1
|
Wu X, Wang J, Hao Z, Zhen H, Hu J, Liu X, Li S, Zhao F, Li M, Zhao Z, Shi B, Ren C. Circular RNA_015343 sponges microRNA-25 to regulate viability, proliferation, and milk fat synthesis of ovine mammary epithelial cells via INSIG1. J Cell Physiol 2024; 239:e31332. [PMID: 38828915 DOI: 10.1002/jcp.31332] [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: 03/11/2024] [Revised: 04/18/2024] [Accepted: 05/16/2024] [Indexed: 06/05/2024]
Abstract
In our previous study, circ_015343 was found to inhibit the viability and proliferation of ovine mammary epithelial cells (OMECs) and the expression levels of milk fat synthesis marker genes, but the regulatory mechanism underlying the processes is still unclear. Accordingly in this study, the target relationships between circ_015343 with miR-25 and between miR-25 with insulin induced gene 1 (INSIG1) were verified, and the functions of miR-25 and INSIG1 were investigated in OMECs. The dual-luciferase reporter assay revealed that miR-25 mimic remarkably decreased the luciferase activity of circ_015343 in HEK293T cells cotransfected with a wild-type vector, while it did not change the activity of circ_015343 in HEK293T cells cotransfected with a mutant vector. These suggest that cic_015343 can adsorb and bind miR-25. The miR-25 increased the viability and proliferation of OMECs, and the content of triglycerides in OMECs. In addition, INSIG1 was found to be a target gene of miR-25 using a dual-luciferase reporter assay. Overexpression of INSIG1 decreased the viability, proliferation, and level of triglycerides of OMECs. In contrast, the inhibition of INSIG1 in expression had the opposite effect on activities and triglycerides of OMECs with overexpressed INSIG1. A rescue experiment revealed that circ_015343 alleviated the inhibitory effect of miR-25 on the mRNA and protein abundance of INSIG1. These results indicate that circ_015343 sponges miR-25 to inhibit the activities and content of triglycerides of OMECs by upregulating the expression of INSIG1 in OMECs. This study provided new insights for understanding the genetic molecular mechanism of lactation traits in sheep.
Collapse
Affiliation(s)
- Xinmiao Wu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jiqing Wang
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zhiyun Hao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Huimin Zhen
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiu Liu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shaobin Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Fangfang Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Mingna Li
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Bingang Shi
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Chunyan Ren
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
2
|
Venkatachalam A, Correia C, Peterson KL, Hou X, Schneider PA, Strathman AR, Flatten KS, Sine CC, Balczewski EA, McGehee CD, Larson MC, Duffield LN, Meng XW, Vincelette ND, Ding H, Oberg AL, Couch FJ, Swisher EM, Li H, Weroha SJ, Kaufmann SH. Proapoptotic activity of JNK-sensitive BH3-only proteins underpins ovarian cancer response to replication checkpoint inhibitors. Mol Cancer 2024; 23:224. [PMID: 39375715 PMCID: PMC11457406 DOI: 10.1186/s12943-024-02125-5] [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: 05/23/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Abstract
Recent studies indicate that replication checkpoint modulators (RCMs) such as inhibitors of CHK1, ATR, and WEE1 have promising monotherapy activity in solid tumors, including platinum-resistant high grade serous ovarian cancer (HGSOC). However, clinical response rates are generally below 30%. While RCM-induced DNA damage has been extensively examined in preclinical and clinical studies, the link between replication checkpoint interruption and tumor shrinkage remains incompletely understood. Here we utilized HGSOC cell lines and patient-derived xenografts (PDXs) to study events leading from RCM treatment to ovarian cancer cell death. These studies show that RCMs increase CDC25A levels and CDK2 signaling in vitro, leading to dysregulated cell cycle progression and increased replication stress in HGSOC cell lines independent of homologous recombination status. These events lead to sequential activation of JNK and multiple BH3-only proteins, including BCL2L11/BIM, BBC3/PUMA and the BMF, all of which are required to fully initiate RCM-induced apoptosis. Activation of the same signaling pathway occurs in HGSOC PDXs that are resistant to poly(ADP-ribose) polymerase inhibitors but respond to RCMs ex vivo with a decrease in cell number in 3-dimensional culture and in vivo with xenograft shrinkage or a significantly diminished growth rate. These findings identify key cell death-initiating events that link replication checkpoint inhibition to antitumor response in ovarian cancer.
Collapse
Affiliation(s)
- Annapoorna Venkatachalam
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Cristina Correia
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Kevin L Peterson
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Xianon Hou
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Paula A Schneider
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Annabella R Strathman
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Karen S Flatten
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Chance C Sine
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Emily A Balczewski
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Present Address: Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Cordelia D McGehee
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Melissa C Larson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Laura N Duffield
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - X Wei Meng
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Nicole D Vincelette
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Present Address: H. Lee Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Husheng Ding
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Fergus J Couch
- Division of Experimental Pathology, Department of Laboratory Medicine, and Pathology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Elizabeth M Swisher
- Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195, USA
| | - Hu Li
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - S John Weroha
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA.
- Department of Oncology, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA.
- Division of Hematology, Department of Medicine, Mayo Clinic, 200 First Street, S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
3
|
Wang H, Wang X, Wang L, Wang H, Zhang Y. Plant‐Derived Phytochemicals and Their Nanoformulations for Inducing Programed Cell Death in Cancer. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202400197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Indexed: 01/05/2025]
Abstract
AbstractPhytochemicals are a diverse class of compounds found in various plant‐based foods and beverages that have displayed the capacity to exert powerful anticancer effects through the induction of programed cell death (PCD) in malignancies. PCD is a sophisticated process that maintains in upholding tissue homeostasis and eliminating injured or neoplastic cells. Phytochemicals have shown the potential to induce PCD in malignant cells through various mechanisms, including modulation of cell signaling pathways, regulation of reactive oxygen species (ROS), and interaction with critical targets in cells such as DNA. Moreover, recent studies have suggested that nanomaterials loaded with phytochemicals may enhance cell death in tumors, which can also stimulate antitumor immunity. In this review, a comprehensive overview of the current understanding of the anticancer effects of phytochemicals and their potential as a promising approach to cancer therapy, is provided. The impacts of phytochemicals such as resveratrol, curcumin, apigenin, quercetin, and some approved plant‐derived drugs, such as taxanes on the regulation of some types of PCD, including apoptosis, pyroptosis, anoikis, autophagic cell death, ferroptosis, and necroptosis, are discussed. The underlying mechanisms and the potential of nanomaterials loaded with phytochemicals to enhance PCD in tumors are also explained.
Collapse
Affiliation(s)
- Haoyu Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Xiaoyang Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Long Wang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| | - Haifan Wang
- Department of Orthopedics The Second Affiliated Hospital Xi'an Jiaotong University Xi'an Shaanxi 710004 China
| | - Yuxing Zhang
- Medical College Xijing University Xi'an Shaanxi 710123 China
| |
Collapse
|
4
|
Han Z, Huang Q, Lv M, Ma M, Zhang W, Feng W, Hu R, Sun X, Li J, Zhong X, Zhou X. Qizhu Anti-Cancer Recipe promotes anoikis of hepatocellular carcinoma cells by activating the c-Jun N-terminal kinase pathway. Heliyon 2023; 9:e22089. [PMID: 38053871 PMCID: PMC10694164 DOI: 10.1016/j.heliyon.2023.e22089] [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: 08/17/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023] Open
Abstract
Background Qizhu Anti-Cancer Recipe (QACR) is a traditional Chinese medicine widely used in treating several liver diseases. However, its function and the relevant mechanism underlying its effect in treating hepatocellular carcinoma (HCC) remain unknown. The aim of this study was to explore the effect of QACR in HCC, which are expected to be a potential therapeutic scheme for HCC. Materials and methods The chemical compositions of QACR were determined by liquid chromatography/quadrupole time-of-fight mass spectrometry (LC-QTOF-MS). The anoikis-resistant HCC cell proliferation and angiopoiesis were detected using the cell counting kit 8 (CCK8) assay, trypan blue, calcein AM/EthD-1, flow cytometer, Western blot, and tube formation assays. An orthotopic xenograft mouse model was established to evaluate the in vivo effects of the QACR. The expression of proliferating cell nuclear antigen (PCNA), Bcl-2, CD31, caspase-3, caspase-8, caspase-9, PARP-1, DFF40, phospho-c-Jun NH2-terminal kinase (p-JNK), and JNK was assessed using Western blot and immunohistochemical analysis. Results QACR reduced the growth and tube formation of anoikis-resistant HCC cells and enhanced cell apoptosis in vitro. In the orthotopic xenograft mouse models, QACR suppressed the tumorigenesis of HCC in vivo. Mechanistically, QACR modulated the JNK pathway. The JNK inhibitor (SP600125) reverses the inhibitory effects of QACR on anoikis-resistant HCC cell proliferation and angiopoiesis. Conclusion Our study suggests that QACR suppresses the proliferation and angiopoiesis of anoikis-resistant HCC cells by activating the JNK pathway. Therefore, QACR is a promising new therapeutic strategy for treating hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zhiyi Han
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Qi Huang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Mengqing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Wei Zhang
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Wenxing Feng
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Rui Hu
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Xinfeng Sun
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Jing Li
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Xin Zhong
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| | - Xiaozhou Zhou
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, No.1 Fuhua Road, Futian District, Shenzhen, 518000, China
- Department of Liver Disease, the Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1 Fuhua Road, Futian District, Shenzhen, 518000, China
| |
Collapse
|
5
|
Empitu MA, Kikyo M, Shirata N, Yamada H, Makino SI, Kadariswantiningsih IN, Aizawa M, Patrakka J, Nishimori K, Asanuma K. Inhibition of Importin- α -Mediated Nuclear Localization of Dendrin Attenuates Podocyte Loss and Glomerulosclerosis. J Am Soc Nephrol 2023; 34:1222-1239. [PMID: 37134307 PMCID: PMC10356163 DOI: 10.1681/asn.0000000000000150] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
SIGNIFICANCE STATEMENT Nuclear translocation of dendrin is observed in injured podocytes, but the mechanism and its consequence are unknown. In nephropathy mouse models, dendrin ablation attenuates proteinuria, podocyte loss, and glomerulosclerosis. The nuclear translocation of dendrin promotes c-Jun N -terminal kinase phosphorylation in podocytes, altering focal adhesion and enhancing cell detachment-induced apoptosis. We identified mediation of dendrin nuclear translocation by nuclear localization signal 1 (NLS1) sequence and adaptor protein importin- α . Inhibition of importin- α prevents nuclear translocation of dendrin, decreases podocyte loss, and attenuates glomerulosclerosis in nephropathy models. Thus, inhibiting importin- α -mediated nuclear translocation of dendrin is a potential strategy to halt podocyte loss and glomerulosclerosis. BACKGROUND Nuclear translocation of dendrin is observed in the glomeruli in numerous human renal diseases, but the mechanism remains unknown. This study investigated that mechanism and its consequence in podocytes. METHODS The effect of dendrin deficiency was studied in adriamycin (ADR) nephropathy model and membrane-associated guanylate kinase inverted 2 ( MAGI2 ) podocyte-specific knockout ( MAGI2 podKO) mice. The mechanism and the effect of nuclear translocation of dendrin were studied in podocytes overexpressing full-length dendrin and nuclear localization signal 1-deleted dendrin. Ivermectin was used to inhibit importin- α . RESULTS Dendrin ablation reduced albuminuria, podocyte loss, and glomerulosclerosis in ADR-induced nephropathy and MAGI2 podKO mice. Dendrin deficiency also prolonged the lifespan of MAGI2 podKO mice. Nuclear dendrin promoted c-Jun N -terminal kinase phosphorylation that subsequently altered focal adhesion, reducing cell attachment and enhancing apoptosis in cultured podocytes. Classical bipartite nuclear localization signal sequence and importin- α mediate nuclear translocation of dendrin. The inhibition of importin- α / β reduced dendrin nuclear translocation and apoptosis in vitro as well as albuminuria, podocyte loss, and glomerulosclerosis in ADR-induced nephropathy and MAGI2 podKO mice. Importin- α 3 colocalized with nuclear dendrin in the glomeruli of FSGS and IgA nephropathy patients. CONCLUSIONS Nuclear translocation of dendrin promotes cell detachment-induced apoptosis in podocytes. Therefore, inhibiting importin- α -mediated dendrin nuclear translocation is a potential strategy to prevent podocyte loss and glomerulosclerosis.
Collapse
Affiliation(s)
- Maulana A. Empitu
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Mitsuhiro Kikyo
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Kanagawa, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naritoshi Shirata
- Sohyaku, Innovative Research Division, Mitsubishi Tanabe Pharmaceutical Corporation, Kanagawa, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Yamada
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Nephrology, Kyoto University Hospital, Kyoto, Japan
| | - Shin-ichi Makino
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Nephrology, Kyoto University Hospital, Kyoto, Japan
| | - Ika N. Kadariswantiningsih
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Faculty of Medicine, Airlangga University, Surabaya, Indonesia
| | - Masashi Aizawa
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jaakko Patrakka
- Karolinska Institute/AstraZeneca Integrated Cardio Metabolic Center (ICMC), Huddinge, Sweden
- Division of Pathology, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Huddinge, Sweden
| | - Katsuhiko Nishimori
- Department of Bioregulation and Pharmacological Medicine and Department of Obesity and Internal Inflammation, Fukushima Medical University, Fukushima, Japan
| | - Katsuhiko Asanuma
- Department of Nephrology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Medical Innovation Center, TMK Project, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
6
|
Jiang Y, Song L, Lin Y, Nowialis P, Gao Q, Li T, Li B, Mao X, Song Q, Xing C, Zheng G, Huang S, Jin L. ROS-mediated SRMS activation confers platinum resistance in ovarian cancer. Oncogene 2023; 42:1672-1684. [PMID: 37020040 PMCID: PMC10231978 DOI: 10.1038/s41388-023-02679-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023]
Abstract
Ovarian cancer is the leading cause of death among gynecological malignancies. Checkpoint blockade immunotherapy has so far only shown modest efficacy in ovarian cancer and platinum-based chemotherapy remains the front-line treatment. Development of platinum resistance is one of the most important factors contributing to ovarian cancer recurrence and mortality. Through kinome-wide synthetic lethal RNAi screening combined with unbiased datamining of cell line platinum response in CCLE and GDSC databases, here we report that Src-Related Kinase Lacking C-Terminal Regulatory Tyrosine And N-Terminal Myristylation Sites (SRMS), a non-receptor tyrosine kinase, is a novel negative regulator of MKK4-JNK signaling under platinum treatment and plays an important role in dictating platinum efficacy in ovarian cancer. Suppressing SRMS specifically sensitizes p53-deficient ovarian cancer cells to platinum in vitro and in vivo. Mechanistically, SRMS serves as a "sensor" for platinum-induced ROS. Platinum treatment-induced ROS activates SRMS, which inhibits MKK4 kinase activity by directly phosphorylating MKK4 at Y269 and Y307, and consequently attenuates MKK4-JNK activation. Suppressing SRMS leads to enhanced MKK4-JNK-mediated apoptosis by inhibiting MCL1 transcription, thereby boosting platinum efficacy. Importantly, through a "drug repurposing" strategy, we uncovered that PLX4720, a small molecular selective inhibitor of B-RafV600E, is a novel SRMS inhibitor that can potently boost platinum efficacy in ovarian cancer in vitro and in vivo. Therefore, targeting SRMS with PLX4720 holds the promise to improve the efficacy of platinum-based chemotherapy and overcome chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Yunhan Jiang
- Department of Molecular Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Lina Song
- Department of Molecular Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Yizhu Lin
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Pawel Nowialis
- Department of Molecular Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Qiongmei Gao
- Department of Molecular Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Tao Li
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Bin Li
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Xiaobo Mao
- Institute for Cell Engineering, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Shuang Huang
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Lingtao Jin
- Department of Molecular Medicine, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
7
|
Abstract
The HER2+ subtype of human breast cancer is associated with the malignant transformation of luminal ductal cells of the mammary epithelium. The sequence analysis of tumor DNA identifies loss of function mutations and deletions of the MAP2K4 and MAP2K7 genes that encode direct activators of the JUN NH2-terminal kinase (JNK). We report that in vitro studies of human mammary epithelial cells with CRISPR-induced mutations in the MAPK and MAP2K components of the JNK pathway caused no change in growth in 2D culture, but these mutations promoted epithelial cell proliferation in 3D culture. Analysis of gene expression signatures in 3D culture demonstrated similar changes caused by HER2 activation and JNK pathway loss. The mechanism of signal transduction cross-talk may be mediated, in part, by JNK-suppressed expression of integrin α6β4 that binds HER2 and amplifies HER2 signaling. These data suggest that HER2 activation and JNK pathway loss may synergize to promote breast cancer. To test this hypothesis, we performed in vivo studies using a mouse model of HER2+ breast cancer with Cre/loxP-mediated ablation of genes encoding JNK (Mapk8 and Mapk9) and the MAP2K (Map2k4 and Map2k7) that activate JNK in mammary epithelial cells. Kaplan-Meier analysis of tumor development demonstrated that JNK pathway deficiency promotes HER2+-driven breast cancer. Collectively, these data identify JNK pathway genes as potential suppressors for HER2+ breast cancer.
Collapse
|
8
|
Snedden M, Singh L, Kyathanahalli C, Hirsch E. Toxic effects of trace phenol/guanidine isothiocyanate (P/GI) on cells cultured nearby in covered 96-well plates. BMC Biotechnol 2022; 22:35. [PMID: 36434619 PMCID: PMC9700959 DOI: 10.1186/s12896-022-00766-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/08/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND A mixture of phenol and guanidine isothiocyanate ("P/GI", the principal components of TRIzol™ and similar products) is routinely used to isolate RNA, DNA, and proteins from a single specimen. In time-course experiments of cells grown in tissue culture, replicate wells are often harvested sequentially and compared, with the assumption that in-well lysis and complete aspiration of P/GI has no effect on continuing cultures in nearby wells. METHODS To test this assumption, we investigated morphology and function of RAW 264.7 cells (an immortalized mouse macrophage cell line) cultured in covered 96-well plates for 4, 8, or 24 h at varying distances from a single control well or a well into which P/GI had been deposited and immediately aspirated completely. RESULTS Time- and distance-dependent disruptions resulting from proximity to a single well containing trace residual P/GI were seen in cell morphology (blebbing, cytoplasmic disruption, and accumulation of intracellular vesicles), cell function (pH of culture medium), and expression of genes related to inflammation (Tnfα) and autophagy (Lc3b). There was no transcriptional change in the anti-apoptotic gene Mcl1, nor the pro-apoptotic gene Hrk, nor in P/GI-unexposed control cultures. LPS-stimulated cells incubated near P/GI had lower expression of the cytokine Il6. These effects were seen as early as 4 h of exposure and at a distance of up to 3 well units from the P/GI-exposed well. CONCLUSIONS Exposure to trace residual quantities of P/GI in covered tissue culture plates leads to substantial disruption of cell morphology and function in as little as 4 h, possibly through induction of autophagy but not apoptosis. This phenomenon should be considered when planning time-course experiments in multi-well covered tissue culture plates.
Collapse
Affiliation(s)
- Madeline Snedden
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, 2650 Ridge Ave, Suite 1538, Evanston, IL 60201 USA
| | - Lavisha Singh
- Department of Statistics, NorthShore University HealthSystem, Evanston, IL USA
| | - Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, 2650 Ridge Ave, Suite 1538, Evanston, IL 60201 USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, IL USA
| | - Emmet Hirsch
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, 2650 Ridge Ave, Suite 1538, Evanston, IL 60201 USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, IL USA
| |
Collapse
|
9
|
Valencia-Expósito A, Gómez-Lamarca MJ, Widmann TJ, Martín-Bermudo MD. Integrins Cooperate With the EGFR/Ras Pathway to Preserve Epithelia Survival and Architecture in Development and Oncogenesis. Front Cell Dev Biol 2022; 10:892691. [PMID: 35769262 PMCID: PMC9234701 DOI: 10.3389/fcell.2022.892691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Adhesion to the extracellular matrix (ECM) is required for normal epithelial cell survival. Disruption of this interaction leads to a specific type of apoptosis known as anoikis. Yet, there are physiological and pathological situations in which cells not connected to the ECM are protected from anoikis, such as during cell migration or metastasis. The main receptors transmitting signals from the ECM are members of the integrin family. However, although integrin-mediated cell-ECM anchorage has been long recognized as crucial for epithelial cell survival, the in vivo significance of this interaction remains to be weighed. In this work, we have used the Drosophila wing imaginal disc epithelium to analyze the importance of integrins as survival factors during epithelia morphogenesis. We show that reducing integrin expression in the wing disc induces caspase-dependent cell death and basal extrusion of the dead cells. In this case, anoikis is mediated by the activation of the JNK pathway, which in turn triggers expression of the proapoptotic protein Hid. In addition, our results strongly suggest that, during wing disc morphogenesis, the EGFR pathway protects cells undergoing cell shape changes upon ECM detachment from anoikis. Furthermore, we show that oncogenic activation of the EGFR/Ras pathway in integrin mutant cells rescues them from apoptosis while promoting their extrusion from the epithelium. Altogether, our results support the idea that integrins promote cell survival during normal tissue morphogenesis and prevent the extrusion of transformed cells.
Collapse
Affiliation(s)
| | - M. Jesús Gómez-Lamarca
- Centro Andaluz de Biología del Desarrollo CSIC-Universidad Pablo de Olavide, Sevilla, Spain
- Departamento de Biología Celular, Universidad de Sevilla, Sevilla, Spain
| | | | - María D. Martín-Bermudo
- Centro Andaluz de Biología del Desarrollo CSIC-Universidad Pablo de Olavide, Sevilla, Spain
- *Correspondence: María D. Martín-Bermudo,
| |
Collapse
|
10
|
Non-canonical phosphorylation of Bmf by p38 MAPK promotes its apoptotic activity in anoikis. Cell Death Differ 2022; 29:323-336. [PMID: 34462553 PMCID: PMC8817011 DOI: 10.1038/s41418-021-00855-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 02/03/2023] Open
Abstract
Bmf contributes to the onset of anoikis by translocating from cytoskeleton to mitochondria when cells lose attachment to the extracellular matrix. However, the structural details of Bmf cytoskeleton tethering and the control of Bmf release upon loss of anchorage remained unknown. Here we showed that cell detachment induced rapid and sustained activation of p38 MAPK in mammary epithelial cell lines. Inhibition of p38 signaling or Bmf knockdown rescued anoikis. Activated p38 MAPK could directly phosphorylate Bmf at multiple sites including a non-proline-directed site threonine 72 (T72). Crystallographic studies revealed that Bmf T72 directly participated in DLC2 binding and its phosphorylation would block Bmf/DLC2 interaction through steric hindrance. Finally, we showed that phosphomimetic mutation of T72 enhanced Bmf apoptotic activity in vitro and in a knock-in mouse model. This work unraveled a novel regulatory mechanism of Bmf activity during anoikis and provided structural basis for Bmf cytoskeleton tethering and dissociation.
Collapse
|
11
|
Ye F, Dan G, Zhao Y, Yu W, Cheng J, Chen M, Sai Y, Zou Z. Small-interfering RNA for c-Jun attenuates cell death by preventing JNK-dependent PARP1 cleavage and DNA fragmentation in nitrogen mustard-injured immortalized human bronchial epithelial cells. Toxicol Res (Camb) 2021; 10:1034-1044. [PMID: 34733488 DOI: 10.1093/toxres/tfab081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 11/14/2022] Open
Abstract
Sulfur mustard (a type of vesicant) can directly damage lung bronchial epithelium via aerosol inhalation, and prevalent cell death is an early event that obstructs the respiratory tract. JNK/c-Jun is a stress response pathway, but its role in cell death of the injured cells is not clear. Here, we report that JNK/c-Jun was activated in immortalized human bronchial epithelial (HBE) cells exposed to a lethal dose (20 μM) of nitrogen mustard (NM, a sulfur mustard analog). c-Jun silencing using small-interfering RNA (siRNA) rendered the cells resistant to NM-mediated cell death by blocking poly(ADP-ribose) polymerase 1 (PARP1) cleavage and DNA fragmentation. In addition, the transduction of upstream extrinsic (Fasl-Fas-caspase-8) and intrinsic (loss of Bcl-2 and mitochondrial membrane potential, ΔΨm) apoptosis pathways, as well as phosphorylated (p)-H2AX (Ser139), an epigenetic marker contributing to DNA fragmentation and PARP1 activity, was partially suppressed. To mimic the detachment of cells by NM, HBE cells were trypsinized and seeded on culture plates that were pre-coated with poly-HEMA to prevent cell adhesion. The JNK/c-Jun pathway was found to be activated in the detached cells. In conclusion, our results indicate that JNK/c-Jun pathway activation is necessary for NM-caused HBE cell death and further suggest that c-Jun silencing may be a potential approach to protect HBE cells from vesicant damage.
Collapse
Affiliation(s)
- Feng Ye
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Guorong Dan
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuanpeng Zhao
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wenpei Yu
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jin Cheng
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mingliang Chen
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yan Sai
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhongmin Zou
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
12
|
Vaz S, Ferreira FJ, Macedo JC, Leor G, Ben-David U, Bessa J, Logarinho E. FOXM1 repression increases mitotic death upon antimitotic chemotherapy through BMF upregulation. Cell Death Dis 2021; 12:542. [PMID: 34035233 PMCID: PMC8149823 DOI: 10.1038/s41419-021-03822-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 11/28/2022]
Abstract
Inhibition of spindle microtubule (MT) dynamics has been effectively used in cancer treatment. Although the mechanisms by which MT poisons elicit mitotic arrest are fairly understood, efforts are still needed towards elucidating how cancer cells respond to antimitotic drugs owing to cytotoxicity and resistance side effects. Here, we identified the critical G2/M transcription factor Forkhead box M1 (FOXM1) as a molecular determinant of cell response to antimitotics. We found FOXM1 repression to increase death in mitosis (DiM) due to upregulation of the BCL-2 modifying factor (BMF) gene involved in anoikis, an apoptotic process induced upon cell detachment from the extracellular matrix. FOXM1 binds to a BMF intronic cis-regulatory element that interacts with both the BMF and the neighbor gene BUB1B promoter regions, to oppositely regulate their expression. This mechanism ensures that cells treated with antimitotics repress BMF and avoid DiM when FOXM1 levels are high. In addition, we show that this mechanism is partly disrupted in anoikis/antimitotics-resistant tumor cells, with resistance correlating with lower BMF expression but in a FOXM1-independent manner. These findings provide a stratification biomarker for antimitotic chemotherapy response.
Collapse
Affiliation(s)
- Sara Vaz
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Aging and Aneuploidy Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.,Programa doutoral em Biologia Molecular e Celular, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313, Porto, Portugal
| | - Fábio J Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Aging and Aneuploidy Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.,Vertebrate Development and Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.,Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313, Porto, Portugal
| | - Joana C Macedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Aging and Aneuploidy Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
| | - Gil Leor
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ben-David
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - José Bessa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal.,Vertebrate Development and Regeneration Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal
| | - Elsa Logarinho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal. .,Aging and Aneuploidy Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
13
|
Zeineh N, Nagler RM, Gabay M, Obeid F, Kahana M, Weizman A, Gavish M. The TSPO Ligands MGV-1 and 2-Cl-MGV-1 Differentially Inhibit the Cigarette Smoke-Induced Cytotoxicity to H1299 Lung Cancer Cells. BIOLOGY 2021; 10:biology10050395. [PMID: 34063262 PMCID: PMC8147464 DOI: 10.3390/biology10050395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/17/2021] [Accepted: 04/29/2021] [Indexed: 12/03/2022]
Abstract
Simple Summary In this study, we investigated the impact of CS on various TSPO-related mitochondrial processes, and the protective ability of our novel TSPO ligands against such CS-induced cellular damages. Our results support the previously reported role of TSPO in apoptotic cell death. Moreover, the present data demonstrate the protective effect of our TSPO ligands against CS-induced cellular damage. Abstract TSPO is involved in cigarette smoke (CS)-induced cellular toxicity, which may result in oral and pulmonary diseases and lung cancer. H1299 lung cancer cells were exposed directly to CS. The H1299 cells were pretreated with our TSPO ligands MGV-1 and 2-Cl-MGV-1 (Ki = 825 nM for both) at a concentration of 25 µM 24 h prior to CS exposure. Cell death and apoptotic markers were measured, in addition to TSPO expression levels, ATP synthase activity, generation of reactive oxygen species (ROS), depolarization of mitochondrial membrane potential (ΔΨm), cAMP and LDH levels. Pretreatment with MGV-1 and 2-Cl-MGV-1 (25 µM), 24 h prior to CS exposure, differentially attenuated the CS-induced cellular insult as well as cell death in H1299 lung cancer cells. These protective effects included prevention of ATP synthase reversal, ROS generation, depolarization of the mitochondrial membrane and elevation in LDH. The preventive efficacy of 2-Cl-MGV-1 was superior to that achieved by MGV-1. Both ligands did not prevent the elevation in cAMP. These findings may indicate a mild protective effect of these TSPO ligands in CS-related pulmonary and keratinocyte cellular pathology.
Collapse
Affiliation(s)
- Nidal Zeineh
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel; (N.Z.); (R.M.N.); (M.G.); (F.O.); (M.K.)
| | - Rafael M. Nagler
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel; (N.Z.); (R.M.N.); (M.G.); (F.O.); (M.K.)
| | - Martin Gabay
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel; (N.Z.); (R.M.N.); (M.G.); (F.O.); (M.K.)
| | - Fadi Obeid
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel; (N.Z.); (R.M.N.); (M.G.); (F.O.); (M.K.)
| | - Meygal Kahana
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel; (N.Z.); (R.M.N.); (M.G.); (F.O.); (M.K.)
| | - Abraham Weizman
- Research Unit, Geha Mental Health Center and Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva 4910002, Israel;
- Departments of Psychiatry, Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Moshe Gavish
- Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel; (N.Z.); (R.M.N.); (M.G.); (F.O.); (M.K.)
- Correspondence: ; Tel.: +972-4829-5275; Fax: +972-4829-5330
| |
Collapse
|
14
|
Stulpinas A, Uzusienis T, Imbrasaite A, Krestnikova N, Unguryte A, Kalvelyte AV. Cell-cell and cell-substratum contacts in the regulation of MAPK and Akt signalling: Importance in therapy, biopharmacy and bioproduction. Cell Signal 2021; 84:110034. [PMID: 33933583 DOI: 10.1016/j.cellsig.2021.110034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022]
Abstract
The use of cultured cells as a tool for research, precision medicine, biopharmacy, and biomanufacturing is constantly increasing. In parallel, the role of cell-cell and cell-substratum contacts in cell functioning is increasingly validated. Adhesion signalling plays a key role here. The activity of cell fate-regulating signalling molecules is an important factor in determining cell behaviour, as well as their response to treatment, depending on cell phenotypic status and location in the body. Three cellular state models (adherent, single cells in suspension, and aggregated cells) were compared for cell signalling, including focal adhesion (FAK), mitogen-activated (MAPK), as well as Akt protein kinases, and transcription factor cJun, by using lung adenocarcinoma A549, muscle-derived stem Myo, as well as primary lung cancer cell lines. Survival of both A549 and Myo cells was dependent on kinases Akt and ERK in detached conditions. Intercellular contacts in aggregates promoted activation of Akt and ERK, and cell survival. Loss of contacts with the substrate increased phosphorylation of MAP kinases JNK and p38, while decreased Akt phosphorylation by processes involving FAK. Unexpectedly, detachment increased phosphorylation of antiapoptotic kinase ERK in A549, while in Myo stem cells ERK phosphorylation was downregulated. JNK target transcription factor cJun protein level was markedly diminished by contacts between cells possibly involving mechanism of proteasomal degradation. Furthermore, studies revealed the opposite dependence of molecules of the same signalling pathway - phospho-cJun and phospho-JNK - on cell culture density. Differences in ERK activation under detachment conditions indicate that targeting of prosurvival kinases during anoikis should be different in different cells. Moreover, the outcome of JNK activation in cells may depend on the amount of cJun, which is determined by cell-cell contacts.
Collapse
Affiliation(s)
- Aurimas Stulpinas
- Dept. of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Centre, Vilnius University, Saulėtekio al. 7, LT-10257, Lithuania
| | - Tomas Uzusienis
- Dept. of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Centre, Vilnius University, Saulėtekio al. 7, LT-10257, Lithuania
| | - Ausra Imbrasaite
- Dept. of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Centre, Vilnius University, Saulėtekio al. 7, LT-10257, Lithuania
| | - Natalija Krestnikova
- Dept. of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Centre, Vilnius University, Saulėtekio al. 7, LT-10257, Lithuania
| | - Ausra Unguryte
- Centre for Innovative Medicine, Santariškių g. 5, LT-08406, Lithuania
| | - Audrone V Kalvelyte
- Dept. of Molecular Cell Biology, Institute of Biochemistry, Life Sciences Centre, Vilnius University, Saulėtekio al. 7, LT-10257, Lithuania.
| |
Collapse
|
15
|
Herrera-Melle L, Crespo M, Leiva M, Sabio G. Stress-activated kinases signaling pathways in cancer development. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2020.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
16
|
Madan E, Peixoto ML, Dimitrion P, Eubank TD, Yekelchyk M, Talukdar S, Fisher PB, Mi QS, Moreno E, Gogna R. Cell Competition Boosts Clonal Evolution and Hypoxic Selection in Cancer. Trends Cell Biol 2020; 30:967-978. [PMID: 33160818 DOI: 10.1016/j.tcb.2020.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
The comparison of fitness between cells leads to the elimination of less competent cells in the presence of more competent neighbors via cell competition (CC). This phenomenon has been linked with several cancer-related genes and thus may play an important role in cancer. Various processes are involved in the regulation of tumor initiation and growth, including tumor hypoxia, clonal stem cell selection, and immune cell response, all of which have been recently shown to have a potential connection with the mechanisms involved in CC. This review aims to unravel the relation between these processes and competitive cell interactions and how this affects disease progression.
Collapse
Affiliation(s)
- Esha Madan
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | | | - Peter Dimitrion
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI, USA
| | - Timothy D Eubank
- In Vivo Multifunctional Magnetic Resonance Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA; Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Michail Yekelchyk
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Sarmistha Talukdar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, USA; Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, USA; Department of Biochemistry, Microbiology and Immunology, Wayne State University Medical School, Detroit, MI, USA
| | - Eduardo Moreno
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal.
| | - Rajan Gogna
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal.
| |
Collapse
|
17
|
Identification and Characterization of MAPK Signaling Pathway Genes and Associated lncRNAs in the Ileum of Piglets Infected by Clostridium perfringens Type C. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8496872. [PMID: 32855971 PMCID: PMC7443001 DOI: 10.1155/2020/8496872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/16/2020] [Accepted: 07/17/2020] [Indexed: 01/18/2023]
Abstract
Clostridium perfringens type C (C. perfringens type C) is one of the main microbial pathogens responsible for piglet diarrhea worldwide, causing substantial economic losses for pig-rearing industries. The mitogen-activated protein kinase (MAPK) signaling pathway is a key regulator of inflammatory bowel disease, especially necrotic enteritis. However, whether and how the MAPK signaling pathway is involved in regulating the process of piglet diarrhea when challenged by C. perfringens type C are still unknown. Here, we screened 38 differentially expressed genes (DEGs) in piglets' ileum tissues experimentally infected with C. perfringens type C that were enriched in the Sus scrofa MAPK signaling pathway, based on our previous transcriptome data. Of these DEGs, 12 genes (TRAF2, MAPK8, and GADD45G, among others) were upregulated whereas 26 genes (MAPK1, TP53, and CHUK, among others) were downregulated in the infected group. Our results showed that MAPK1, TP53, MAPK8, MYC, and CHUK were in the core nodes of the PPI network. Additionally, we obtained 35 lncRNAs from the sequencing data, which could be trans-targeted to MAPK signaling pathway genes and were differentially expressed in the ileum tissues infected with C. perfringens. We used qRT-PCR to verify the expression levels of genes and lncRNAs related to the MAPK signaling pathway; their expression patterns were consistent with RNA sequencing data. Our results provide strong support for deeply exploring the role of the MAPK signaling pathway in diarrhea caused by C. perfringens type C.
Collapse
|
18
|
UFM1-Specific Ligase 1 Ligating Enzyme 1 Mediates Milk Protein and Fat Synthesis-Related Gene Expression via the JNK Signaling Pathway in Mouse Mammary Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4045674. [PMID: 32655766 PMCID: PMC7321527 DOI: 10.1155/2020/4045674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/04/2020] [Accepted: 04/30/2020] [Indexed: 01/25/2023]
Abstract
Ubiquitin-like modifier 1 ligating enzyme 1 (UFL1) has been characterized as a ubiquitin-like (Ubl) protein that affects a range of cellular processes across various pathways. In this study, mouse mammary epithelial cells (HC11 cell line) and UFL1 knockout (KO) mice were used to establish UFL1 knockdown models to explore the influence of UFL1 on milk protein and fat synthesis in the mouse mammary gland and the underlying mechanisms. This is the first study to show UFL1 localization in mouse mammary epithelial cells. UFL1 depletion by transfected UFL1 siRNA (siUFL1) caused aggravated apoptosis. In addition, UFL1 depletion suppressed milk protein synthesis-related protein level in vivo and in vitro. Conversely, ACACA and FASN expressions increased in UFL1-deficient mice. Moreover, UFL1 depletion increased triglyceride synthesis levels and inhibited the p-JNK expression. Importantly, the expression of proteins related to milk protein synthesis was decreased in JNK- and UFL1-deficient cells, whereas proteins related to milk fat synthesis showed the opposite trend, indicating that UFL1 affects milk protein and fat synthesis via the suppression of JNK activation. Overall, our findings indicate that UFL1 plays a key role in mammary milk and fat synthesis via JNK activation.
Collapse
|
19
|
Pu Y, Liu YQ, Zhou Y, Qi YF, Liao SP, Miao SK, Zhou LM, Wan LH. Dual role of RACK1 in airway epithelial mesenchymal transition and apoptosis. J Cell Mol Med 2020; 24:3656-3668. [PMID: 32064783 PMCID: PMC7131927 DOI: 10.1111/jcmm.15061] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/04/2020] [Accepted: 01/21/2020] [Indexed: 02/05/2023] Open
Abstract
Airway epithelial apoptosis and epithelial mesenchymal transition (EMT) are two crucial components of asthma pathogenesis, concomitantly mediated by TGF‐β1. RACK1 is the downstream target gene of TGF‐β1 shown to enhancement in asthma mice in our previous study. Balb/c mice were sensitized twice and challenged with OVA every day for 7 days. Transformed human bronchial epithelial cells, BEAS‐2B cells were cultured and exposed to recombinant soluble human TGF‐β1 to induced apoptosis (30 ng/mL, 72 hours) and EMT (10 ng/mL, 48 hours) in vitro, respectively. siRNA and pharmacological inhibitors were used to evaluate the regulation of RACK1 protein in apoptosis and EMT. Western blotting analysis and immunostaining were used to detect the protein expressions in vivo and in vitro. Our data showed that RACK1 protein levels were significantly increased in OVA‐challenged mice, as well as TGF‐β1‐induced apoptosis and EMT of BEAS‐2B cells. Knockdown of RACK1 (siRACK1) significantly inhibited apoptosis and decreased TGF‐β1 up‐regulated EMT related protein levels (N‐cadherin and Snail) in vitro via suppression of JNK and Smad3 activation. Moreover, siSmad3 or siJNK impaired TGF‐β1‐induced N‐cadherin and Snail up‐regulation in vitro. Importantly, JNK gene silencing (siERK) also impaired the regulatory effect of TGF‐β1 on Smad3 activation. Our present data demonstrate that RACK1 is a concomitant regulator of TGF‐β1 induces airway apoptosis and EMT via JNK/Smad/Snail signalling axis. Our findings may provide a new insight into understanding the regulation mechanism of RACK1 in asthma pathogenesis.
Collapse
Affiliation(s)
- Yue Pu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yuan-Qi Liu
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Yan Zhou
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yi-Fan Qi
- Grade 2015, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Shi-Ping Liao
- Functional Laboratory, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Shi-Kun Miao
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Li-Ming Zhou
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| | - Li-Hong Wan
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
20
|
Porter EG, Dhiman A, Chowdhury B, Carter BC, Lin H, Stewart JC, Kazemian M, Wendt MK, Dykhuizen EC. PBRM1 Regulates Stress Response in Epithelial Cells. iScience 2019; 15:196-210. [PMID: 31077944 PMCID: PMC6514269 DOI: 10.1016/j.isci.2019.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 03/10/2019] [Accepted: 04/19/2019] [Indexed: 12/16/2022] Open
Abstract
Polybromo1 (PBRM1) is a chromatin remodeler subunit highly mutated in cancer, particularly clear cell renal carcinoma. PBRM1 is a member of the SWI/SNF subcomplex, PBAF (PBRM1-Brg1/Brm-associated factors), and is characterized by six tandem bromodomains. Here we establish a role for PBRM1 in epithelial cell maintenance through the expression of genes involved in cell adhesion, metabolism, stress response, and apoptosis. In support of a general role for PBRM1 in stress response and apoptosis, we observe that loss of PBRM1 results in an increase in reactive oxygen species generation and a decrease in cellular viability under stress conditions. We find that loss of PBRM1 promotes cell growth under favorable conditions but is required for cell survival under conditions of cellular stress. PBRM1 facilitates the expression of stress response genes in epithelial cells Deletion of PBRM1 promotes growth under low-stress conditions PBRM1 restrains ROS generation and induces apoptosis under high-stress conditions Under H2O2 stress, PBRM1 cooperates with cJun and NRF2 to induce gene expression
Collapse
Affiliation(s)
- Elizabeth G Porter
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Alisha Dhiman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Basudev Chowdhury
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Benjamin C Carter
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Hang Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Jane C Stewart
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Majid Kazemian
- Department of Biochemistry, Purdue University, West Lafayette, IN 47906, USA
| | - Michael K Wendt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA
| | - Emily C Dykhuizen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47906, USA.
| |
Collapse
|
21
|
Oudenaarden CRL, van de Ven RAH, Derksen PWB. Re-inforcing the cell death army in the fight against breast cancer. J Cell Sci 2018; 131:131/16/jcs212563. [DOI: 10.1242/jcs.212563] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
ABSTRACT
Metastatic breast cancer is responsible for most breast cancer-related deaths. Disseminated cancer cells have developed an intrinsic ability to resist anchorage-dependent apoptosis (anoikis). Anoikis is caused by the absence of cellular adhesion, a process that underpins lumen formation and maintenance during mammary gland development and homeostasis. In healthy cells, anoikis is mostly governed by B-cell lymphoma-2 (BCL2) protein family members. Metastatic cancer cells, however, have often developed autocrine BCL2-dependent resistance mechanisms to counteract anoikis. In this Review, we discuss how a pro-apoptotic subgroup of the BCL2 protein family, known as the BH3-only proteins, controls apoptosis and anoikis during mammary gland homeostasis and to what extent their inhibition confers tumor suppressive functions in metastatic breast cancer. Specifically, the role of the two pro-apoptotic BH3-only proteins BCL2-modifying factor (BMF) and BCL2-interacting mediator of cell death (BIM) will be discussed here. We assess current developments in treatment that focus on mimicking the function of the BH3-only proteins to induce apoptosis, and consider their applicability to restore normal apoptotic responses in anchorage-independent disseminating tumor cells.
Collapse
Affiliation(s)
- Clara R. L. Oudenaarden
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Lund University, Department of Experimental Oncology, Scheelevägen 2, 22363 Lund, Sweden
| | - Robert A. H. van de Ven
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
- Harvard Medical School, Department of Cell Biology, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Patrick W. B. Derksen
- UMC Utrecht, Department of Pathology, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| |
Collapse
|
22
|
Girnius N, Edwards YJ, Garlick DS, Davis RJ. The cJUN NH 2-terminal kinase (JNK) signaling pathway promotes genome stability and prevents tumor initiation. eLife 2018; 7:36389. [PMID: 29856313 PMCID: PMC5984035 DOI: 10.7554/elife.36389] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022] Open
Abstract
Breast cancer is the most commonly diagnosed malignancy in women. Analysis of breast cancer genomic DNA indicates frequent loss-of-function mutations in components of the cJUN NH2-terminal kinase (JNK) signaling pathway. Since JNK signaling can promote cell proliferation by activating the AP1 transcription factor, this apparent association of reduced JNK signaling with tumor development was unexpected. We examined the effect of JNK deficiency in the murine breast epithelium. Loss of JNK signaling caused genomic instability and the development of breast cancer. Moreover, JNK deficiency caused widespread early neoplasia and rapid tumor formation in a murine model of breast cancer. This tumor suppressive function was not mediated by a role of JNK in the growth of established tumors, but by a requirement of JNK to prevent tumor initiation. Together, these data identify JNK pathway defects as ‘driver’ mutations that promote genome instability and tumor initiation. As cells in our body grow and divide, their DNA can experience changes or damage. Most of these ‘mutations’ are harmless, or quickly fixed by the body. Yet, sometimes a mutation can trigger a chain of genetic events that drives the cells to multiply uncontrollably, which leads to tumors. Identifying these ‘driver mutations’ is complex, but key to understanding how cancers start and can be fought. Breast cancer is the most common type of cancer diagnosed in women worldwide. Large studies have focused on sequencing the DNA of cancerous breast cells to try to identify the mutations that started the cancer. Results show that, in these cells, a biological mechanism called the JNK signaling pathway is often inactivated because mutations affect the molecules that take part in this process. Like a chain reaction, the proteins of the JNK pathway act on each other until the last one, called JNK, gets switched on. This protein then goes on to participate in a number of cellular processes such as DNA repair. Is it possible that mutations in this pathway actually drive cancer, and if so, how? Girnius et al. addressed these questions by inactivating the JNK pathway in the breast cells of mice. Over the next year and a half, the JNK-deficient animals were more likely to get breast cancer than normal mice. Further experiments showed that, in breast cells, the JNK protein prevented tumors from appearing. However, once the tumors were present, it was less effective at stopping them from growing. The DNA of the breast cancer cells with no JNK protein also contained more genetic changes and mistakes. This suggests that the JNK signaling pathway helps to keep the genetic information ‘healthy’. This may be because, normally, the JNK protein activates processes that fix DNA mutations. Taken together, the results presented by Girnius et al. show that genetic changes which inactivate the JNK pathway can drive the development of breast cancer. Certain anti-cancer drugs kill cancerous cells by damaging their DNA. Breast tumor cells with inactive JNK pathways are less able to repair their genetic information, and so these drugs could potentially work well on them. Future experiments will be needed to test this hypothesis.
Collapse
Affiliation(s)
- Nomeda Girnius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Yvonne Jk Edwards
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - David S Garlick
- Histo-Scientific Research Laboratories, Mount Jackson, United States
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States.,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, United States
| |
Collapse
|
23
|
Grainyhead-like 2 (GRHL2) knockout abolishes oral cancer development through reciprocal regulation of the MAP kinase and TGF-β signaling pathways. Oncogenesis 2018; 7:38. [PMID: 29735981 PMCID: PMC5938237 DOI: 10.1038/s41389-018-0047-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/25/2018] [Accepted: 04/05/2018] [Indexed: 01/01/2023] Open
Abstract
Grainyhead-Like 2 (GRHL2) is an epithelial-specific transcription factor that regulates epithelial morphogenesis and differentiation. Prior studies suggested inverse regulation between GRHL2 and TGF-β in epithelial plasticity and potential carcinogenesis. Here, we report the role of GRHL2 in oral carcinogenesis in vivo using a novel Grhl2 knockout (KO) mouse model and the underlying mechanism involving its functional interaction with TGF-β signaling. We developed epithelial-specific Grhl2 conditional KO mice by crossing Grhl2 floxed mice with those expressing CreER driven by the K14 promoter. After induction of Grhl2 KO, we confirmed the loss of GRHL2 and its target proteins, while Grhl2 KO strongly induced TGF-β signaling molecules. When exposed to 4-nitroquinoline 1-oxide (4-NQO), a strong chemical carcinogen, Grhl2 wild-type (WT) mice developed rampant oral tongue tumors, while Grhl2 KO mice completely abolished tumor development. In cultured oral squamous cell carcinoma (OSCC) cell lines, TGF-β signaling was notably induced by GRHL2 knockdown while being suppressed by GRHL2 overexpression. GRHL2 knockdown or KO in vitro and in vivo, respectively, led to loss of active p-Erk1/2 and p-JNK MAP kinase levels; moreover, ectopic overexpression of GRHL2 strongly induced the MAP kinase activation. Furthermore, the suppressive effect of GRHL2 on TGF-β signaling was diminished in cells exposed to Erk and JNK inhibitors. These data indicate that GRHL2 activates the Erk and JNK MAP kinases, which in turn suppresses the TGF -β signaling. This novel signaling represents an alternative pathway by which GRHL2 regulates carcinogenesis, and is distinct from the direct transcriptional regulation by GRHL2 binding at its target gene promoters, e.g., E-cadherin, hTERT, p63, and miR-200 family genes. Taken together, the current study provides the first genetic evidence to support the role of GRHL2 in carcinogenesis and the underlying novel mechanism that involves the functional interaction between GRHL2 and TGF-β signaling through the MAPK pathways.
Collapse
|
24
|
Xu Y, Wang W, Wang M, Liu X, Lee MH, Wang M, Zhang H, Li H, Chen W. High Salt Intake Attenuates Breast Cancer Metastasis to Lung. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:3386-3392. [PMID: 29553743 DOI: 10.1021/acs.jafc.7b05923] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Diet-related factors are thought to modify the risk of cancers, while the influence of high salt intake remains largely uncharacterized. Breast cancer is the most common cancer in women worldwide. In the present study, we examined the effect of salt intake on breast cancer by using a 4T1 mouse mammary tumor model. Unexpectedly, both the fitness and the survival rate of the tumor-bearing mice were improved by high salt intake. Similarly, high salt intake suppressed the primary tumor growth as well as metastasis to lung in mice. Mechanistically, high salt intake greatly reduced food intake and thus might exert antitumor effect through mimicking calorie restriction. Immunoblotting showed the lower proliferation marker Ki-67 and the higher expression of the tumor suppressor gene p53 in tumors of high salt intake mice. Importantly, high salt intake might induce hyperosmotic stress, which sensitized breast cancer cells to p53-dependent anoikis. Collectively, our findings raise the possibility that endogenous salt deposition might act as the first-line defense system against breast cancer progression as well as metastasis.
Collapse
Affiliation(s)
- Yijuan Xu
- State Key Laboratory of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
| | - Wenzhe Wang
- State Key Laboratory of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
| | - Minmin Wang
- State Key Laboratory of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
| | - Xuejiao Liu
- The China-US (Henan) Hormel Cancer Institute , Zhengzhou 450008 , China
| | - Mee-Hyun Lee
- The China-US (Henan) Hormel Cancer Institute , Zhengzhou 450008 , China
| | - Mingfu Wang
- School of Biological Sciences , The University of Hong Kong , Hong Kong , China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
| | - Haitao Li
- State Key Laboratory of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- School of Food Science and Technology , Jiangnan University , Wuxi 214122 , China
- Beijing Innovation Centre of Food Nutrition and Human Health , Beijing Technology & Business University , Beijing 100048 , China
| |
Collapse
|
25
|
Rybczynska AA, Boersma HH, de Jong S, Gietema JA, Noordzij W, Dierckx RAJO, Elsinga PH, van Waarde A. Avenues to molecular imaging of dying cells: Focus on cancer. Med Res Rev 2018. [PMID: 29528513 PMCID: PMC6220832 DOI: 10.1002/med.21495] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful treatment of cancer patients requires balancing of the dose, timing, and type of therapeutic regimen. Detection of increased cell death may serve as a predictor of the eventual therapeutic success. Imaging of cell death may thus lead to early identification of treatment responders and nonresponders, and to “patient‐tailored therapy.” Cell death in organs and tissues of the human body can be visualized, using positron emission tomography or single‐photon emission computed tomography, although unsolved problems remain concerning target selection, tracer pharmacokinetics, target‐to‐nontarget ratio, and spatial and temporal resolution of the scans. Phosphatidylserine exposure by dying cells has been the most extensively studied imaging target. However, visualization of this process with radiolabeled Annexin A5 has not become routine in the clinical setting. Classification of death modes is no longer based only on cell morphology but also on biochemistry, and apoptosis is no longer found to be the preponderant mechanism of cell death after antitumor therapy, as was earlier believed. These conceptual changes have affected radiochemical efforts. Novel probes targeting changes in membrane permeability, cytoplasmic pH, mitochondrial membrane potential, or caspase activation have recently been explored. In this review, we discuss molecular changes in tumors which can be targeted to visualize cell death and we propose promising biomarkers for future exploration.
Collapse
Affiliation(s)
- Anna A Rybczynska
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Genetics, University of Groningen, Groningen, the Netherlands
| | - Hendrikus H Boersma
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Clinical Pharmacy & Pharmacology, University of Groningen, Groningen, the Netherlands
| | - Steven de Jong
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University of Groningen, Groningen, the Netherlands
| | - Walter Noordzij
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudi A J O Dierckx
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.,Department of Nuclear Medicine, Ghent University, Ghent, Belgium
| | - Philip H Elsinga
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Aren van Waarde
- Molecular Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
26
|
Girnius N, Edwards YJK, Davis RJ. The cJUN NH 2-terminal kinase (JNK) pathway contributes to mouse mammary gland remodeling during involution. Cell Death Differ 2018; 25:1702-1715. [PMID: 29511338 PMCID: PMC6143629 DOI: 10.1038/s41418-018-0081-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 11/18/2022] Open
Abstract
Involution returns the lactating mammary gland to a quiescent state after weaning. The mechanism of involution involves collapse of the mammary epithelial cell compartment. To test whether the cJUN NH2-terminal kinase (JNK) signal transduction pathway contributes to involution, we established mice with JNK deficiency in the mammary epithelium. We found that JNK is required for efficient involution. JNK deficiency did not alter the STAT3/5 or SMAD2/3 signaling pathways that have been previously implicated in this process. Nevertheless, JNK promotes the expression of genes that drive involution, including matrix metalloproteases, cathepsins, and BH3-only proteins. These data demonstrate that JNK has a key role in mammary gland involution post lactation.
Collapse
Affiliation(s)
- Nomeda Girnius
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Yvonne J K Edwards
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA. .,Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
27
|
Ohsawa S, Vaughen J, Igaki T. Cell Extrusion: A Stress-Responsive Force for Good or Evil in Epithelial Homeostasis. Dev Cell 2018; 44:284-296. [PMID: 29408235 DOI: 10.1016/j.devcel.2018.01.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/08/2018] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Epithelial tissues robustly respond to internal and external stressors via dynamic cellular rearrangements. Cell extrusion acts as a key regulator of epithelial homeostasis by removing apoptotic cells, orchestrating morphogenesis, and mediating competitive cellular battles during tumorigenesis. Here, we delineate the diverse functions of cell extrusion during development and disease. We emphasize the expanding role for apoptotic cell extrusion in exerting morphogenetic forces, as well as the strong intersection of cell extrusion with cell competition, a homeostatic mechanism that eliminates aberrant or unfit cells. While cell competition and extrusion can exert potent, tumor-suppressive effects, dysregulation of either critical homeostatic program can fuel cancer progression.
Collapse
Affiliation(s)
- Shizue Ohsawa
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - John Vaughen
- Department of Developmental Biology, Stanford School of Medicine, Beckman Center, 279 Campus Drive B300, Stanford, CA 94305, USA
| | - Tatsushi Igaki
- Laboratory of Genetics, Graduate School of Biostudies, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|