1
|
Dai Y, Yuan Z, Fan W, Lin Z. Molecular mechanism of aberrant decidualization in adenomyosis leading to reduced endometrial receptivity. Front Endocrinol (Lausanne) 2025; 15:1435177. [PMID: 39886033 PMCID: PMC11779606 DOI: 10.3389/fendo.2024.1435177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/18/2024] [Indexed: 02/01/2025] Open
Abstract
Patients with adenomyosis not only experience a decrease in quality of life as a result of dysmenorrhea and severe monthly flow but they are also rendered infertile. Pregnancy rates are still low among women with adenomyosis, even with assisted reproduction. According to the current study, endometrial receptivity is primarily responsible for the lower conception rate among patients with adenomyosis. Decidualization of endometrial stromal cells is the fundamental requirement for endometrial receptivity and the maintenance of a normal pregnancy, even though endometrial receptivity is made up of a variety of cells, including immune cells, endometrial epithelial cells, and endometrial stromal cells. Our overview reveals that endometriosis deficiencies are present in patients with adenomyosis. These flaws may be linked to aberrant pathways in endometrial stromal cells, such as PI3K/Akt, JAK2/STAT3, and hedgehog. Correcting the abnormal expression of molecules in endometrial stromal cells in the endometrium of patients with adenomyosis may become the focus of research to improve endometrial receptivity and increase the pregnancy rate.
Collapse
Affiliation(s)
- Yuanquan Dai
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zheng Yuan
- Department of Gynecology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Weisen Fan
- Department of Gynecology, Guang anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiheng Lin
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Yang M, Wang K, Zhang L, Zhang H, Zhang C. DCAF2 is essential for the development of uterine epithelia and mouse fertility. Front Cell Dev Biol 2024; 12:1474660. [PMID: 39364135 PMCID: PMC11446810 DOI: 10.3389/fcell.2024.1474660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/09/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction The successful outcome of a pregnancy depends on the proper functioning uterine epithelium. DNA damage binding protein 1 and cullin 4-associated factor 2 (DCAF2), a conserved substrate receptor for the cullin 4-RING E3 ubiquitin ligase (CRL4) complex, is essential for maintaining genome stability by facilitating ubiquitin-mediated degradation of substrates. Methods To better understand the physiological role of DCAF2 in female reproduction, we conducted a study using mice with conditional knockout (cKO) of DCAF2 in the uterus using the progesterone receptor Cre (Pgr Cre/+) mouse model. Results Our results showed the cKO mice were completely infertile, despite having ovarian function. The cKO mice exhibited severely thin uteri, demonstrating notable defects in both the uterine epithelium and a lack of glands. In addition, there were impaired proliferation and differentiation of epithelial cells in the cKO mice, ultimately resulting in failed implantation. Moreover, through deciphering the uterine transcriptome of cKO mice, we revealed crucial differentially expressed genes associated with steroid signaling. Further experiments have demonstrated cKO mice exhibit elevated uterine PGR signaling and reduced estrogen receptor signaling, although the levels of progesterone and estrogen remained unaltered. These alterations may contribute to defects in epithelium. Discussion Overall, our findings highlight a previously unrecognized but indispensable role for DCAF2 in the development of uterine luminal and glandular epithelium by orchestrating PGR and estrogen receptor responses. Its deficiency in the uterus leads to mouse infertility.
Collapse
Affiliation(s)
- Man Yang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Kaixuan Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Liang Zhang
- Research Center of Translational Medicine, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongya Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
3
|
Aljubran F, Schumacher K, Graham A, Gunewardena S, Marsh C, Lydic M, Holoch K, Nothnick WB. Uterine cyclin A2-deficient mice as a model of female early pregnancy loss. J Clin Invest 2024; 134:e163796. [PMID: 39264721 PMCID: PMC11563677 DOI: 10.1172/jci163796] [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: 06/21/2023] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
Proper action of the female sex steroids 17β-estradiol (E2) and progesterone (P4) on the endometrium is essential for fertility. Beyond its role in regulating the cell cycle, cyclin A2 (CCNA2) also mediates E2 and P4 signaling in vitro, but a potential role in modulating steroid action for proper endometrial tissue development and function is unknown. To fill this gap in our knowledge, we examined human endometrial tissue from fertile and infertile cisgender women for CCNA2 expression and correlated this with pregnancy outcome. Functional assessment of CCNA2 was validated in vivo using a conditional Ccna2 uterine-deficient mouse model, while in vitro function was assessed using human cell culture models. We found that CCNA2 expression was significantly reduced in endometrial tissue, specifically the stromal cells, from women undergoing in vitro fertilization who failed to achieve pregnancy. Conditional deletion of Ccna2 from mouse uterine tissue resulted in an inability to achieve pregnancy, which appeared to be due to alterations in the process of decidualization, which was confirmed using in vitro models. From these studies, we conclude that CCNA2 expression during the proliferative/regenerative stage of the menstrual cycle allows for proper steroid responsiveness, decidualization, and pregnancy. When CCNA2 expression levels are insufficient, there is impaired endometrial responsiveness, aberrant decidualization, and loss of pregnancy.
Collapse
Affiliation(s)
| | | | | | | | - Courtney Marsh
- Department of Cell Biology and Physiology
- Department of Obstetrics and Gynecology
- Center for Reproductive Sciences
| | - Michael Lydic
- Department of Obstetrics and Gynecology
- Center for Reproductive Sciences
| | | | - Warren B. Nothnick
- Department of Cell Biology and Physiology
- Department of Obstetrics and Gynecology
- Center for Reproductive Sciences
- Department of Cancer Biology
- Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
4
|
Li MY, Shen HH, Cao XY, Gao XX, Xu FY, Ha SY, Sun JS, Liu SP, Xie F, Li MQ. Targeting a mTOR/autophagy axis: a double-edged sword of rapamycin in spontaneous miscarriage. Biomed Pharmacother 2024; 177:116976. [PMID: 38906022 DOI: 10.1016/j.biopha.2024.116976] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Immune dysfunction is a primary culprit behind spontaneous miscarriage (SM). To address this, immunosuppressive agents have emerged as a novel class of tocolytic drugs, modulating the maternal immune system's tolerance towards the embryo. Rapamycin (PubChem CID:5284616), a dual-purpose compound, functions as an immunosuppressive agent and triggers autophagy by targeting the mTOR pathway. Its efficacy in treating SM has garnered significant research interest in recent times. Autophagy, the cellular process of self-degradation and recycling, plays a pivotal role in numerous health conditions. Research indicates that autophagy is integral to endometrial decidualization, trophoblast invasion, and the proper functioning of decidual immune cells during a healthy pregnancy. Yet, in cases of SM, there is a dysregulation of the mTOR/autophagy axis in decidual stromal cells or immune cells at the maternal-fetal interface. Both in vitro and in vivo studies have highlighted the potential benefits of low-dose rapamycin in managing SM. However, given mTOR's critical role in energy metabolism, inhibiting it could potentially harm the pregnancy. Moreover, while low-dose rapamycin has been deemed safe for treating recurrent implant failure, its potential teratogenic effects remain uncertain due to insufficient data. In summary, rapamycin represents a double-edged sword in the treatment of SM, balancing its impact on autophagy and immune regulation. Further investigation is warranted to fully understand its implications.
Collapse
Affiliation(s)
- Meng-Ying Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Hui-Hui Shen
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Yan Cao
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Xiao Gao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China
| | - Feng-Yuan Xu
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Si-Yao Ha
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510235, China
| | - Jian-Song Sun
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, People's Republic of China
| | - Song-Ping Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China.
| | - Feng Xie
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China.
| | - Ming-Qing Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Department of Gynecologic Endocrinology and Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, People's Republic of China.
| |
Collapse
|
5
|
Huang Y, Wang Z, Li B, Ke L, Xiong Y, Zhang Y. Loss of KLF15 impairs endometrial receptivity by inhibiting EMT in endometriosis. J Endocrinol 2024; 261:e230319. [PMID: 38513352 PMCID: PMC11056958 DOI: 10.1530/joe-23-0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 03/20/2024] [Indexed: 03/23/2024]
Abstract
The impaired endometrial receptivity is a major factor contributing to infertility in patients with endometriosis (EM), but the underlying mechanism remains unclear. Our study aimed to investigate the role of Kruppel-like factor 15 (KLF15) in endometrial receptivity and its regulation in EM. We observed a significant decrease in KLF15 expression in the mid-secretory epithelial endometrial cells of EM patients compared to normal females without EM. To confirm the role of KLF15 in endometrial receptivity, we found a significantly reduced KLF15 expression and a significant decrease in embryo implantation number in the rat model via uterine horn infection with siRNA. This highlights the importance of KLF15 as a regulator receptivity. Furthermore, through ChIP-qPCR, we discovered that the progesterone receptor (PR) directly binds to KLF15 promoter regions, indicating that progesterone resistance may mediate the decrease in KLF15 expression in EM patients. Additionally, we found that the mid-secretory endometrium of EM patients exhibited impaired epithelial-mesenchymal transition (EMT). Knockdown of KLF15 upregulated E-cadherin and downregulated vimentin expression, leading to inhibited invasiveness and migration of Ishikawa cells. Overexpression KLF15 promotes EMT, invasiveness, and migration ability, and increases the attachment rate of JAR cells to Ishikawa cells. Through RNA-seq analysis, we identified TWIST2 as a downstream gene of KLF15. We confirmed that KLF15 directly binds to the promoter region of TWIST2 via ChIP-qPCR, promoting epithelial cell EMT during the establishment of endometrial receptivity. Our study reveals the involvement of KLF15 in the regulation of endometrial receptivity and its downstream effects on EMT. These findings provide valuable insights into potential therapeutic approaches for treating non-receptive endometrium in patients with EM.
Collapse
Affiliation(s)
- Yaxiong Huang
- Department of Reproductive Medicine center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, PR China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan Hubei Province, PR China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan Hubei Province, PR China
- Department of Gynaecology and Obstetrics, Sinopharm Dongfeng Hospital, Hubei University of Medicine, Shiyan, Hubei Province, PR China
| | - Zihan Wang
- Department of Reproductive Medicine center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, PR China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan Hubei Province, PR China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan Hubei Province, PR China
| | - Bin Li
- Department of Gynaecology and Obstetrics, Sinopharm Dongfeng Hospital, Hubei University of Medicine, Shiyan, Hubei Province, PR China
| | - Lina Ke
- Department of Gynaecology and Obstetrics, Sinopharm Dongfeng Hospital, Hubei University of Medicine, Shiyan, Hubei Province, PR China
| | - Yao Xiong
- Department of Reproductive Medicine center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, PR China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan Hubei Province, PR China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan Hubei Province, PR China
| | - Yuanzhen Zhang
- Department of Reproductive Medicine center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, PR China
- Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan Hubei Province, PR China
- Wuhan Clinical Research Center for Reproductive Science and Birth Health, Wuhan Hubei Province, PR China
| |
Collapse
|
6
|
Hogan MP, Holding ML, Nystrom GS, Colston TJ, Bartlett DA, Mason AJ, Ellsworth SA, Rautsaw RM, Lawrence KC, Strickland JL, He B, Fraser P, Margres MJ, Gilbert DM, Gibbs HL, Parkinson CL, Rokyta DR. The genetic regulatory architecture and epigenomic basis for age-related changes in rattlesnake venom. Proc Natl Acad Sci U S A 2024; 121:e2313440121. [PMID: 38578985 PMCID: PMC11032440 DOI: 10.1073/pnas.2313440121] [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: 08/08/2023] [Accepted: 03/13/2024] [Indexed: 04/07/2024] Open
Abstract
Developmental phenotypic changes can evolve under selection imposed by age- and size-related ecological differences. Many of these changes occur through programmed alterations to gene expression patterns, but the molecular mechanisms and gene-regulatory networks underlying these adaptive changes remain poorly understood. Many venomous snakes, including the eastern diamondback rattlesnake (Crotalus adamanteus), undergo correlated changes in diet and venom expression as snakes grow larger with age, providing models for identifying mechanisms of timed expression changes that underlie adaptive life history traits. By combining a highly contiguous, chromosome-level genome assembly with measures of expression, chromatin accessibility, and histone modifications, we identified cis-regulatory elements and trans-regulatory factors controlling venom ontogeny in the venom glands of C. adamanteus. Ontogenetic expression changes were significantly correlated with epigenomic changes within genes, immediately adjacent to genes (e.g., promoters), and more distant from genes (e.g., enhancers). We identified 37 candidate transcription factors (TFs), with the vast majority being up-regulated in adults. The ontogenetic change is largely driven by an increase in the expression of TFs associated with growth signaling, transcriptional activation, and circadian rhythm/biological timing systems in adults with corresponding epigenomic changes near the differentially expressed venom genes. However, both expression activation and repression contributed to the composition of both adult and juvenile venoms, demonstrating the complexity and potential evolvability of gene regulation for this trait. Overall, given that age-based trait variation is common across the tree of life, we provide a framework for understanding gene-regulatory-network-driven life-history evolution more broadly.
Collapse
Affiliation(s)
- Michael P. Hogan
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Matthew L. Holding
- Department of Biological Science, Florida State University, Tallahassee, FL32306
- Life Sciences Institute, University of Michigan, Ann Arbor, MI48109
| | - Gunnar S. Nystrom
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Timothy J. Colston
- Department of Biological Science, Florida State University, Tallahassee, FL32306
- Department of Biology, University of Puerto Rico at Mayagüez, Mayagüez, PR00681
| | - Daniel A. Bartlett
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Andrew J. Mason
- Department of Biological Sciences, Clemson University, Clemson, SC29634
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, Columbus, OH43210
| | - Schyler A. Ellsworth
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Rhett M. Rautsaw
- Department of Biological Sciences, Clemson University, Clemson, SC29634
- Department of Integrative Biology, University of South Florida, Tampa, FL33620
- School of Biological Sciences, Washington State University, Pullman, WA99164
| | - Kylie C. Lawrence
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Jason L. Strickland
- Department of Biological Sciences, Clemson University, Clemson, SC29634
- Department of Biology, University of South Alabama, Mobile, AL36688
| | - Bing He
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Peter Fraser
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| | - Mark J. Margres
- Department of Integrative Biology, University of South Florida, Tampa, FL33620
| | - David M. Gilbert
- Laboratory of Chromosome Replication and Epigenome Regulation, San Diego Biomedical Research Institute, San Diego, CA92121
| | - H. Lisle Gibbs
- Department of Evolution, Ecology and Organismal Biology, The Ohio State University, Columbus, OH43210
| | - Christopher L. Parkinson
- Department of Biological Sciences, Clemson University, Clemson, SC29634
- Department of Forestry and Environmental Conservation, Clemson University, Clemson, SC29634
| | - Darin R. Rokyta
- Department of Biological Science, Florida State University, Tallahassee, FL32306
| |
Collapse
|
7
|
Chadchan SB, Popli P, Liao Z, Andreas E, Dias M, Wang T, Gunderson SJ, Jimenez PT, Lanza DG, Lanz RB, Foulds CE, Monsivais D, DeMayo FJ, Yalamanchili HK, Jungheim ES, Heaney JD, Lydon JP, Moley KH, O'Malley BW, Kommagani R. A GREB1-steroid receptor feedforward mechanism governs differential GREB1 action in endometrial function and endometriosis. Nat Commun 2024; 15:1947. [PMID: 38431630 PMCID: PMC10908778 DOI: 10.1038/s41467-024-46180-4] [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: 12/01/2022] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Cellular responses to the steroid hormones, estrogen (E2), and progesterone (P4) are governed by their cognate receptor's transcriptional output. However, the feed-forward mechanisms that shape cell-type-specific transcriptional fulcrums for steroid receptors are unidentified. Herein, we found that a common feed-forward mechanism between GREB1 and steroid receptors regulates the differential effect of GREB1 on steroid hormones in a physiological or pathological context. In physiological (receptive) endometrium, GREB1 controls P4-responses in uterine stroma, affecting endometrial receptivity and decidualization, while not affecting E2-mediated epithelial proliferation. Of mechanism, progesterone-induced GREB1 physically interacts with the progesterone receptor, acting as a cofactor in a positive feedback mechanism to regulate P4-responsive genes. Conversely, in endometrial pathology (endometriosis), E2-induced GREB1 modulates E2-dependent gene expression to promote the growth of endometriotic lesions in mice. This differential action of GREB1 exerted by a common feed-forward mechanism with steroid receptors advances our understanding of mechanisms that underlie cell- and tissue-specific steroid hormone actions.
Collapse
Affiliation(s)
- Sangappa B Chadchan
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Pooja Popli
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Zian Liao
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Eryk Andreas
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michelle Dias
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Tianyuan Wang
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Stephanie J Gunderson
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Patricia T Jimenez
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Denise G Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Rainer B Lanz
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Charles E Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Hari Krishna Yalamanchili
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Emily S Jungheim
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Obstetrics and Gynecology, Fienberg School of Medicine, Chicago, IL, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Kelle H Moley
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Ramakrishna Kommagani
- Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Pavličev M, McDonough-Goldstein CE, Zupan AM, Muglia L, Hu YC, Kong F, Monangi N, Dagdas G, Zupančič N, Maziarz J, Sinner D, Zhang G, Wagner G, Muglia L. A common allele increases endometrial Wnt4 expression, with antagonistic implications for pregnancy, reproductive cancers, and endometriosis. Nat Commun 2024; 15:1152. [PMID: 38346980 PMCID: PMC10861470 DOI: 10.1038/s41467-024-45338-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/20/2024] [Indexed: 02/15/2024] Open
Abstract
The common human SNP rs3820282 is associated with multiple phenotypes including gestational length and likelihood of endometriosis and cancer, presenting a paradigmatic pleiotropic variant. Deleterious pleiotropic mutations cause the co-occurrence of disorders either within individuals, or across population. When adverse and advantageous effects are combined, pleiotropy can maintain high population frequencies of deleterious alleles. To reveal the causal molecular mechanisms of this pleiotropic SNP, we introduced this substitution into the mouse genome by CRISPR/Cas 9. Previous work showed that rs3820282 introduces a high-affinity estrogen receptor alpha-binding site at the Wnt4 locus. Here, we show that this mutation upregulates Wnt4 transcription in endometrial stroma, following the preovulatory estrogen peak. Effects on uterine transcription include downregulation of epithelial proliferation and induction of progesterone-regulated pro-implantation genes. We propose that these changes increase uterine permissiveness to embryo invasion, whereas they decrease resistance to invasion by cancer and endometriotic foci in other estrogen-responsive tissues.
Collapse
Affiliation(s)
- Mihaela Pavličev
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria.
- Complexity Science Hub, Vienna, Austria.
| | | | | | - Lisa Muglia
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yueh-Chiang Hu
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fansheng Kong
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nagendra Monangi
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Gülay Dagdas
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
| | - Nina Zupančič
- University Medical Center Ljubljana, Department of Cardiovascular Surgery, Ljubljana, Slovenia
| | - Jamie Maziarz
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Debora Sinner
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ge Zhang
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Günter Wagner
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Yale Systems Biology Institute, Yale University, West Haven, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, USA
| | - Louis Muglia
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Burroughs Wellcome Fund, Research Triangle Park, NC, Durham, USA
| |
Collapse
|
9
|
Chen H, Li LL, Du Y. Krüppel-like factor 15 in liver diseases: Insights into metabolic reprogramming. Front Pharmacol 2023; 14:1115226. [PMID: 36937859 PMCID: PMC10017497 DOI: 10.3389/fphar.2023.1115226] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Liver diseases, characterized by metabolic disorder, have become a global public health problem with high morbidity and mortality. Krüppel-like factor 15 (KLF15) is a zinc-finger transcription factor mainly enriched in liver. Increasing evidence suggests that hepatic KLF15 is activated rapidly during fasting, and contributes to the regulation of gluconeogenesis, lipid, amino acid catabolism, bile acids, endobiotic and xenobiotic metabolism. This review summarizes the latest advances of KLF15 in metabolic reprogramming, and explore the function of KLF15 in acute liver injury, hepatitis B virus, and autoimmune hepatitis. which aims to evaluate the potential of KLF15 as a therapeutic target and prognostic biomarker for liver diseases.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Lan-Lan Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Yan Du
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
- *Correspondence: Yan Du,
| |
Collapse
|
10
|
GCN5 participates in KLF4-VEGFA feedback to promote endometrial angiogenesis. iScience 2022; 25:104509. [PMID: 35733790 PMCID: PMC9207667 DOI: 10.1016/j.isci.2022.104509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 11/22/2022] Open
Abstract
Endometrial angiogenesis is necessary for good endometrial receptivity. Krüppel-like factor 4 (KLF4) is a transcription factor that is essential for regulating angiogenesis. Here we found that vascular endothelial growth factor A (VEGFA) can form a positive feedback loop with KLF4 to promote the proliferation and migration of human endometrial microvascular endothelial cells (HEMECs) and inhibit cell apoptosis. General control non-derepressible 5 (GCN5) is also time-dependent on VEGFA and participates in the KLF4-VEGFA loop. In addition, we found that GCN5 is a succinyltransferase that modulates the succinylation of histones and nonhistones. GCN5 interacts with KLF4 and is recruited to the KLF4-binding site of the VEGFA promoter to succinylate H3K79, which initiates gene transcription epigenetically. For nonhistones, GCN5 succinylates KLF4 that is activated by ERK signaling in HEMECs treated with VEGFA to increase its transcription activity. These results demonstrate KLF4-VEGFA positive feedback loop is regulated by epigenetics, which contributes to endometrial angiogenesis. KLF4 mediates VEGFA-induced endometrial angiogenesis VEGFA increases the interaction between KLF4 and GCN5 VEGFA promotes H3K79 succinylation by upregulating KLF4 and GCN5 VEGFA succinylates KLF4 and promotes interaction of KLF4 and GCN5 via ERK pathway
Collapse
|
11
|
Hashimoto K, Kodama A, Ohira M, Kimoto M, Nakagawa R, Usui Y, Ujihara Y, Hanashima A, Mohri S. Postnatal expression of cell cycle promoter Fam64a causes heart dysfunction by inhibiting cardiomyocyte differentiation through repression of Klf15. iScience 2022; 25:104337. [PMID: 35602953 PMCID: PMC9118685 DOI: 10.1016/j.isci.2022.104337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/07/2022] [Accepted: 04/26/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction of fetal cell cycle genes into damaged adult hearts has emerged as a promising strategy for stimulating proliferation and regeneration of postmitotic adult cardiomyocytes. We have recently identified Fam64a as a fetal-specific cell cycle promoter in cardiomyocytes. Here, we analyzed transgenic mice maintaining cardiomyocyte-specific postnatal expression of Fam64a when endogenous expression was abolished. Despite an enhancement of cardiomyocyte proliferation, these mice showed impaired cardiomyocyte differentiation during postnatal development, resulting in cardiac dysfunction in later life. Mechanistically, Fam64a inhibited cardiomyocyte differentiation by repressing Klf15, leading to the accumulation of undifferentiated cardiomyocytes. In contrast, introduction of Fam64a in differentiated adult wildtype hearts improved functional recovery upon injury with augmented cell cycle and no dedifferentiation in cardiomyocytes. These data demonstrate that Fam64a inhibits cardiomyocyte differentiation during early development, but does not induce de-differentiation in once differentiated cardiomyocytes, illustrating a promising potential of Fam64a as a cell cycle promoter to attain heart regeneration. Overexpression of cell cycle promoter Fam64a in cardiomyocytes causes heart failure Fam64a inhibits cardiomyocyte differentiation during development by repressing Klf15 Transient and local induction of Fam64a in adult hearts improves recovery upon injury Fam64a activates cardiomyocyte cell cycle without dedifferentiation upon injury
Collapse
Affiliation(s)
- Ken Hashimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Aya Kodama
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Momoko Ohira
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Misaki Kimoto
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Reiko Nakagawa
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe 650-0047, Japan
| | - Yuu Usui
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Yoshihiro Ujihara
- Department of Electrical and Mechanical Engineering, Nagoya Institute of Technology, Nagoya 466-8555, Japan
| | - Akira Hanashima
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| | - Satoshi Mohri
- First Department of Physiology, Kawasaki Medical School, Kurashiki 701-0192, Japan
| |
Collapse
|
12
|
Expression and Prognosis Value of the KLF Family Members in Colorectal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6571272. [PMID: 35345512 PMCID: PMC8957442 DOI: 10.1155/2022/6571272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 12/17/2022]
Abstract
Krüppel-like factors (KLFs) are some kind of transcriptional regulator that regulates a broad range of cellular functions and has been linked to the development of certain malignancies. KLF expression patterns and prognostic values in colorectal cancer (CRC) have, however, been investigated rarely. To investigate the differential expression, predictive value, and gene mutations of KLFs in CRC patients, we used various online analytic tools, including ONCOMINE, TCGA, cBioPortal, and the TIMER database. KLF2-6, KLF8-10, KLF12-15, and KLF17 mRNA expression levels were dramatically downregulated in CRC tissues, but KLF1, KLF7, and KLF16 mRNA expression levels were significantly elevated in CRC tissues. According to the findings of Cox regression analysis, upregulation of KLF3, KLF5, and KLF6 and downregulation of KLF15 were linked with a better prognosis in CRC. For functional enrichment, our findings revealed that KLF members are involved in a variety of cancer-related biological processes. In colon cancer and rectal cancer, KLFs were also shown to be associated with a variety of immune cells. The findings of this research reveal that KLF family members' mRNA expression levels are possible prognostic indicators for patients with CRC.
Collapse
|
13
|
Yang D, Ran Y, Li X, Jiang X, Chen J, Sun J, Tian L, Teerds K, Bai W. Cyanidin-3-O-glucoside ameliorates cadmium induced uterine epithelium proliferation in mice. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:127571. [PMID: 34986559 DOI: 10.1016/j.jhazmat.2021.127571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 10/09/2021] [Accepted: 10/19/2021] [Indexed: 06/14/2023]
Abstract
Cadmium (Cd) is an environmental pollutant and endocrine disrupter, abundantly present in water, food, and soil. Accumulation of Cd in the body can negatively affect female reproduction; especially the uterus is exceptionally sensitive to the toxic actions of Cd. The anthocyanin cyanidin-3-O-glucoside (C3G) is a naturally occurring phenolic compound in fruits and plants that can antagonize the toxic effects of Cd. This capacity makes C3G a possible candidate to prevent Cd-induced female infertility. The present study aimed to investigate: 1) whether C3G intake could prevent Cd-induced female reproductive toxicity, and 2) the underlying mechanisms responsible for this protective effect. The results of our study indicated that Cd exposure did not affect ovarian function, but induced hypertrophy of the uterine endometrium. Oral intake of C3G markedly reduced the effects of Cd exposure on the thickness of the uterine epithelium cells. Transcriptomic analysis of the endometrium revealed that C3G intake had anti-estrogenic effects, attenuating Cd-induced endometrial epithelial cell proliferation by inhibiting estrogen-responsive genes, enhancing epithelial progesterone receptor expression, and regulating Klf4 expression. The current findings implicate that C3G has the potential to be used as a dietary supplement based on its capacity to intervene in Cd-induced female reproductive toxicity.
Collapse
Affiliation(s)
- Dacheng Yang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, 510632, PR China; Department of Bioengineering, College of life science and technology, Jinan University, Guangzhou, 510632, PR China
| | - Yanhong Ran
- Department of Bioengineering, College of life science and technology, Jinan University, Guangzhou, 510632, PR China
| | - Xusheng Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, 510632, PR China
| | - Xinwei Jiang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, 510632, PR China
| | - Jiali Chen
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, 510632, PR China
| | - Jianxia Sun
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, 510006, PR China
| | - Lingmin Tian
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, 510632, PR China
| | - Katia Teerds
- Department of Animal Sciences, Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
14
|
Yoo JY, Kim TH, Shin JH, Marquardt RM, Müller U, Fazleabas AT, Young SL, Lessey BA, Yoon HG, Jeong JW. Loss of MIG-6 results in endometrial progesterone resistance via ERBB2. Nat Commun 2022; 13:1101. [PMID: 35232969 PMCID: PMC8888616 DOI: 10.1038/s41467-022-28608-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/27/2021] [Indexed: 01/17/2023] Open
Abstract
Female subfertility is highly associated with endometriosis. Endometrial progesterone resistance is suggested as a crucial element in the development of endometrial diseases. We report that MIG-6 is downregulated in the endometrium of infertile women with endometriosis and in a non-human primate model of endometriosis. We find ERBB2 overexpression in the endometrium of uterine-specific Mig-6 knockout mice (Pgrcre/+Mig-6f/f; Mig-6d/d). To investigate the effect of ERBB2 targeting on endometrial progesterone resistance, fertility, and endometriosis, we introduce Erbb2 ablation in Mig-6d/d mice (Mig-6d/dErbb2d/d mice). The additional knockout of Erbb2 rescues all phenotypes seen in Mig-6d/d mice. Transcriptomic analysis shows that genes differentially expressed in Mig-6d/d mice revert to their normal expression in Mig-6d/dErbb2d/d mice. Together, our results demonstrate that ERBB2 overexpression in endometrium with MIG-6 deficiency causes endometrial progesterone resistance and a nonreceptive endometrium in endometriosis-related infertility, and ERBB2 targeting reverses these effects. Female subfertility is highly associated with endometriosis. Here the authors show that progesterone-induced MIG-6 is reduced in endometrium of infertile women and non-human primates with endometriosis, and in a mouse model find that Erbb2 is the key mediator of Mig-6 loss induced endometriosis-related infertility.
Collapse
Affiliation(s)
- Jung-Yoon Yoo
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.,Department of Biomedical Laboratory Science, Yonsei University Mirae Campus, Wonju, South Korea
| | - Tae Hoon Kim
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA
| | - Jung-Ho Shin
- Division of Reproductive Endocrinology, Department of Obstetrics & Gynecology, Guro Hospital, Korea University Medical Center, Seoul, South Korea
| | - Ryan M Marquardt
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| | - Ulrich Müller
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Asgerally T Fazleabas
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA
| | - Steven L Young
- Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, NC, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| | - Jae-Wook Jeong
- Department of Obstetrics,Gynecology & Reproductive Biology, Michigan State University, College of Human Medicine, Grand Rapids, MI, USA.
| |
Collapse
|
15
|
Zhu KY, Tian Y, Li YX, Meng QX, Ge J, Cao XC, Zhang T, Yu Y. The functions and prognostic value of Krüppel-like factors in breast cancer. Cancer Cell Int 2022; 22:23. [PMID: 35033064 PMCID: PMC8760734 DOI: 10.1186/s12935-022-02449-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Krüppel‐like factors (KLFs) are zinc finger proteins which participate in transcriptional gene regulation. Although increasing evidence indicate that KLFs are involved in carcinogenesis and progression, its clinical significance and biological function in breast cancer are still limited. Methods We investigated all the expression of KLFs (KLF1-18) at transcriptional levels by using Oncomine and Gene Expression Profiling Interactive Analysis (GEPIA). The mRNA and protein expression levels of KLFs were also determined by using RT-qPCR and immunohistochemistry, respectively. CBioPortal, GeneMANIA and STRING were used to comprehensive analysis of the molecular characteristics of KLFs. The clinical value of prognostic prediction based on the expression of KLFs was determined by using the KM plotter. The relevant molecular pathways of KLFs were further analyzed by using Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. Finally, we investigated the effect of KLF2 and KLF15 on biological behavior of breast cancer cells in vitro. Results The expression of KLF2/4/6/8/9/11/15 was significantly down-regulated in breast cancer. The patients with high KLF2, KLF4 or KLF15 expression had a better outcome, while patients with high KLF8 or KLF11 had a poor prognosis. Furthermore, our results showed that KLF2 or KLF15 can be used as a prognostic factor independent on the other KLFs in patients with breast cancer. Overexpression of KLF2 or KLF15 inhibited cell proliferation and migration, and blocked cell cycle at G0/G1 phase, resulting in cell apoptosis. Conclusions KLF2 and KLF15 function as tumor suppressors in breast cancer and are potential biomarkers for prognostic prediction in patients with breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02449-6.
Collapse
Affiliation(s)
- Ke-Yun Zhu
- Department of Hepatobiliary Surgery, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yao Tian
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ying-Xi Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Qing-Xiang Meng
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Department of Radiobiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jie Ge
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Xu-Chen Cao
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
| | - Ti Zhang
- Department of Hepatobiliary Surgery, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China. .,Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Yue Yu
- Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
| |
Collapse
|
16
|
Cheng J, Liang J, Li Y, Gao X, Ji M, Liu M, Tian Y, Feng G, Deng W, Wang H, Kong S, Lu Z. Shp2 in uterine stromal cells critically regulates on time embryo implantation and stromal decidualization by multiple pathways during early pregnancy. PLoS Genet 2022; 18:e1010018. [PMID: 35025868 PMCID: PMC8791483 DOI: 10.1371/journal.pgen.1010018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 01/26/2022] [Accepted: 01/05/2022] [Indexed: 11/19/2022] Open
Abstract
Approximately 75% of failed pregnancies are considered to be due to embryo implantation failure or defects. Nevertheless, the explicit signaling mechanisms governing this process have not yet been elucidated. Here, we found that conditional deletion of the Shp2 gene in mouse uterine stromal cells deferred embryo implantation and inhibited the decidualization of stromal cells, which led to embryonic developmental delay and to the death of numerous embryos mid-gestation, ultimately reducing female fertility. The absence of Shp2 in stromal cells increased the proliferation of endometrial epithelial cells, thereby disturbing endometrial epithelial remodeling. However, Shp2 deletion impaired the proliferation and polyploidization of stromal cells, which are distinct characteristics of decidualization. In human endometrial stromal cells (hESCs), Shp2 expression gradually increased during the decidualization process. Knockout of Shp2 blocked the decidual differentiation of hESCs, while Shp2 overexpression had the opposite effect. Shp2 knockout inhibited the proliferation of hESCs during decidualization. Whole gene expression profiling analysis of hESCs during the decidualization process showed that Shp2 deficiency disrupted many signaling transduction pathways and gene expression. Analyses of hESCs and mouse uterine tissues confirmed that the signaling pathways extracellular regulated protein kinases (ERK), protein kinase B (AKT), signal transducer and activator of transcription 3 (STAT3) and their downstream transcription factors CCAAT/enhancer binding protein β (C/EBPβ) and Forkhead box transcription factor O1 (FOXO-1) were involved in the Shp2 regulation of decidualization. In summary, these results demonstrate that Shp2 plays a crucial role in stromal decidualization by mediating and coordinating multiple signaling pathways in uterine stromal cells. Our discovery possibly provides a novel key regulator of embryo implantation and novel therapeutic target for pregnancy failure. Embryo implantation includes the establishment of uterine receptivity, blastocyst attachment, and endometrial decidualization. Disorders of this process usually induce pregnancy failure, resulting in women infertility. But the signaling mechanisms governing this process remain unclear. Here, using gene knockout mouse model and human endometrial stromal cells (hESCs), we identified a novel key regulator of embryo implantation, Shp2, which plays a crucial role in stromal decidualization by mediating and coordinating multiple signaling pathways in uterine stromal cells. Shp2 deficiency in mouse uterine stromal cells inhibited the uterine stromal decidualization, disturbing embryo implantation and embryonic development, ultimately reducing female fertility. The absence of Shp2 in hESCs also blocked the decidual differentiation. Our findings not only promote the understanding of peri-implantation biology, but may also provide a critical target for more effectively diagnose and/or treat women with recurrent implantation failure or early pregnancy loss.
Collapse
Affiliation(s)
- Jianghong Cheng
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Jia Liang
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Yingzhe Li
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Xia Gao
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Mengjun Ji
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Mengying Liu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Yingpu Tian
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
| | - Gensheng Feng
- Department of Pathology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Wenbo Deng
- Reproductive Medical Centre, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
| | - Haibin Wang
- Reproductive Medical Centre, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
- * E-mail: (HW); (SK); (ZL)
| | - Shuangbo Kong
- Reproductive Medical Centre, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
- * E-mail: (HW); (SK); (ZL)
| | - Zhongxian Lu
- School of Pharmaceutical Sciences, State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, China
- * E-mail: (HW); (SK); (ZL)
| |
Collapse
|
17
|
Qu X, Liu X, Zhang Y, Shi Z, Wang X. Depletion of Kruppel-like factor 15 sensitized gliomas to temozolomide cytotoxicity through O 6-methylguanine-DNA methyl-transferase. Biochem Biophys Rep 2021; 27:101058. [PMID: 34222684 PMCID: PMC8242961 DOI: 10.1016/j.bbrep.2021.101058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 11/29/2022] Open
Abstract
Temozolomide (TMZ)-based chemotherapy is a standard strategy for gliomas, although chemoresistance remains a major therapeutic challenge. The chemical mechanism by which TMZ induces cell death is DNA methylation, leading to double-stranded breaks (DSBs) and thus to apoptosis. However, TMZ-induced N6-meG sites are efficiently repaired and mediated by the DNA repair protein O 6-methylguanine-DNA methyl-transferase (MGMT), leading to TMZ resistance. KLF15, a member of the Kruppel-like factors family, mainly functions as transcription factor and potential suppressor gene by inhibiting proliferation, migration, and inducing apoptosis. However, the roles and regulatory mechanisms of KLF15 in glioma tumorigenesis and chemoresistance are poorly understood. In this study, KLF15 expression was upregulated in glioma tissues and cell lines upon TMZ treatment. Knockdown of KLF15 amplified TMZ-induced repression of cell proliferation, while KLF15 overexpression reversed this process. Mechanistically, KLF15 functioned as a transcriptional activator of MGMT. Moreover, KLF15 knockdown sensitized tumors to TMZ treatment in vivo. Taken together, these results suggested that KLF15 up-regulated MGMT through direct binding to the promoter of MGMT, which plays an important role in glioma resistance to TMZ, and which may be a potential target for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Xinjuan Qu
- Department of Neurosurgery, Linyi Hospital of Traditional Chinese Medicine, Shandong, 276002, China
| | - Xuelai Liu
- Department of Neurosurgery, Linyi Hospital of Traditional Chinese Medicine, Shandong, 276002, China
| | - Yumei Zhang
- Department of Neurology, Linyi Hospital of Traditional Chinese Medicine, Shandong, 276002, China
| | - Zhan Shi
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing, 210002, China
| | - Xiaohua Wang
- Department of General Internal Medicine, Linyi People's Hospital, Shandong, 276003, China
| |
Collapse
|
18
|
Oestreich AK, Chadchan SB, Medvedeva A, Lydon JP, Jungheim ES, Moley KH, Kommagani R. The autophagy protein, FIP200 (RB1CC1) mediates progesterone responses governing uterine receptivity and decidualization†. Biol Reprod 2021; 102:843-851. [PMID: 31901086 DOI: 10.1093/biolre/ioz234] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/08/2019] [Accepted: 12/27/2019] [Indexed: 12/16/2022] Open
Abstract
Successful establishment of pregnancy depends on steroid hormone-driven cellular changes in the uterus during the peri-implantation period. To become receptive to embryo implantation, uterine endometrial stromal cells (ESCs) must transdifferentiate into decidual cells that secrete factors necessary for embryo survival and trophoblast invasion. Autophagy is a key homeostatic process vital for cellular homeostasis. Although the uterus undergoes major cellular changes during early pregnancy, the precise role of autophagy in uterine function is unknown. Here, we report that conditional knockout of the autophagy protein FIP200 in the reproductive tract of female mice results in reduced fecundity due to an implantation defect. In the absence of FIP200, aberrant progesterone signaling results in sustained uterine epithelial proliferation and failure of stromal cells to decidualize. Additionally, loss of FIP200 impairs decidualization of human ESCs. We conclude that the autophagy protein FIP200 plays a crucial role in uterine receptivity, decidualization, and fertility. These data establish autophagy as a major cellular pathway required for uterine receptivity and decidualization in both mice and human ESCs.
Collapse
Affiliation(s)
- Arin K Oestreich
- Department Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA and
| | - Sangappa B Chadchan
- Department Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA and
| | - Alexandra Medvedeva
- Department Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA and
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Emily S Jungheim
- Department Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA and
| | - Kelle H Moley
- Department Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA and
| | - Ramakrishna Kommagani
- Department Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of Medicine, St. Louis, MO, 63110, USA and
| |
Collapse
|
19
|
Sun CX, Liu BJ, Su Y, Shi GW, Wang Y, Chi JF. MiR-181a promotes cell proliferation and migration through targeting KLF15 in papillary thyroid cancer. Clin Transl Oncol 2021; 24:66-75. [PMID: 34312797 DOI: 10.1007/s12094-021-02670-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/10/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Papillary thyroid cancer (PTC) is the predominant histological type of thyroid cancer, accounting for 80% of thyroid cancers. MiR-181a is a novel microRNA that is usually upregulated in multiple cancers. This study aims to explore the role and underlying mechanism of miR-181a in PTC. METHODS CCK8 and Transwell assays were performed to evaluate cell viability and migration. The mRNA level of miR-181a and KLF15 was calculated by qRT-PCR. The protein level of E-Cadherin, N-Cadherin and GAPDH was evaluated by western blot. Dual luciferase assay was conducted to validate that miR-181a directly targeting the 3'-UTR of KLF15 mRNA in TPC-1 cells. RESULTS We observed that miR-181a was overexpressed and KLF15 was low expressed in PTC tissues and cell lines. Upregulation of miR-181a or downregulation of KLF15 predicted poor outcomes in PTC patients. MiR-181a improved cell growth of PTC, migration and epithelial-mesenchymal transition (EMT) in TPC-1 cells. KLF15 was a target gene of miR-181a and its expression was mediated by miR-181a. KLF15 partially reversed the facilitating effect of miR-181a on cell proliferation and migration in TPC-1 cells. CONCLUSION We discovered that miR-181a served as an oncogene downregulating KLF15, thereby inhibiting cell proliferation, migration and the EMT. These findings demonstrate that miR-181a plays a significant role in PTC progression and could be a therapeutic target for PTC.
Collapse
Affiliation(s)
- C X Sun
- Department of Endocrinology, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - B J Liu
- Operation Room, Rizhao Hospital of TCM, Rizhao, 276800, Shandong, China
| | - Y Su
- Operation Room, Qingdao Hospital of Traditional Chinese Medicine, Qingdao Hiser Hospital, Qingdao, 266033, Shandong, China
| | - G W Shi
- Health Management Center, Zhangqiu District People's Hospital, Jinan, 250200, Shandong, China
| | - Y Wang
- Health Management Center, Zhangqiu District People's Hospital, Jinan, 250200, Shandong, China
| | - J F Chi
- Department of Endocrinology, Jinan Central Hospital, 105 Jiefang Road, Lixia District, Jinan, 250013, Shandong, China.
| |
Collapse
|
20
|
Endometrial receptivity and implantation require uterine BMP signaling through an ACVR2A-SMAD1/SMAD5 axis. Nat Commun 2021; 12:3386. [PMID: 34099644 PMCID: PMC8184938 DOI: 10.1038/s41467-021-23571-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
During early pregnancy in the mouse, nidatory estrogen (E2) stimulates endometrial receptivity by activating a network of signaling pathways that is not yet fully characterized. Here, we report that bone morphogenetic proteins (BMPs) control endometrial receptivity via a conserved activin receptor type 2 A (ACVR2A) and SMAD1/5 signaling pathway. Mice were generated to contain single or double conditional deletion of SMAD1/5 and ACVR2A/ACVR2B receptors using progesterone receptor (PR)-cre. Female mice with SMAD1/5 deletion display endometrial defects that result in the development of cystic endometrial glands, a hyperproliferative endometrial epithelium during the window of implantation, and impaired apicobasal transformation that prevents embryo implantation and leads to infertility. Analysis of Acvr2a-PRcre and Acvr2b-PRcre pregnant mice determined that BMP signaling occurs via ACVR2A and that ACVR2B is dispensable during embryo implantation. Therefore, BMPs signal through a conserved endometrial ACVR2A/SMAD1/5 pathway that promotes endometrial receptivity during embryo implantation. Building on the known role of BMP signalling in implantation, the authors define the role of uterine ACVR2A and ALK3 (via SMAD1/5) in vivo in regulating murine endometrial receptivity and embryo implantation.
Collapse
|
21
|
Cai Z, Yang C, Liao J, Song H, Zhang S. Sex-biased genes and metabolites explain morphologically sexual dimorphism and reproductive costs in Salix paraplesia catkins. HORTICULTURE RESEARCH 2021; 8:125. [PMID: 34059667 PMCID: PMC8166972 DOI: 10.1038/s41438-021-00566-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 05/10/2023]
Abstract
Dioecious species evolved from species with monomorphic sex systems in order to achieve overall fitness gains by separating male and female functions. As reproductive organs, unisexual flowers have different reproductive roles and exhibit conspicuous sexual dimorphism. To date, little is known about the temporal variations in and molecular mechanisms underlying the morphology and reproductive costs of dioecious flowers. We investigated male and female flowers of Salix paraplesia in three flowering stages before pollination (the early, blooming and late stages) via transcriptional sequencing as well as metabolite content and phenotypic analysis. We found that a large number of sex-biased genes, rather than sex-limited genes, were responsible for sexual dimorphism in S. paraplesia flowers and that the variation in gene expression in male flowers intensified this situation throughout flower development. The temporal dynamics of sex-biased genes derived from changes in reproductive function during the different flowering stages. Sexually differentiated metabolites related to respiration and flavonoid biosynthesis exhibited the same bias directions as the sex-biased genes. These sex-biased genes were involved mainly in signal transduction, photosynthesis, respiration, cell proliferation, phytochrome biosynthesis, and phenol metabolism; therefore, they resulted in more biomass accumulation and higher energy consumption in male catkins. Our results indicated that sex-biased gene expression in S. paraplesia flowers is associated with different reproductive investments in unisexual flowers; male flowers require a greater reproductive investment to meet their higher biomass accumulation and energy consumption needs.
Collapse
Affiliation(s)
- Zeyu Cai
- Key Laboratory of Mountain Surface Processes and Ecological Regulation, Institute of Mountain Hazards and Environment, Chinese Academy of Sciences, Chengdu, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Congcong Yang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jun Liao
- College of Geography and Tourism, Chongqing Normal University, Chongqing, China
| | - Haifeng Song
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Sheng Zhang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
McAvey B, Kuokkanen S, Zhu L, Pollard JW. The selective progesterone receptor modulator, telapristone acetate, is a mixed antagonist/agonist in the human and mouse endometrium and inhibits pregnancy in mice. F&S SCIENCE 2021; 2:59-70. [PMID: 35559765 DOI: 10.1016/j.xfss.2021.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/30/2020] [Accepted: 01/08/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To investigate the effect of the selective progesterone receptor modulator, telapristone acetate (CDB-4124), on endometrial biology and reproductive outcomes. Ovariectomized and hormone-treated CD1 female mice, CD1 female mice with xenotransplants of reconstructed human endometrial tissue, mated wildtype female mice, and cultured human endometrial stromal cells (hESCs) were treated with CDB-4124, followed by the assessment of endometrial cell deoxyribonucleic acid (DNA) proliferation, stromal decidual response, and embryo implantation. DESIGN Experimental study. SETTING Academic research laboratory. PATIENTS Healthy volunteer women from the community were recruited for endometrial biopsies. ANIMALS CD1 out-bred mice (Charles River Laboratories) and nude mice, NU/J (Jackson Laboratories, Bar Harbor, ME). INTERVENTION Treatment of mice and hESCs with CDB-4124. MAIN OUTCOME MEASURE The effect of CDB-4124 on endometrial cell morphology and DNA synthesis, decidual response, and mouse embryo implantation. RESULTS CDB-4124 inhibited estradiol-induced epithelial DNA synthesis in the mouse uterus and xenotransplanted human endometrium. This antiproliferative effect was less than that of progesterone (P4) and was observed when CDB-4124 was administered alone or concomitantly with P4. In the uterine epithelium, CDB-4124 acted as a P4 agonist and partial antagonist. In contrast, CDB-4124 acted as a complete P4 antagonist in the uterine stroma, where it blocked P4's action to induce a decidual response in the pseudopregnant mouse uterus and wildtype mouse uterus after copulation. In mated female mice, CDB-4124 impaired embryo implantation. Similarly, CDB-4124 inhibited the morphological and biochemical transformations of hESCs to decidual cells in vitro. CONCLUSION CDB-4124 exerts mixed P4 antagonistic/agonistic effects in the human and mouse endometrium, which result in failed embryo implantation because of the absence of stromal decidualization.
Collapse
Affiliation(s)
- Beth McAvey
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York; Ichan School of Medicine, RMA, New York
| | - Satu Kuokkanen
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York; NYU Langone Reproductive Specialists of NY, NYU Langone School of Medicine, NYU Langone Long Island School of Medicine, Mineola, New York
| | - Liyin Zhu
- Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey W Pollard
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York; Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburg, Edinburgh, United Kingdom.
| |
Collapse
|
23
|
Shi X, Zheng Y, Jiang L, Zhou B, Yang W, Li L, Ding L, Huang M, Gery S, Lin DC, Koeffler HP. EWS-FLI1 regulates and cooperates with core regulatory circuitry in Ewing sarcoma. Nucleic Acids Res 2020; 48:11434-11451. [PMID: 33080033 PMCID: PMC7672457 DOI: 10.1093/nar/gkaa901] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/22/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022] Open
Abstract
Core regulatory circuitry (CRC)-dependent transcriptional network is critical for developmental tumors in children and adolescents carrying few gene mutations. However, whether and how CRC contributes to transcription regulation in Ewing sarcoma is unknown. Here, we identify and functionally validate a CRC 'trio' constituted by three transcription factors (TFs): KLF15, TCF4 and NKX2-2, in Ewing sarcoma cells. Epigenomic analyses demonstrate that EWS-FLI1, the primary fusion driver for this cancer, directly establishes super-enhancers of each of these three TFs to activate their transcription. In turn, KLF15, TCF4 and NKX2-2 co-bind to their own and each other's super-enhancers and promoters, forming an inter-connected auto-regulatory loop. Functionally, CRC factors contribute significantly to cell proliferation of Ewing sarcoma both in vitro and in vivo. Mechanistically, CRC factors exhibit prominent capacity of co-regulating the epigenome in cooperation with EWS-FLI1, occupying 77.2% of promoters and 55.6% of enhancers genome-wide. Downstream, CRC TFs coordinately regulate gene expression networks in Ewing sarcoma, controlling important signaling pathways for cancer, such as lipid metabolism pathway, PI3K/AKT and MAPK signaling pathways. Together, molecular characterization of the oncogenic CRC model advances our understanding of the biology of Ewing sarcoma. Moreover, CRC-downstream genes and signaling pathways may contain potential therapeutic targets for this malignancy.
Collapse
Affiliation(s)
- Xianping Shi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; Sino-French Hoffmann institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 510120, P.R. China
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yueyuan Zheng
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Liling Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; Sino-French Hoffmann institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 510120, P.R. China
| | - Bo Zhou
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wei Yang
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Liyan Li
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lingwen Ding
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117600, Singapore
| | - Moli Huang
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, P.R. China
| | - Sigal Gery
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - De-Chen Lin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - H Phillip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Hospital Singapore, Singapore 119074, Singapore
| |
Collapse
|
24
|
Alshamrani AA. Roles of microRNAs in Ovarian Cancer Tumorigenesis: Two Decades Later, What Have We Learned? Front Oncol 2020; 10:1084. [PMID: 32850313 PMCID: PMC7396563 DOI: 10.3389/fonc.2020.01084] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/29/2020] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is one of the top gynecological malignancies that cause deaths among females in the United States. At the molecular level, significant progress has been made in our understanding of ovarian cancer development and progression. MicroRNAs (miRNAs) are short, single-stranded, highly conserved non-coding RNA molecules (19–25 nucleotides) that negatively regulate target genes post-transcriptionally. Over the last two decades, mounting evidence has demonstrated the aberrant expression of miRNAs in different human malignancies, including ovarian carcinomas. Deregulated miRNAs can have profound impacts on various cancer hallmarks by repressing tumor suppressor genes. This review will discuss up-to-date knowledge of how the aberrant expression of miRNAs and their targeted genes drives ovarian cancer initiation, proliferation, survival, and resistance to chemotherapies. Understanding the mechanisms by which these miRNAs affect these hallmarks should allow the development of novel therapeutic strategies to treat these lethal malignancies.
Collapse
Affiliation(s)
- Ali A Alshamrani
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
25
|
Cheng L, Shi L, Dai H. Bioinformatics analysis of potential prognostic biomarkers among Krüppel-like transcription Factors (KLFs) in breast cancer. Cancer Biomark 2019; 26:411-420. [PMID: 31640084 DOI: 10.3233/cbm-190199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Lin Cheng
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Liang Shi
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hong Dai
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
26
|
de Oliveira V, Schaefer J, Calder M, Lydon JP, DeMayo FJ, Bhattacharya M, Radovick S, Babwah AV. Uterine Gα q/11 signaling, in a progesterone-dependent manner, critically regulates the acquisition of uterine receptivity in the female mouse. FASEB J 2019; 33:9374-9387. [PMID: 31091422 PMCID: PMC6662978 DOI: 10.1096/fj.201900026r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/23/2019] [Indexed: 12/20/2022]
Abstract
A nonreceptive uterus is a major cause of embryo implantation failure. This study examined the importance of the Gαq/11-coupled class of GPCRs as regulators of uterine receptivity. Mice were created lacking uterine Gαq and Gα11; as a result, signaling by all uterine Gαq/11-coupled receptors was disrupted. Reproductive profiling of the knockout females revealed that on d 4 of pregnancy, despite adequate serum progesterone (P4) levels and normal P4 receptor (PR) expression, there was no evidence of PR signaling. This resulted in the down-regulation of heart and neural crest derivatives expressed 2, Kruppel-like factor 15, and cyclin G1 and the subsequent persistent proliferation of the luminal epithelium. Aquaporin (Aqp) 11 was also potently down-regulated, whereas Aqp5/AQP5 expression persisted, resulting in the inhibition of luminal closure. Hypertrophy of the myometrial longitudinal muscle was also dramatically diminished, likely contributing to the observed implantation failure. Further analyses revealed that a major mechanism via which uterine Gαq/11 signaling induces PR signaling is through the transcriptional up-regulation of leucine-rich repeat-containing GPCR 4 (Lgr4). LGR4 was previously identified as a trigger of PR activation and signaling. Overall, this study establishes that Gαq/11 signaling, in a P4-dependent manner, critically regulates the acquisition of uterine receptivity in the female mouse, and disruption of such signaling results in P4 resistance.-de Oliveira, V., Schaefer, J., Calder, M., Lydon, J. P., DeMayo, F. J., Bhattacharya, M., Radovick, S., Babwah, A. V. Uterine Gαq/11 signaling, in a progesterone-dependent manner, critically regulates the acquisition of uterine receptivity in the female mouse.
Collapse
Affiliation(s)
- Vanessa de Oliveira
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Jennifer Schaefer
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Michele Calder
- Department of Obstetrics and Gynaecology, Western University, London, Ontario, Canada
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Francesco J. DeMayo
- Reproductive and Developmental Biology Laboratory, The National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Moshmi Bhattacharya
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Sally Radovick
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| | - Andy V. Babwah
- Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
- Child Health Institute of New Jersey, New Brunswick, New Jersey, USA
| |
Collapse
|
27
|
Transcriptome Changes in the Mink Uterus during Blastocyst Dormancy and Reactivation. Int J Mol Sci 2019; 20:ijms20092099. [PMID: 31035421 PMCID: PMC6540205 DOI: 10.3390/ijms20092099] [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: 03/14/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 12/29/2022] Open
Abstract
Embryo implantation in the mink follows the pattern of many carnivores, in that preimplantation embryo diapause occurs in every gestation. Details of the gene expression and regulatory networks that terminate embryo diapause remain poorly understood. Illumina RNA-Seq was used to analyze global gene expression changes in the mink uterus during embryo diapause and activation leading to implantation. More than 50 million high quality reads were generated, and assembled into 170,984 unigenes. A total of 1684 differential expressed genes (DEGs) in uteri with blastocysts in diapause were compared to the activated embryo group (p < 0.05). Among these transcripts, 1527 were annotated as known genes, including 963 up-regulated and 564 down-regulated genes. The gene ontology terms for the observed DEGs, included cellular communication, phosphatase activity, extracellular matrix and G-protein couple receptor activity. The KEGG pathways, including PI3K-Akt signaling pathway, focal adhesion and extracellular matrix (ECM)-receptor interactions were the most enriched. A protein-protein interaction (PPI) network was constructed, and hub nodes such as VEGFA, EGF, AKT, IGF1, PIK3C and CCND1 with high degrees of connectivity represent gene clusters expected to play an important role in embryo activation. These results provide novel information for understanding the molecular mechanisms of maternal regulation of embryo activation in mink.
Collapse
|
28
|
Wang X, Mittal P, Castro CA, Rajkovic G, Rajkovic A. Med12 regulates ovarian steroidogenesis, uterine development and maternal effects in the mammalian egg. Biol Reprod 2019; 97:822-834. [PMID: 29126187 DOI: 10.1093/biolre/iox143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/07/2017] [Indexed: 12/21/2022] Open
Abstract
The transcriptional factor MED12 is part of the essential mediator transcriptional complex that acts as a transcriptional coactivator in all eukaryotes. Missense gain-of-function mutations in human MED12 are associated with uterine leiomyomas, yet the role of MED12 deficiency in tumorigenesis and reproductive biology has not been fully explored. We generated a Med12 reproductive conditional knockout mouse model to evaluate its role in uterine mesenchyme, granulosa cells, and oocytes. Mice heterozygous for Med12 deficiency in granulosa cells and uterus (Med12fl/+ Amhr2-Cre) were subfertile, while mice homozygous for Med12 deficiency in granulosa cells and uterus (Med12fl/fl Amhr2-Cre) were infertile. Morphological and histological analysis of the Med12fl/fl Amhr2-Cre reproductive tract revealed atrophic uteri and hyperchromatic granulosa cells with disrupted expression of Lhcgr, Esr1, and Esr2. Med12fl/fl Amhr2-Cre mice estrous cycle was disrupted, and serum analysis showed blunted rise in estradiol in response to pregnant mare serum gonadotropin. Uterine atrophy was partially rescued by exogenous steroid supplementation with dysregulation of Notch1 and Smo expression in steroid supplemented Med12fl/fl Amhr2-Cre uteri, indicating intrinsic uterine defects. Oocyte-specific ablation of Med12 caused infertility without disrupting normal folliculogenesis and ovulation, consistent with maternal effects of Med12 in early embryo development. These results show the critical importance of Med12 in reproductive tract development and that Med12 loss of function does not cause tumorigenesis in reproductive tissues.
Collapse
Affiliation(s)
- Xinye Wang
- Tsinghua MD Program, Tsinghua University School of Medicine, Beijing, China.,Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Priya Mittal
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Carlos A Castro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Gabriel Rajkovic
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA
| | - Aleksandar Rajkovic
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, Pittsburgh, Pennsylvania, USA.,Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
29
|
Rytkönen KT, Erkenbrack EM, Poutanen M, Elo LL, Pavlicev M, Wagner GP. Decidualization of Human Endometrial Stromal Fibroblasts is a Multiphasic Process Involving Distinct Transcriptional Programs. Reprod Sci 2018; 26:323-336. [PMID: 30309298 DOI: 10.1177/1933719118802056] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Decidual stromal cells differentiate from endometrial stromal fibroblasts (ESFs) under the influence of progesterone and cyclic adenosine monophosphate (cAMP) and are essential for implantation and the maintenance of pregnancy. They evolved in the stem lineage of placental (eutherian) mammals coincidental with the evolution of implantation. Here we use the well-established in vitro decidualization protocol to compare early (3 days) and late (8 days) gene transcription patterns in immortalized human ESF. We document extensive, dynamic changes in the early and late decidual cell transcriptomes. The data suggest the existence of an early signal transducer and activator of transcription (STAT) pathway dominated state and a later nuclear factor κB (NFKB) pathway regulated state. Transcription factor expression in both phases is characterized by putative or known progesterone receptor ( PGR) target genes, suggesting that both phases are under progesterone control. Decidualization leads to proliferative quiescence, which is reversible by progesterone withdrawal after 3 days but to a lesser extent after 8 days of decidualization. In contrast, progesterone withdrawal induces cell death at comparable levels after short or long exposure to progestins and cAMP. We conclude that decidualization is characterized by a biphasic gene expression dynamic that likely corresponds to different phases in the establishment of the fetal-maternal interface.
Collapse
Affiliation(s)
- Kalle T Rytkönen
- 1 Yale Systems Biology Institute, West Haven, CT, USA.,2 Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA.,3 Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu, Finland.,4 Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Eric M Erkenbrack
- 1 Yale Systems Biology Institute, West Haven, CT, USA.,2 Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Matti Poutanen
- 3 Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu, Finland
| | - Laura L Elo
- 4 Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mihaela Pavlicev
- 5 Cincinnati Children's Hospital and Medical Center, Cincinnati, OH, USA
| | - Günter P Wagner
- 1 Yale Systems Biology Institute, West Haven, CT, USA.,2 Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA.,6 Department of Obstetrics, Yale Medical School, New Haven, CT, USA.,7 Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
30
|
Abstract
Cancer cells reprogramme metabolism to maximize the use of nitrogen and carbon for the anabolic synthesis of macromolecules that are required during tumour proliferation and growth. To achieve this aim, one strategy is to reduce catabolism and nitrogen disposal. The urea cycle (UC) in the liver is the main metabolic pathway to convert excess nitrogen into disposable urea. Outside the liver, UC enzymes are differentially expressed, enabling the use of nitrogen for the synthesis of UC intermediates that are required to accommodate cellular needs. Interestingly, the expression of UC enzymes is altered in cancer, revealing a revolutionary mechanism to maximize nitrogen incorporation into biomass. In this Review, we discuss the metabolic benefits underlying UC deregulation in cancer and the relevance of these alterations for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Rom Keshet
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Peter Szlosarek
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK
- Barts Health NHS Trust, St Bartholomew's Hospital, London, UK
| | - Arkaitz Carracedo
- CIC bioGUNE, Bizkaia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country, Bilbao, Spain
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
31
|
Mahadevappa R, Neves H, Yuen SM, Jameel M, Bai Y, Yuen HF, Zhang SD, Zhu Y, Lin Y, Kwok HF. DNA Replication Licensing Protein MCM10 Promotes Tumor Progression and Is a Novel Prognostic Biomarker and Potential Therapeutic Target in Breast Cancer. Cancers (Basel) 2018; 10:cancers10090282. [PMID: 30135378 PMCID: PMC6162382 DOI: 10.3390/cancers10090282] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is one of the most common malignancies in women worldwide. In breast cancer, the cell proliferation rate is known to influence the cancer malignancy. Recent studies have shown that DNA replication initiation/licensing factors are involved in cancer cell proliferation as well as cancer cell migration and invasion. Licensing factors have also been reported as important prognostic markers in lung, prostrate, and bladder cancers. Here, we studied the role of MCM10, a novel licensing factor, in breast cancer progression. From the public database, NCBI, we investigated six independent breast cancer patient cohorts, totaling 1283 patients. We observed a significant association between high MCM10 mRNA expression with tumor grading and patients’ survival time. Most importantly, using breast cancer cohorts with available treatment information, we also demonstrated that a high level of MCM10 is associated with a better response to conventional treatment. Similarly, in in vitro studies, the expression level of MCM10 in breast cancer cell lines is significantly higher compared to paired normal breast epithelium cells. Knockdown of MCM10 expression in the cancer cell line showed significantly decreased tumorigenic properties such as cell proliferation, migration and anchorage independence. The MCF7 breast cancer cell line, after MCM10 expression knockdown, showed significantly decreased tumorigenic properties such as cell proliferation, migration, and anchorage independent growth. Mechanistically, MCM10 expression is observed to be regulated by an Estrogen Receptor (ER) signaling pathway, where its expression is suppressed by the inhibition of the ER or serum withdrawal. Our results suggest that MCM10 plays an important role in breast cancer progression and is a potential prognostic/predictive biomarker and therapeutic target for breast cancer patients.
Collapse
Affiliation(s)
- Ravikiran Mahadevappa
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Henrique Neves
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Shun Ming Yuen
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Muhammad Jameel
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Yuchen Bai
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| | - Hiu-Fung Yuen
- Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore.
| | - Shu-Dong Zhang
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Londonderry BT47 6SB, UK.
| | - Youzhi Zhu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China.
| | - Yao Lin
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China.
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau, China.
| |
Collapse
|
32
|
Huang C, Sun H, Wang Z, Liu Y, Cheng X, Liu J, Jiang R, Zhang X, Zhen X, Zhou J, Chen L, Ding L, Yan G, Jiang Y. Increased Krüppel-like factor 12 impairs embryo attachment via downregulation of leukemia inhibitory factor in women with recurrent implantation failure. Cell Death Discov 2018; 4:23. [PMID: 30109142 PMCID: PMC6079092 DOI: 10.1038/s41420-018-0088-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/26/2018] [Accepted: 07/13/2018] [Indexed: 02/07/2023] Open
Abstract
Recurrent implantation failure (RIF) caused by various etiological factors remains a challenge for fertility clinicians using assisted reproductive technology (ART) worldwide. Dysregulation of leukemia inhibitory factor (LIF) in the endometria of women with RIF is involved in impaired endometrial receptivity and embryo adhesion. However, the mechanism through which LIF expression is regulated in women with RIF is still poorly understood. Our previous study noted that the abnormally increased endometrial Krüppel-like factor 12 (KLF12) in RIF women led to impaired decidualization and embryo implantation. Here, we further found that KLF12 inhibited embryo adhesion in vivo and in vitro by repressing LIF expression. Mechanistically, KLF12 bound to conserved sites (CAGTGGG, -6771 to -6765 and -7115 to -7109) within the LIF promoter region and repressed LIF transcription directly. Exogenous LIF significantly reversed the KLF12-mediated repression of BeWo spheroid adhesion. KLF12 expression was reduced significantly in Ishikawa cells treated with progestogen, which was due to the activation of Akt signaling. These findings may provide novel potential therapeutic regimens for patients with RIF and disrupted endometrial receptivity.
Collapse
Affiliation(s)
- Chenyang Huang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Haixiang Sun
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Zhilong Wang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Yang Liu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Xi Cheng
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Jingyu Liu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Ruiwei Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Xindong Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Xin Zhen
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Jidong Zhou
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Linjun Chen
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Lijun Ding
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Guijun Yan
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| | - Yue Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 China
| |
Collapse
|
33
|
Hoa N, Ge L, Korach KS, Levin ER. Estrogen receptor beta maintains expression of KLF15 to prevent cardiac myocyte hypertrophy in female rodents. Mol Cell Endocrinol 2018; 470:240-250. [PMID: 29127073 PMCID: PMC6242344 DOI: 10.1016/j.mce.2017.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/26/2017] [Accepted: 11/06/2017] [Indexed: 12/28/2022]
Abstract
Maintaining a healthy, anti-hypertrophic state in the heart prevents progression to cardiac failure. In humans, angiotensin II (AngII) indirectly and directly stimulates hypertrophy and progression, while estrogens acting through estrogen receptor beta (ERβ) inhibit these AngII actions. The KLF15 transcription factor has been purported to provide anti-hypertrophic action. In cultured neonatal rat cardiomyocytes, we found AngII inhibited KLF1 expression and nuclear localization, substantially prevented by estradiol (E2) or β-LGND2 (β-LGND2), an ERβ agonist. AngII stimulation of transforming growth factor beta expression in the myocytes activated p38α kinase via TAK1 kinase, inhibiting KLF15 expression. All was comparably reduced by E2 or β-LGND2. Knockdown of KLF15 in the myocytes induced myocyte hypertrophy and limited the anti-hypertrophic actions of E2 and β-LGND2. Key aspects were confirmed in an in-vivo model of cardiac hypertrophy. Our findings define additional anti-hypertrophic effects of ERβ supporting testing specific receptor agonists in humans to prevent progression of cardiac disease.
Collapse
Affiliation(s)
- Neil Hoa
- Division of Endocrinology, Department of Veterans Affairs Medical Center, Long Beach, CA, 90822, USA
| | - Lisheng Ge
- Division of Endocrinology, Department of Veterans Affairs Medical Center, Long Beach, CA, 90822, USA
| | | | - Ellis R Levin
- Division of Endocrinology, Department of Veterans Affairs Medical Center, Long Beach, CA, 90822, USA; Department of Medicine, University of California, Irvine, CA, 92717, USA; Department of Biochemistry, University of California, Irvine, CA, 92717, USA.
| |
Collapse
|
34
|
Wang X, He M, Li J, Wang H, Huang J. KLF15 suppresses cell growth and predicts prognosis in lung adenocarcinoma. Biomed Pharmacother 2018; 106:672-677. [PMID: 29990857 DOI: 10.1016/j.biopha.2018.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/01/2018] [Accepted: 07/01/2018] [Indexed: 02/07/2023] Open
Abstract
Krüppel-like factors (KLFs) are transcription factors containing three different C2H2-type zinc finger domains in their carboxy-terminal regions which have been identified to play important roles in a variety of cancers. However, little is known about KLF15 in lung adenocarcinoma (LAUD). Our study demonstrated that the expression levels of KLF15 were observably down-regulated in LAUD tissues compared to paired adjacent normal tissues. LUAD patients with low expression levels of KLF15 have worse prognosis than those with high expression levels of KLF15. KLF15 could suppress cell growth, which was partly via up-regulating CDKN1 A/p21 and CDKN2A/p15. Our findings suggested that KLF15 showed a significant role in LAUD progression and may shed light on a promising novel therapeutic target for blocking progression of LAUD.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, PR China; Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, PR China
| | - Mingqing He
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, PR China
| | - Jianzhong Li
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, PR China
| | - Haiying Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, PR China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, PR China.
| |
Collapse
|
35
|
Qi S, Yan L, Liu Z, Mu YL, Li M, Zhao X, Chen ZJ, Zhang H. Melatonin inhibits 17β-estradiol-induced migration, invasion and epithelial-mesenchymal transition in normal and endometriotic endometrial epithelial cells. Reprod Biol Endocrinol 2018; 16:62. [PMID: 29935526 PMCID: PMC6015458 DOI: 10.1186/s12958-018-0375-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 06/04/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Melatonin is a potential therapeutic agent for endometriosis, but its molecular mechanism is unclear. Here, we investigated the effect of melatonin on the epithelial-mesenchymal transition (EMT) in endometriotic endometrial epithelial cells and explored the pathway that might be involved. METHODS This hospital-based study included 60 women of reproductive age using the endometrium for immunohistochemistry, 6 women of reproductive age undergoing bilateral tubal ligation and 6 patients with endometriosis for isolation of endometrial epithelial cells or subsequent analysis, respectively. We examined the expression of Notch1/Numb signaling and EMT markers by immunohistochemistry analysis and western blot analysis, the invasion and migration of endometrial epithelial cells by transwell assays, and the cell proliferation by CCK8 assays. RESULTS Compared with normal endometrium, the endometriotic eutopic endometrium showed increased expression of Notch1, Slug, Snail, and N-cadherin, and decreased expression of E-cadherin and Numb. Melatonin or Notch inhibition by specific inhibitor blocked 17β-estradiol-induced cell proliferation, invasion, migration and EMT-related markers in both normal and endometriotic epithelial cells. CONCLUSIONS Our data suggest that aberrant expression of Notch1/Numb signaling and the EMT is present in endometriotic endometrium. Melatonin may block 17β-estradiol-induced migration, invasion and EMT in normal and endometriotic epithelial cells by upregulating Numb expression and decreasing the activity of the Notch signaling pathway.
Collapse
Affiliation(s)
- Shasha Qi
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, People's Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250021, People's Republic of China
- The Key laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, 250021, People's Republic of China
| | - Lei Yan
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, People's Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250021, People's Republic of China
- The Key laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, 250021, People's Republic of China
| | - Zhao Liu
- Department of Urology, Qilu Hospital of Shandong University, 107 Wenhua Xi Road, Jinan, 250012, People's Republic of China
| | - Yu-Lan Mu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Mingjiang Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Xingbo Zhao
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, People's Republic of China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250021, People's Republic of China
- The Key laboratory for Reproductive Endocrinology, Shandong University, Ministry of Education, Jinan, 250021, People's Republic of China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200030, People's Republic of China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, People's Republic of China
| | - Hui Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021, People's Republic of China.
| |
Collapse
|
36
|
Tsuji M, Tanaka T, Nagashima R, Sagisaka Y, Tousen Y, Nishide Y, Ishimi Y, Ishimi Y. Effect of daidzein and equol on DNA replication in MCF-7 cells. J Biochem 2018; 163:371-380. [PMID: 29346578 DOI: 10.1093/jb/mvy006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/30/2017] [Indexed: 11/13/2022] Open
Abstract
It has been reported that daidzein and equol stimulate DNA replication and proliferation of MCF-7 cells. However, their molecular mechanisms of action are still unclear. We examined the effects of daidzein and equol on DNA replication of MCF-7 cells, focusing on MCM2-7 proteins, which function as the replicative helicase. In the presence of either 1 μM of daidzein or equol, the number of cells in S-phase, which was determined by detecting bromodeoxyuridine incorporated into replicated DNA, almost doubled. The total amounts of MCM7 protein and chromatin-bound MCM7 protein increased in the presence of daidzein. The data suggest that phytoestrogens facilitate cell cycle progression in G1-phase by increasing the level of MCM proteins. In the presence of phytoestrogens, phosphorylation of Rb and levels of MCM2, 3 and 7 mRNA increased, suggesting that stimulation of MCM2-7 transcription is involved in the cell cycle progression. Under the same conditions, double-stranded DNA breakage in logarithmically growing MCF-7 cells, which was detected using anti-γ-H2AX antibodies, did not increase in the presence of equol.
Collapse
Affiliation(s)
- Mako Tsuji
- Ibaraki University, Mito, Ibaraki 310-8512, Japan
| | | | | | | | - Yuko Tousen
- National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan
| | - Yoriko Nishide
- National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan
| | - Yoshiko Ishimi
- National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan
| | - Yukio Ishimi
- Ibaraki University, Mito, Ibaraki 310-8512, Japan
| |
Collapse
|
37
|
Chen Y, Cao XY, Li YN, Qiu YY, Li YN, Li W, Wang H. Reversal of cisplatin resistance by microRNA-139-5p-independent RNF2 downregulation and MAPK inhibition in ovarian cancer. Am J Physiol Cell Physiol 2018; 315:C225-C235. [PMID: 29719173 DOI: 10.1152/ajpcell.00283.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Some microRNAs (miRs) are dysregulated in cancers, and aberrant miR expression has been reported to correlate with chemoresistance of cancer cells. Therefore, the present study aims at investigating the effects of microRNA-139-5p (miR-139-5p) on cisplatin resistance of ovarian cancer (OC) with involvement of ring finger protein 2 (RNF2) and the mitogen-activated protein kinase (MAPK) signaling pathway. OC tissues were obtained from 66 primary OC patients. The cisplatin-sensitive A2780 and cisplatin-resistant A2780/DDP cell lines were collected for construction of RNF2 silencing and overexpressed plasmids. Cell vitality and apoptosis were detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and annexin V-FITC/propidium iodide double-staining, respectively. Next, expression of RNF2, extracellular signal-related kinase, and p38 was determined by quantitative reverse transcription-quantitative polymerase chain reaction and Western blot analysis. Finally, the volume of xenograft tumors in BALB/c nude mice was detected. RNF2 and miR-139-5p were identified to be involved in OC. In addition, MAPK activation and RNF2 were related to cisplatin resistance of OC. miR-139-5p was downregulated in cisplatin-resistant OC tissues, and miR-139-5p overexpression could inhibit cell vitality, reduce cisplatin resistance, and promote apoptosis of OC cells. Furthermore, miR-139-5p combined with MAPK inhibitors more obviously reduced cisplatin resistance of OC. Taken together, this study demonstrated that miR-139-5p overexpression combined with inactivation of the MAPK signaling pathway can reverse the cisplatin resistance of OC by suppressing RNF2. Thus, miR-139-5p overexpression might be a future therapeutic strategy for OC.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, People's Republic of China
| | - Xiao-Yun Cao
- Medical Insurance Management Office, Economic and Technological Development Zone, People's Hospital of Linyi, Linyi, People's Republic of China
| | - Ying-Ni Li
- Department of Obstetrics and Gynecology, Economic and Technological Development Zone, People's Hospital of Linyi, Linyi, People's Republic of China
| | - Yu-Yan Qiu
- Department of Obstetrics and Gynecology, Economic and Technological Development Zone, People's Hospital of Linyi, Linyi, People's Republic of China
| | - Ying-Na Li
- Department of Obstetrics and Gynecology, Economic and Technological Development Zone, People's Hospital of Linyi, Linyi, People's Republic of China
| | - Wen Li
- Department of Reproductive Medicine, Linyi People's Hospital, Linyi, People's Republic of China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Linyi, People's Republic of China
| |
Collapse
|
38
|
Sun P, Wang J, Guo X, Chen Y, Xing C, Gao A. Benzene and its metabolite decreases cell proliferation via LncRNA-OBFC2A-mediated anti-proliferation effect involving NOTCH1 and KLF15. Oncotarget 2018; 8:40857-40871. [PMID: 28388563 PMCID: PMC5522231 DOI: 10.18632/oncotarget.16588] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/09/2017] [Indexed: 12/18/2022] Open
Abstract
LncRNA has been considered to play a crucial role in the progression of several diseases by affecting cell proliferation. However, its role in benzene toxicity remains unclear. Our study showed that the expression of lncRNA-OBFC2A increased accompanied with the change of cell proliferation related-genes in benzene-exposed workers. In vitro experiments, 1,4-Benzoquinone dose-dependently inhibited cell proliferation and simultaneously caused the decrease of NOTCH1 expression and the increase of KLF15 in AHH-1 cell lines. Meanwhile, 1, 4-Benzoquinone obviously increased the expression of lncRNA-OBFC2A, which was consistent with our previous population results. Therefore, we propose that lncRNA-OBFC2A is involved in benzene toxicity by regulating cell proliferation. Further, we successfully constructed a lentivirus model of interfering the expression of lncRNA-OBFC2A. After interfering lncRNA-OBFC2A, the cell proliferation inhibition and the expression of NOTCH1 and KLF15 induced by 1, 4-Benzoquinone were reversed. Subsequently, RNA fluorescence in situ Hybridization assay showed that lncRNA-OBFC2A was located in cell nuclei. These results suggest that benzene and its metabolite decreases cell proliferation via LncRNA-OBFC2A-mediated anti-proliferation effect involving NOTCH1 and KLF15. LncRNA-OBFC2A can be a potential biomarker for benzene toxicity.
Collapse
Affiliation(s)
- Pengling Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Jing Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiaoli Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yujiao Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Caihong Xing
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ai Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
Sun P, Guo X, Chen Y, Zhang W, Duan H, Gao A. VNN3, a potential novel biomarker for benzene toxicity, is involved in 1, 4-benzoquinone induced cell proliferation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 233:323-330. [PMID: 29096305 DOI: 10.1016/j.envpol.2017.10.087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 06/07/2023]
Abstract
Benzene is widely employed in the field of production, and its toxicity on biological systems has received increasing attention. Cell proliferation is a major life characteristic of living organisms. KLF15 and NOTCH1 are mature and classical genes in cell proliferation studies, particularly in the area of tumor investigation. The aim of this study was to investigate the effect and mechanism of VNN3 on cell proliferation induced by 1,4-benzoquinone (1,4-BQ), an important metabolite of benzene, and obtain a sensitive biomarker for the hazard screening and health care of benzene exposure. Normally growing AHH-1 cells were cultured in vitro and were incubated with different concentrations of 1,4-BQ (0, 10, 20, and 40 μM) for 24 h. A CCK-8 assay was used to assess the cell viability, whereas EdU was used to detect the cell proliferation of AHH-1 cells. The expression of VNN3, KLF15 and NOTCH1 was detected by real-time PCR. Moreover, a lentiviral model was constructed in AHH-1 cells to interfere with VNN3 expression. The results showed that 1,4-BQ clearly increased the expression of VNN3. Moreover, 1,4-BQ dose-dependently inhibited cell proliferation and caused increased KLF15 expression; in contrast, the NOTCH1 expression decreased in AHH-1 cells. Furthermore, following interference with the VNN3 expression, the cell proliferation inhibition and the expression of KLF15 and NOTCH1 were rescued. To further investigate the action of VNN3 in benzene hematotoxicity, we assessed it in benzene-exposed workers. The results showed that there was a remarkable correlation between the VNN3 expression and hemogram, which included RBC, NEUT and HGB. In addition, analysis of the KLF15 and NOTCH1 expression showed that the VNN3 expression was related to cell proliferation, which was consistent with the in vitro results. In conclusion, VNN3 influences cell proliferation induced by 1,4-BQ by regulating the expression of KLF15 and NOTCH1. VNN3 may represent a potential biomarker of benzene toxicity.
Collapse
Affiliation(s)
- Pengling Sun
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xiaoli Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yujiao Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Wei Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Huawei Duan
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ai Gao
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
40
|
Zheng XR, Pan X, Zhang J, Cao X. Hyperinsulinemia-induced PAX6 expression promotes endometrial epithelial cell proliferation via negatively modulating p27 signaling. Biomed Pharmacother 2018; 97:802-808. [DOI: 10.1016/j.biopha.2017.10.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/22/2017] [Accepted: 10/26/2017] [Indexed: 12/12/2022] Open
|
41
|
Kuokkanen S, Zhu L, Pollard JW. Xenografted tissue models for the study of human endometrial biology. Differentiation 2017; 98:62-69. [PMID: 29156254 DOI: 10.1016/j.diff.2017.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 01/10/2023]
Abstract
The human endometrium undergoes extensive morphological, biochemical and molecular changes under the influence of female sex steroid hormones. Besides the fact that estrogen stimulates endometrial cell proliferation and progesterone inhibits this proliferation and induces differentiation, there is limited knowledge about precise molecular mechanisms underlying human endometrial biology. The importance of paracrine signaling in endometrial physiology explains why in vitro culture of endometrial cells has been challenging. Researchers, therefore, have developed alternative experimental in vivo models for the study of endometrial biology. The objective of this review is to summarize the recent developments and work on these in vivo endometrial research models. The in vivo recombinant tissue models in which wild-type endometrial cells are combined with endometrial cells from a gene-targeted mouse strain followed by xenografting to host mice have been critical in confirming the significance of paracrine signaling between the epithelium and stroma in the growth regulation of the endometrium. Additionally, these studies have uncovered differences between the mouse and human, emphasizing the need for the development of experimental models specifically of the human endometrium. Recently, xenotransplants of human endometrial fragments into the subcutaneous space of host mice and endometrial xenografts of dissociated and recombined epithelial and stromal cells beneath the kidney capsule of immunodeficient host mice have proven to be highly promising tools for in vivo research of endometrial functions. For the first time, the latter approach provides an immense opportunity for the application of genome engineering, such as targeted ablation of endometrial genes for example by using CRISPR/CAS9 system. This research will begin to elucidate the functional role of specific genes in this complex tissue. Another advantage of xenotransplantation and xenograft models of the human endometrium is their use to investigate endometrial effects of new compounds and drugs without needing to give them to women. Underpinning the molecular mechanisms underlying endometrial functions is critical to ultimately advance our understanding of endometrial pathophysiology and develop targeted therapies to prevent and cure endometrial pathologies as well as enhance endometrial function when it is desired for fertility.
Collapse
Affiliation(s)
- Satu Kuokkanen
- Department of Obstetrics and Gynecology&Women's Health, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States
| | - Liyin Zhu
- Department of Obstetrics and Gynecology&Women's Health, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States; Department of Developmental&Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jeffrey W Pollard
- Department of Obstetrics and Gynecology&Women's Health, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, United States; Department of Developmental&Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, United States; MRC Centre for Reproductive Health, University of Edinburgh, UK.
| |
Collapse
|
42
|
Gao L, Qiu H, Liu J, Ma Y, Feng J, Qian L, Zhang J, Liu Y, Bian T. KLF15 promotes the proliferation and metastasis of lung adenocarcinoma cells and has potential as a cancer prognostic marker. Oncotarget 2017; 8:109952-109961. [PMID: 29299121 PMCID: PMC5746356 DOI: 10.18632/oncotarget.21972] [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: 03/27/2017] [Accepted: 09/24/2017] [Indexed: 12/19/2022] Open
Abstract
Lung adenocarcinoma (LADC)is a general form of non-small cell lung cancer that represents a significant threat to public health worldwide. The 5-year-survival rate for LADC is currently below 15%. The transcription factor KLF15, also called kidney-enriched KLF (KKLF), has been proven to play a role in inhibiting proliferation and diversification of carcinoma cells, including those of the endometrium, pancreas and breast, but the involvement of KLF15 in LADC has not previously been studied. In this study, we compared the in vitro expression of KLF15 in human LADC tissues and adjacent normal lung tissues. Expression of KLF15 was found to be abnormally high in LADC tissues and cells compared with adjacent non-tumorous tissues, and was correlated with tumor TNM stage and tumor differentiation (P = 0.003, P = 0.001, respectively). The effect of KLF15 on cell growth and migration were explored in vitro by Western Blotting, CCK8 and colony formation assays, flow cytometry analysis and transwell migration assays, and in vivo by analysis of tumorigenesis in 5-week old BALB/c nude mice. Knockdown of KLF15 significantly upregulated the protein levels of cleaved caspase-3, caspase-7, caspase-8 and PARP, thereby inducing apoptosis. Downregulation of KLF15 in A549 and NCI-H1650 cell lines resulted in these cell lines exhibiting markedly slower growth rates when injected subcutaneously into the flank of nude mice, compared with the comparator control groups (P < 0.05). Collectively, our findings suggest that KLF15 may have a significant effect on LADC cell survival, and that it represents a potential therapeutic and preventive biomarker for LADC prognosis and treatment.
Collapse
Affiliation(s)
- Lihua Gao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Hongmei Qiu
- Department of Respiration, Nantong Geriatric Rehabilitation Hospital, Branch of Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jian Liu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yuzhen Ma
- Centre of Reproductive Medicine, Inner Mongolia Hospital, Inner Mongolia, Hohhot, 010021, China
| | - Jia Feng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Li Qian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| |
Collapse
|
43
|
Monsivais D, Matzuk MM, Pangas SA. The TGF-β Family in the Reproductive Tract. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022251. [PMID: 28193725 DOI: 10.1101/cshperspect.a022251] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The transforming growth factor β (TGF-β) family has a profound impact on the reproductive function of various organisms. In this review, we discuss how highly conserved members of the TGF-β family influence the reproductive function across several species. We briefly discuss how TGF-β-related proteins balance germ-cell proliferation and differentiation as well as dauer entry and exit in Caenorhabditis elegans. In Drosophila melanogaster, TGF-β-related proteins maintain germ stem-cell identity and eggshell patterning. We then provide an in-depth analysis of landmark studies performed using transgenic mouse models and discuss how these data have uncovered basic developmental aspects of male and female reproductive development. In particular, we discuss the roles of the various TGF-β family ligands and receptors in primordial germ-cell development, sexual differentiation, and gonadal cell development. We also discuss how mutant mouse studies showed the contribution of TGF-β family signaling to embryonic and postnatal testis and ovarian development. We conclude the review by describing data obtained from human studies, which highlight the importance of the TGF-β family in normal female reproductive function during pregnancy and in various gynecologic pathologies.
Collapse
Affiliation(s)
- Diana Monsivais
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030
| | - Martin M Matzuk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Texas 77030.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030.,Department of Pharmacology, Baylor College of Medicine, Houston, Texas 77030
| | - Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Texas 77030
| |
Collapse
|
44
|
Zhou J, Wang F, Xu C, Zhou Z, Zhang W. KLF15 regulates dopamine D2 receptor and participates in mouse models of neuropathic pain. Biochem Biophys Res Commun 2017; 492:269-274. [DOI: 10.1016/j.bbrc.2017.08.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/18/2017] [Indexed: 02/06/2023]
|
45
|
Winuthayanon W, Lierz SL, Delarosa KC, Sampels SR, Donoghue LJ, Hewitt SC, Korach KS. Juxtacrine Activity of Estrogen Receptor α in Uterine Stromal Cells is Necessary for Estrogen-Induced Epithelial Cell Proliferation. Sci Rep 2017; 7:8377. [PMID: 28827707 PMCID: PMC5566397 DOI: 10.1038/s41598-017-07728-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 07/04/2017] [Indexed: 01/11/2023] Open
Abstract
Aberrant regulation of uterine cell growth can lead to endometrial cancer and infertility. To understand the molecular mechanisms of estrogen-induced uterine cell growth, we removed the estrogen receptor α (Esr1) from mouse uterine stromal cells, where the embryo is implanted during pregnancy. Without ESR1 in neighboring stroma cells, epithelial cells that line the inside of the uterus are unable to grow due to a lack of growth factors secreted from adjacent stromal cells. Moreover, loss of stromal ESR1 caused mice to deliver fewer pups due in part due to inability of some embryos to implant in the uterus, indicating that stromal ESR1 is crucial for uterine cell growth and pregnancy.
Collapse
Affiliation(s)
- Wipawee Winuthayanon
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, 99164, United States.
| | - Sydney L Lierz
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, 27709, United States
| | - Karena C Delarosa
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, 99164, United States
| | - Skylar R Sampels
- School of Molecular Biosciences, Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, 99164, United States
| | - Lauren J Donoghue
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, 27709, United States
| | - Sylvia C Hewitt
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, 27709, United States
| | - Kenneth S Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina, 27709, United States
| |
Collapse
|
46
|
Sun C, Ma P, Wang Y, Liu W, Chen Q, Pan Y, Zhao C, Qian Y, Liu J, Li W, Shu Y. KLF15 Inhibits Cell Proliferation in Gastric Cancer Cells via Up-Regulating CDKN1A/p21 and CDKN1C/p57 Expression. Dig Dis Sci 2017; 62:1518-1526. [PMID: 28421457 DOI: 10.1007/s10620-017-4558-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/28/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Krüppel-like factors (KLFs) have been identified in multi-cancers and act as oncogenes or tumor suppressors. The function of KLF15, one member of KLFs, has not been well elucidated, especially in gastric cancer (GC). AIMS This study was designed to investigate the prognostic value and biological functions of KLF15 in GC. METHODS KLF15 protein expression in GC patients was evaluated by immunohistochemistry assays in 50 paired GC tissues and adjacent normal tissues, and correlations between KLF15 expression and clinicopathological characteristics and prognosis were analyzed. Then, we investigated the over-expression of KLF15 on cell proliferation and its mechanism in GC cells. RESULTS KLF15 expression levels were significantly down-regulated in GC tissues compared to adjacent normal tissues. And KLF15 expression was negatively correlated with clinical stage, lymphatic metastasis, and distant metastasis. Furthermore, KLF15 expression could predict prognosis in patients with GC. Moreover, over-expression of KLF15 could inhibit cell proliferation partly via regulating CDKN1A/p21 and CDKN1C/p57. CONCLUSION These findings demonstrate that KLF15 plays a significant role in GC progression and could be a therapeutic target for GC.
Collapse
Affiliation(s)
- Chongqi Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China
| | - Pei Ma
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China
| | - Yanfen Wang
- Department of Pathology, Yangzhou No.1 People's Hospital, Yangzhou, People's Republic of China
| | - Weitao Liu
- Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qinnan Chen
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yutian Pan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China
| | - Chenhui Zhao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China
| | - Yingchen Qian
- Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jie Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China. .,Department of Medical Oncology, Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, People's Republic of China.
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, No.300 Guangzhou Road, Nanjing, People's Republic of China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, People's Republic of China. .,Department of Medical Oncology, Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, People's Republic of China.
| |
Collapse
|
47
|
Monsivais D, Clementi C, Peng J, Fullerton PT, Prunskaite-Hyyryläinen R, Vainio SJ, Matzuk MM. BMP7 Induces Uterine Receptivity and Blastocyst Attachment. Endocrinology 2017; 158:979-992. [PMID: 28324064 PMCID: PMC5460793 DOI: 10.1210/en.2016-1629] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023]
Abstract
In women, the window of implantation is limited to a brief 2- to 3-day period characterized by optimal levels of circulating ovarian hormones and a receptive endometrium. Although the window of implantation is assumed to occur 8 to 10 days after ovulation in women, molecular markers of endometrial receptivity are necessary to determine optimal timing prior to embryo transfer. Previous studies showed that members of the bone morphogenetic protein (BMP) family are expressed in the uterus necessary for female fertility; however, the role of BMP7 during implantation and in late gestation is not known. To determine the contribution of BMP7 to female fertility, we generated Bmp7flox/flox-Pgr-cre+/- [BMP7 conditional knockout (cKO)] mice. We found that absence of BMP7 in the female reproductive tract resulted in subfertility due to uterine defects. At the time of implantation, BMP7 cKO females displayed a nonreceptive endometrium with elevated estrogen-dependent signaling. These implantation-related defects also affected decidualization and resulted in decreased expression of decidual cell markers such as Wnt4, Cox2, Ereg, and Bmp2. We also observed placental abnormalities in pregnant Bmp7 cKO mice, including excessive parietal trophoblast giant cells and absence of a mature placenta at 10.5 days post coitum. To establish possible redundant roles of BMP5 and BMP7 during pregnancy, we generated double BMP5 knockout/BMP7 cKO [BMP5/7 double knockout (DKO)] mice; however, we found that the combined deletion had no additive disruptive effect on fertility. Our studies indicate that BMP7 is an important factor during the process of implantation that contributes to healthy embryonic development.
Collapse
Affiliation(s)
- Diana Monsivais
- Departments of Pathology and Immunology
- Reproductive Medicine, and
| | - Caterina Clementi
- Departments of Pathology and Immunology
- Molecular and Cellular Biology, and
- Reproductive Medicine, and
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jia Peng
- Departments of Pathology and Immunology
- Molecular and Human Genetics
- Reproductive Medicine, and
| | - Paul T. Fullerton
- Departments of Pathology and Immunology
- Molecular and Human Genetics
- Reproductive Medicine, and
| | - Renata Prunskaite-Hyyryläinen
- Departments of Pathology and Immunology
- Reproductive Medicine, and
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Aapistie 5A, 90014 Oulu, Finland
| | - Seppo J. Vainio
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Aapistie 5A, 90014 Oulu, Finland
| | - Martin M. Matzuk
- Departments of Pathology and Immunology
- Molecular and Human Genetics
- Molecular and Cellular Biology, and
- Pharmacology, Centers for
- Drug Discovery and
- Reproductive Medicine, and
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
48
|
Huang C, Jiang Y, Zhou J, Yan Q, Jiang R, Cheng X, Xing J, Ding L, Sun J, Yan G, Sun H. Increased Krüppel-like factor 12 in recurrent implantation failure impairs endometrial decidualization by repressing Nur77 expression. Reprod Biol Endocrinol 2017; 15:25. [PMID: 28359310 PMCID: PMC5374626 DOI: 10.1186/s12958-017-0243-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Decidualization is a prerequisite for successful implantation and the establishment of pregnancy. A critical role of impaired decidualization in subfertility has been established. In human endometrial stromal cells (hESCs), Krüppel-like factor 12 (KLF12) and Nur77 are novel regulators of decidualization. We investigated whether KLF12 impaired the decidualization of hESCs in recurrent implantation failure (RIF) patients. METHODS Endometrial tissues and hESCs were collected from RIF patients (n = 34) and fertile controls (n = 30) for in vitro analysis. Primary hESCs isolated from RIF endometrial tissues were used to evaluate the biological functions of KLF12 and Nur77. In addition, their molecular mechanisms were investigated by adenovirus-mediated overexpression. Gene expression regulation was examined by real-time-quantitative PCR (qRT-PCR), immunostaining and luciferase reporter assay. Further, blastocyst-like spheroid (BLS) and blastocyst implantation models were performed to examine the roles of KLF12 and Nur77 during embryo expansion on hESCs. RESULTS hESCs from the RIF patients showed a poor decidual response, mainly characterized by decreased decidual prolactin (dPRL) secretion, impaired transformation and limited BLS expansion. In addition, KLF12 expression was increased in endometrial tissues from the RIF patients compared with those from the fertile controls, especially in stromal compartments. The opposite results were observed for Nur77 expression in these tissues. KLF12 repressed hESC decidualization by decreasing Nur77 expression. Mechanistically, KLF12 bound to a conserved site in the Nur77 promoter region. Nur77 overexpression significantly reversed the KLF12-mediated repression of dPRL expression, decidual transformation and BLS/blastocyst expansion. CONCLUSIONS KLF12 impairs endometrial decidualization by transcriptionally repressing Nur77, and Nur77 overexpression reverses the poor decidual response of hESCs in RIF patients.
Collapse
Affiliation(s)
- Chenyang Huang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Yue Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Jianjun Zhou
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Qiang Yan
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Ruiwei Jiang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Xi Cheng
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Jun Xing
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Lijun Ding
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA USA
| | - Guijun Yan
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| | - Haixiang Sun
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
- Collaborative Innovation Platform for Reproductive Biology and Technology of Nanjing University Medical School, Nanjing, 210008 People’s Republic of China
| |
Collapse
|
49
|
Kong S, Han X, Cui T, Zhou C, Jiang Y, Zhang H, Wang B, Wang H, Zhang S. MCM2 mediates progesterone-induced endometrial stromal cell proliferation and differentiation in mice. Endocrine 2016; 53:595-606. [PMID: 26910396 DOI: 10.1007/s12020-016-0894-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 02/05/2016] [Indexed: 12/17/2022]
Abstract
Uterine decidualization characterized by stromal cell proliferation and differentiation is critical to the establishment of pregnancy in many species. Progesterone is a key factor in regulating endometrial cell decidualization, however, the molecular basis involved in mediating the effects of progesterone during decidualization remains largely unknown. We report here that the DNA replication licensing factor MCM2, one of the conserved set of six-related proteins (MCM complex: MCM2-7) essential for eukaryotic DNA replication, is dynamically expressed in both proliferative and differentiated stromal cells during mouse periimplantation uterus. Applying PR-knockout mouse model and pharmacological strategy, we further found that the expression of Mcm2 is induced by progesterone action in the mouse uterine stroma. Employing a primary cell culture system, we further demonstrated that siRNA-mediated silencing of MCM2 arrests the cell cycle at G1-S transition during stromal cell proliferation. Moreover, the downregulation of Mcm2 could also compromise stromal cell differentiation. Collectively, our studies uncovered the role of a unique DNA replication licensing molecule MCM2 in mediating Progesterone-induced stromal cell decidualization in mouse uterus.
Collapse
Affiliation(s)
- Shuangbo Kong
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Xue Han
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Tongtong Cui
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Chan Zhou
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Yufei Jiang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Hangxiao Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Bingyan Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Haibin Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China
| | - Shuang Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, People's Republic of China.
| |
Collapse
|
50
|
Kelley DR, Snoek J, Rinn JL. Basset: learning the regulatory code of the accessible genome with deep convolutional neural networks. Genome Res 2016; 26:990-9. [PMID: 27197224 PMCID: PMC4937568 DOI: 10.1101/gr.200535.115] [Citation(s) in RCA: 562] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 04/26/2016] [Indexed: 12/22/2022]
Abstract
The complex language of eukaryotic gene expression remains incompletely understood. Despite the importance suggested by many noncoding variants statistically associated with human disease, nearly all such variants have unknown mechanisms. Here, we address this challenge using an approach based on a recent machine learning advance-deep convolutional neural networks (CNNs). We introduce the open source package Basset to apply CNNs to learn the functional activity of DNA sequences from genomics data. We trained Basset on a compendium of accessible genomic sites mapped in 164 cell types by DNase-seq, and demonstrate greater predictive accuracy than previous methods. Basset predictions for the change in accessibility between variant alleles were far greater for Genome-wide association study (GWAS) SNPs that are likely to be causal relative to nearby SNPs in linkage disequilibrium with them. With Basset, a researcher can perform a single sequencing assay in their cell type of interest and simultaneously learn that cell's chromatin accessibility code and annotate every mutation in the genome with its influence on present accessibility and latent potential for accessibility. Thus, Basset offers a powerful computational approach to annotate and interpret the noncoding genome.
Collapse
Affiliation(s)
- David R Kelley
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Jasper Snoek
- School of Engineering and Applied Science, Harvard University, Cambridge, Massachusetts 02138, USA
| | - John L Rinn
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|