stabilization in S-phase and during checkpoint activation. Together, these results demonstrate a positive feedback loop involving Rad9A-dependend activation of Chk1, coupled with Chk1-dependent stabilization of Rad9A that is critical for checkpoint regulation.
Collapse
|
91
|
Hyun M, Choi S, Stevnsner T, Ahn B. The Caenorhabditis elegans Werner syndrome protein participates in DNA damage checkpoint and DNA repair in response to CPT-induced double-strand breaks. Cell Signal 2015; 28:214-223. [PMID: 26691982 DOI: 10.1016/j.cellsig.2015.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/01/2015] [Accepted: 12/11/2015] [Indexed: 12/16/2022]
Abstract
The RecQ helicases play roles in maintenance of genomic stability in species ranging from Escherichia coli to humans and interact with proteins involved in DNA metabolic pathways such as DNA repair, recombination, and replication. Our previous studies found that the Caenorhabditis elegans WRN-1 RecQ protein (a human WRN ortholog) exhibits ATP-dependent 3'-5' helicase activity and that the WRN-1 helicase is stimulated by RPA-1 on a long forked DNA duplex. However, the role of WRN-1 in response to S-phase associated with DSBs is unclear. We found that WRN-1 is involved in the checkpoint response to DSBs after CPT, inducing cell cycle arrest, is recruited to DSBs by RPA-1 and functions upstream of ATL-1 and ATM-1 for CHK-1 phosphorylation in the S-phase checkpoint. In addition, WRN-1 and RPA-1 recruitments to the DSBs require MRE-11, suggesting that DSB processing controlled by MRE-11 is important for WRN-1 at DSBs. The repair of CPT-induced DSBs is greatly reduced in the absence of WRN-1. These observations suggest that WRN-1 functions downstream of RPA-1 and upstream of CHK-1 in the DSB checkpoint pathway and is also required for the repair of DSB.
Collapse
Affiliation(s)
- Moonjung Hyun
- Department of Life Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Seoyun Choi
- Department of Life Sciences, University of Ulsan, Ulsan, Republic of Korea
| | - Tinna Stevnsner
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Byungchan Ahn
- Department of Life Sciences, University of Ulsan, Ulsan, Republic of Korea.
| |
Collapse
|
92
|
UV Damage-Induced Phosphorylation of HBO1 Triggers CRL4DDB2-Mediated Degradation To Regulate Cell Proliferation. Mol Cell Biol 2015; 36:394-406. [PMID: 26572825 DOI: 10.1128/mcb.00809-15] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
Histone acetyltransferase binding to ORC-1 (HBO1) is a critically important histone acetyltransferase for forming the prereplicative complex (pre-RC) at the replication origin. Pre-RC formation is completed by loading of the MCM2-7 heterohexameric complex, which functions as a helicase in DNA replication. HBO1 recruited to the replication origin by CDT1 acetylates histone H4 to relax the chromatin conformation and facilitates loading of the MCM complex onto replication origins. However, the acetylation status and mechanism of regulation of histone H3 at replication origins remain elusive. HBO1 positively regulates cell proliferation under normal cell growth conditions. Whether HBO1 regulates proliferation in response to DNA damage is poorly understood. In this study, we demonstrated that HBO1 was degraded after DNA damage to suppress cell proliferation. Ser50 and Ser53 of HBO1 were phosphorylated in an ATM/ATR DNA damage sensor-dependent manner after UV treatment. ATM/ATR-dependently phosphorylated HBO1 preferentially interacted with DDB2 and was ubiquitylated by CRL4(DDB2). Replacement of endogenous HBO1 in Ser50/53Ala mutants maintained acetylation of histone H3K14 and impaired cell cycle regulation in response to UV irradiation. Our findings demonstrate that HBO1 is one of the targets in the DNA damage checkpoint. These results show that ubiquitin-dependent control of the HBO1 protein contributes to cell survival during UV irradiation.
Collapse
|
93
|
Massey AJ, Stokes S, Browne H, Foloppe N, Fiumana A, Scrace S, Fallowfield M, Bedford S, Webb P, Baker L, Christie M, Drysdale MJ, Wood M. Identification of novel, in vivo active Chk1 inhibitors utilizing structure guided drug design. Oncotarget 2015; 6:35797-812. [PMID: 26437226 PMCID: PMC4742142 DOI: 10.18632/oncotarget.5929] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/14/2015] [Indexed: 12/13/2022] Open
Abstract
Chk1 kinase is a critical component of the DNA damage response checkpoint especially in cancer cells and targeting Chk1 is a potential therapeutic opportunity for potentiating the anti-tumor activity of DNA damaging chemotherapy drugs. Fragment elaboration by structure guided design was utilized to identify and develop a novel series of Chk1 inhibitors culminating in the identification of V158411, a potent ATP-competitive inhibitor of the Chk1 and Chk2 kinases. V158411 abrogated gemcitabine and camptothecin induced cell cycle checkpoints, resulting in the expected modulation of cell cycle proteins and increased cell death in cancer cells. V158411 potentiated the cytotoxicity of gemcitabine, cisplatin, SN38 and camptothecin in a variety of p53 deficient human tumor cell lines in vitro, p53 proficient cells were unaffected. In nude mice, V158411 showed minimal toxicity as a single agent and in combination with irinotecan. In tumor bearing animals, V158411 was detected at high levels in the tumor with a long elimination half-life; no pharmacologically significant in vivo drug-drug interactions with irinotecan were identified through analysis of the pharmacokinetic profiles. V158411 potentiated the anti-tumor activity of irinotecan in a variety of human colon tumor xenograft models without additional systemic toxicity. These results demonstrate the opportunity for combining V158411 with standard of care chemotherapeutic agents to potentiate the therapeutic efficacy of these agents without increasing their toxicity to normal cells. Thus, V158411 would warrant further clinical evaluation.
Collapse
Affiliation(s)
| | | | | | | | | | - Simon Scrace
- Vernalis Research, Granta Park, Cambridge, UK
- Horizon Discovery, Cambridge Research Park, Waterbeach, Cambridge, UK
| | | | | | - Paul Webb
- Vernalis Research, Granta Park, Cambridge, UK
| | - Lisa Baker
- Vernalis Research, Granta Park, Cambridge, UK
| | | | - Martin J. Drysdale
- Vernalis Research, Granta Park, Cambridge, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow, UK
| | - Mike Wood
- Vernalis Research, Granta Park, Cambridge, UK
| |
Collapse
|
94
|
Serine–Threonine Kinase 38 regulates CDC25A stability and the DNA damage-induced G2/M checkpoint. Cell Signal 2015; 27:1569-75. [DOI: 10.1016/j.cellsig.2015.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/10/2015] [Accepted: 04/26/2015] [Indexed: 12/19/2022]
|
95
|
Cloning and expression of a Chk1 gene in Daphnia pulex during different modes of reproduction. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0310-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
96
|
Abstract
In order to maintain genomic stability, cells have developed sophisticated signalling pathways to enable DNA damage or DNA replication stress to be resolved. Key mediators of this DNA damage response (DDR) are the ATM and ATR kinases, which induce cell cycle arrest and facilitate DNA repair via their downstream targets. Inhibiting the DDR has become an attractive therapeutic concept in cancer therapy, since (i) resistance to genotoxic therapies has been associated with increased DDR signalling, and (ii) many cancers have defects in certain components of the DDR rendering them highly dependent on the remaining DDR pathways for survival. ATM and ATR act as the apical regulators of the response to DNA double strand breaks and replication stress, respectively, with overlapping but non-redundant activities. Highly selective small molecule inhibitors of ATM and ATR are currently in preclinical and clinical development, respectively. Preclinical data have provided a strong rationale for clinical testing of these compounds both in combination with radio- or chemotherapy, and in synthetic lethal approaches to treat tumours with deficiencies in certain DDR components. Whole genome sequencing studies have reported that mutations in DDR genes occur with a high frequency in many common tumour types, suggesting that a synthetic lethal approach with ATM or ATR inhibitors could have widespread utility, providing that appropriate biomarkers are developed.
Collapse
Affiliation(s)
- Anika Maria Weber
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, The Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Anderson Joseph Ryan
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, The Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
97
|
Yuan Z, Guo W, Yang J, Li L, Wang M, Lei Y, Wan Y, Zhao X, Luo N, Cheng P, Liu X, Nie C, Peng Y, Tong A, Wei Y. PNAS-4, an Early DNA Damage Response Gene, Induces S Phase Arrest and Apoptosis by Activating Checkpoint Kinases in Lung Cancer Cells. J Biol Chem 2015; 290:14927-44. [PMID: 25918161 DOI: 10.1074/jbc.m115.658419] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Indexed: 02/05/2023] Open
Abstract
PNAS-4, a novel pro-apoptotic gene, was activated during the early response to DNA damage. Our previous study has shown that PNAS-4 induces S phase arrest and apoptosis when overexpressed in A549 lung cancer cells. However, the underlying action mechanism remains far from clear. In this work, we found that PNAS-4 expression in lung tumor tissues is significantly lower than that in adjacent lung tissues; its expression is significantly increased in A549 cells after exposure to cisplatin, methyl methane sulfonate, and mitomycin; and its overexpression induces S phase arrest and apoptosis in A549 (p53 WT), NCI-H460 (p53 WT), H526 (p53 mutation), and Calu-1 (p53(-/-)) lung cancer cells, leading to proliferation inhibition irrespective of their p53 status. The S phase arrest is associated with up-regulation of p21(Waf1/Cip1) and inhibition of the Cdc25A-CDK2-cyclin E/A pathway. Up-regulation of p21(Waf1/Cip1) is p53-independent and correlates with activation of ERK. We further showed that the intra-S phase checkpoint, which occurs via DNA-dependent protein kinase-mediated activation of Chk1 and Chk2, is involved in the S phase arrest and apoptosis. Gene silencing of Chk1/2 rescues, whereas that of ATM or ATR does not affect, S phase arrest and apoptosis. Furthermore, human PNAS-4 induces DNA breaks in comet assays and γ-H2AX staining. Intriguingly, caspase-dependent cleavage of Chk1 has an additional role in enhancing apoptosis. Taken together, our findings suggest a novel mechanism by which elevated PNAS-4 first causes DNA-dependent protein kinase-mediated Chk1/2 activation and then results in inhibition of the Cdc25A-CDK2-cyclin E/A pathway, ultimately causing S phase arrest and apoptosis in lung cancer cells.
Collapse
Affiliation(s)
- Zhu Yuan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China,
| | - Wenhao Guo
- the Department of Abdominal Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, No. 37, Guoxue Road, Chengdu 610041, Sichuan Province, China, and
| | - Jun Yang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Lei Li
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Meiliang Wang
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Yi Lei
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Yang Wan
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Xinyu Zhao
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Na Luo
- the Nankai University School of Medicine/Collaborative Innovation Center of Biotherapy, Tianjin 300071, China
| | - Ping Cheng
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Xinyu Liu
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Chunlai Nie
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Yong Peng
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| | - Aiping Tong
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China,
| | - Yuquan Wei
- From the State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, 17 People's South Road, Chengdu 610041, China
| |
Collapse
|
98
|
Kartal-Yandim M, Adan-Gokbulut A, Baran Y. Molecular mechanisms of drug resistance and its reversal in cancer. Crit Rev Biotechnol 2015; 36:716-26. [DOI: 10.3109/07388551.2015.1015957] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Melis Kartal-Yandim
- Department of Molecular Biology and Genetics, İzmir Institute of Technology, Urla, İzmir, Turkey and
| | - Aysun Adan-Gokbulut
- Department of Molecular Biology and Genetics, İzmir Institute of Technology, Urla, İzmir, Turkey and
| | - Yusuf Baran
- Department of Molecular Biology and Genetics, İzmir Institute of Technology, Urla, İzmir, Turkey and
- Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, Turkey
| |
Collapse
|
99
|
Gomha SM, Abdulla MM, Abou-Seri SM. Identification of novel aminothiazole and aminothiadiazole conjugated cyanopyridines as selective CHK1 inhibitors. Eur J Med Chem 2015; 92:459-70. [PMID: 25594740 DOI: 10.1016/j.ejmech.2015.01.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/08/2014] [Accepted: 01/09/2015] [Indexed: 10/24/2022]
Abstract
Inhibitors of checkpoint kinase 1 (CHK1) are of current interest as potential anti-tumor agents. Novel series of cyanopyridyl-aminothiadiazoles (synthesized from reaction of 1-(3-cyano-4,6-diphenylpyridin-2-yl)-3-phenylthiourea (14) with hydrazonoyl halides) and cyanopyridyl-aminothiazolyl-thiadiazoles (synthesized from treatment of 14 with ethyl chloroacetate followed by reaction of the obtained cyanopyridyl-aminothiazole with hydrazonoyl halides) were synthesized and evaluated for their CHK1 inhibitory potential using a cell-based assay cascade. The tested compounds exhibited a potent and selective CHK1 inhibitory activity at nanomolar levels that reflected their ability to abrogate cell cycle arrest and potentiate the cytotoxic effect of the genotoxic drug gemcitabine in colon cancer cells. Molecular modeling simulation revealed that, the most active compound 28a docked well into the enzyme active site and their complex is stabilized by a key H-bonding with the backbone amide of Cys-87 as well as multiple favorable hydrophobic interactions with different hydrophobic binding regions of the enzyme.
Collapse
Affiliation(s)
- Sobhi M Gomha
- Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt.
| | | | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Egypt
| |
Collapse
|
100
|
Pessach I, Papoudou-Bai A, Goussia A, Kamina S, Kyrtsonis MC, Bourantas KL, Kanavaros P. Immunohistochemical expression of cell cycle proteins in multiple myeloma. Leuk Lymphoma 2015; 56:2720-3. [PMID: 25573203 DOI: 10.3109/10428194.2015.1004171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ilias Pessach
- a Hematology Clinic, University Hospital of Ioannina , Ioannina , Greece
| | | | - Anna Goussia
- b Department of Pathology , University of Ioannina , Ioannina , Greece
| | - Sevasti Kamina
- b Department of Pathology , University of Ioannina , Ioannina , Greece
| | - Marie-Christine Kyrtsonis
- c Hematology Section, First Department of Propedeutic Internal Medicine , Laikon University Hospital , Athens , Greece
| | | | - Panagiotis Kanavaros
- d Department of Anatomy-Histology-Embryology , School of Medicine, University of Ioannina , Ioannina , Greece
| |
Collapse
|