1
|
Wang JM, Zhang FH, Liu ZX, Tang YJ, Li JF, Xie LP. Cancer on motors: How kinesins drive prostate cancer progression? Biochem Pharmacol 2024; 224:116229. [PMID: 38643904 DOI: 10.1016/j.bcp.2024.116229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/02/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Prostate cancer causes numerous male deaths annually. Although great progress has been made in the diagnosis and treatment of prostate cancer during the past several decades, much about this disease remains unknown, especially its pathobiology. The kinesin superfamily is a pivotal group of motor proteins, that contains a microtubule-based motor domain and features an adenosine triphosphatase activity and motility characteristics. Large-scale sequencing analyses based on clinical samples and animal models have shown that several members of the kinesin family are dysregulated in prostate cancer. Abnormal expression of kinesins could be linked to uncontrolled cell growth, inhibited apoptosis and increased metastasis ability. Additionally, kinesins may be implicated in chemotherapy resistance and escape immunologic cytotoxicity, which creates a barrier to cancer treatment. Here we cover the recent advances in understanding how kinesins may drive prostate cancer progression and how targeting their function may be a therapeutic strategy. A better understanding of kinesins in prostate cancer tumorigenesis may be pivotal for improving disease outcomes in prostate cancer patients.
Collapse
Affiliation(s)
- Jia-Ming Wang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Feng-Hao Zhang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zi-Xiang Liu
- Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, People's Republic of China
| | - Yi-Jie Tang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jiang-Feng Li
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
| | - Li-Ping Xie
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.
| |
Collapse
|
2
|
Wu F, Chen R, Li Y, Wan Y, Hu J. Unregistered Hexaphenoxycyclotriphosphazene and Its Metabolite Antagonize Retinoic Acid and Retinoic X Receptors and Cause Early Developmental Damage. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:20551-20558. [PMID: 38037888 DOI: 10.1021/acs.est.3c07997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Hexaphenoxycyclotriphosphazene (HPCTP), an unregistered chemical, has been used as a substitute for triphenyl phosphate in flame retardants and plasticizers. Here, we identified its metabolite, pentaphenoxycyclotriphosphazene (PPCTP) in the liver of Japanese medaka exposed to HPCTP. When sexually mature female medaka were exposed to HPCTP at 37.0, 90.4, and 465.4 ng/L for 35 days, the HPCTP concentration (642.1-2531.9 ng/g lipid weight [lw]) in the embryos considerably exceeded that (34.7-298.1 ng/g lw) in the maternal muscle, indicating remarkable maternal transfer. During 0-9 days postfertilization, the HPCTP concentration in the embryos decreased continuously, while the PPCTP concentration increased. HPCTP and PPCTP antagonized the retinoic X receptor with 50% inhibitory concentrations (IC50) of 34.8 and 21.2 μM, respectively, and PPCTP also antagonized the retinoic acid receptor with IC50 of 2.79 μM. Such antagonistic activities may contribute to eye deformity (4.7% at 465.4 ng/L), body malformation (2.1% at 90.4 ng/L and 6.8% at 465.4 ng/L), and early developmental mortality (11.6-21.7% in all exposure groups) of the embryos. HPCTP was detected in a main tributary of the Yangtze River Basin. Thus, HPCTP poses a risk to wild fish populations, given the developmental toxicities associated with this chemical and its metabolite.
Collapse
Affiliation(s)
- Feifan Wu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Ruichao Chen
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Yu Li
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Yi Wan
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| | - Jianying Hu
- MOE Laboratory for Earth Surface Process, College of Urban and Environmental Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
3
|
Yue C, Chai Z, Hu Z, Shang L, Deng Y, Tang YZ. Deficiency of nitrogen but not phosphorus triggers the life cycle transition of the dinoflagellate Scrippsiella acuminata from vegetative growth to resting cyst formation. HARMFUL ALGAE 2022; 118:102312. [PMID: 36195426 DOI: 10.1016/j.hal.2022.102312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 06/16/2023]
Abstract
Nitrogen (N) and phosphorus (P) are essential elements for algal growth. When N and P are deficient, dinoflagellates will take a series of measures to achieve population continuation including formation of resting cysts, an important ecological strategy of dinoflagellates that plays a key role in the initiation and termination of harmful algal blooms (HABs). How the deficiency of N and P affects algal growth and cyst formation has been investigated in some dinoflagellate species, but how it affects the life cycle transition in dinoflagellates has been poorly understood. In this study, we further explored the effect of N and P deficiency on the algal growth and resting cyst production in the cosmopolitan HABs-causing species Scrippsiella acuminata via refining the N and P concentration gradients. Further, we tracked the expression patterns of one CyclinB and one CDK1 genes of S. acuminata at different growth stages under three deficiency concentrations (1/1000 dilutions of N, P, and both N and P). The results suggest that N deficiency always triggered the cyst formation but P deficiency mainly inhibited the vegetative growth instead of inducing cyst formation. We also observed the highest cyst production when S. acuminata was cultured in the f/2-Si medium that was a one-thousandth dilution of N and P (N∼ 0.882 μM; P∼ 0.0362 μM). Our results for the expressions of CyclinB and CDK1 were well consistent with the results of algal growth and cyst formation at different deficiencies of N and P in terms of that higher expressions of these two genes were corresponding to higher rates of vegetative cell growth, while their expressions in resting cysts maintained to be moderate but significantly lower than that in fast-growing vegetative cells. Although we are still not sure whether the changing expressions of the two genes did regulate the transition of life cycle (i.e. cyst formation), or happened as parallels to the expressions of other truly regulating genes, our observations are surely inspirational for further investigations on the genetic regulation of life cycle transition in dinoflagellates. Our work will provide clues to probe the physiological and molecular mechanisms underlying the nutrient deficiency-induced alternation between life cycle stages in dinoflagellates.
Collapse
Affiliation(s)
- Caixia Yue
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaoyang Chai
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Zhangxi Hu
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Lixia Shang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China
| | - Yunyan Deng
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China.
| | - Ying Zhong Tang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, 266071, China.
| |
Collapse
|
4
|
Kim S, Leem J, Oh JS, Kim JS. Cytotoxicity of 9,10-Phenanthrenequinone Impairs Mitotic Progression and Spindle Assembly Independent of ROS Production in HeLa Cells. TOXICS 2022; 10:toxics10060327. [PMID: 35736935 PMCID: PMC9227850 DOI: 10.3390/toxics10060327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022]
Abstract
The polycyclic aromatic hydrocarbon quinone derivative 9,10-phenanthrenequinone (9,10-PQ) is one of the most abundant and toxic components found in diesel exhaust particles (DEPs). These DEPs are created during diesel fuel combustion and are considered the main source of urban air pollution. As 9,10-PQ can produce excessive reactive oxygen species (ROS) through redox cycling, it has been shown to exert potent cytotoxic effects against various cell types. However, the mechanisms underlying this cytotoxicity remain unclear. In this study, we showed that 9,10-PQ exerts cytotoxicity by impairing mitotic progression and spindle assembly in HeLa cells. Exposure to 9,10-PQ impaired spindle assembly and chromosome alignment, resulting in delayed mitotic entry and progression in HeLa cells. Furthermore, 9,10-PQ exposure decreased the CEP192 and p-Aurora A levels at the spindle poles. Notably, these mitotic defects induced by 9,10-PQ were not rescued by scavenging ROS, implying the ROS-independent activity of 9,10-PQ. Therefore, our results provide the first evidence that 9,10-PQ exerts its cytotoxicity through specific inhibition of mitotic progression and spindle assembly, independent of ROS.
Collapse
Affiliation(s)
- Seul Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
| | - Jiyeon Leem
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Jeong Su Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (J.S.O.); (J.-S.K.)
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea;
- Correspondence: (J.S.O.); (J.-S.K.)
| |
Collapse
|
5
|
Qu J, Wu X, Wang Q, Wang J, Sun X, Ji D, Li Y. Effect of miR-101 on the Proliferation and Apoptosis of Goat Hair Follicle Stem Cells. Genes (Basel) 2022; 13:genes13061035. [PMID: 35741797 PMCID: PMC9222262 DOI: 10.3390/genes13061035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 02/04/2023] Open
Abstract
The Yangtze River Delta white goat is a rare goat species capable of producing high-quality brush hair. Dual specificity protein phosphatase 1 (DUSP1) may play a role in the formation of high-quality brush hair, as evidenced by our previous research. We investigated the potential mechanisms that regulate the proliferation and apoptosis of goat hair follicle stem cells. We particularly focused on the relationship between DUSP1 and miR-101, which directly targets DUSP1, predicted and screened through bioinformatics websites. Then, fluorescence assays, flow cytometry, RT-qPCR, and Western blotting were used to investigate the effects of miR-101 on the proliferation and apoptosis of hair follicle stem cells. We found that miR-101 overexpression significantly decreased (p < 0.01) apoptosis and promoted the proliferation of hair follicle stem cells. Furthermore, the overexpression of miR-101 increased (p < 0.05) the mRNA and protein expression levels of the proliferation-related gene (PCNA) and anti-apoptotic gene (Bcl-2), and it decreased (p < 0.05) the mRNA and protein expression levels of the apoptotic gene (Bax). In conclusion, miR-101 can promote the proliferation of and inhibit the apoptosis of hair follicle stem cells by targeting DUSP1, which provides a theoretical basis for further elucidating the molecular mechanism that regulates the production of high-quality brush hair of Yangtze River Delta white goats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yongjun Li
- Correspondence: ; Tel.: +86-514-8799-6481
| |
Collapse
|
6
|
Aiba Y, Kim J, Imamura A, Okumoto K, Nakajo N. Regulation of Myt1 kinase activity via its N-terminal region in Xenopus meiosis and mitosis. Cells Dev 2021; 169:203754. [PMID: 34695617 DOI: 10.1016/j.cdev.2021.203754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/28/2022]
Abstract
Immature animal oocytes are naturally arrested at the first meiotic prophase (Pro-I), which corresponds to the G2 phase of the cell cycle. In Xenopus oocytes, Myt1 kinase phosphorylates and inactivates cyclin-dependent kinase 1 (Cdk1) at Pro-I, thereby preventing oocytes from entering meiosis I (MI) prematurely. Previous studies have shown that, upon resuming MI, Cdk1 and p90rsk, which is a downstream kinase of the Mos-MAPK pathway, in turn phosphorylate the C-terminal region of Myt1, to suppress its activity, thereby ensuring high Cdk1 activity during M phase. However, the roles of the N-terminal region of Myt1 during meiosis and mitosis remain to be elucidated. In the present study, we show that the N-terminal region of Myt1 participates in the regulation of Myt1 activity in the Xenopus cell cycle. In particular, we found that a short, conserved sequence in the N-terminal region, termed here as the PAYF motif, is required for the normal activity of Myt1 in oocytes. Furthermore, multiple phosphorylations by Cdk1 at the Myt1 N-terminal region were found to be involved in the negative regulation of Myt1. In particular, phosphorylations at Thr11 and Thr16 of Myt1, which are adjacent to the PAYF motif, were found to be important for the inactivation of Myt1 in the M phase of the cell cycle. These results suggest that in addition to the regulation of Myt1 activity via the C-terminal region, the N-terminal region of Myt1 also plays an important role in the regulation of Myt1 activity.
Collapse
Affiliation(s)
- Yukito Aiba
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan.
| | - Jihoon Kim
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan.
| | - Arata Imamura
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan.
| | - Kanji Okumoto
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan; Department of Biology, Graduate School of Sciences, Kyushu University, Fukuoka, Japan.
| | - Nobushige Nakajo
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan; Department of Biology, Graduate School of Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
7
|
Murase Y, Ono H, Ogawa K, Yoshioka R, Ishikawa Y, Ueda H, Akahoshi K, Ban D, Kudo A, Tanaka S, Tanabe M. Inhibitor library screening identifies ispinesib as a new potential chemotherapeutic agent for pancreatic cancers. Cancer Sci 2021; 112:4641-4654. [PMID: 34510663 PMCID: PMC8586681 DOI: 10.1111/cas.15134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022] Open
Abstract
Screening custom‐made libraries of inhibitors may reveal novel drugs for treating pancreatic cancer. In this manner, we identified ispinesib as a candidate and attempted to determine its clinical efficacy and the biological significance of its functional target Eg5 in pancreatic cancer. One hundred compounds in our library were screened for candidate drugs using cell cytotoxicity assays. Ispinesib was found to mediate effective antitumor effects in pancreatic cancer. The clinical significance of the expression of the ispinesib target Eg5 was investigated in 165 pancreatic cancer patients by immunohistochemical staining and in Eg5‐positive pancreatic cancer patient‐derived xenograft (PDX) mouse models. Patients with Eg5‐positive tumors experienced significantly poorer clinical outcomes than those not expressing Eg5 (overall survival; P < .01, recurrence‐free survival; P < .01). Ispinesib or Eg5 inhibition with specific siRNA significantly suppressed cell proliferation and induced apoptosis in pancreatic cancer cell lines. Mechanistically, ispinesib acted by inducing incomplete mitosis with nuclear disruption, resulting in multinucleated monoastral spindle cells. In the PDX mouse model, ispinesib dramatically reduced tumor growth relative to vehicle control (652.2 mm3 vs 18.1 mm3 in mean tumor volume, P < .01 by ANOVA; 545 mg vs 28 mg in tumor weight, P < .01, by ANOVA). Ispinesib, identified by inhibitor library screening, could be a promising novel therapeutic agent for pancreatic cancer. The expression of its target Eg5 is associated with poorer postoperative prognosis and is important for the clinical efficacy of ispinesib in pancreatic cancer.
Collapse
Affiliation(s)
- Yoshiki Murase
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroaki Ono
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kosuke Ogawa
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Risa Yoshioka
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiya Ishikawa
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroki Ueda
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichi Akahoshi
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Division of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepatobiliary and Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
8
|
Yang J, Virostko J, Hormuth DA, Liu J, Brock A, Kowalski J, Yankeelov TE. An experimental-mathematical approach to predict tumor cell growth as a function of glucose availability in breast cancer cell lines. PLoS One 2021; 16:e0240765. [PMID: 34255770 PMCID: PMC8277046 DOI: 10.1371/journal.pone.0240765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
We present the development and validation of a mathematical model that predicts how glucose dynamics influence metabolism and therefore tumor cell growth. Glucose, the starting material for glycolysis, has a fundamental influence on tumor cell growth. We employed time-resolved microscopy to track the temporal change of the number of live and dead tumor cells under different initial glucose concentrations and seeding densities. We then constructed a family of mathematical models (where cell death was accounted for differently in each member of the family) to describe overall tumor cell growth in response to the initial glucose and confluence conditions. The Akaikie Information Criteria was then employed to identify the most parsimonious model. The selected model was then trained on 75% of the data to calibrate the system and identify trends in model parameters as a function of initial glucose concentration and confluence. The calibrated parameters were applied to the remaining 25% of the data to predict the temporal dynamics given the known initial glucose concentration and confluence, and tested against the corresponding experimental measurements. With the selected model, we achieved an accuracy (defined as the fraction of measured data that fell within the 95% confidence intervals of the predicted growth curves) of 77.2 ± 6.3% and 87.2 ± 5.1% for live BT-474 and MDA-MB-231 cells, respectively.
Collapse
Affiliation(s)
- Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Jack Virostko
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Oncology, The University of Texas at Austin, Austin, Texas, United States of America
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America
| | - David A. Hormuth
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Junyan Liu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Oncology, The University of Texas at Austin, Austin, Texas, United States of America
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America
| | - Jeanne Kowalski
- Department of Oncology, The University of Texas at Austin, Austin, Texas, United States of America
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America
| | - Thomas E. Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Oncology, The University of Texas at Austin, Austin, Texas, United States of America
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas, United States of America
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas, United States of America
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| |
Collapse
|
9
|
Li Q, Yang F, Shi X, Bian S, Shen F, Wu Y, Zhu C, Fu F, Wang J, Zhou J, Chen Y. MTHFD2 promotes ovarian cancer growth and metastasis via activation of the STAT3 signaling pathway. FEBS Open Bio 2021; 11:2845-2857. [PMID: 34231329 PMCID: PMC8487042 DOI: 10.1002/2211-5463.13249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 12/02/2022] Open
Abstract
Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) is a bifunctional enzyme located in the mitochondria. MTHFD2 has been reported to be overexpressed in several malignant tumors and is implicated in cancer development. This study aimed to investigate the effect of MTHFD2 on ovarian cancer progression. The expression of MTHFD2 was detected by bioinformatic analysis, immunohistochemistry, RT‐qPCR (real‐time quantitative PCR analysis), and western blot analysis. The effects of MTHFD2 depletion on cell proliferation, migration, and invasion were determined through in vitro experiments. Cell cycle progression and apoptosis were accessed by flow cytometry. The related signaling pathway protein expression was determined by western blot analysis. We found that MTHFD2 is highly expressed in both ovarian cancer tissues and cell lines. MTHFD2 deletion suppressed cell proliferation and metastasis. Knockdown of MTHFD2 induces cell apoptosis and G2/M arrest, whereas the number of cells in S phase increased with MTHFD2 overexpression. Mechanically, our results indicate that an inhibitory effect of MTHFD2 knockdown may be mediated by the downregulation of cyclin B1/Cdc2 complex and the inhibitory effect on its activity. Additionally, MTHFD2 could regulate cell growth and aggressiveness via activation of STAT3 and the STAT3‐induced epithelial–mesenchymal transition signaling pathway. These findings indicate that MTHFD2 is overexpressed in ovarian cancer and regulates cell proliferation and metastasis, presenting an attractive therapeutic target.
Collapse
Affiliation(s)
- Qiutong Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Fang Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,The Second Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Xiu Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Shimin Bian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Fangrong Shen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Yuhong Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Chenjie Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Fengqing Fu
- Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Juan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Clinical Research Center of Obstetrics and Gynecology, Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China.,Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
10
|
Pharmacological relevance of CDK inhibitors in Alzheimer's disease. Neurochem Int 2021; 148:105115. [PMID: 34182065 DOI: 10.1016/j.neuint.2021.105115] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/11/2022]
Abstract
Evidence suggests that cell cycle activation plays a role in the pathophysiology of neurodegenerative diseases. Alzheimer's disease is a progressive, terminal neurodegenerative disease that affects memory and other important mental functions. Intracellular deposition of Tau protein, a hyperphosphorylated form of a microtubule-associated protein, and extracellular aggregation of Amyloid β protein, which manifests as neurofibrillary tangles (NFT) and senile plaques, respectively, characterize this condition. In recent years, however, several studies have concluded that cell cycle re-entry is one of the key causes of neuronal death in the pathogenesis of Alzheimer's disease. The eukaryotic cell cycle is well-coordinated machinery that performs critical functions in cell replenishment, such as DNA replication, cell creation, repair, and the birth of new daughter cells from the mother cell. The complex interplay between the levels of various cyclins and cyclin-dependent kinases (CDKs) at different checkpoints is needed for cell cycle synchronization. CDKIs (cyclin-dependent kinase inhibitors) prevent cyclin degradation and CDK inactivation. Different external and internal factors regulate them differently, and they have different tissue expression and developmental functions. The checkpoints ensure that the previous step is completed correctly before starting the new cell cycle phase, and they protect against the transfer of defects to the daughter cells. Due to the development of more selective and potent ATP-competitive CDK inhibitors, CDK inhibitors appear to be on the verge of having a clinical impact. This avenue is likely to yield new and effective medicines for the treatment of cancer and other neurodegenerative diseases. These new methods for recognizing CDK inhibitors may be used to create non-ATP-competitive agents that target CDK4, CDK5, and other CDKs that have been recognized as important therapeutic targets in Alzheimer's disease treatment.
Collapse
|
11
|
Extracellular Vesicles from Plants: Current Knowledge and Open Questions. Int J Mol Sci 2021; 22:ijms22105366. [PMID: 34065193 PMCID: PMC8160738 DOI: 10.3390/ijms22105366] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
The scientific interest in the beneficial properties of natural substances has been recognized for decades, as well as the growing attention in extracellular vesicles (EVs) released by different organisms, in particular from animal cells. However, there is increasing interest in the isolation and biological and functional characterization of these lipoproteic structures in the plant kingdom. Similar to animal vesicles, these plant-derived extracellular vesicles (PDEVs) exhibit a complex content of small RNAs, proteins, lipids, and other metabolites. This sophisticated composition enables PDEVs to be therapeutically attractive. In this review, we report and discuss current knowledge on PDEVs in terms of isolation, characterization of their content, biological properties, and potential use as drug delivery systems. In conclusion, we outline controversial issues on which the scientific community shall focus the attention shortly.
Collapse
|
12
|
Grapefruit-Derived Micro and Nanovesicles Show Distinct Metabolome Profiles and Anticancer Activities in the A375 Human Melanoma Cell Line. Cells 2020; 9:cells9122722. [PMID: 33371199 PMCID: PMC7766354 DOI: 10.3390/cells9122722] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Fruit juice is one of the most easily accessible resources for the isolation of plant-derived vesicles. Here we found that micro- and nano-sized vesicles (MVs and NVs) from four Citrus species, C. sinensis, C. limon, C. paradisi and C. aurantium, specifically inhibit the proliferation of lung, skin and breast cancer cells, with no substantial effect on the growth of non-cancer cells. Cellular and molecular analyses demonstrate that grapefruit-derived vesicles cause cell cycle arrest at G2/M checkpoint associated with a reduced cyclins B1 and B2 expression levels and the upregulation of cell cycle inhibitor p21. Further data suggest the inhibition of Akt and ERK signalling, reduced intercellular cell adhesion molecule-1 and cathepsins expressions, and the presence of cleaved PARP-1, all associated with the observed changes at the cellular level. Gas chromatography-mass spectrometry-based metabolomics reveals distinct metabolite profiles for the juice and vesicle fractions. NVs exhibit a high relative amount of amino acids and organic acids whereas MVs and fruit juice are characterized by a high percentage of sugars and sugar derivatives. Grapefruit-derived NVs are in particular rich in alpha–hydroxy acids and leucine/isoleucine, myo-inositol and doconexent, while quininic acid was detected in MVs. Our findings reveal the metabolite signatures of grapefruit-derived vesicles and substantiate their potential use in new anticancer strategies.
Collapse
|
13
|
Cuijpers SAG, Willemstein E, Ruppert JG, van Elsland DM, Earnshaw WC, Vertegaal ACO. Chromokinesin KIF4A teams up with stathmin 1 to regulate abscission in a SUMO-dependent manner. J Cell Sci 2020; 133:jcs248591. [PMID: 32591481 PMCID: PMC7390632 DOI: 10.1242/jcs.248591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022] Open
Abstract
Cell division ends when two daughter cells physically separate via abscission, the cleavage of the intercellular bridge. It is not clear how the anti-parallel microtubule bundles bridging daughter cells are severed. Here, we present a novel abscission mechanism. We identified chromokinesin KIF4A, which is adjacent to the midbody during cytokinesis, as being required for efficient abscission. KIF4A is regulated by post-translational modifications. We evaluated modification of KIF4A by the ubiquitin-like protein SUMO. We mapped lysine 460 in KIF4A as the SUMO acceptor site and employed CRISPR-Cas9-mediated genome editing to block SUMO conjugation of endogenous KIF4A. Failure to SUMOylate this site in KIF4A delayed cytokinesis. SUMOylation of KIF4A enhanced the affinity for the microtubule destabilizer stathmin 1 (STMN1). We here present a new level of abscission regulation through the dynamic interactions between KIF4A and STMN1 as controlled by SUMO modification of KIF4A.
Collapse
Affiliation(s)
- Sabine A G Cuijpers
- Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Edwin Willemstein
- Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jan G Ruppert
- Wellcome Centre for Cell Biology, University of Edinburgh, EH9 3JR Edinburgh, Scotland, UK
| | - Daphne M van Elsland
- Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - William C Earnshaw
- Wellcome Centre for Cell Biology, University of Edinburgh, EH9 3JR Edinburgh, Scotland, UK
| | - Alfred C O Vertegaal
- Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
14
|
McBeath E, Parker-Thornburg J, Fujii Y, Aryal N, Smith C, Hofmann MC, Abe JI, Fujiwara K. Rapid Evaluation of CRISPR Guides and Donors for Engineering Mice. Genes (Basel) 2020; 11:E628. [PMID: 32521708 PMCID: PMC7349695 DOI: 10.3390/genes11060628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/27/2022] Open
Abstract
Although the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/ CRISPR associated protein 9 (Cas9) technique has dramatically lowered the cost and increased the speed of generating genetically engineered mice, success depends on using guide RNAs and donor DNAs which direct efficient knock-out (KO) or knock-in (KI). By Sanger sequencing DNA from blastocysts previously injected with the same CRISPR components intended to produce the engineered mice, one can test the effectiveness of different guide RNAs and donor DNAs. We describe in detail here a simple, rapid (three days), inexpensive protocol, for amplifying DNA from blastocysts to determine the results of CRISPR point mutation KIs. Using it, we show that (1) the rate of KI seen in blastocysts is similar to that seen in mice for a given guide RNA/donor DNA pair, (2) a donor complementary to the variable portion of a guide integrated in a more all-or-none fashion, (3) donor DNAs can be used simultaneously to integrate two different mutations into the same locus, and (4) by placing silent mutations about every 6 to 10 bp between the Cas9 cut site and the desired mutation(s), the desired mutation(s) can be incorporated into genomic DNA over 30 bp away from the cut at the same high efficiency as close to the cut.
Collapse
Affiliation(s)
- Elena McBeath
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Jan Parker-Thornburg
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (C.S.)
| | - Yuka Fujii
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.F.); (J.-i.A.); (K.F.)
| | - Neeraj Aryal
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (C.S.)
| | - Chad Smith
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.A.); (C.S.)
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Jun-ichi Abe
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.F.); (J.-i.A.); (K.F.)
| | - Keigi Fujiwara
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.F.); (J.-i.A.); (K.F.)
| |
Collapse
|
15
|
Lemonnier T, Dupré A, Jessus C. The G2-to-M transition from a phosphatase perspective: a new vision of the meiotic division. Cell Div 2020; 15:9. [PMID: 32508972 PMCID: PMC7249327 DOI: 10.1186/s13008-020-00065-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022] Open
Abstract
Cell division is orchestrated by the phosphorylation and dephosphorylation of thousands of proteins. These post-translational modifications underlie the molecular cascades converging to the activation of the universal mitotic kinase, Cdk1, and entry into cell division. They also govern the structural events that sustain the mechanics of cell division. While the role of protein kinases in mitosis has been well documented by decades of investigations, little was known regarding the control of protein phosphatases until the recent years. However, the regulation of phosphatase activities is as essential as kinases in controlling the activation of Cdk1 to enter M-phase. The regulation and the function of phosphatases result from post-translational modifications but also from the combinatorial association between conserved catalytic subunits and regulatory subunits that drive their substrate specificity, their cellular localization and their activity. It now appears that sequential dephosphorylations orchestrated by a network of phosphatase activities trigger Cdk1 activation and then order the structural events necessary for the timely execution of cell division. This review discusses a series of recent works describing the important roles played by protein phosphatases for the proper regulation of meiotic division. Many breakthroughs in the field of cell cycle research came from studies on oocyte meiotic divisions. Indeed, the meiotic division shares most of the molecular regulators with mitosis. The natural arrests of oocytes in G2 and in M-phase, the giant size of these cells, the variety of model species allowing either biochemical or imaging as well as genetics approaches explain why the process of meiosis has served as an historical model to decipher signalling pathways involved in the G2-to-M transition. The review especially highlights how the phosphatase PP2A-B55δ critically orchestrates the timing of meiosis resumption in amphibian oocytes. By opposing the kinase PKA, PP2A-B55δ controls the release of the G2 arrest through the dephosphorylation of their substrate, Arpp19. Few hours later, the inhibition of PP2A-B55δ by Arpp19 releases its opposing kinase, Cdk1, and triggers M-phase. In coordination with a variety of phosphatases and kinases, the PP2A-B55δ/Arpp19 duo therefore emerges as the key effector of the G2-to-M transition.
Collapse
Affiliation(s)
- Tom Lemonnier
- Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Sorbonne Université, CNRS, 75005 Paris, France
| | - Aude Dupré
- Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Sorbonne Université, CNRS, 75005 Paris, France
| | - Catherine Jessus
- Laboratoire de Biologie du Développement-Institut de Biologie Paris Seine, LBD-IBPS, Sorbonne Université, CNRS, 75005 Paris, France
| |
Collapse
|
16
|
Germacrone exerts anti-cancer effects on gastric cancer through induction of cell cycle arrest and promotion of apoptosis. BMC Complement Med Ther 2020; 20:21. [PMID: 32020876 PMCID: PMC7076853 DOI: 10.1186/s12906-019-2810-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
Background Germacrone is one of the natural bioactive compounds found in Rhizoma curcuma essential oils. In this study, the potential anti-cancer effect of germacrone in gastric cancer cell line BGC823 was investigated. Methods The cell viability and proliferative activity were assessed, and cell cycle analysis was also performed. Hoechst 33258 and Annexin V/PI double staining was used for detection of cell apoptosis. Protein profiles of cell cycle-related and apoptosis-related proteins were assessed. Results MTT assay revealed that germacrone had marked cytotoxicity on BGC823 cells. Germacrone induced cell cycle arrest in the G2/M phase via remarkably decreased expression levels of cyclin B1, cdc 2 and cdc 25c. In addition, the treatment with germacrone induced caspase-3 activity and PARP cleavage. These findings demonstrated the effects of germacrone on inhibiting cell proliferation through induction of G2/M phase cell cycle arrest and promotion of cell apoptosis. It also indicated that germacrone functioned through modulations of cell cycle-associated protein expression and mitochondria-mediated apoptosis. Conclusion These findings will be valuable as the molecular basis for the germacrone-mediated anti-cancer effect against gastric cancer.
Collapse
|
17
|
Ku JM, Hong SH, Kim HI, Kim MJ, Kim SK, Kim M, Choi SY, Park J, Kim HK, Kim JH, Seo HS, Shin YC, Ko SG. Synergistic anticancer effect of combined use of Trichosanthes kirilowii with cisplatin and pemetrexed enhances apoptosis of H1299 non-small-cell lung cancer cells via modulation of ErbB3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 66:153109. [PMID: 31790894 DOI: 10.1016/j.phymed.2019.153109] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/02/2019] [Accepted: 10/06/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Lung cancer is one of the most common malignancies worldwide. To treat lung cancer, various anticancer drugs were developed and tested, but they failed because of drug resistance. In the present study, we tested herbal medicines, such as TK and CuD, as anticancer drugs to decrease side effects and resistance. METHODS Cell viability was measured by an MTT assay. Analysis of cell cycle arrest was performed by flow cytometry. Induction of apoptosis by cucurbitacin D was measured by an annexin V-FITC/PI assay. We performed RTK kit analysis. Levels of p-ErbB3, p-STAT3, p-NF-κB, and caspases were measured by western blot analysis. Nuclear staining of ErbB3 was measured by immunocytochemistry. Transcriptional activity of STAT3 and NF-κB was detected by STAT3 and NF-κB luciferase reporter gene assays. RESULTS We found a synergistic effect of TK with CDDP and PXD in primary culture of human NSCLC tumor cells. The combination of CDDP/PXD and TK or CuD inhibited the proliferation of H1299 cells. The combination of CDDP/PXD and TK or CuD induced sub-G1 and G2/M cell cycle arrest in H1299 cells. The combination of CDDP/PXD and TK or CuD induced apoptosis, regulated apoptotic molecules, caused morphological changes and inhibited colony formation in H1299 cells. We found that TK suppresses p-ErbB3 expression and signaling. The combination of CDDP/PXD and TK or CuD inhibited p-AKT, p-Erk, and p-JNK signaling and suppressed Stat3 and NF-κB transcriptional activity in H1299 cells. More importantly, the combination of CDDP/PXD and TK or CuD inhibited p-ErbB3 and downstream molecules in H1299 cells. The combination of CDDP/PXD and TK or CuD inhibited ErbB2/ErbB3 dimerization. Our results clearly demonstrate that the synergistic effect of CDDP/PXD and TK or CuD inhibits cell growth and induces apoptosis by inhibiting ErbB3 signaling. CONCLUSION The combination of CDDP/PXD and TK or CuD decreases cell proliferation and induces apoptosis by inhibiting ErbB3 signaling in H1299 lung cancer cells. TK or CuD could be useful as a compound to treat lung cancer. Additionally, targeting ErbB3 may also be useful for treating lung cancer.
Collapse
Affiliation(s)
- Jin Mo Ku
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Se Hyang Hong
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Hyo In Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Min Jeong Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Kyoung Kim
- Department of Applied Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Minkyu Kim
- Department of Applied Korean Medicine, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Seok Young Choi
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jeongkoo Park
- MetaBio Institute, 9, Olympic-ro 78-gil, Gangdong-gu, Seoul 05327, Republic of Korea
| | - Hyun Koo Kim
- Department of Thoracic and Cardiovascular Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Ji Hye Kim
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Hye Sook Seo
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Yong Cheol Shin
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea.
| |
Collapse
|
18
|
Liu R, Qu Z, Lin Y, Lee CS, Tai WCS, Chen S. Brevilin A Induces Cell Cycle Arrest and Apoptosis in Nasopharyngeal Carcinoma. Front Pharmacol 2019; 10:594. [PMID: 31178739 PMCID: PMC6544084 DOI: 10.3389/fphar.2019.00594] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common malignant cancers in Southeast Asia and Southern China. Centipeda minima extract (CME) had previously demonstrated anti-cancer effects in human NPC. Brevilin A, a sesquiterpene lactone isolated from C. minima, has been reported to exhibit biological activities. In this study, we investigated its anti-NPC effect and further explored its molecular mechanisms. The effects of brevilin A were tested in the NPC cell lines CNE-1, CNE-2, SUNE-1, HONE1, and C666-1. Effects of brevilin A on cell viability were determined by MTT assay, and cell cycle and apoptosis were detected by flow cytometry. The molecular mechanism of cell cycle regulation and apoptosis were investigated via Western blot. Results showed that brevilin A inhibited NPC cell viability in a concentration- and time-dependent manner. Brevilin A induced cell cycle arrest at G2/M and induced apoptosis. Western blot results demonstrated that brevilin A could down-regulate cyclin D3, cdc2, p-PI3K, p-AKT, p-mTOR, and p-STAT3, while up-regulating cleaved PARP, cleaved caspase 9, and Bax. Regulation of cyclin B1, cdk6, and Bcl-2 expression by brevilin A showed dynamic changes according to dose and time. In the tumor xenograft model, brevilin A could reduce tumor growth, at a similar magnitude to cisplatin. However, notably, whereas cisplatin treatment led to significant weight loss in treated mice, treatment with brevilin A did not, indicating its relative lack of toxicity. Taken together, brevilin A regulated cell cycle, activated the caspase signaling pathway, and inhibited PI3K/AKT/mTOR and STAT3 signaling pathways in vitro, and exhibited similar efficacy to the common chemotherapeutic cisplatin in vivo, without its associated toxicity. These findings provide a framework for the preclinical development of brevilin A as a chemotherapeutic for NPC.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Zhao Qu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Yushan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Chi-Sing Lee
- Department of Chemistry, The Hong Kong Baptist University, Hong Kong, China
| | - William Chi-Shing Tai
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong, China
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Department of Chemistry, The Hong Kong Baptist University, Hong Kong, China.,Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Yoshitome S, Aiba Y, Yuge M, Furuno N, Watanabe M, Nakajo N. Involvement of Myt1 kinase in the G2 phase of the first cell cycle in Xenopus laevis. Biochem Biophys Res Commun 2019; 515:139-144. [PMID: 31128913 DOI: 10.1016/j.bbrc.2019.05.104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 10/26/2022]
Abstract
During cleavage of Xenopus laevis, the first mitotic cell cycle immediately following fertilization is approximately 90 min and consists of S, G2, and M phases. In contrast, the subsequent eleven cell cycles are approximately 30 min and consist mostly of S and M phases. The balance between Cdc25 and Wee1A/Myt1 is thought to be crucial for Xenopus first cell cycle progression; however, the role of Myt1 in this period has not been fully investigated. In this study, we examined the roles of Myt1, Wee1A, and Cdc25A in the first cell cycle of Xenopus laevis. Inhibition of Cdc25A with antisense morpholino oligonucleotides lengthened the duration of the first cell cycle to some extent, whereas it was slightly shortened by ectopic Cdc25A expression, suggesting that the low concentration of Cdc25A during the first cell cycle does not fully account for the long duration of this cycle. Using the Wee1A antisense morpholino oligonucleotide and neutralizing antibody against Myt1, we found that Myt1 phosphorylates and inhibits Cdk1 much more effectively than Wee1A during the first cell cycle in Xenopus. Taken together, these results suggest that the activity of Myt1 is predominantly responsible for the duration of the long G2 phase in the first mitotic cell cycle in Xenopus.
Collapse
Affiliation(s)
- Satoshi Yoshitome
- Department of Biology, Graduate School of Sciences, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan; Department of Enviromental Science, International College of Arts and Sciences, Fukuoka Women's University, Kasumigaoka 1-1-1, Higashi-Ku, Fukuoka, 813-8529, Japan.
| | - Yukito Aiba
- Department of Biology, Graduate School of Sciences, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Masahiro Yuge
- Department of Enviromental Science, International College of Arts and Sciences, Fukuoka Women's University, Kasumigaoka 1-1-1, Higashi-Ku, Fukuoka, 813-8529, Japan
| | - Nobuaki Furuno
- Amphibian Research Center, Hiroshima University, Kagamiyama 1-3-1, Higashihiroshima, 739-8526, Japan
| | - Minoru Watanabe
- Institute of Liberal Arts and Sciences, Tokushima University, Minamijosanjima-cho 1-1, Tokushima, 770-8502, Japan
| | - Nobushige Nakajo
- Department of Biology, Graduate School of Sciences, Kyushu University, Motooka 744, Nishi-ku, Fukuoka, 819-0395, Japan
| |
Collapse
|
20
|
Liu R, Dow Chan B, Mok DKW, Lee CS, Tai WCS, Chen S. Arnicolide D, from the herb Centipeda minima, Is a Therapeutic Candidate against Nasopharyngeal Carcinoma. Molecules 2019; 24:molecules24101908. [PMID: 31108969 PMCID: PMC6571971 DOI: 10.3390/molecules24101908] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/29/2019] [Accepted: 05/10/2019] [Indexed: 02/08/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a high morbidity and mortality cancer with an obvious racial and geographic bias, particularly endemic to Southeast China. Our previous studies demonstrated that Centipeda minima extract (CME) exhibited anti-cancer effects in human NPC cell lines. Arnicolide C and arnicolide D are sesquiterpene lactones isolated from Centipeda minima. In this study, for the first time, we investigated their anti-NPC effects and further explored the related molecular mechanisms. The effects of both arnicolide C and arnicolide D were tested in NPC cells CNE-1, CNE-2, SUNE-1, HONE1, and C666-1. The results showed that the two compounds inhibited NPC cell viability in a concentration- and time-dependent manner. As the inhibitory effect of arnicolide D was the more pronounced of the two, our following studies focused on this compound. Arnicolide D could induce cell cycle arrest at G2/M, and induce cell apoptosis. The molecular mechanism of cell cycle regulation and apoptosis induction was investigated, and the results showed that arnicolide D could downregulate cyclin D3, cdc2, p-PI3K, p-AKT, p-mTOR, and p-STAT3, and upregulate cleaved PARP, cleaved caspase 9, and Bax. Regulation of cyclin B1, cdk6, and Bcl-2 expression by arnicolide D showed dynamic changes according to dose and time. Taken together, arnicolide D modulated the cell cycle, activated the caspase signaling pathway, and inhibited the PI3K/AKT/mTOR and STAT3 signaling pathways. These findings provide a solid base of evidence for arnicolide D as a lead compound for further development, and act as proof for the viability of drug development from traditional Chinese medicines.
Collapse
Affiliation(s)
- Rui Liu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China.
| | - Brandon Dow Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, China.
| | - Daniel Kam-Wah Mok
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China.
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, China.
| | - Chi-Sing Lee
- Department of Chemistry, The Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China.
| | - William Chi-Shing Tai
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China.
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, China.
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518057, China.
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom 999077, Hong Kong, China.
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
21
|
C/EBPβ Is a Transcriptional Regulator of Wee1 at the G₂/M Phase of the Cell Cycle. Cells 2019; 8:cells8020145. [PMID: 30754676 PMCID: PMC6407104 DOI: 10.3390/cells8020145] [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: 01/18/2019] [Revised: 02/06/2019] [Accepted: 02/09/2019] [Indexed: 12/25/2022] Open
Abstract
The CCAAT/enhancer-binding protein β (C/EBPβ) is a transcription factor that regulates cellular proliferation, differentiation, apoptosis and tumorigenesis. Although the pro-oncogenic roles of C/EBPβ have been implicated in various human cancers, how it contributes to tumorigenesis or tumor progression has not been determined. Immunohistochemistry with human non-small cell lung cancer (NSCLC) tissues revealed that higher levels of C/EBPβ protein were expressed compared to normal lung tissues. Knockdown of C/EBPβ by siRNA reduced the proliferative capacity of NSCLC cells by delaying the G2/M transition in the cell cycle. In C/EBPβ-knockdown cells, a prolonged increase in phosphorylation of cyclin dependent kinase 1 at tyrosine 15 (Y15-pCDK1) was displayed with simultaneously increased Wee1 and decreased Cdc25B expression. Chromatin immunoprecipitation (ChIP) analysis showed that C/EBPβ bound to distal promoter regions of WEE1 and repressed WEE1 transcription through its interaction with histone deacetylase 2. Treatment of C/EBPβ-knockdown cells with a Wee1 inhibitor induced a decrease in Y15-pCDK1 and recovered cells from G2/M arrest. In the xenograft tumors, the depletion of C/EBPβ significantly reduced tumor growth. Taken together, these results indicate that Wee1 is a novel transcription target of C/EBPβ that is required for the G2/M phase of cell cycle progression, ultimately regulating proliferation of NSCLC cells.
Collapse
|
22
|
Basu P, Maier C. Phytoestrogens and breast cancer: In vitro anticancer activities of isoflavones, lignans, coumestans, stilbenes and their analogs and derivatives. Biomed Pharmacother 2018; 107:1648-1666. [DOI: 10.1016/j.biopha.2018.08.100] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/17/2018] [Accepted: 08/17/2018] [Indexed: 01/11/2023] Open
|
23
|
Zhai B, Steinø A, Bacha J, Brown D, Daugaard M. Dianhydrogalactitol induces replication-dependent DNA damage in tumor cells preferentially resolved by homologous recombination. Cell Death Dis 2018; 9:1016. [PMID: 30283085 PMCID: PMC6170372 DOI: 10.1038/s41419-018-1069-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 08/30/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
1,2:5,6-Dianhydrogalactitol (DAG) is a bifunctional DNA-targeting agent causing N7-guanine alkylation and inter-strand DNA crosslinks currently in clinical trial for treatment of glioblastoma. While preclinical studies and clinical trials have demonstrated antitumor activity of DAG in a variety of malignancies, understanding the molecular mechanisms underlying DAG-induced cytotoxicity is essential for proper clinical qualification. Using non-small cell lung cancer (NSCLC) as a model system, we show that DAG-induced cytotoxicity materializes when cells enter S phase with unrepaired N7-guanine DNA crosslinks. In S phase, DAG-mediated DNA crosslink lesions translated into replication-dependent DNA double-strand breaks (DSBs) that subsequently triggered irreversible cell cycle arrest and loss of viability. DAG-treated NSCLC cells attempt to repair the DSBs by homologous recombination (HR) and inhibition of the HR repair pathway sensitized NSCLC cells to DAG-induced DNA damage. Accordingly, our work describes a molecular mechanism behind N7-guanine crosslink-induced cytotoxicity in cancer cells and provides a rationale for using DAG analogs to treat HR-deficient tumors.
Collapse
Affiliation(s)
- Beibei Zhai
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada.,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Anne Steinø
- DelMar Pharmaceuticals, Inc., Vancouver, BC, V5Z 1K5, Canada.,DelMar Pharmaceuticals, Inc., Menlo Park, CA, 94025, USA
| | - Jeffrey Bacha
- DelMar Pharmaceuticals, Inc., Vancouver, BC, V5Z 1K5, Canada.,DelMar Pharmaceuticals, Inc., Menlo Park, CA, 94025, USA
| | - Dennis Brown
- DelMar Pharmaceuticals, Inc., Vancouver, BC, V5Z 1K5, Canada.,DelMar Pharmaceuticals, Inc., Menlo Park, CA, 94025, USA
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, BC, V6H 3Z6, Canada. .,Department of Urologic Sciences, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
24
|
Feng S, Zhu J, Xia K, Yu W, Wang Y, Wang J, Li F, Yang Z, Yang X, Liu B, Tao H, Liang C. Cantharidin Inhibits Anti-Apoptotic Bcl-2 Family Proteins and Induces Apoptosis in Human Osteosarcoma Cell Lines MG-63 and MNNG/HOS via Mitochondria-Dependent Pathway. Med Sci Monit 2018; 24:6742-6749. [PMID: 30248086 PMCID: PMC6180936 DOI: 10.12659/msm.910294] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Cantharidin (CTD) is one of the major active ingredients of blister beetles and has significant antitumor activity in many cancer cell lines. The aim of our study was to evaluate the effect of CTD on the apoptosis of human osteosarcoma cells MG-63 and MNNG/HOS, and to explore the possible molecular mechanism. Material/Methods Osteosarcoma cells MG-63 and MNNG/HOS were treated with varying concentrations of CTD. The proliferation inhibition of cells was detected by MTS. Flow cytometry and Hoechst 33258 staining were used to determine cell cycle arrest and apoptosis, and apoptosis-related protein levels were analyzed by Western blotting. Results Our current findings suggest that CTD could inhibit the proliferation of these 2 osteosarcoma cells. The cells treated with CTD showed an obvious apoptotic morphology, and CTD promoted cells apoptosis in a dose-dependent manner. In addition, cantharidin-induced apoptosis was accompanied by increased expression of Bax and PARP and decreased expression of Bcl-2, p-Akt, and p-Cdc2. Conclusions CTD accelerates the apoptosis of MG-63 and MNNG/HOS cells in a concentration-dependent manner through the mitochondria-dependent pathway, suggesting that use of CTD is a novel approach for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Shoumin Feng
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Jian Zhu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Wei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Yitian Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Junjie Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Zhengming Yang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Xiaobo Yang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Bing Liu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Huimin Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland).,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
25
|
Shen Y, Sherman JW, Chen X, Wang R. Phosphorylation of CDC25C by AMP-activated protein kinase mediates a metabolic checkpoint during cell-cycle G 2/M-phase transition. J Biol Chem 2018; 293:5185-5199. [PMID: 29467227 PMCID: PMC5892595 DOI: 10.1074/jbc.ra117.001379] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/01/2018] [Indexed: 12/30/2022] Open
Abstract
From unicellular to multicellular organisms, cell-cycle progression is tightly coupled to biosynthetic and bioenergetic demands. Accumulating evidence has demonstrated the G1/S-phase transition as a key checkpoint where cells respond to their metabolic status and commit to replicating the genome. However, the mechanism underlying the coordination of metabolism and the G2/M-phase transition in mammalian cells remains unclear. Here, we show that the activation of AMP-activated protein kinase (AMPK), a highly conserved cellular energy sensor, significantly delays mitosis entry. The cell-cycle G2/M-phase transition is controlled by mitotic cyclin-dependent kinase complex (CDC2-cyclin B), which is inactivated by WEE1 family protein kinases and activated by the opposing phosphatase CDC25C. AMPK directly phosphorylates CDC25C on serine 216, a well-conserved inhibitory phosphorylation event, which has been shown to mediate DNA damage–induced G2-phase arrest. The acute induction of CDC25C or suppression of WEE1 partially restores mitosis entry in the context of AMPK activation. These findings suggest that AMPK-dependent phosphorylation of CDC25C orchestrates a metabolic checkpoint for the cell-cycle G2/M-phase transition.
Collapse
Affiliation(s)
- Yuqing Shen
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and.,the Department of Microbiology and Immunology, Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Medical School, Southeast University, Nanjing 210009, China
| | - John William Sherman
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and
| | - Xuyong Chen
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and
| | - Ruoning Wang
- From the Center for Childhood Cancer and Blood Diseases, Hematology/Oncology and BMT, Research Institute at Nationwide Children's Hospital, Ohio State University, Columbus, Ohio 43205 and
| |
Collapse
|
26
|
Anti-Tumor and Radiosensitization Effects of N-Butylidenephthalide on Human Breast Cancer Cells. Molecules 2018; 23:molecules23020240. [PMID: 29370116 PMCID: PMC6017952 DOI: 10.3390/molecules23020240] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 11/17/2022] Open
Abstract
N-Butylidenephthalide (BP), which is extracted from a traditional Chinese medicine, Radix Angelica Sinensis (danggui), displays antitumor activity against various cancer cell lines. The purpose of this study was to investigate the cytotoxic and radiosensitizing effect of BP and the underlying mechanism of action in human breast cancer cells. BP induces apoptosis in breast cancer cells, which was revealed by the TUNEL assay; the activation of caspase-9 and PARP was detected by western blot. In addition, BP-induced G2/M arrest was examined by flow cytometry and the expression levels of the G2/M regulatory protein were detected by western blot. BP also suppresses the migration and invasion of breast cancer cells, which was tested by wound healing and the matrigel invasion assay; the involvement of EMT-related gene expressions was detected by real-time PCR. Furthermore, BP enhanced the radiosensitivity of breast cancer cells, which was measured by the colony formation assay and comet assay, where the foci of γ-H2AX after radiation significantly increased in BP pretreated cells and was evidenced by immunocytochemistry staining and western blot. The homologous recombination (HR) repair protein Rad51 was down-regulated after BP pretreatment. These results indicate that BP might be a potential chemotherapeutic and radiosensitizing agent for breast cancer therapy.
Collapse
|
27
|
Deng Y, Hu Z, Shang L, Peng Q, Tang YZ. Transcriptomic Analyses of Scrippsiella trochoidea Reveals Processes Regulating Encystment and Dormancy in the Life Cycle of a Dinoflagellate, with a Particular Attention to the Role of Abscisic Acid. Front Microbiol 2017; 8:2450. [PMID: 29312167 PMCID: PMC5732363 DOI: 10.3389/fmicb.2017.02450] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022] Open
Abstract
Due to the vital importance of resting cysts in the biology and ecology of many dinoflagellates, a transcriptomic investigation on Scrippsiella trochoidea was conducted with the aim to reveal the molecular processes and relevant functional genes regulating encystment and dormancy in dinoflagellates. We identified via RNA-seq 3,874 (out of 166,575) differentially expressed genes (DEGs) between resting cysts and vegetative cells; a pause of photosynthesis (confirmed via direct measurement of photosynthetic efficiency); an active catabolism including β-oxidation, glycolysis, glyoxylate pathway, and TCA in resting cysts (tested via measurements of respiration rate); 12 DEGs encoding meiotic recombination proteins and members of MEI2-like family potentially involved in sexual reproduction and encystment; elevated expressions in genes encoding enzymes responding to pathogens (chitin deacetylase) and ROS stress in cysts; and 134 unigenes specifically expressed in cysts. We paid particular attention to genes pertaining to phytohormone signaling and identified 4 key genes regulating abscisic acid (ABA) biosynthesis and catabolism, with further characterization based on their full-length cDNA obtained via RACE-PCR. The qPCR results demonstrated elevated biosynthesis and repressed catabolism of ABA during the courses of encystment and cyst dormancy, which was significantly enhanced by lower temperature (4 ± 1°C) and darkness. Direct measurements of ABA using UHPLC-MS/MS and ELISA in vegetative cells and cysts both fully supported qPCR results. These results collectively suggest a vital role of ABA in regulating encystment and maintenance of dormancy, akin to its function in seed dormancy of higher plants. Our results provided a critical advancement in understanding molecular processes in resting cysts of dinoflagellates.
Collapse
Affiliation(s)
- Yunyan Deng
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhangxi Hu
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Lixia Shang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Quancai Peng
- Research Center of Analysis and Measurement, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Ying Zhong Tang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory of Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
28
|
Zhao Y, Wen H, Teotia S, Du Y, Zhang J, Li J, Sun H, Tang G, Peng T, Zhao Q. Suppression of microRNA159 impacts multiple agronomic traits in rice (Oryza sativa L.). BMC PLANT BIOLOGY 2017; 17:215. [PMID: 29162059 PMCID: PMC5699021 DOI: 10.1186/s12870-017-1171-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 11/10/2017] [Indexed: 05/18/2023]
Abstract
BACKGROUND microRNAs (miRNAs) are important regulators in plant growth and development. miR159 is a conserved miRNA among different plant species and has various functions in plants. Studies on miR159 are mostly done on model plant, Arabidopsis thaliana. In rice, studies on miR159 were either based upon genome-wide expression analyses focused upon responses to different nitrogen forms and abiotic stress or upon phenotypic studies of transgenic plants overexpressing its precursor. STTM (Short Tandem Target Mimic) is an effective tool to block the activity of endogenous mature miRNA activity in plant. Therefore, specific roles of miR159 in rice could be explored by down regulating miR159 through STTM. RESULTS In this study, expression of mature miR159 was successfully suppressed by STTM which resulted in the increased expressions of its two targets genes, OsGAMYB and OsGAMYBL1 (GAMYB-LIKE 1). Overall, STTM159 plants exhibited short stature along with smaller organ size and reduction in stem diameter, length of flag leaf, main panicle, spikelet hulls and grain size. Histological analysis of stem, leaf and mature spikelet hull showed the reduced number of small vascular bundles (SVB), less number of small veins (SV) between two big veins (LV) and less cell number in outer parenchyma. Gene Ontology (GO) enrichment analysis of differentially expressed genes between wild type plants and STTM159 transgenic plants showed that genes involved in cell division, auxin, cytokinin (CK) and brassinosteroids (BRs) biosynthesis and signaling are significantly down-regulated in STTM159 plants. CONCLUSION Our data suggests that in rice, miR159 positively regulates organ size, including stem, leaf, and grain size due to the promotion of cell division. Further analysis from the RNA-seq data showed that the decreased cell divisions in STTM159 transgenic plants may result, at least partly from the lower expression of the genes involved in cell cycle and hormone homeostasis, which provides new insights of rice miR159-specific functions.
Collapse
Affiliation(s)
- Yafan Zhao
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Huili Wen
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Sachin Teotia
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931 USA
| | - Yanxiu Du
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Jing Zhang
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Junzhou Li
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Hongzheng Sun
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Guiliang Tang
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931 USA
| | - Ting Peng
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| | - Quanzhi Zhao
- Collaborative Innovation Center of Henan Grain Crops, Henan Agricultural University, Zhengzhou, 450002 China
- Key Laboratory of Rice Biology in Henan Province, Henan Agricultural University, Zhengzhou, 450002 China
| |
Collapse
|
29
|
Sun S, Xie F, Xu X, Cai Q, Zhang Q, Cui Z, Zheng Y, Zhou J. Advanced oxidation protein products induce S-phase arrest of hepatocytes via the ROS-dependent, β-catenin-CDK2-mediated pathway. Redox Biol 2017; 14:338-353. [PMID: 29032312 PMCID: PMC5975226 DOI: 10.1016/j.redox.2017.09.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 09/18/2017] [Indexed: 01/16/2023] Open
Abstract
Liver regeneration has important clinical importance in the setting of partial hepatectomy (PH). Following PH, quiescent hepatocytes can reenter cell cycle to restore liver mass. Hepatocyte cell cycle progression, as the basic motivations of liver regeneration, can be disrupted by multiple pathological factors such as oxidative stress. This study aimed to evaluate the role of advanced oxidation protein products (AOPP) in S-phase arrest in hepatocytes. Serum AOPP level were measured during the perioperative period of PH in 33 patients with hepatocellular carcinoma (HCC). Normal Sprague Dawley rats, human and murine liver cell line (HL-7702 and AML-12) were challenged with AOPP prepared by incubation of rat serum albumin (RSA) with hypochlorous acid, and the effect of AOPP on hepatocytes cell cycle progression and liver regeneration was studied after PH. AOPP levels were increased following partial hepatectomy (PH) in patients with primary liver cancer. AOPP treatment impaired liver regeneration in rats following 70% partial hepatectomy. S-phase arrest was induced by AOPP administration in hepatocytes derived from the remnant liver at controlled times following partial hepatectomy in rats, and in HL-7702 and AML-12 cells. The effect of AOPP on hepatocyte S phase arrest was mainly mediated by a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent reactive oxygen species (ROS) generation, downregulation of downstream β-catenin signaling and decreased cyclin-dependent kinase 2 (CDK2) expression, which inhibited S-phase progression in hepatocytes. This study provides preliminary evidence that AOPP can induce S-phase arrest in hepatocytes via the ROS-dependent, β-catenin-CDK2-mediated pathway. These findings suggest a novel pathogenic role of AOPP contributing to the impaired liver regeneration and may provide the basis for developing new strategies to improve liver regeneration in patients undergoing PH.
Collapse
Affiliation(s)
- Shibo Sun
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Fang Xie
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Xiaoping Xu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qing Cai
- Department of Hepatobiliary Surgery and Liver Transplantation Center, Guangzhou General Hospital of Guangzhou Military Area, Guangzhou, Guangdong 510515, China
| | - Qifan Zhang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhonglin Cui
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yujian Zheng
- Department of Hepatobiliary Surgery and Liver Transplantation Center, Guangzhou General Hospital of Guangzhou Military Area, Guangzhou, Guangdong 510515, China
| | - Jie Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
30
|
Guo H, Xu W, Chen J, Yan L, Ding J, Hou Y, Chen X. Positively charged polypeptide nanogel enhances mucoadhesion and penetrability of 10-hydroxycamptothecin in orthotopic bladder carcinoma. J Control Release 2017; 259:136-148. [DOI: 10.1016/j.jconrel.2016.12.041] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 12/21/2016] [Accepted: 12/30/2016] [Indexed: 02/06/2023]
|
31
|
Beaujois R, Ottoni E, Zhang X, Gagnon C, Hassine S, Mollet S, Viranaicken W, DesGroseillers L. The M-phase specific hyperphosphorylation of Staufen2 involved the cyclin-dependent kinase CDK1. BMC Cell Biol 2017; 18:25. [PMID: 28705199 PMCID: PMC5513041 DOI: 10.1186/s12860-017-0142-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/10/2017] [Indexed: 01/21/2023] Open
Abstract
Background Staufen2 (STAU2) is an RNA-binding protein involved in the post-transcriptional regulation of gene expression. This protein was shown to be required for organ formation and cell differentiation. Although STAU2 functions have been reported in neuronal cells, its role in dividing cells remains deeply uncharacterized. Especially, its regulation during the cell cycle is completely unknown. Results In this study, we showed that STAU2 isoforms display a mitosis-specific slow migration pattern on SDS-gels in all tested transformed and untransformed cell lines. Deeper analyses in hTert-RPE1 and HeLa cells further indicated that the slow migration pattern of STAU2 isoforms is due to phosphorylation. Time course studies showed that STAU2 phosphorylation occurs before prometaphase and terminates as cells exit mitosis. Interestingly, STAU2 isoforms were phosphorylated on several amino acid residues in the C-terminal half via the cyclin-dependent kinase 1 (Cdk1), an enzyme known to play crucial roles during mitosis. Introduction of phospho-mimetic or phospho-null mutations in STAU2 did not impair its RNA-binding capacity, its stability, its interaction with protein co-factors or its sub-cellular localization, suggesting that STAU2 phosphorylation in mitosis does not regulate these functions. Similarly, STAU2 phosphorylation is not likely to be crucial for cell cycle progression since expression of phosphorylation mutants in hTert-RPE1 cells did not impair cell proliferation. Conclusions Altogether, these results indicate that STAU2 isoforms are phosphorylated during mitosis and that the phosphorylation process involves Cdk1. The meaning of this post-translational modification is still elusive. Electronic supplementary material The online version of this article (doi:10.1186/s12860-017-0142-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rémy Beaujois
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Elizabeth Ottoni
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Xin Zhang
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Christina Gagnon
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Sami Hassine
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Stéphanie Mollet
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Wildriss Viranaicken
- Present address: UMR PIMIT, Processus Infectieux en Milieu Insulaire Tropical, Université de la Réunion, 97490 Sainte Clotilde, La Réunion, France
| | - Luc DesGroseillers
- Département de biochimie et médecine moléculaire, Faculté de médecine, Université de Montréal, 2900 Edouard Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
32
|
Omar UM, Al Doghaither HA, Rahimulddin SA, Al Zahrani SM, Al-Ghafari AB. In Vitro Cytotoxic and Anticancer Effects of Zamzam Water in Human Lung Cancer (A594) Cell Line. Malays J Med Sci 2017; 24:15-25. [PMID: 28814929 DOI: 10.21315/mjms2017.24.3.3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 04/05/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Zamzam water is naturally alkaline and rich in a variety of minerals which may represent a powerful tool for cancer therapy. In this research, the cytotoxic effects of Zamzam water were investigated in human lung cancer (A549) cell line and compared with human skin fibroblasts (HSF). METHODS Two different preparations of Zamzam water were used: Z1, with pH adjusted to 7.2 and Z2, with no pH adjustment. The effects of both treatments on the morphology of the A549 and HSF cell lines were investigated. The cell viability of HSF and A549 cells was identified by the MTT assay and trypan blue exclusion. Detection of apoptotic cells and cell cycle analyses were determined using flow cytometry. Moreover, reactive oxygen species (ROS) were measured for both cell lines. RESULTS Both Zamzam water treatments, Z1 and Z2 showed reductions in the cell viability of A549 cells. Cell death occurred via necrosis among cells treated with Z2. Cell cycle arrest occurred in the G0/G1 phases for cells treated with Z2. Cellular and mitochondrial ROS productions were not affected by either treatment. CONCLUSION Our findings indicate that Zamzam water might have potential therapeutic efficacy for lung cancer.
Collapse
Affiliation(s)
- Ulfat M Omar
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sawsan A Rahimulddin
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shiekhah M Al Zahrani
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
33
|
Antineoplastic Effects of Honokiol on Melanoma. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5496398. [PMID: 28194418 PMCID: PMC5282456 DOI: 10.1155/2017/5496398] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/01/2016] [Indexed: 12/13/2022]
Abstract
Honokiol, a plant lignan has been shown to have antineoplastic effects against nonmelanoma skin cancer developments in mice. In this study, antineoplastic effects of honokiol were investigated in malignant melanoma models. In vitro effects of honokiol treatment on SKMEL-2 and UACC-62 melanoma cells were evaluated by measuring the cell viability, proliferation, apoptosis, cell cycle analysis, and expressions of various proteins associated with cell cycle progression and apoptosis. For the in vivo study, male nude mice inoculated with SKMEL-2 or UACC-62 cells received injections of sesame oil or honokiol for two to seven weeks. In vitro honokiol treatment caused significant decrease in cell viability, proliferation, cell cycle arrest, increased apoptosis, and modulation of apoptotic and cell cycle regulatory proteins. Honokiol caused an accumulation of cells in the G2/M phase of the cell cycle in SKMEL-2 and G0/G1 phase in UACC-62 cells. An elevated level of caspases and PARP were observed in both cell lines treated with honokiol. A decrease in the expression of various cell cycle regulatory proteins was also observed in honokiol treated cells. Honokiol caused a significant reduction of tumor growth in SKMEL-2 and UACC-62 melanoma xenografts. These findings suggest that honokiol is a good candidate for further studies as a possible treatment for malignant melanoma.
Collapse
|
34
|
Wang Y, Shi F, Xing GH, Xie P, Zhao N, Yin YF, Sun SY, He J, Wang Y, Xuan SY. Protein Regulator of Cytokinesis PRC1 Confers Chemoresistance and Predicts an Unfavorable Postoperative Survival of Hepatocellular Carcinoma Patients. J Cancer 2017; 8:801-808. [PMID: 28382142 PMCID: PMC5381168 DOI: 10.7150/jca.17640] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023] Open
Abstract
Background: PRC1, a microtubules(MTs)-associated protein, is essential in the mitosis and cell cycle regulation. It has been recently linked to chemoresistance and tumorigenesis. The current study sought to explore the role of PRC1 on chemoresistance and postoperative prognosis of hepatocellular carcinoma(HCC). Methods: PRC1 was transfected into HCC cells to detect its effects of chemoresistance to 5-fluorouracil in vitro and in vivo. This study also investigated the impact of PRC1 on 5-FU-induced G2/M phase arrest and the potential molecular mechanism. Surgical specimens from HCC patients were examined immunohistochemically for PRC1 expression. Results: Ectopic expression of PRC1 significantly increased the chemoresistance, promoted the tumor growth and abrogated 5-FU-induced G2/M phase arrest via p21/p27-pRBs pathway. In clinical specimens, high expression of PRC1(immunostaining score≥3) in HCC cells predicted an unfavorable postoperative survival of HCC patients(P=0.019), especially for whom received postoperative chemotherapy(P=0.002). In multivariate Cox analyses, high PRC1 expression significantly predicted an unfavorable postoperative prognosis, not dependent of TNM stage. Conclusion: High PRC1 expression in HCC cells increased chemoresistance, attenuated 5-FU-induced apoptosis, abrogated 5-FU-induced G2/M phase arrest, and predicts an unfavorable survival, especially for the patients who received chemotherapy. PRC1 might be a novel prognostic and predictive marker and therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Yu Wang
- Department of Gastroenterology, Qingdao Municipal Hospital, Medical College, Qingdao University, Qingdao, Shandong, China
- Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China
- ✉ Corresponding authors: Shi-Ying Xuan, Department of Gastroenterology, Qingdao Municipal Hospital, Medical College, Qingdao University, Qingdao, Shandong, China. E-mail: ; Yu Wang, Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China. E-mail: ; Ying Wang, Department of Cardiovascular Surgery of the First Affiliated Hospital& Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China. ; and Jing He, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China. E-mail:
| | - Feng Shi
- Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Guo-Hui Xing
- Department of Digestive Medicine, Rizhao Traditional Chinese Medicine Hospital, Wanghai Road, Rizhao, Shandong, China
| | - Ping Xie
- Department of Gynaecology and Obstetrics, Rizhao Maternal and Child Health Hospital, Rizhao, Shandong, China
| | - Na Zhao
- Endoscope Center, Rizhao Traditional Chinese Medicine Hospital, Wanghai Road, Rizhao, Shandong, China
| | - Yu-Feng Yin
- Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Shu-Yan Sun
- Department of Pathology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
- ✉ Corresponding authors: Shi-Ying Xuan, Department of Gastroenterology, Qingdao Municipal Hospital, Medical College, Qingdao University, Qingdao, Shandong, China. E-mail: ; Yu Wang, Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China. E-mail: ; Ying Wang, Department of Cardiovascular Surgery of the First Affiliated Hospital& Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China. ; and Jing He, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China. E-mail:
| | - Ying Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital& Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China
- ✉ Corresponding authors: Shi-Ying Xuan, Department of Gastroenterology, Qingdao Municipal Hospital, Medical College, Qingdao University, Qingdao, Shandong, China. E-mail: ; Yu Wang, Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China. E-mail: ; Ying Wang, Department of Cardiovascular Surgery of the First Affiliated Hospital& Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China. ; and Jing He, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China. E-mail:
| | - Shi-Ying Xuan
- Department of Gastroenterology, Qingdao Municipal Hospital, Medical College, Qingdao University, Qingdao, Shandong, China
- ✉ Corresponding authors: Shi-Ying Xuan, Department of Gastroenterology, Qingdao Municipal Hospital, Medical College, Qingdao University, Qingdao, Shandong, China. E-mail: ; Yu Wang, Department of Gastroenterology, Rizhao People's Hospital, Rizhao, Shandong, China. E-mail: ; Ying Wang, Department of Cardiovascular Surgery of the First Affiliated Hospital& Institute for Cardiovascular Science, Soochow University, Suzhou, Jiangsu, China. ; and Jing He, Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China. E-mail:
| |
Collapse
|
35
|
An B, Zhang S, Yan J, Huang L, Li X. Synthesis, in vitro and in vivo evaluation of new hybrids of millepachine and phenstatin as potent tubulin polymerization inhibitors. Org Biomol Chem 2017; 15:852-862. [DOI: 10.1039/c6ob02507b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this paper, a series of millepachine derivatives were synthesized and evaluated as tubulin polymerization inhibitors.
Collapse
Affiliation(s)
- Baijiao An
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Shun Zhang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Jun Yan
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Ling Huang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Xingshu Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| |
Collapse
|
36
|
Meghavarnam AK, Salah M, Sreepriya M, Janakiraman S. Growth inhibitory and proapoptotic effects ofl-asparaginase fromFusarium culmorumASP-87 on human leukemia cells (Jurkat). Fundam Clin Pharmacol 2016; 31:292-300. [DOI: 10.1111/fcp.12257] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/16/2016] [Accepted: 11/22/2016] [Indexed: 11/26/2022]
Affiliation(s)
- Anil K. Meghavarnam
- Department of Microbiology and Biotechnology; Bangalore University; Jnana Bharathi Campus Bangaluru 560056 Karnataka India
| | - Maryam Salah
- Centre of Advanced Study in Marine Biology; Faculty of Marine Sciences; Annamalai University; Parangipettai 608502 Tamil Nadu India
| | - Meenakshisundaram Sreepriya
- Department of Microbiology and Biotechnology; Bangalore University; Jnana Bharathi Campus Bangaluru 560056 Karnataka India
| | - Savitha Janakiraman
- Department of Microbiology and Biotechnology; Bangalore University; Jnana Bharathi Campus Bangaluru 560056 Karnataka India
| |
Collapse
|
37
|
Ahmadian N, Pashaei-Asl R, Samadi N, Rahmati-Yamchi M, Rashidi MR, Ahmadian M, Esmaeili M, Salamat F, Besharat S, Joshaghani HR. Hesa-A Effects on Cell Cycle Signaling in Esophageal Carcinoma Cell Line. Middle East J Dig Dis 2016; 8:297-302. [PMID: 27957293 PMCID: PMC5145297 DOI: 10.15171/mejdd.2016.39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Hesa-A is a natural compound with anticancer properties. The exact mechanism of its action in esophageal cancer is not clear, yet. The aim of this study was to evaluate the cell toxicity effect of Hesa-A on the esophageal carcinoma cell lines, KYSE-30, and cell cycle genes expression. METHODS In this study, we tested cell toxicity with MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) assay and flow cytometry to evaluatet he cell cycle arrest. Real time polymerase chain reaction was used to assess the expression of P53, P16, P21, cyclin D1, and cyclin B1 genes. RESULTS Our results showed that Hesa-A is effective in the expression of cell cycling check point proteins. Hesa-A induced an arrest in G2 phase of esophageal cell cycle. The levels of P53 (>13 times), P21 (>21 times), P16, cyclin B1, and cyclin D1 genes were increased 48 hours after Hesa-A treatment. CONCLUSION P21 and P16 expression were the potential mechanisms for G2 arrest of KYSE-30 esophageal cancer cell line by Hesa-A.
Collapse
Affiliation(s)
- Nasser Ahmadian
- Faculty of Advanced Medical Science Technology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Roghiyeh Pashaei-Asl
- Faculty of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Faculty of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad-Reza Rashidi
- Research Center for Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masomeh Ahmadian
- Faculty of Advanced Medical Science Technology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Moosa Esmaeili
- Women's Reproductive Health Research Center, Alzahra Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Salamat
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sima Besharat
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hamid Reza Joshaghani
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran
| |
Collapse
|
38
|
Willmer T, Peres J, Mowla S, Abrahams A, Prince S. The T-Box factor TBX3 is important in S-phase and is regulated by c-Myc and cyclin A-CDK2. Cell Cycle 2016; 14:3173-83. [PMID: 26266831 DOI: 10.1080/15384101.2015.1080398] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The transcription factor, TBX3, is critical for the formation of, among other structures, the heart, limbs and mammary glands and haploinsufficiency of the human TBX3 gene result in ulnar-mammary syndrome which is characterized by hypoplasia of these structures. On the other hand, the overexpression of TBX3 is a feature of a wide range of cancers and it has been implicated in several aspects of the oncogenic process. This includes its ability to function as an immortalizing gene and to promote proliferation through actively repressing negative cell cycle regulators. Together this suggests that TBX3 levels may need to be tightly regulated during the cell cycle. Here we demonstrate that this is indeed the case and that TBX3 mRNA and protein levels peak at S-phase and that the TBX3 protein is predominantly localized to the nucleus of S-phase cells. The increased levels of TBX3 in S-phase are shown to occur transcriptionally through activation by c-Myc at E-box motifs located at -1210 and -701 bps and post-translationally by cyclin A-CDK2 phosphorylation. Importantly, when TBX3 is depleted by shRNA the cells accumulate in S-phase. These results suggest that TBX3 is required for cells to transit through S-phase and that this function may be linked to its role as a pro-proliferative factor.
Collapse
Affiliation(s)
- Tarryn Willmer
- a Department of Human Biology ; Faculty of Health Sciences; University of Cape Town ; Cape Town , South Africa
| | - Jade Peres
- a Department of Human Biology ; Faculty of Health Sciences; University of Cape Town ; Cape Town , South Africa
| | - Shaheen Mowla
- a Department of Human Biology ; Faculty of Health Sciences; University of Cape Town ; Cape Town , South Africa
| | - Amaal Abrahams
- a Department of Human Biology ; Faculty of Health Sciences; University of Cape Town ; Cape Town , South Africa
| | - Sharon Prince
- a Department of Human Biology ; Faculty of Health Sciences; University of Cape Town ; Cape Town , South Africa
| |
Collapse
|
39
|
Gui H, Shen Z. Concentrate diet modulation of ruminal genes involved in cell proliferation and apoptosis is related to combined effects of short-chain fatty acid and pH in rumen of goats. J Dairy Sci 2016; 99:6627-6638. [PMID: 27236768 DOI: 10.3168/jds.2015-10446] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/28/2016] [Indexed: 01/20/2023]
Abstract
Short-chain fatty acids (SCFA) regulate cell proliferation and cell apoptosis in gastrointestinal tissue in vitro and in vivo. We have tested the hypothesis that a medium-concentrate intake induces mRNA abundance alterations of genes involved in cell proliferation and cell apoptosis in the rumen epithelium of goats, and that these changes in mRNA abundance are related to ruminal SCFA concentration and ruminal pH. Goats (n=16) were randomly allocated to 2 groups and fed either a low-concentrate (LC) diet (10% concentrate; n=8) or a medium-concentrate (MC) diet (35% concentrate; n=8) in 2 equal portions daily. The individually housed goats were fed separately with their respective diet for 3wk and were slaughtered 6h after the morning feed on d 22. In vivo, goats receiving the MC treatment exhibited a greater ruminal SCFA concentration (73.7mM) compared with those receiving the LC treatment (53.2mM), and the pH decreased from 6.9 to 6.5. The expression of proliferative genes of cyclin A, cyclin B1, cyclin D1, cyclin E1, CDK1, CDK2, CDK4, and CDK6 mRNA in the MC group was enhanced. The gene expression of apoptosis genes (caspase 3, caspase 8, caspase 9, p53, and Bax) was significantly higher, and the ratio of Bcl-2 to Bax (Bcl-2/Bax) expression was lower in the MC group than in the LC group. The same trend was observed in the population of apoptotic cells analyzed by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay. The cell density in the stratum germinativum of the MC group was significantly increased compared with that in the LC group. During primary culture of rumen epithelial cells, SCFA or pH treatment alone of the culture medium had significant effects on the expression of most of the genes tested in the present study. Furthermore, SCFA and pH exerted combined effects on the expression of cyclin A, cyclin B1, cyclin E1, CDK6, p53, Bcl-2, and Bcl-2/Bax. Thus, the MC diet induces alteration of gene expression of the genes that regulate both cell proliferation and apoptosis. These genes are regulated by combined effect of ruminal SCFA and ruminal pH.
Collapse
Affiliation(s)
- Hongbing Gui
- Laboratory of Animal Physiology and Biochemistry, Nanjing Agriculture University, Nanjing, China, 210095
| | - Zanming Shen
- Laboratory of Animal Physiology and Biochemistry, Nanjing Agriculture University, Nanjing, China, 210095.
| |
Collapse
|
40
|
Katagi A, Sui L, Kamitori K, Suzuki T, Katayama T, Hossain A, Noguchi C, Dong Y, Yamaguchi F, Tokuda M. Inhibitory effect of isoamericanol A from Jatropha curcas seeds on the growth of MCF-7 human breast cancer cell line by G2/M cell cycle arrest. Heliyon 2016; 2:e00055. [PMID: 27441238 PMCID: PMC4945895 DOI: 10.1016/j.heliyon.2015.e00055] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/13/2015] [Accepted: 12/10/2015] [Indexed: 01/03/2023] Open
Abstract
Although various parts of J. curcas (Jatropha curcas L., Euphorbiaceae) have long been used as traditional folk medicines for their antiviral, analgesic, and/or antidotal efficacies, we are the first to investigate the role of anti-carcinogenicity of isoamericanol A (IAA) from the seed extract. Our results showed that IAA is capable of inhibiting cell proliferation in a dose-dependent manner on the human cancer cell lines of MCF-7, MDA-MB231, HuH-7, and HeLa. Flow cytometry analysis showed IAA significantly induces cell cycle arrest at G2/M on MCF-7 cells. At both protein and mRNA levels examined by western blot and real-time PCR, the results revealed increased expression of BTG2 (B-cell translocation gene 2), p21 (p21WAF1/CIPI), and GADD45A (growth arrest and DNA-damage-inducible, alpha) after IAA treatment, but inversed expression in CDK1 (cyclin-dependent kinase 1) and cyclins B1 and B2. All these effects contribute to G2/M cell cycle arrest. Furthermore, these results coincide with the changes in molecular expressions determined by DNA-microarray analysis. Our findings indicate that IAA has an inhibitory effect on cell proliferation of MCF-7 through cell cycle arrest, giving it great potential as a future therapeutic reagent for cancers.
Collapse
Affiliation(s)
- Ayako Katagi
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Li Sui
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Kazuyo Kamitori
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Toshisada Suzuki
- Laboratory of Biomass Chemistry, Biological Molecular Chemistry, Kagawa University, Faculty of Agriculture / Graduate School of Agriculture, 2393 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Takeshi Katayama
- Laboratory of Biomass Chemistry, Biological Molecular Chemistry, Kagawa University, Faculty of Agriculture / Graduate School of Agriculture, 2393 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Akram Hossain
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Chisato Noguchi
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Youyi Dong
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Fuminori Yamaguchi
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| | - Masaaki Tokuda
- Department of Cell Physiology, Kagawa University, Faculty of Medicine / Graduate School of Medicine, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793 Japan
| |
Collapse
|
41
|
Hong F, Zhao X, Si W, Ze Y, Wang L, Zhou Y, Hong J, Yu X, Sheng L, Liu D, Xu B, Zhang J. Decreased spermatogenesis led to alterations of testis-specific gene expression in male mice following nano-TiO2 exposure. JOURNAL OF HAZARDOUS MATERIALS 2015; 300:718-728. [PMID: 26296075 DOI: 10.1016/j.jhazmat.2015.08.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 05/28/2023]
Abstract
Although TiO2 nanoparticles (NPs) exposure has been demonstrated to cross blood-testis barrier and accumulate in the testis resulting in the reduction of sperm numbers, limited data with respect to the molecular mechanism of decreased spermatogenesis caused by TiO2 NP exposure. In this research, testicular damage, sperm number and alterations in testis-specific gene expressions in male mice induced by intragastric administration with TiO2 NPs for six months were investigated. It was found out that TiO2 NPs could migrate to cells, deposit in the testis and epididymis and thus cause damages to relevant organs, which are, to be more specific, the reductions of total sperm concentrations and sperm motility and an enhancement in the number of abnormal sperms in the cauda epididymis. Furthermore, the individual expression regarding to the mRNAs and proteins of testis-specific genes, including Cdc2, Cyclin B1, Dmcl, TERT, Tesmin, TESP-1, XPD and XRCCI, were significantly declined, whereas Gsk3-β and PGAM4 expressions were greatly elevated in mouse testis due to the exposures, which in fact implied that the reduced spermatogenesis may be involved in the alternated testis-specific gene expressions in those exposed male mice.
Collapse
Affiliation(s)
- Fashui Hong
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China; Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian 223300, China; School of Life Sciences, Huaiyin Normal University, Huaian 223300, China.
| | - Xiaoyang Zhao
- Medical College of Soochow University, Suzhou 215123, China
| | - Wenhui Si
- Key Laboratory of Agricultural and Animal Products Processing and Quality Control, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China; Suzhou Polytechnic Institute of Agriculture, Suzhou 215008, China
| | - Yuguan Ze
- Medical College of Soochow University, Suzhou 215123, China
| | - Ling Wang
- Library of Soochow University, Suzhou 215123, China
| | - Yingjun Zhou
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China; Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian 223300, China; School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Jie Hong
- Medical College of Soochow University, Suzhou 215123, China
| | - Xiaohong Yu
- Medical College of Soochow University, Suzhou 215123, China
| | - Lei Sheng
- Medical College of Soochow University, Suzhou 215123, China
| | - Dong Liu
- Medical College of Soochow University, Suzhou 215123, China
| | - Bingqing Xu
- Medical College of Soochow University, Suzhou 215123, China
| | - Jianhao Zhang
- Key Laboratory of Agricultural and Animal Products Processing and Quality Control, Ministry of Agriculture, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
42
|
Hu Q, Chen J, Zhang J, Xu C, Yang S, Jiang H. IOX1, a JMJD2A inhibitor, suppresses the proliferation and migration of vascular smooth muscle cells induced by angiotensin II by regulating the expression of cell cycle-related proteins. Int J Mol Med 2015; 37:189-96. [PMID: 26530537 DOI: 10.3892/ijmm.2015.2393] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 10/01/2015] [Indexed: 11/06/2022] Open
Abstract
The epigenetic modification of vascular smooth muscle cell (VSMC) phenotypic switching, proliferation, migration, apoptosis and extracellular matrix synthesis is known to occur in atherosclerosis. The aim of the present study was to investigate the effects of IOX1, a Jumonji domain-containing 2A (JMJD2A) inhibitor, on regulation of the cell cycle in angiotensin II (Ang II)-stimulated VSMCs and to elucidate the possible mechanisms involved. The proliferation and migration of the Ang II-stimulated VSMCs in the presence or absence of IOX1 were evaluated in vitro. Flow cytometric analysis was used to determine the effects of IOX1 on cell cycle progression. RT-qPCR and western blot analysis were carried out to measure the expression levels of cell cycle-related genes. The trimethylation of histone H3 lysine 9 (H3K9me3) at the promoters of these genes was detected by chromatin immunoprecipitation (ChIP) assay. We confirmed that the JMJD2A levels were increased, whereas the H3K9me3 levels were decreased in the Ang II-stimulated VSMCs. The inhibition of JMJD2A by IOX1 suppressed the Ang II-induced cell proliferation, migration and cell cycle progression by inhibiting cyclin D1 expression and increasing p21 expression. The underlying mechanisms were related to the restoration of the H3K9me3 levels at the promoters of these genes. In conclusion, the findings of our study indicate that IOX1 exerts its anti-proliferative and anti-migratory effects by regulating the expression of the cell cycle-related proteins, cyclin D1 and p21.
Collapse
Affiliation(s)
- Qi Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Changwu Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shuo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
43
|
Hahm SW, Park J, Oh SY, Lee CW, Park KY, Kim H, Son YS. Anticancer properties of extracts from Opuntia humifusa against human cervical carcinoma cells. J Med Food 2015; 18:31-44. [PMID: 25379883 DOI: 10.1089/jmf.2013.3096] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In this study, we found that the total polyphenol and ascorbic acid levels in the fruit of Opuntia humifusa are higher than those in other parts of the plant. We further hypothesized that antioxidants in O. humifusa might affect the growth or survival of cancer cells. Hexane extracts of seeds and ethyl acetate extracts of fruits and stems significantly suppressed the proliferation of HeLa cervical carcinoma cells, but did not affect the proliferation of normal human BJ fibroblasts. Additionally, the extracts of O. humifusa induced G1 phase arrest in HeLa cells. The O. humifusa extracts reduced the levels of G1 phase-associated cyclin D1, cyclin-dependent kinase 4 (Cdk4), and phosphorylated retinoblastoma proteins. Moreover, p21(WAF1/Cip1) and p53 expression significantly increased after treatment. We examined the effects of ethyl acetate extracts of O. humifusa fruit (OHF) on HeLa cells xenograft tumor growth. OHF treatment significantly reduced tumor volume and this decrease was correlated with decreased Cdk4 and cyclin D1 expression. Furthermore, flavonoids, trans Taxifolin, and dihydrokaempferol, were isolated from OHF. Thus, this extract may be a promising candidate for treating human cervical carcinoma.
Collapse
Affiliation(s)
- Sahng-Wook Hahm
- 1 Institute of Life Science and Natural Resources, College of Life Sciences and Biotechnology, Korea University , Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
44
|
Cathepsin L suppression increases the radiosensitivity of human glioma U251 cells via G2/M cell cycle arrest and DNA damage. Acta Pharmacol Sin 2015; 36:1113-25. [PMID: 26095040 DOI: 10.1038/aps.2015.36] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/13/2015] [Indexed: 01/08/2023] Open
Abstract
AIM Cathepsin L is a lysosomal cysteine protease that plays important roles in cancer tumorigenesis, proliferation and chemotherapy resistance. The aim of this study was to determine how cathepsin L regulated the radiosensitivity of human glioma cells in vitro. METHODS Human glioma U251 cells (harboring the mutant type p53 gene) and U87 cells (harboring the wide type p53 gene) were irradiated with X-rays. The expression of cathepsin L was analyzed using Western blot and immunofluorescence assays. Cell survival and DNA damage were evaluated using clonogenic and comet assays, respectively. Flow cytometry was used to detect the cell cycle distribution. Apoptotic cells were observed using Hoechst 33258 staining and fluorescence microscopy. RESULTS Irradiation significantly increased the cytoplasmic and nuclear levels of cathepsin L in U251 cells but not in U87 cells. Treatment with the specific cathepsin L inhibitor Z-FY-CHO (10 μmol/L) or transfection with cathepsin L shRNA significantly increased the radiosensitivity of U251 cells. Both suppression and knockdown of cathepsin L in U251 cells increased irradiation-induced DNA damage and G2/M phase cell cycle arrest. Both suppression and knockdown of cathepsin L in U251 cells also increased irradiation-induced apoptosis, as shown by the increased levels of Bax and decreased levels of Bcl-2. CONCLUSION Cathepsin L is involved in modulation of radiosensitivity in human glioma U251 cells (harboring the mutant type p53 gene) in vitro.
Collapse
|
45
|
WANG JIN, YIN HAILIN, PANANDIKAR ASHWINI, GANDHI VARSHA, SEN SUBRATA. Elevated cyclin A associated kinase activity promotes sensitivity of metastatic human cancer cells to DNA antimetabolite drug. Int J Oncol 2015; 47:782-90. [PMID: 26058363 PMCID: PMC4501665 DOI: 10.3892/ijo.2015.3037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/04/2015] [Indexed: 11/16/2022] Open
Abstract
Drug resistance is a major obstacle in successful systemic therapy of metastatic cancer. We analyzed the involvement of cell cycle regulatory proteins in eliciting response to N (phosphonoacetyl)-L-aspartate (PALA), an inhibitor of de novo pyrimidine synthesis, in two metastatic variants of human cancer cell line MDA-MB-435 isolated from lung (L-2) and brain (Br-1) in nude mouse, respectively. L-2 and Br-l cells markedly differed in their sensitivity to PALA. While both cell types displayed an initial S phase delay/arrest, Br-l cells proliferated but most L-2 cells underwent apoptosis. There was distinct elevation in cyclin A, and phosphorylated Rb proteins concomitant with decreased expression of bcl-2 protein in the PALA treated L-2 cells undergoing apoptosis. Markedly elevated cyclin A associated and cdk2 kinase activities together with increased E2F1-DNA binding were detected in these L-2 cells. Induced ectopic cyclin A expression sensitized Br-l cells to PALA by activating an apoptotic pathway. Our findings demonstrate that elevated expression of cyclin A and associated kinase can activate an apoptotic pathway in cells exposed to DNA antimetabolites. Abrogation of this pathway can lead to resistance against these drugs in metastatic variants of human carcinoma cells.
Collapse
Affiliation(s)
- JIN WANG
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - HAILIN YIN
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - ASHWINI PANANDIKAR
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - VARSHA GANDHI
- Department of Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - SUBRATA SEN
- Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Program in Human and Molecular Genetics, University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
46
|
Cucurbitacin D induces cell cycle arrest and apoptosis by inhibiting STAT3 and NF-κB signaling in doxorubicin-resistant human breast carcinoma (MCF7/ADR) cells. Mol Cell Biochem 2015; 409:33-43. [PMID: 26169986 PMCID: PMC4589559 DOI: 10.1007/s11010-015-2509-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 07/04/2015] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most common cancer for women and is a major cause of mortality in women. Doxorubicin is a generally used chemotherapy drug for breast cancer. However, multidrug resistance of breast cancer interferes with the chemotherapy. We examined whether cucurbitacin D affects doxorubicin resistance of MCF7/ADR breast cancer cells. Cell viability was measured by MTT assay. Levels of p-STAT3, p-NF-κB, IκB, and caspases were measured by Western blot analysis. Nuclear staining of Stat3 and NF-κB was measured by immunocytochemistry. STAT3 and NF-κB transcriptional activity was detected by STAT3 and NF-κB luciferase reporter gene assays. Analysis of cell cycle arrest was performed by flow cytometry. Induction of apoptosis by cucurbitacin D was measured by Annexin V-FITC/propidium iodide assay. More than 90 % of MCF7/ADR cells lived upon treatment with doxorubicin for 24 h. However, upon treatment with cucurbitacin D, cell death was more than 60 %. Co-administration of cucurbitacin D and doxorubicin induced apoptosis, and G2/M cell cycle arrest, and inhibited upregulated Stat3 by doxorubicin on MCF7/ADR cells. Additionally, cucurbitacin D led to an increase in the IκBα level in the cytosol and a decrease in the p-NF-κB level in the nucleus. Finally, cucurbitacin D inhibited translocation of Stat3 and NF-κB and decreased transcriptional activity in the nucleus. Cucurbitacin D decreases cell proliferation and induces apoptosis by inhibiting Stat3 and NF-κB signaling in doxorubicin-resistant breast cancer cells. Cucurbitacin D could be used as a useful compound to treat adriamycin-resistant patients.
Collapse
|
47
|
Phosphanegold(I) thiolates, Ph3PAu[SC(OR)=NC6H4Me-4] for R = Me, Et and iPr, induce apoptosis, cell cycle arrest and inhibit cell invasion of HT-29 colon cancer cells through modulation of the nuclear factor-κB activation pathway and ubiquitination. J Biol Inorg Chem 2015; 20:855-73. [DOI: 10.1007/s00775-015-1271-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/10/2015] [Indexed: 11/27/2022]
|
48
|
Platelet-activating factor induces cell cycle arrest and disrupts the DNA damage response in mast cells. Cell Death Dis 2015; 6:e1745. [PMID: 25950475 PMCID: PMC4669695 DOI: 10.1038/cddis.2015.115] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 12/17/2022]
Abstract
Platelet-activating factor (PAF) is a potent phospholipid modulator of inflammation that has diverse physiological and pathological functions. Previously, we demonstrated that PAF has an essential role in ultraviolet (UV)-induced immunosuppression and reduces the repair of damaged DNA, suggesting that UV-induced PAF is contributing to skin cancer initiation by inducing immune suppression and also affecting a proper DNA damage response. The exact role of PAF in modulating cell proliferation, differentiation or transformation is unclear. Here, we investigated the mechanism(s) by which PAF affects the cell cycle and impairs early DNA damage response. PAF arrests proliferation in transformed and nontransformed human mast cells by reducing the expression of cyclin-B1 and promoting the expression of p21. PAF-treated cells show a dose-dependent cell cycle arrest mainly at G2–M, and a decrease in the DNA damage response elements MCPH1/BRIT-1 and ataxia telangiectasia and rad related (ATR). In addition, PAF disrupts the localization of p-ataxia telangiectasia mutated (p-ATM), and phosphorylated-ataxia telangiectasia and rad related (p-ATR) at the site of DNA damage. Whereas the potent effect on cell cycle arrest may imply a tumor suppressor activity for PAF, the impairment of proper DNA damage response might implicate PAF as a tumor promoter. The outcome of these diverse effects may be dependent on specific cues in the microenvironment.
Collapse
|
49
|
Wang YR, Xu Y, Jiang ZZ, Guerram M, Wang B, Zhu X, Zhang LY. Deoxypodophyllotoxin induces G2/M cell cycle arrest and apoptosis in SGC-7901 cells and inhibits tumor growth in vivo. Molecules 2015; 20:1661-75. [PMID: 25608854 PMCID: PMC6272175 DOI: 10.3390/molecules20011661] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Revised: 01/04/2015] [Accepted: 01/13/2015] [Indexed: 11/21/2022] Open
Abstract
Deoxypodophyllotoxin (DPT), a natural microtubule destabilizer, was isolated from Anthriscus sylvestris, and a few studies have reported its anti-cancer effect. However, the in vivo antitumor efficacy of DPT is currently indeterminate. In this study, we investigated the anti-gastric cancer effects of DPT both in vitro and in vivo. Our data showed that DPT inhibited cancer cell proliferation and induced G2/M cell cycle arrest accompanied by an increase in apoptotic cell death in SGC-7901 cancer cells. In addition, DPT caused cyclin B1, Cdc2 and Cdc25C to accumulate, decreased the expression of Bcl-2 and activated caspase-3 and PARP, suggesting that caspase-mediated pathways were involved in DPT-induced apoptosis. Animal studies revealed that DPT significantly inhibited tumor growth and decreased microvessel density (MVD) in a xenograft model of gastric cancer. Taken together, our findings provide a framework for further exploration of DPT as a novel chemotherapeutic for human gastric cancer.
Collapse
Affiliation(s)
- Yu-Rong Wang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Yuan Xu
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Zhen-Zhou Jiang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Mounia Guerram
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Bin Wang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiong Zhu
- Medical and Chemical Institute, China Pharmaceutical University, Nanjing 210009, China.
| | - Lu-Yong Zhang
- Jiangsu Center for Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
50
|
Chaffee BR, Shang F, Chang ML, Clement TM, Eddy EM, Wagner BD, Nakahara M, Nagata S, Robinson ML, Taylor A. Nuclear removal during terminal lens fiber cell differentiation requires CDK1 activity: appropriating mitosis-related nuclear disassembly. Development 2014; 141:3388-98. [PMID: 25139855 DOI: 10.1242/dev.106005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lens epithelial cells and early lens fiber cells contain the typical complement of intracellular organelles. However, as lens fiber cells mature they must destroy their organelles, including nuclei, in a process that has remained enigmatic for over a century, but which is crucial for the formation of the organelle-free zone in the center of the lens that assures clarity and function to transmit light. Nuclear degradation in lens fiber cells requires the nuclease DNase IIβ (DLAD) but the mechanism by which DLAD gains access to nuclear DNA remains unknown. In eukaryotic cells, cyclin-dependent kinase 1 (CDK1), in combination with either activator cyclins A or B, stimulates mitotic entry, in part, by phosphorylating the nuclear lamin proteins leading to the disassembly of the nuclear lamina and subsequent nuclear envelope breakdown. Although most post-mitotic cells lack CDK1 and cyclins, lens fiber cells maintain these proteins. Here, we show that loss of CDK1 from the lens inhibited the phosphorylation of nuclear lamins A and C, prevented the entry of DLAD into the nucleus, and resulted in abnormal retention of nuclei. In the presence of CDK1, a single focus of the phosphonuclear mitotic apparatus is observed, but it is not focused in CDK1-deficient lenses. CDK1 deficiency inhibited mitosis, but did not prevent DNA replication, resulting in an overall reduction of lens epithelial cells, with the remaining cells possessing an abnormally large nucleus. These observations suggest that CDK1-dependent phosphorylations required for the initiation of nuclear membrane disassembly during mitosis are adapted for removal of nuclei during fiber cell differentiation.
Collapse
Affiliation(s)
- Blake R Chaffee
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Fu Shang
- Laboratory for Nutrition and Vision Research, Human Nutrition Research Center on Aging, Nutrition &Vision Res-USDA-HNRCA, Tufts University, Boston 02111, MA, USA
| | - Min-Lee Chang
- Laboratory for Nutrition and Vision Research, Human Nutrition Research Center on Aging, Nutrition &Vision Res-USDA-HNRCA, Tufts University, Boston 02111, MA, USA
| | - Tracy M Clement
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Edward M Eddy
- National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | - Brad D Wagner
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Masaki Nakahara
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigekazu Nagata
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Allen Taylor
- Laboratory for Nutrition and Vision Research, Human Nutrition Research Center on Aging, Nutrition &Vision Res-USDA-HNRCA, Tufts University, Boston 02111, MA, USA Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|