1
|
Li XZ, Song W, Zhao ZH, Lu YH, Xu GL, Yang LJ, Yin S, Sun QY, Chen LN. Flavopiridol induces cell cycle arrest and apoptosis by interfering with CDK1 signaling pathway in human ovarian granulosa cells. Sci Rep 2024; 14:26239. [PMID: 39482384 PMCID: PMC11528022 DOI: 10.1038/s41598-024-77032-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
Several clinical trials have been conducted to evaluate the use of flavopiridol (FP) to treat a variety of cancers, and almost all cancer drugs were found to be associated with toxicity and side effects. It is not clear whether the use of FP will affect the female reproductive system. Granulosa cells, as the important cells that constitute the follicle, play a crucial role in determining the reproductive ability of females. In this study, we investigated whether different concentrations of FP have a toxic effect on the growth of immortalized human ovarian granulosa cells. The results showed that FP had an inhibitory effect on cell proliferation at a level of nanomole concentration. FP reduced cell proliferation and induced apoptosis by inducing mitochondrial dysfunction and oxidative stress, as well as increasing BAX/BCL2 and pCDK1 levels. These results suggest that toxicity to the reproductive system should be considered when FP is used in clinical applications.
Collapse
Affiliation(s)
- Xiao-Zhen Li
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Song
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zheng-Hui Zhao
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - You-Hui Lu
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gen-Lu Xu
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Li-Jia Yang
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shen Yin
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Lei-Ning Chen
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
2
|
Borlak J, Ciribilli Y, Bisio A, Selvaraj S, Inga A, Oh JH, Spanel R. The Abl1 tyrosine kinase is a key player in doxorubicin-induced cardiomyopathy and its p53/p73 cell death mediated signaling differs in atrial and ventricular cardiomyocytes. J Transl Med 2024; 22:845. [PMID: 39285385 PMCID: PMC11403941 DOI: 10.1186/s12967-024-05623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/16/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Doxorubicin is an important anticancer drug, however, elicits dose-dependently cardiomyopathy. Given its mode of action, i.e. topoisomerase inhibition and DNA damage, we investigated genetic events associated with cardiomyopathy and searched for mechanism-based possibilities to alleviate cardiotoxicity. We treated rats at clinically relevant doses of doxorubicin. Histopathology and transmission electron microscopy (TEM) defined cardiac lesions, and transcriptomics unveiled cardiomyopathy-associated gene regulations. Genomic-footprints revealed critical components of Abl1-p53-signaling, and EMSA-assays evidenced Abl1 DNA-binding activity. Gene reporter assays confirmed Abl1 activity on p53-targets while immunohistochemistry/immunofluorescence microscopy demonstrated Abl1, p53&p73 signaling. RESULTS Doxorubicin treatment caused dose-dependently toxic cardiomyopathy, and TEM evidenced damaged mitochondria and myofibrillar disarray. Surviving cardiomyocytes repressed Parkin-1 and Bnip3-mediated mitophagy, stimulated dynamin-1-like dependent mitochondrial fission and induced anti-apoptotic Bag1 signaling. Thus, we observed induced mitochondrial biogenesis. Transcriptomics discovered heterogeneity in cellular responses with minimal overlap between treatments, and the data are highly suggestive for distinct cardiomyocyte (sub)populations which differed in their resilience and reparative capacity. Genome-wide footprints revealed Abl1 and p53 enriched binding sites in doxorubicin-regulated genes, and we confirmed Abl1 DNA-binding activity in EMSA-assays. Extraordinarily, Abl1 signaling differed in the heart with highly significant regulations of Abl1, p53 and p73 in atrial cardiomyocytes. Conversely, in ventricular cardiomyocytes, Abl1 solely-modulated p53-signaling that was BAX transcription-independent. Gene reporter assays established Abl1 cofactor activity for the p53-reporter PG13-luc, and ectopic Abl1 expression stimulated p53-mediated apoptosis. CONCLUSIONS The tyrosine kinase Abl1 is of critical importance in doxorubicin induced cardiomyopathy, and we propose its inhibition as means to diminish risk of cardiotoxicity.
Collapse
Affiliation(s)
- Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Yari Ciribilli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alberto Inga
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
3
|
Xu X, Wang Z, Lv L, Liu C, Wang L, Sun YN, Zhao Z, Shi B, Li Q, Hao GM. Molecular regulation of DNA damage and repair in female infertility: a systematic review. Reprod Biol Endocrinol 2024; 22:103. [PMID: 39143547 PMCID: PMC11323701 DOI: 10.1186/s12958-024-01273-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
DNA damage is a key factor affecting gametogenesis and embryo development. The integrity and stability of DNA are fundamental to a woman's successful conception, embryonic development, pregnancy and the production of healthy offspring. Aging, reactive oxygen species, radiation therapy, and chemotherapy often induce oocyte DNA damage, diminished ovarian reserve, and infertility in women. With the increase of infertility population, there is an increasing need to study the relationship between infertility related diseases and DNA damage and repair. Researchers have tried various methods to reduce DNA damage in oocytes and enhance their DNA repair capabilities in an attempt to protect oocytes. In this review, we summarize recent advances in the DNA damage response mechanisms in infertility diseases such as PCOS, endometriosis, diminished ovarian reserve and hydrosalpinx, which has important implications for fertility preservation.
Collapse
Affiliation(s)
- Xiuhua Xu
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
- Cardiovascular platform, Institute of Health and Disease, Hebei Medical University, Shijiazhuang, 050000, China
| | - Ziwei Wang
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Luyi Lv
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ci Liu
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lili Wang
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ya-Nan Sun
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Zhiming Zhao
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Baojun Shi
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Qian Li
- Cardiovascular platform, Institute of Health and Disease, Hebei Medical University, Shijiazhuang, 050000, China.
| | - Gui-Min Hao
- Hebei Key Laboratory of Infertility and Genetics, Hebei Clinical Research Center for Birth Defects, Hebei Medical Key discipline of Reproductive Medicine, Hebei Collaborative Innovation Center of Integrated Traditional and Western Medicine on Reproductive Disease, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
4
|
Markowska A, Antoszczak M, Markowska J, Huczyński A. Gynotoxic Effects of Chemotherapy and Potential Protective Mechanisms. Cancers (Basel) 2024; 16:2288. [PMID: 38927992 PMCID: PMC11202309 DOI: 10.3390/cancers16122288] [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: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Chemotherapy is one of the leading cancer treatments. Unfortunately, its use can contribute to several side effects, including gynotoxic effects in women. Ovarian reserve suppression and estrogen deficiency result in reduced quality of life for cancer patients and are frequently the cause of infertility and early menopause. Classic alkylating cytostatics are among the most toxic chemotherapeutics in this regard. They cause DNA damage in ovarian follicles and the cells they contain, and they can also induce oxidative stress or affect numerous signaling pathways. In vitro tests, animal models, and a few studies among women have investigated the effects of various agents on the protection of the ovarian reserve during classic chemotherapy. In this review article, we focused on the possible beneficial effects of selected hormones (anti-Müllerian hormone, ghrelin, luteinizing hormone, melatonin), agents affecting the activity of apoptotic pathways and modulating gene expression (C1P, S1P, microRNA), and several natural (quercetin, rapamycin, resveratrol) and synthetic compounds (bortezomib, dexrazoxane, goserelin, gonadoliberin analogs, imatinib, metformin, tamoxifen) in preventing gynotoxic effects induced by commonly used cytostatics. The presented line of research appears to provide a promising strategy for protecting and/or improving the ovarian reserve in the studied group of cancer patients. However, well-designed clinical trials are needed to unequivocally assess the effects of these agents on improving hormonal function and fertility in women treated with ovotoxic anticancer drugs.
Collapse
Affiliation(s)
- Anna Markowska
- Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| | - Janina Markowska
- Gynecological Oncology Center, Poznańska 58A, 60-850 Poznań, Poland;
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| |
Collapse
|
5
|
Dose-dependent effects of ghrelin and aberrant anti-Mullerian hormone levels in the prevention of ovarian damage caused by cisplatin in Wistar-albino rats. Arch Gynecol Obstet 2021; 305:1003-1009. [PMID: 34687336 DOI: 10.1007/s00404-021-06292-4] [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: 04/17/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE Ghrelin has previously been proven to have anti-inflammatory and antioxidant properties in preventing cisplatin-induced ovarian damage. The aim of this study was to evaluate the potential effects of this hormone in preventing this damage in rats using histopathological and biochemical methods. METHODS Twenty-eight Wistar-albino rats were randomly divided into four groups. While no drug was given to Group 1 (sham group), acylated ghrelin was intraperitoneally administered to Group 2 at 0.5 nmol/kg and Group 3 at 2 nmol/kg for 21 days. Group 4 received only saline solution. On the 15th day, a single dose of 5 mg/kg cisplatin was intraperitoneally administered to each rat in Groups 2, 3 and 4. Serum anti-Mullerian hormone (AMH) values were measured on days 0, 15 and 21. Then, laparotomy and bilateral oophorectomy were performed, and the ovaries were histopathologically examined. RESULTS The number of primordial and primary follicles was significantly higher in Group 3 than in the saline solution + cisplatin group. In Group 4, cisplatin caused significantly higher follicle damage in the primordial, primary and secondary phases compared to the sham group. The AMH level of the SF + cisplatin group was significantly lower than that of the sham group and the high-dose ghrelin + cisplatin group, and the AMH level of the sham group was significantly higher than that of the low-dose ghrelin + cisplatin group. CONCLUSION High-dose ghrelin was effective in preventing cisplatin-induced ovarian damage by preserving the number of primordial and primary follicles. Larger randomized studies are needed to determine the optimal dosage and duration of ghrelin.
Collapse
|
6
|
Lee JH, Choi YS. The role of gonadotropin-releasing hormone agonists in female fertility preservation. Clin Exp Reprod Med 2021; 48:11-26. [PMID: 33648041 PMCID: PMC7943347 DOI: 10.5653/cerm.2020.04049] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/13/2020] [Indexed: 11/19/2022] Open
Abstract
Advances in anticancer treatments have resulted in increasing survival rates among cancer patients. Accordingly, the quality of life after treatment, particularly the preservation of fertility, has gradually emerged as an essential consideration. Cryopreservation of embryos or unfertilized oocytes has been considered as the standard method of fertility preservation among young women facing gonadotoxic chemotherapy. Other methods, including ovarian suppression and ovarian tissue cryopreservation, have been considered experimental. Recent large-scale randomized controlled trials have demonstrated that temporary ovarian suppression using gonadotropin-releasing hormone agonists (GnRHa) during chemotherapy is beneficial for preventing chemotherapy-induced premature ovarian insufficiency in breast cancer patients. It should also be emphasized that GnRHa use during chemotherapy does not replace established fertility preservation methods. All young women facing gonadotoxic chemotherapy should be counseled about and offered various options for fertility preservation, including both GnRHa use and cryopreservation of embryos, oocytes, and/or ovarian tissue.
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Young Sik Choi
- Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
7
|
Yildirim E, Yaba A. Determination of c-Abl tyrosine kinase and mTERT catalytic subunit of telomerase expression level during prenatal-postnatal mouse ovary-testis development. Reprod Biol 2020; 20:555-567. [PMID: 33191142 DOI: 10.1016/j.repbio.2020.10.004] [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: 05/25/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 11/26/2022]
Abstract
Expression levels of genes involved in the development of germ cells vary throughout the process from bipotential gonadal period to adult gonadal formation. In mice, developments of female and male reproductive system are regulated by germ cell-specific factors and hormones, and determinative days in this regulation are very important. c-Abl is a non-receptor tyrosine kinase with cellular functions including cell proliferation, growth and development. mTERT is involved in maintaining telomerase activity and proliferation of surviving cells. We suggested that c-Abl and mTERT might be important for the healthy development of prenatal and postnatal mouse ovary and testis. We aim to demonstrate localization and expressions of c-Abl and mTERT in crucial days of ovary and testis development in prenatal and postnatal period in mouse by immunofluorescence staining and qRT-PCR, respectively. The importance of c-Abl and mTERT expressions during the healthy gonadal development is indicated in the prenatal and postnatal gonadal development. Also, protein expression levels were detected by Western Blot in only postnatal ovary and testis. Determining the functions of the c-Abl and mTERT throughout the process will be important in terms of understanding the infertility cases in the female and male with future studies.
Collapse
Affiliation(s)
- Ecem Yildirim
- Yeditepe University School of Medicine, Department of Histology and Embryology, 34755, Istanbul, Turkey
| | - Aylin Yaba
- Yeditepe University School of Medicine, Department of Histology and Embryology, 34755, Istanbul, Turkey.
| |
Collapse
|
8
|
Lin M, Guo L, Cheng Z, Huan X, Huang Y, Xu K. Association of hMSH5 C85T polymorphism with radiation sensitivity of testicular cell lines GC-1, GC-2, TM3, and TM4. Andrology 2020; 8:1174-1183. [PMID: 32306546 DOI: 10.1111/andr.12803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/12/2020] [Accepted: 04/14/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND The hMSH5 C85T polymorphism, which encodes hMSH5 P29S, is associated with individual differences in spermatogenic abnormalities caused by ionizing radiation (IR), but the molecular mechanisms remain unclear. OBJECTIVES This manuscript aims to explore the role of hMSH5 C85T polymorphism in IR-induced individual differences in spermatogenic abnormalities. MATERIAL AND METHODS We transfected pcDNA-hMSH5P29S vector into mouse spermatogonia GC-1, mouse spermatocytes GC-2, mouse testicular mesenchymal cells TM3, and mouse testicular support cells TM4. After radiation, we evaluated cell survival with colony formation assay, apoptosis with TUNEL assay and caspase-3 activity assay, DNA damage with comet assay and an in vivo NHEJ activity assay. RESULTS Results showed that only spermatocytes GC-2 transfected with pcDNA-hMSH5P29S vector had significant differences in IR-induced cell survival and apoptosis when compared to that transfected with pcDNA empty vector and pcDNA-wild-hMSH5 vector, while there was no statistical difference in GC-1, TM3, and TM4. In addition, comet assay showed that the DNA damage of GC-2 transfected with pcDNA-hMSH5P29S vector increased significantly compared to that transfected with pcDNA empty vector and pcDNA-wild-hMSH5 vector after IR. And in vivo NHEJ activity assay showed that the NHEJ activity of GC-2 transfected with pcDNA-hMSH5P29S vector was statistically higher than that transfected with pcDNA empty vector and pcDNA-wild-hMSH5 vector. CONCLUSION Our study indicates that the hMSH5 C85T polymorphism leads to an abnormal increase in apoptosis and lessen the control on error-prone NHEJ of spermatocyte GC-2, thereby altering the difference of radiation sensitivity of spermatogenesis.
Collapse
Affiliation(s)
- Mingyue Lin
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lihuang Guo
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhenbo Cheng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Xisha Huan
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yue Huang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| | - Keqian Xu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
9
|
Lopes F, Liu J, Morgan S, Matthews R, Nevin L, Anderson RA, Spears N. Single and combined effects of cisplatin and doxorubicin on the human and mouse ovary in vitro. Reproduction 2019; 159:193-204. [PMID: 31821159 PMCID: PMC6993208 DOI: 10.1530/rep-19-0279] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 12/09/2019] [Indexed: 01/06/2023]
Abstract
Chemotherapy drugs are administered to patients using combination regimens, and as such the possibility of multiplicative effects between drugs need to be investigated. This study examines the individual and combined effects of the chemotherapy drugs cisplatin and doxorubicin on the human ovary. Although cisplatin and doxorubicin are known to affect female fertility, there is limited information about their direct effects on the human ovary, and none examining the possibility of combined, multiplicative effects of co-exposure to these drugs. Here, human ovarian biopsies were obtained from 14 women at the time of caesarean section, with 38 mouse ovaries also obtained from neonatal C57Bl/6J mice. Tissue was cultured for 6 days prior to analyses, with chemotherapy drugs added to culture medium on the second day of culture only. Treatment groups of a single (5 μg/mL human; 0.5 μg/mL mouse) or double (10 μg/mL human; 1.0 μg/mL mouse) dose of cisplatin, a single (1 μg/mL human; 0.05 μg/mL mouse) or double (2 μg/mL human; 0.1 μg/mL mouse) dose of doxorubicin or a combination of a single dose of both drugs together were compared to controls without drug exposure. Exposure to cisplatin or doxorubicin significantly decreased follicle health in human and mouse, supporting the suitability of the mouse as a model for the human ovary. There was also a significant reduction of mouse follicle number. Human ovarian stromal tissue exhibited increased apoptosis and decreased cell proliferation. Crucially, there was no evidence indicating the occurrence of multiplicative effects between cisplatin and doxorubicin.
Collapse
Affiliation(s)
- Federica Lopes
- F Lopes, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Jin Liu
- J Liu, Department of Public Health, Fujian Medical University, Fuzhou, China
| | - Stephanie Morgan
- S Morgan, Biological Sciences, University of Southampton, Southampton, United Kingdom of Great Britain and Northern Ireland
| | - Rebecca Matthews
- R Matthews, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Lucy Nevin
- L Nevin, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Richard A Anderson
- R Anderson, MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Norah Spears
- N Spears, Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
10
|
Tucker EJ, Jaillard S, Grover SR, van den Bergen J, Robevska G, Bell KM, Sadedin S, Hanna C, Dulon J, Touraine P, Sinclair AH. TP63-truncating variants cause isolated premature ovarian insufficiency. Hum Mutat 2019; 40:886-892. [PMID: 30924587 DOI: 10.1002/humu.23744] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/05/2019] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
Abstract
Premature ovarian insufficiency involves amenorrhea and elevated follicle-stimulating hormone before age 40, and its genetic basis is poorly understood. Here, we study 13 premature ovarian insufficiency (POI) patients using whole-exome sequencing. We identify PREPL and TP63 causative variants, and variants in other potentially novel POI genes. PREPL deficiency is a known cause of syndromic POI, matching the patients' phenotype. A role for TP63 in ovarian biology has previously been proposed but variants have been described in multiorgan syndromes, and not isolated POI. One patient with isolated POI harbored a de novo nonsense TP63 variant in the terminal exon and an unrelated patient had a different nonsense variant in the same exon. These variants interfere with the repression domain while leaving the activation domain intact. We expand the phenotypic spectrum of TP63-related disorders, provide a new genotype:phenotype correlation for TP63 and identify a new genetic cause of isolated POI.
Collapse
Affiliation(s)
- Elena J Tucker
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Sylvie Jaillard
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,CHU Rennes, Service de Cytogénétique et Biologie Cellulaire, F-35033, Rennes, France.,INSERM U1085-IRSET, Université de Rennes1, Frances Pathologies Gynécologiques Rares, F-35042, Rennes, Paris, France
| | - Sonia R Grover
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,Department of Paediatric and Adolescent Gynaecology, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Jocelyn van den Bergen
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Gorjana Robevska
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Katrina M Bell
- Bioinformatics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Simon Sadedin
- Bioinformatics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Chloe Hanna
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatric and Adolescent Gynaecology, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Jérôme Dulon
- Department of Endocrinology and Reproductive Medicine, AP-HP, Sorbonne University Medicine, Centre de Référence desMaladies Endocriniennes Rares de laCroissance et du Développement, Centre des Pathologies Gynécologiques Rares, Paris, France
| | - Philippe Touraine
- Department of Endocrinology and Reproductive Medicine, AP-HP, Sorbonne University Medicine, Centre de Référence desMaladies Endocriniennes Rares de laCroissance et du Développement, Centre des Pathologies Gynécologiques Rares, Paris, France
| | - Andrew H Sinclair
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Kim SY, Nair DM, Romero M, Serna VA, Koleske AJ, Woodruff TK, Kurita T. Transient inhibition of p53 homologs protects ovarian function from two distinct apoptotic pathways triggered by anticancer therapies. Cell Death Differ 2018; 26:502-515. [PMID: 29988075 DOI: 10.1038/s41418-018-0151-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/22/2022] Open
Abstract
Platinum-based chemotherapies can result in ovarian insufficiency by reducing the ovarian reserve, a reduction believed to result from apoptosis of immature oocytes via activation/phosphorylation of TAp63α by multiple kinases including CHEK2, CK1, and ABL1. Here we demonstrate that cisplatin (CDDP) induces oocyte apoptosis through a novel pathway and that temporary repression of this pathway fully preserves ovarian function in vivo. Although ABL kinase inhibitors effectively block CDDP-induced apoptosis of oocytes, oocytic ABL1, and ABL2 are dispensable for damage-induced apoptosis. Instead, CDDP activates TAp63α through the ATR > CHEK1 pathway independent of TAp63α hyper-phosphorylation, whereas X-irradiation activates the ATM > CHEK2 > TAp63α-hyper-phosphorylation pathway. Furthermore, oocyte-specific deletion of Trp73 partially protects oocytes from CDDP but not from X-ray, highlighting the fundamental differences of two pathways. Nevertheless, temporary repression of DNA damage response by a kinase inhibitor that attenuates phosphorylation of ATM, ATR, CHEK1, and CHEK2 fully preserves fertility in female mice against CDDP as well as X-ray. Our current study establishes the molecular basis and feasibility of adjuvant therapies to protect ovarian function against two distinctive gonadotoxic therapeutics, CDDP, and ionizing radiation.
Collapse
Affiliation(s)
- So-Youn Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Devi M Nair
- Department of Cancer Biology and Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Megan Romero
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Vanida A Serna
- Department of Cancer Biology and Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Teresa K Woodruff
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Takeshi Kurita
- Department of Cancer Biology and Genetics, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Developmental effects of imatinib mesylate on follicle assembly and early activation of primordial follicle pool in postnatal rat ovary. Reprod Biol 2017; 17:25-33. [DOI: 10.1016/j.repbio.2016.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022]
|
13
|
Abstract
The Abelson (ABL) tyrosine kinases were identified as drivers of leukemia in mice and humans. Emerging data has shown a role for the ABL family kinases, ABL1 and ABL2, in the progression of several solid tumors. This review will focus on recent reports of the involvement of the ABL kinases in tumor progression using mouse models as well as recent data generated from genomic and proteomic studies linking enhanced expression and hyper-activation of the ABL kinases to some human cancers. Preclinical studies on small molecule inhibitors of the ABL kinases suggest that their use may have beneficial effects for the treatment of selected solid tumors.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710 USA
| |
Collapse
|
14
|
Impaired steroidogenesis and apoptosis of granulosa-luteal cells in primary culture induced by cis-platinum. Am J Obstet Gynecol 2014; 210:252.e1-7. [PMID: 24215857 DOI: 10.1016/j.ajog.2013.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 10/08/2013] [Accepted: 11/04/2013] [Indexed: 11/20/2022]
Abstract
OBJECTIVE The purpose of this study was to test the hypothesis that the cytotoxic drug cis-platinum (CP) induces premature ovarian failure by reducing the viability of human granulosa cells. STUDY DESIGN We incubated cultured human granulosa-luteal cells (GLCs) with varying concentrations of CP for 48 hours. Steroidogenesis and apoptosis were assessed by progesterone and estradiol, annexin V/propidium iodide, phase contrast, and transmission electron microscopy. RESULTS CP caused impaired production of progesterone and estradiol in a dose- and a time-dependent fashion. The estradiol production was more pronounced than progesterone for each concentration of CP that was studied. The phase contrast microscopy of CP-treated GLCs showed loss of cell number with condensed nuclei. CP-induced apoptosis was maximum at 20 μg/mL compared with a 10-μg/mL concentration (79.9% ± 4.6% vs 58.3% ± 3.9%; P < .01). The hallmark of apoptosis (ie, nuclear condensation, cell size shrinkage) was seen in CP-treated cells by transmission electron microscopy. CONCLUSION CP induces apoptosis of human GLCs in culture with impaired steroidogenesis, which may be one mechanism by which a CP-containing regime induces premature ovarian failure.
Collapse
|
15
|
Li Y, Chen X, Shi M, Wang H, Cao W, Wang X, Li C, Feng W. Proteomic-based identification of Apg-2 as a therapeutic target for chronic myeloid leukemia. Cell Signal 2013; 25:2604-12. [DOI: 10.1016/j.cellsig.2013.08.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 08/23/2013] [Indexed: 12/28/2022]
|
16
|
Morgan S, Lopes F, Gourley C, Anderson RA, Spears N. Cisplatin and doxorubicin induce distinct mechanisms of ovarian follicle loss; imatinib provides selective protection only against cisplatin. PLoS One 2013; 8:e70117. [PMID: 23922929 PMCID: PMC3726485 DOI: 10.1371/journal.pone.0070117] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 06/17/2013] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Chemotherapy treatment in premenopausal women has been linked to ovarian follicle loss and premature ovarian failure; the exact mechanism by which this occurs is uncertain. Here, two commonly used chemotherapeutic agents (cisplatin and doxorubicin) were added to a mouse ovary culture system, to compare the sequence of events that leads to germ cell loss. The ability of imatinib mesylate to protect the ovary against cisplatin or doxorubicin-induced ovarian damage was also examined. EXPERIMENTAL DESIGN Newborn mouse ovaries were cultured for a total of six days, exposed to a chemotherapeutic agent on the second day: this allowed for the examination of the earliest stages of follicle development. Cleaved PARP and TUNEL were used to assess apoptosis following drug treatment. Imatinib was added to cultures with cisplatin and doxorubicin to determine any protective effect. RESULTS Histological analysis of ovaries treated with cisplatin showed oocyte-specific damage; in comparison doxorubicin preferentially caused damage to the granulosa cells. Cleaved PARP expression significantly increased for cisplatin (16 fold, p<0.001) and doxorubicin (3 fold, p<0.01). TUNEL staining gave little evidence of primordial follicle damage with either drug. Imatinib had a significant protective effect against cisplatin-induced follicle damage (p<0.01) but not against doxorubicin treatment. CONCLUSION Cisplatin and doxorubicin both induced ovarian damage, but in a markedly different pattern, with imatinib protecting the ovary against damage by cisplatin but not doxorubicin. Any treatment designed to block the effects of chemotherapeutic agents on the ovary may need to be specific to the drug(s) the patient is exposed to.
Collapse
Affiliation(s)
- Stephanie Morgan
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Federica Lopes
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Charlie Gourley
- Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Norah Spears
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
17
|
Ciccone S, Maiani E, Bellusci G, Diederich M, Gonfloni S. Parkinson's disease: a complex interplay of mitochondrial DNA alterations and oxidative stress. Int J Mol Sci 2013; 14:2388-409. [PMID: 23348931 PMCID: PMC3587993 DOI: 10.3390/ijms14022388] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/14/2013] [Accepted: 01/21/2013] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is one of the most common age-related neurodegenerative diseases. This pathology causes a significant loss of dopaminergic neurons in the Substantia Nigra. Several reports have claimed a role of defective nuclear and mitochondrial DNA repair pathways in PD etiology, in particular, of the Base Excision Repair (BER) system. In addition, recent findings, related to PD progression, indicate that oxidative stress pathways involving c-Abl and GST could also be implicated in this pathology. This review focuses on recently described networks most likely involved in an integrated manner in the course of PD.
Collapse
Affiliation(s)
- Sarah Ciccone
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy; E-Mails: (S.C.); (E.M.); (G.B.)
| | - Emiliano Maiani
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy; E-Mails: (S.C.); (E.M.); (G.B.)
| | - Giovanna Bellusci
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy; E-Mails: (S.C.); (E.M.); (G.B.)
| | - Marc Diederich
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Kirchberg Hospital, 9 Rue Edward Steichen, 2540 Luxembourg, Luxembourg; E-Mail:
- College of Pharmacy, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul 151-742, Korea
| | - Stefania Gonfloni
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 00133 Rome, Italy; E-Mails: (S.C.); (E.M.); (G.B.)
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Kirchberg Hospital, 9 Rue Edward Steichen, 2540 Luxembourg, Luxembourg; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +39-06-72594319; Fax: +39-06-2023500
| |
Collapse
|
18
|
Targeting DNA damage response: threshold, chromatin landscape and beyond. Pharmacol Ther 2013; 138:46-52. [PMID: 23291058 DOI: 10.1016/j.pharmthera.2012.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 12/13/2012] [Indexed: 12/24/2022]
Abstract
Cells are continually exposed to DNA assaults from exogenous and endogenous sources. To maintain genomic integrity, cells have evolved a highly conserved mechanism for repairing DNA lesions and, in particular, DNA double strand breaks (DSBs). Emerging evidence indicates that DNA repair/signaling machinery acts in an integrated fashion with chromatin structure at damaged sites. This review focuses on the interplay between histone modifications and the chromatin-mediated response to DNA damage.
Collapse
|
19
|
Katsumata R, Ishigaki S, Katsuno M, Kawai K, Sone J, Huang Z, Adachi H, Tanaka F, Urano F, Sobue G. c-Abl inhibition delays motor neuron degeneration in the G93A mouse, an animal model of amyotrophic lateral sclerosis. PLoS One 2012; 7:e46185. [PMID: 23049975 PMCID: PMC3458026 DOI: 10.1371/journal.pone.0046185] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 08/28/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive death of motor neurons. Although the pathogenesis of ALS remains unclear, several cellular processes are known to be involved, including apoptosis. A previous study revealed the apoptosis-related gene c-Abl to be upregulated in sporadic ALS motor neurons. METHODOLOGY/FINDINGS We investigated the possibility that c-Abl activation is involved in the progression of ALS and that c-Abl inhibition is potentially a therapeutic strategy for ALS. Using a mouse motor neuron cell line, we found that mutation of Cu/Zn-superoxide dismutase-1 (SOD1), which is one of the causative genes of familial ALS, induced the upregulation of c-Abl and decreased cell viability, and that the c-Abl inhibitor dasatinib inhibited cytotoxicity. Activation of c-Abl with a concomitant increase in activated caspase-3 was observed in the lumbar spine of G93A-SOD1 transgenic mice (G93A mice), a widely used model of ALS. The survival of G93A mice was improved by oral administration of dasatinib, which also decreased c-Abl phosphorylation, inactivated caspase-3, and improved the innervation status of neuromuscular junctions. In addition, c-Abl expression in postmortem spinal cord tissues from sporadic ALS patients was increased by 3-fold compared with non-ALS patients. CONCLUSIONS/SIGNIFICANCE The present results suggest that c-Abl is a potential therapeutic target for ALS and that the c-Abl inhibitor dasatinib has neuroprotective properties in vitro and in vivo.
Collapse
Affiliation(s)
- Ryu Katsumata
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Shinsuke Ishigaki
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Kaori Kawai
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Jun Sone
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Zhe Huang
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Hiroaki Adachi
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Fumiaki Tanaka
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Fumihiko Urano
- Program in Gene Function and Expression, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Tsurumai-cho, Showa-ku, Nagoya, Japan
- Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
20
|
Morgan S, Anderson RA, Gourley C, Wallace WH, Spears N. How do chemotherapeutic agents damage the ovary? Hum Reprod Update 2012; 18:525-35. [PMID: 22647504 DOI: 10.1093/humupd/dms022] [Citation(s) in RCA: 287] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Chemotherapy treatment in premenopausal women is associated with an increased risk of premature ovarian failure (POF) but the exact mechanism through which this occurs is uncertain. In this review we examine the current evidence for the direct action of chemotherapeutic agents on the ovary and discuss possible molecular pathways through which follicle loss may occur. METHODS A systemic search of the databases, PubMed and Google Scholar, was made for all English language articles through to 2011 in each subject area discussed. RESULTS POF results from the loss of primordial follicles but this is not necessarily a direct effect of the chemotherapeutic agents. Instead, the disappearance of primordial follicles could be due to an increased rate of growth initiation to replace damaged developing follicles. Likewise, the loss of oocytes need not necessarily be a direct result of damage: evidence suggests that chemotherapy drugs can also induce oocyte death indirectly via damage to somatic cells. Specific molecular mechanisms and likely ovarian targets are discussed for some of the anti-cancer drugs most commonly used to treat premenopausal women. Finally, we consider current and prospective methods of preserving fertility. CONCLUSIONS It is likely that different chemotherapeutic drugs act through a range of mechanisms and on different target cells. More research into the cellular mechanisms underpinning chemotherapy-induced follicle loss could lead to the generation of treatments specifically designed to prevent POF.
Collapse
Affiliation(s)
- S Morgan
- Centre for Integrative Physiology, University of Edinburgh, George Square, Edinburgh EH8 9XD, UK
| | | | | | | | | |
Collapse
|
21
|
Matta A, Siu KWM, Ralhan R. 14-3-3 zeta as novel molecular target for cancer therapy. Expert Opin Ther Targets 2012; 16:515-23. [DOI: 10.1517/14728222.2012.668185] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Ishii T, Warabi E, Yanagawa T. Novel roles of peroxiredoxins in inflammation, cancer and innate immunity. J Clin Biochem Nutr 2012; 50:91-105. [PMID: 22448089 PMCID: PMC3303482 DOI: 10.3164/jcbn.11-109] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 09/20/2011] [Indexed: 02/06/2023] Open
Abstract
Peroxiredoxins possess thioredoxin or glutathione peroxidase and chaperone-like activities and thereby protect cells from oxidative insults. Recent studies, however, reveal additional functions of peroxiredoxins in gene expression and inflammation-related biological reactions such as tissue repair, parasite infection and tumor progression. Notably, peroxiredoxin 1, the major mammalian peroxiredoxin family protein, directly interacts with transcription factors such as c-Myc and NF-κB in the nucleus. Additionally, peroxiredoxin 1 is secreted from some cells following stimulation with TGF-β and other cytokines and is thus present in plasma and body fluids. Peroxiredoxin 1 is now recognized as one of the pro-inflammatory factors interacting with toll-like receptor 4, which triggers NF-κB activation and other signaling pathways to evoke inflammatory reactions. Some cancer cells release peroxiredoxin 1 to stimulate toll-like receptor 4-mediated signaling for their progression. Interestingly, peroxiredoxins expressed in protozoa and helminth may modulate host immune responses partly through toll-like receptor 4 for their survival and progression in host. Extracellular peroxiredoxin 1 and peroxiredoxin 2 are known to enhance natural killer cell activity and suppress virus-replication in cells. Peroxiredoxin 1-deficient mice show reduced antioxidant activities but also exhibit restrained tissue inflammatory reactions under some patho-physiological conditions. Novel functions of peroxiredoxins in inflammation, cancer and innate immunity are the focus of this review.
Collapse
Affiliation(s)
- Tetsuro Ishii
- Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | | | | |
Collapse
|
23
|
DNA damage response: The emerging role of c-Abl as a regulatory switch? Biochem Pharmacol 2011; 82:1269-76. [DOI: 10.1016/j.bcp.2011.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Revised: 06/30/2011] [Accepted: 07/01/2011] [Indexed: 12/20/2022]
|
24
|
Jiang Z, Kamath R, Jin S, Balasubramani M, Pandita TK, Rajasekaran B. Tip60-mediated acetylation activates transcription independent apoptotic activity of Abl. Mol Cancer 2011; 10:88. [PMID: 21781306 PMCID: PMC3157453 DOI: 10.1186/1476-4598-10-88] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 07/22/2011] [Indexed: 11/19/2022] Open
Abstract
Background The proto-oncogene, c-Abl encodes a ubiquitously expressed tyrosine kinase that critically governs the cell death response induced by genotoxic agents such as ionizing radiation and cisplatin. The catalytic function of Abl, which is essential for executing DNA damage response (DDR), is normally tightly regulated but upregulated several folds upon IR exposure due to ATM-mediated phosphorylation on S465. However, the mechanism/s leading to activation of Abl's apoptotic activity is currently unknown. Results We investigated the role of acetyl modification in regulating apoptotic activity of Abl and the results showed that DNA strand break-inducing agents, ionizing radiation and bleomycin induced Abl acetylation. Using mass spectrophotometry and site-specific acetyl antibody, we identified Abl K921, located in the DNA binding domain, and conforming to one of the lysine residue in the consensus acetylation motif (KXXK--X3-5--SGS) is acetylated following DNA damage. We further observed that the S465 phosphorylated Abl is acetyl modified during DNA damage. Signifying the modification, cells expressing the non acetylatable K921R mutant displayed attenuated apoptosis compared to wild-type in response to IR or bleomycin treatment. WT-Abl induced apoptosis irrespective of new protein synthesis. Furthermore, upon γ-irradiation K921R-Abl displayed reduced chromatin binding compared to wild type. Finally, loss of Abl K921 acetylation in Tip60-knocked down cells and co-precipitation of Abl with Tip60 in DNA damaged cells identified Tip60 as an Abl acetylase. Conclusion Collective data showed that DNA damage-induced K921 Abl acetylation, mediated by Tip60, stimulates transcriptional-independent apoptotic activity and chromatin-associative property thereby defining a new regulatory mechanism governing Abl's DDR function.
Collapse
Affiliation(s)
- Zhihua Jiang
- Department of Microbiology and Molecular Genetics, Pittsburgh, PA 15240, USA
| | | | | | | | | | | |
Collapse
|
25
|
Topology and dynamics of the interaction between 5-nitroimidazole radiosensitizers and duplex DNA studied by a combination of docking, molecular dynamic simulations and NMR spectroscopy. J Mol Struct 2011. [DOI: 10.1016/j.molstruc.2011.02.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|