1
|
Zhang Y, Saha K, Nandani R, Yuan J, Dey M, Gu Z. Isothiocyanates attenuate heparin-induced proliferation of colon cancer cells in vitro. Food Sci Nutr 2024; 12:7842-7853. [PMID: 39479720 PMCID: PMC11521738 DOI: 10.1002/fsn3.4296] [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: 03/09/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 11/02/2024] Open
Abstract
Isothiocyanates (ITCs), prevalent in cruciferous vegetables, are known for their anticarcinogenic properties. Prior research has indicated that heparin can stimulate the growth of colon cancer cells. However, the implications of ITCs in the diet of cancer patients receiving heparin-based therapies have yet to be fully understood. This exploratory in vitro study examines the proliferative effects of low-molecular-weight heparin (LMWH) on human colon cancer cells and assesses the antiproliferative potential of four ITC compounds, exploring possible epidermal growth factor family of receptor tyrosine kinases (Erb-B) related mechanisms. We evaluated cell viability in HCT-116 and HT-29 cell lines following treatment with ITCs alone or combined with LMWH (20 μg/mL) at various concentrations (1-100 μM). Clonogenic and wound-healing assays were performed after 24 h of treatment with 5 μM ITCs. Additionally, messenger RNA (mRNA) and protein expression of Erb-B family genes was measured using quantitative polymerase chain reaction (qPCR) and Western blotting. Statistical analysis was conducted using analysis of variance (ANOVA) with Dunnett's post hoc test. Results indicated that the half-maximal inhibitory concentration (IC50) values for Phenylethyl isothiocyanate (PEITC), Benzyl isothiocyanate (BITC), and Sulforaphane (SFN) were lower than those of Allyl isothiocyanate (AITC) in LMWH-stimulated HCT-116 (20.77, 19.10, and 44.05 μM, respectively) and HT-29 (74.94, 26.77, and 43.49 μM, respectively). PEITC and SFN significantly reduced ErbB1 (epidermal growth factor receptor (EGFR)) and ErbB4 (receptor tyrosine-protein kinase erbB-4) expression, while BITC decreased ErbB2 (receptor tyrosine-protein kinase erbB-2) and transforming growth factor beta (TGF-β) expression in HCT-116 cells (all, p < .05). PEITC, BITC, and SFN also increased proapoptotic Bax expression and decreased the antiapoptotic B-cell lymphoma 2 (Bcl-2) expression (all, p < .05). These findings suggest that specific ITCs may mitigate cancer cell proliferation induced by LMWH in cancer therapies, highlighting their potential therapeutic efficacy.
Collapse
Affiliation(s)
- Yizi Zhang
- Department of Agricultural and Biosystems EngineeringSouth Dakota State UniversityBrookingsSouth DakotaUSA
| | - Karabi Saha
- Department of Pharmaceutical SciencesSouth Dakota State UniversityBrookingsSouth DakotaUSA
| | - Raj Nandani
- Department of Pharmaceutical SciencesSouth Dakota State UniversityBrookingsSouth DakotaUSA
| | - Jiahui Yuan
- Department of Agricultural and Biosystems EngineeringSouth Dakota State UniversityBrookingsSouth DakotaUSA
| | - Moul Dey
- School of Health and Consumer SciencesSouth Dakota State UniversityBrookingsSouth DakotaUSA
| | - Zhengrong Gu
- Department of Agricultural and Biosystems EngineeringSouth Dakota State UniversityBrookingsSouth DakotaUSA
| |
Collapse
|
2
|
Manickasamy MK, Jayaprakash S, Girisa S, Kumar A, Lam HY, Okina E, Eng H, Alqahtani MS, Abbas M, Sethi G, Kumar AP, Kunnumakkara AB. Delineating the role of nuclear receptors in colorectal cancer, a focused review. Discov Oncol 2024; 15:41. [PMID: 38372868 PMCID: PMC10876515 DOI: 10.1007/s12672-023-00808-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/20/2023] [Indexed: 02/20/2024] Open
Abstract
Colorectal cancer (CRC) stands as one of the most prevalent form of cancer globally, causing a significant number of deaths, surpassing 0.9 million in the year 2020. According to GLOBOCAN 2020, CRC ranks third in incidence and second in mortality in both males and females. Despite extensive studies over the years, there is still a need to establish novel therapeutic targets to enhance the patients' survival rate in CRC. Nuclear receptors (NRs) are ligand-activated transcription factors (TFs) that regulate numerous essential biological processes such as differentiation, development, physiology, reproduction, and cellular metabolism. Dysregulation and anomalous expression of different NRs has led to multiple alterations, such as impaired signaling cascades, mutations, and epigenetic changes, leading to various diseases, including cancer. It has been observed that differential expression of various NRs might lead to the initiation and progression of CRC, and are correlated with poor survival outcomes in CRC patients. Despite numerous studies on the mechanism and role of NRs in this cancer, it remains of significant scientific interest primarily due to the diverse functions that various NRs exhibit in regulating key hallmarks of this cancer. Thus, modulating the expression of NRs with their agonists and antagonists, based on their expression levels, holds an immense prospect in the diagnosis, prognosis, and therapeutical modalities of CRC. In this review, we primarily focus on the role and mechanism of NRs in the pathogenesis of CRC and emphasized the significance of targeting these NRs using a variety of agents, which may represent a novel and effective strategy for the prevention and treatment of this cancer.
Collapse
Affiliation(s)
- Mukesh Kumar Manickasamy
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sujitha Jayaprakash
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Huiyan Eng
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117600, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Queenstown, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, 781039, Assam, India.
| |
Collapse
|
3
|
Su P, Mao X, Ma J, Huang L, Yu L, Tang S, Zhuang M, Lu Z, Osafo KS, Ren Y, Wang X, Lin X, Huang L, Huang X, Braicu EI, Sehouli J, Sun P. ERRα promotes glycolytic metabolism and targets the NLRP3/caspase-1/GSDMD pathway to regulate pyroptosis in endometrial cancer. J Exp Clin Cancer Res 2023; 42:274. [PMID: 37864196 PMCID: PMC10588109 DOI: 10.1186/s13046-023-02834-7] [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: 06/06/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Tumor cells can resist chemotherapy-induced pyroptosis through glycolytic reprogramming. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. Herein, we refine the oncogenic role of ERRα in the pyroptosis pathway and glycolytic metabolism. METHODS The interaction between ERRα and HIF-1α was verified using co-immunoprecipitation. The transcriptional binding sites of ERRα and NLRP3 were confirmed using dual-luciferase reporter assay and cleavage under targets and tagmentation (CUT&Tag). Flow cytometry, transmission electron microscopy, scanning electron microscopy, cell mito stress test, and extracellular acidification rate analysis were performed to investigate the effects of ERRα on the pyroptosis pathway and glycolytic metabolism. The results of these experiments were further confirmed in endometrial cancer (EC)-derived organoids and nude mice. In addition, the expression of ERRα-related pyroptosis genes was analyzed using The Cancer Genome Atlas and Gene Expression Omnibus database. RESULTS Triggered by a hypoxic microenvironment, highly expressed ERRα could bind to the promoter of NLRP3 and inhibit caspase-1/GSDMD signaling, which reduced inflammasome activation and increased pyroptosis resistance, thereby resulting in the resistance of cancer cells to cisplatin. Moreover, ERRα activated glycolytic rate-limiting enzyme to bridge glycolytic metabolism and pyroptosis in EC. This phenomenon was further confirmed in EC-derived organoids and nude mice. CUT & Tag sequencing and The Cancer Genome Atlas database analysis showed that ERRα participated in glycolysis and programmed cell death, which resulted in EC progression. CONCLUSIONS ERRα inhibits pyroptosis in an NLRP3-dependent manner and induces glycolytic metabolism, resulting in cisplatin resistance in EC cells.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Jincheng Ma
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Lirui Yu
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Shuting Tang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Mingzhi Zhuang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Zhonglei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Kelvin Stefan Osafo
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xinrui Wang
- Medical Research Center, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
| | - Xite Lin
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Leyi Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaoli Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, FuzhouFujian, 350005, China
| | - Elena Ioana Braicu
- Department of Gynecology and Obstetrics, Charité Virchow University Hospital, Augustenberger Platz1, 13353, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology and Obstetrics, Charité Virchow University Hospital, Augustenberger Platz1, 13353, Berlin, Germany
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China.
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
4
|
Chen L, Zhao W, Li M, Yang Y, Tian C, Zhang D, Chang Z, Zhang Y, Zhao ZJ, Chen Y, Ma L. SHP2 participates in decidualization by activating ERK to maintain normal nuclear localization of progesterone receptor. Reproduction 2023; 166:37-53. [PMID: 37184079 PMCID: PMC10304905 DOI: 10.1530/rep-22-0367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 05/15/2023] [Indexed: 05/16/2023]
Abstract
In brief The establishment and maintenance of embryo implantation and pregnancy require decidualization of endometrial stromal cells. This paper reveals that SHP2 ensures the correct subcellular localization of progesterone receptor, thereby safeguarding the process of decidualization. Abstract Decidualization is the process of conversion of endometrial stromal cells into decidual stromal cells, which is caused by progesterone production that begins during the luteal phase of the menstrual cycle and then increases throughout pregnancy dedicated to support embryonic development. Decidualization deficiency is closely associated with various pregnancy complications, such as recurrent miscarriage (RM). Here, we reported that Src-homology-2-containing phospho-tyrosine phosphatase (SHP2), a key regulator in the signal transduction process downstream of various receptors, plays an indispensable role in decidualization. SHP2 expression was upregulated during decidualization. SHP2 inhibitor RMC-4550 and shRNA-mediated SHP2 reduction resulted in a decreased level of phosphorylation of ERK and aberrant cytoplasmic localization of progesterone receptor (PR), coinciding with reduced expression of IGFBP1 and various other target genes of decidualization. Solely inhibiting ERK activity recapitulated these observations. Administration of RMC-4550 led to decidualization deficiency and embryo absorption in mice. Moreover, reduced expression of SHP2 was detected in the decidua of RM patients. Our results revealed that SHP2 is key to PR's nuclear localization, thereby indispensable for decidualization and that reduced expression of SHP2 might be engaged in the pathogenesis of RM.
Collapse
Affiliation(s)
- Lin Chen
- Center for Reproductive Medicine, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Weijie Zhao
- Center for Reproductive Medicine, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Mengxiong Li
- Department of Gynaecology, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yazhu Yang
- Center for Reproductive Medicine, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Chengzi Tian
- Center for Reproductive Medicine, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunzhe Zhang
- Faculty of Life Sciences and Medicine, Kings College London, London, United Kingdom
| | - Zhizhuang Joe Zhao
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Lin Ma
- Center for Reproductive Medicine, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
5
|
Marani A, Gioacchini H, Paolinelli M, Offidani A, Campanati A. Potential drug-drug interactions with mitogen-activated protein kinase (MEK) inhibitors used to treat melanoma. Expert Opin Drug Metab Toxicol 2023; 19:555-567. [PMID: 37659065 DOI: 10.1080/17425255.2023.2255519] [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: 05/15/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 09/05/2023]
Abstract
INTRODUCTION The management of patients with BRAF-mutated advanced melanoma who are undergoing targeted therapy with MEK inhibitors can be complicated by the co-administration of multiple medications, which can give rise to drug-drug interactions of clinical significance. COVERED AREAS Our review presents a comprehensive analysis of the pharmacokinetic and pharmacodynamic interactions of the three approved for advanced melanoma MEK inhibitor drugs - binimetinib, cobimetinib, and trametinib. MEDLINE (PubMed) was utilized for the literature search, comprising clinical studies, observational studies, and preclinical research. The review discusses the impact of these interactions on efficacy and safety of the treatments and differentiates between interactions supported by pharmacokinetic or pharmacodynamic mechanisms, those encountered in clinical practice, and those observed in preclinical studies. EXPERT OPINION Physicians should be aware about potential benefits, but also increased toxicity caused by drug interactions between MEK inhibitors and other drugs in the management of patients with metastatic melanoma.
Collapse
Affiliation(s)
- A Marani
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - H Gioacchini
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - M Paolinelli
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - A Offidani
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - A Campanati
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| |
Collapse
|
6
|
Musheyev D, Miller E, Birnbaum N, Miller E, Erblich S, Schuck A, Alayev A. Inhibition of ERK signaling for treatment of ERRα positive TNBC. PLoS One 2023; 18:e0283047. [PMID: 37163498 PMCID: PMC10171695 DOI: 10.1371/journal.pone.0283047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/28/2023] [Indexed: 05/12/2023] Open
Abstract
Breast cancer is the second leading cause of cancer-related deaths in women and triple-negative breast cancer (TNBC), in particular, is an aggressive and highly metastatic type of breast cancer that does not respond to established targeted therapies and is associated with poor prognosis and worse survival. Previous studies identified a subgroup of triple-negative breast cancer patients with high expression of estrogen related receptor alpha (ERRα) that has better prognosis when treated with tamoxifen. We therefore set out to identify common targets of tamoxifen and ERRα in the context of TNBC using phosphoproteomic analysis. In this study, we discovered that phosphorylation of mitogen-activated protein kinase 1 (MAPK1) is regulated by tamoxifen as well as ERRα. Additionally, we showed that inhibition of MAPK signaling together with the use of a selective ERRα inverse agonist, XCT-790, leads to a significant upregulation of apoptosis and paves way for the therapeutic use of MAPK inhibitors for treatment of ERRα expressing TNBC.
Collapse
Affiliation(s)
- David Musheyev
- Department of Internal Medicine, Stony Brook Southampton Hospital, Southampton, New York, United States of America
| | - Esther Miller
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Natania Birnbaum
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Elisheva Miller
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Shoshana Erblich
- Department of Mechanical Engineering, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Alyssa Schuck
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| | - Anya Alayev
- Department of Biology, Stern College for Women, Yeshiva University, New York, New York, United States of America
| |
Collapse
|
7
|
Wang Y, Zhou Y, Lin H, Chen H, Wang S. Paeoniflorin Inhibits the Proliferation and Metastasis of Ulcerative Colitis-Associated Colon Cancer by Targeting EGFL7. JOURNAL OF ONCOLOGY 2022; 2022:7498771. [PMID: 36072982 PMCID: PMC9441402 DOI: 10.1155/2022/7498771] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
Abstract
In this study, we studied the therapeutic potential of PF in mouse models of CAC. PF could inhibit the proliferation, migration, invasion, and clone formation and promote the apoptosis of colon cancer cells. Furthermore, PF showed a good effect on inhibiting the aggregation and infiltration of inflammatory cells, protecting the intestinal mucosal barrier, and inhibiting the growth of colon tumors in the AOM/DSS-induced CAC model. PF also significantly inhibited the expression of TNF-α, IL-1β, IL-6, IL-13, and CEA-related inflammatory factors in the AOM/DSS-induced CAC model by inhibiting the TLR4/NF-κB signaling pathway and EGFL7 expression. Therefore, PF showed a potential therapeutic effect on mice in the CAC model by inhibitingTLR4/NF-κB mediated inflammatory response and EGFL7 expression.
Collapse
Affiliation(s)
- Yunxia Wang
- Department of Surgery, Shidong Hospital of Shanghai Yangpu District, Shanghai 200438, China
| | - Yi Zhou
- Department of Surgery, Shidong Hospital of Shanghai Yangpu District, Shanghai 200438, China
| | - Hui Lin
- Department of Surgery, Shidong Hospital of Shanghai Yangpu District, Shanghai 200438, China
| | - Haiyan Chen
- Department of Anesthesiology, Shidong Hospital of Shanghai Yangpu District, Shanghai 200438, China
| | - Shu Wang
- Urology Surgery, Shidong Hospital of Shanghai Yangpu District, Shanghai 200438, China
| |
Collapse
|
8
|
Su P, Yu L, Mao X, Sun P. Role of HIF-1α/ERRα in Enhancing Cancer Cell Metabolism and Promoting Resistance of Endometrial Cancer Cells to Pyroptosis. Front Oncol 2022; 12:881252. [PMID: 35800058 PMCID: PMC9253301 DOI: 10.3389/fonc.2022.881252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
Oxygen is critical to energy metabolism, and tumors are often characterized by a hypoxic microenvironment. Owing to the high metabolic energy demand of malignant tumor cells, their survival is promoted by metabolic reprogramming in the hypoxic microenvironment, which can confer tumor cell resistance to pyroptosis. Pyroptosis resistance can inhibit anti-tumor immunity and promote the development of malignant tumors. Hypoxia inducible factor-1α (HIF-1α) is a key regulator of metabolic reprogramming in tumor cells, and estrogen-related receptor α (ERRα) plays a key role in regulating cellular energy metabolism. Therefore, the close interaction between HIF-1α and ERRα influences the metabolic and functional changes in cancer cells. In this review, we summarize the reprogramming of tumor metabolism involving HIF-1α/ERRα. We review our understanding of the role of HIF-1α/ERRα in promoting tumor growth adaptation and pyroptosis resistance, emphasize its key role in energy homeostasis, and explore the regulation of HIF-1α/ERRα in preventing and/or treating endometrial carcinoma patients. This review provides a new perspective for the study of the molecular mechanisms of metabolic changes in tumor progression.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lirui Yu
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaodan Mao
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
- *Correspondence: Pengming Sun,
| |
Collapse
|
9
|
Tzeng YDT, Tsui KH, Tseng LM, Hou MF, Chu PY, Sheu JJC, Li CJ. Integrated analysis of pivotal biomarker of LSM1, immune cell infiltration and therapeutic drugs in breast cancer. J Cell Mol Med 2022; 26:4007-4020. [PMID: 35692083 PMCID: PMC9279588 DOI: 10.1111/jcmm.17436] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/13/2022] [Accepted: 05/27/2022] [Indexed: 12/29/2022] Open
Abstract
The discovery of early diagnosis and prognostic markers for breast cancer can significantly improve survival and reduce mortality. LSM1 is known to be involved in the general process of mRNA degradation in complexes containing LSm subunits, but the molecular and biological functions in breast cancer remain unclear. Here, the expression of LSM1 mRNA in breast cancer was estimated using The Cancer Genome Atlas (TCGA), Oncomine, TIMER and bc‐GenExMiner databases. We found that functional LSM1 inactivation caused by mutations and profound deletions predicted poor prognosis in breast cancer (BRCA) patients. LSM1 was highly expressed in both BRCA tissues and cells compared to normal breast tissues/cells. High LSM1 expression is associated with poorer overall survival and disease‐free survival. The association between LSM1 and immune infiltration of breast cancer was assessed by TIMER and CIBERSORT algorithms. LSM1 showed a strong correlation with various immune marker sets. Most importantly, pharmacogenetic analysis of BRCA cell lines revealed that LSM1 inactivation was associated with increased sensitivity to refametinib and trametinib. However, both drugs could mimic the effects of LSM1 inhibition and their drug sensitivity was associated with MEK molecules. Therefore, we investigated the clinical application of LSM1 to provide a basis for sensitive diagnosis, prognosis and targeted treatment of breast cancer.
Collapse
Affiliation(s)
- Yen-Dun Tony Tzeng
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ling-Ming Tseng
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Center for Cancer Research, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
Therapeutic Strategies and Potential Actions of Female Sex Steroid Hormones and Their Receptors in Colon Cancer Based on Preclinical Studies. Life (Basel) 2022; 12:life12040605. [PMID: 35455096 PMCID: PMC9032023 DOI: 10.3390/life12040605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
Several epidemiological studies have reported that the use of female sex steroid hormones could reduce the risk of colon cancer (CRC). This review summarizes the available data related to estradiol (E2) and progesterone (P4) single and dual treatments in CRC male and female in vitro and in vivo models, mainly from preclinical studies, alongside their potential molecular mechanisms. Most of the studies showed that E2 exogenous treatment and/or reactivation of its beta receptor (ERβ) significantly inhibited cell proliferation, induced cell cycle arrest, and promoted apoptosis by modulating several molecular pathways. Likewise, the inhibition of ERα receptors produced similar antitumorigenic actions, both in vivo and in vitro, suggesting that E2 could have dual opposing roles in CRC that are dependent on the expression profile of its nuclear receptors. The available studies on P4 are scarce, and the results revealed that in vitro and in vivo treatments with natural and synthetic progesterone were also associated with promising tumoricidal actions. Nevertheless, the combination of E2 with P4 showed enhanced anticancer activities compared with their monotherapy protocols in male–female cell lines and animals. Collectively, the studies suggested that the female sex steroid hormones could provide a novel and effective therapeutic strategy against CRC.
Collapse
|
11
|
Pan Z, Wang K, Wang X, Jia Z, Yang Y, Duan Y, Huang L, Wu ZX, Zhang JY, Ding X. Cholesterol promotes EGFR-TKIs resistance in NSCLC by inducing EGFR/Src/Erk/SP1 signaling-mediated ERRα re-expression. Mol Cancer 2022; 21:77. [PMID: 35303882 PMCID: PMC8932110 DOI: 10.1186/s12943-022-01547-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The use of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) brings remarkable benefits for the survival of patients with advanced NSCLC harboring EGFR mutations. Unfortunately, acquired resistance seems to be inevitable and limits the application of EGFR-TKIs in clinical practice. This study reported a common molecular mechanism sustaining resistance and potential treatment options to overcome EGFR-TKIs resistance. METHODS EGFR-TKIs resistant NSCLC cells were established and confirmed by MTT assay. Cholesterol content was detected and the promotional function of cholesterol on NSCLC growth was determined in vivo. Then, we identified ERRα expression as the downstream factor of cholesterol-mediated drug resistance. To dissect the regulatory mechanism, we conducted experiments, including immunofluorescence, co-immunoprecipitation, luciferase reporter assay and chromatin immunoprecipitation assay. RESULTS Long-term exposure to EGFR-TKIs generate drug resistance with the characteristic of cholesterol accumulation in lipid rafts, which promotes EGFR and Src to interact and lead EGFR/Src/Erk signaling reactivation-mediated SP1 nuclear translocation and ERRα re-expression. Further investigation identifies ERRα as a target gene of SP1. Functionally, re-expression of ERRα sustains cell proliferation by regulating ROS detoxification process. Lovastatin, a drug used to decrease cholesterol level, and XCT790, an inverse agonist of ERRα, overcome gefitinib and osimertinib resistance both in vitro and in vivo. CONCLUSIONS Our study indicates that cholesterol/EGFR/Src/Erk/SP1 axis-induced ERRα re-expression promotes survival of gefitinib and osimertinib-resistant cancer cells. Besides, we demonstrate the potential of lowing cholesterol and downregulation of ERRα as effective adjuvant treatment of NSCLC.
Collapse
Affiliation(s)
- Zhenzhen Pan
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Kai Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Xiniao Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Zhirong Jia
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Yuqi Yang
- College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Yalei Duan
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Lianzhan Huang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Zhuo-Xun Wu
- College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Jian-Ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xuansheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China.
| |
Collapse
|
12
|
Zhang H, Xu H, Tang Q, Bi F. The selective serotonin reuptake inhibitors enhance the cytotoxicity of sorafenib in hepatocellular carcinoma cells. Anticancer Drugs 2021; 32:793-801. [PMID: 33675613 DOI: 10.1097/cad.0000000000001067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Sertraline and fluoxetine are the two most commonly used selective serotonin reuptake inhibitors (SSRIs) in the treatment of depression. Accumulating evidence has revealed that SSRIs can reduce the risk of hepatocellular carcinoma (HCC), but their therapeutic effects in HCC have not yet been elucidated. Previous studies have reported that sertraline and fluoxetine can suppress the growth of gastric carcinoma, melanoma and nonsmall cell lung cancers by inhibiting the mammalian target rapamycin (mTOR) activity. In this study, we found that sertraline and fluoxetine blocked the protein kinase B (AKT)/mTOR pathway and suppressed the growth of HCC cells in vitro, in xenografts and in diethylnitrosamine/carbon tetrachloride (DEN/CCL4)-induced primary liver mouse model. Sertraline and fluoxetine can synergize with sorafenib, the first approved standard therapy for advanced HCC, to inhibit the viability of HCC cells in vitro and in vivo. In addition, the combination of sorafenib and SSRIs synergistically inhibited the effects of the AKT/mTOR pathway. These results reveal novel therapeutic effects of a combination of SSRIs and sorafenib in HCC.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | | | | | | |
Collapse
|
13
|
Zhang H, Xu H, Zhang C, Tang Q, Bi F. Ursodeoxycholic acid suppresses the malignant progression of colorectal cancer through TGR5-YAP axis. Cell Death Discov 2021; 7:207. [PMID: 34365464 PMCID: PMC8349355 DOI: 10.1038/s41420-021-00589-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/20/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
The Hippo/YAP pathway plays an important role in the development of cancers. Previous studies have reported that bile acids can activate YAP (Yes Associated Protein) to promote tumorigenesis and tumor progression. Ursodeoxycholic acid (UDCA) is a long-established old drug used for cholestasis treatment. So far, the effect of UDCA on YAP signaling in colorectal cancer (CRC) is not well defined. This study means to explore relationship of UDCA and YAP in CRC. UDCA suppressed YAP signaling by activating the membrane G-protein-coupled bile acid receptor (TGR5). TGR5 mainly regulated cAMP/PKA signaling pathway to inhibit RhoA activity, thereby suppressing YAP signaling. Moreover, the restoration of YAP expression alleviated the inhibitory effect of UDCA on CRC cell proliferation. In AOM/DSS-induced CRC model, UDCA inhibited tumor growth in a concentration-dependent manner and decreased expression of YAP and Ki67. UDCA plays a distinguished role in regulating YAP signaling and CRC growth from the primary bile acids and partial secondary bile acids, demonstrating the importance of maintaining normal intestinal bile acid metabolism in cancer patients. It also presents a potential therapeutic intervention for CRC.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huanji Xu
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chenliang Zhang
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiulin Tang
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Feng Bi
- Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
14
|
Screening of Human Gut Bacterial Culture Collection Identifies Species That Biotransform Quercetin into Metabolites with Anticancer Properties. Int J Mol Sci 2021; 22:ijms22137045. [PMID: 34208885 PMCID: PMC8269047 DOI: 10.3390/ijms22137045] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/19/2022] Open
Abstract
We previously demonstrated that flavonoid metabolites inhibit cancer cell proliferation through both CDK-dependent and -independent mechanisms. The existing evidence suggests that gut microbiota is capable of flavonoid biotransformation to generate bioactive metabolites including 2,4,6-trihydroxybenzoic acid (2,4,6-THBA), 3,4-dihydroxybenzoic acid (3,4-DHBA), 3,4,5-trihyroxybenzoic acid (3,4,5-THBA) and 3,4-dihydroxyphenylacetic acid (DOPAC). In this study, we screened 94 human gut bacterial species for their ability to biotransform flavonoid quercetin into different metabolites. We demonstrated that five of these species were able to degrade quercetin including Bacillus glycinifermentans, Flavonifractor plautii, Bacteroides eggerthii, Olsenella scatoligenes and Eubacterium eligens. Additional studies showed that B. glycinifermentans could generate 2,4,6-THBA and 3,4-DHBA from quercetin while F. plautii generates DOPAC. In addition to the differences in the metabolites produced, we also observed that the kinetics of quercetin degradation was different between B. glycinifermentans and F. plautii, suggesting that the pathways of degradation are likely different between these strains. Similar to the antiproliferative effects of 2,4,6-THBA and 3,4-DHBA demonstrated previously, DOPAC also inhibited colony formation ex vivo in the HCT-116 colon cancer cell line. Consistent with this, the bacterial culture supernatant of F. plautii also inhibited colony formation in this cell line. Thus, as F. plautii and B. glycinifermentans generate metabolites possessing antiproliferative activity, we suggest that these strains have the potential to be developed into probiotics to improve human gut health.
Collapse
|
15
|
Yang Y, Li S, Li B, Li Y, Xia K, Aman S, Yang Y, Ahmad B, Zhao B, Wu H. FBXL10 promotes ERRα protein stability and proliferation of breast cancer cells by enhancing the mono-ubiquitylation of ERRα. Cancer Lett 2021; 502:108-119. [PMID: 33450359 DOI: 10.1016/j.canlet.2021.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 01/25/2023]
Abstract
The underlying mechanism of orphan nuclear receptor estrogen-related receptor α (ERRα) in breast cancer was investigated by identifying its interaction partners using mass spectrometry. F-box and leucine-rich repeat protein 10 (FBXL10), which modulates various physiological processes, may interact with ERRα in breast cancer. Here, we investigated the interaction between FBXL10 and ERRα, and their protein expression and correlation in breast cancer. Mechanical studies revealed that FBXL10 stabilized ERRα protein levels by reducing its poly-ubiquitylation and promoting its mono-ubiquitylation. The reporter gene assay and examination of ERRα target genes validated the increased transcriptional activity of ERRα due to its increased protein levels by FBXL10. FBXL10 also increased ERRα enrichment at the promoter region of its target genes. Functionally, FBXL10 facilitated the ERRα/peroxisome proliferator-activated receptor gamma coactivator 1 β (PGC1β)-mediated proliferation and tumorigenesis of breast cancer cells in vitro and in vivo. Our results uncovered a molecular mechanism linking the mono-ubiquitylation and protein stability of ERRα to functional interaction with FBXL10. Moreover, a novel regulatory axis of FBXL10 and ERRα regulating the proliferation and tumorigenesis of breast cancer cells was established.
Collapse
Affiliation(s)
- Yangyang Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Bowen Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Yanan Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Kangkai Xia
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Sattout Aman
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Yuxi Yang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Bashir Ahmad
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Binggong Zhao
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, China.
| |
Collapse
|
16
|
Joo MK, Shin S, Ye DJ, An HG, Kwon TU, Baek HS, Kwon YJ, Chun YJ. Combined treatment with auranofin and trametinib induces synergistic apoptosis in breast cancer cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:84-94. [PMID: 33103613 DOI: 10.1080/15287394.2020.1835762] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Auranofin is a gold complex used as an anti-rheumatic agent and may act as a potent anticancer drug against breast tumors. Trametinib is a specific mitogen-activated protein kinase inhibitor, approved for the treatment of metastatic melanoma. The aim of this study was to examine the synergistic effects of auranofin and trametinib on apoptosis in MCF-7 human breast cancer cells. The combination treatment inhibited cancer cell proliferation and induced cell cycle arrest at the sub-G1 phase and apoptosis via poly (ADP-ribose) polymerase cleavage and caspase-3/7 activation. It is noteworthy that this treatment significantly increased p38 mitogen-activated protein kinase (MAPK) phosphorylation to induce mitochondrial stress, subsequently promoting cancer cell apoptosis through release of apoptosis-inducing factor. Further data demonstrated that combined treatment significantly induced increase in nuclear translocation of AIF. These results indicated that activation of the p38 MAPK signaling pathway and mitochondrial apoptosis may contribute to the synergistic consequences in MCF-7 cells. Collectively, our data demonstrated that combined treatment with auranofin and trametinib exhibited synergistic breast cancer cell death and this combination might be utilized as a novel therapeutic strategy for breast cancer.
Collapse
Affiliation(s)
- Min-Kyung Joo
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Sangyun Shin
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Dong-Jin Ye
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Hong-Gyu An
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Tae-Uk Kwon
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Hyoung-Seok Baek
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Yeo-Jung Kwon
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| | - Young-Jin Chun
- Center for Metareceptome Research, College of Pharmacy, Chung-Ang University , Dongjak-gu, Republic of Korea
| |
Collapse
|
17
|
Massaro C, Safadeh E, Sgueglia G, Stunnenberg HG, Altucci L, Dell’Aversana C. MicroRNA-Assisted Hormone Cell Signaling in Colorectal Cancer Resistance. Cells 2020; 10:cells10010039. [PMID: 33396628 PMCID: PMC7823834 DOI: 10.3390/cells10010039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
Despite substantial progress in cancer therapy, colorectal cancer (CRC) is still the third leading cause of cancer death worldwide, mainly due to the acquisition of resistance and disease recurrence in patients. Growing evidence indicates that deregulation of hormone signaling pathways and their cross-talk with other signaling cascades inside CRC cells may have an impact on therapy resistance. MicroRNAs (miRNAs) are small conserved non-coding RNAs thatfunction as negative regulators in many gene expression processes. Key studies have identified miRNA alterations in cancer progression and drug resistance. In this review, we provide a comprehensive overview and assessment of miRNAs role in hormone signaling pathways in CRC drug resistance and their potential as future targets for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Crescenzo Massaro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
| | - Elham Safadeh
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
| | - Giulia Sgueglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
| | | | - Lucia Altucci
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-566-7564 (L.A.); +39-081-566-7566 (C.D.)
| | - Carmela Dell’Aversana
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS)-National Research Council (CNR), Via Sergio Pansini 5, 80131 Naples, Italy
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-566-7564 (L.A.); +39-081-566-7566 (C.D.)
| |
Collapse
|
18
|
Huang X, Ruan G, Liu G, Gao Y, Sun P. Immunohistochemical Analysis of PGC-1α and ERRα Expression Reveals Their Clinical Significance in Human Ovarian Cancer. Onco Targets Ther 2020; 13:13055-13062. [PMID: 33376354 PMCID: PMC7764629 DOI: 10.2147/ott.s288332] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and estrogen-related receptor alpha (ERRα) play a vital role in various human cancers. The purpose of this study was to investigate whether the PGC-1α/ERRα axis could serve as an effective prognostic marker in ovarian cancer (OC). Patients and Methods We investigated the expression of both PGC-1α and ERRα in 42 ovarian cancer and 31 noncancerous ovarian samples by immunohistochemistry (IHC). The relationship between the expression of PGC-1α and ERRα in OC and the clinical characteristics of patients was evaluated. In addition, data from the Human Protein Atlas (HPA) database were collected to validate the prognostic significance of PGC-1α and ERRα mRNA expression in OC. Results PGC-1α and ERRα showed notably higher expression in OC tissues than in noncancerous tissues (P=0.0059, P=0.002). Moreover, in patients with OC, high ERRα and PGC-1α/ERRα expression significantly correlated with tumor differentiation (P=0.027; P=0.04), lymph node status (P=0.023; P=0.021), CA125 (P=0.036; P=0.021), and HE4 (P=0.021; P=0.05), while high PGC-1α expression was only significantly associated with tumor differentiation (P=0.029). The combined analysis of high PGC-1α and ERRα expression revealed a tendency towards poor cancer-specific survival (P=0.1276). Conclusion PGC-1α and ERRα are overexpressed in OC and might be significant prognostic factors for this cancer.
Collapse
Affiliation(s)
- Xiqi Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Guanyu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Guifen Liu
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Yuqin Gao
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
19
|
Liu SL, Wu XS, Li FN, Yao WY, Wu ZY, Dong P, Wang XF, Gong W. ERRα promotes pancreatic cancer progression by enhancing the transcription of PAI1 and activating the MEK/ERK pathway. Am J Cancer Res 2020; 10:3622-3643. [PMID: 33294258 PMCID: PMC7716152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 10/23/2020] [Indexed: 06/12/2023] Open
Abstract
Estrogen-related receptor alpha (ERRα), an orphan nuclear receptor, was reported to be highly associated with the progression and tumorigenesis of several human malignancies. However, the biological role and underlying molecular mechanisms of ERRα in pancreatic cancer (PC) remain unknown. The present study demonstrated that ERRα was significantly overexpressed in PC tissues and cell lines. Its high expression was correlated with tumor size, distant metastasis, TNM stage, tumor differentiation and poor prognosis of PC. Subsequent functional assays showed that ERRα promoted PC cell proliferation, tumor growth, as well as migration and invasion via activating the epithelial-mesenchymal transition. In addition, knockdown of ERRα induced apoptosis and G0/G1 cell cycle arrest in PC cells. Plasminogen activator inhibitor 1 (PAI1) was identified by RNA sequencing, knockdown of which could suppress the cell proliferation, migration and invasion that promoted by ERRα overexpression. Further mechanistic investigation using chromatin immunoprecipitation and dual-luciferase reporter assays revealed that ERRα could bind to the PAI1 promoter region and transcriptionally enhance PAI1 expression. Moreover, our data indicated that ERRα played its oncogenic role in PC via activating the MEK/ERK pathway. Taken together, our study demonstrates that ERRα promotes PC progression by enhancing the transcription of PAI1 and activation of the MEK/ERK pathway, pointing to ERRα as a novel diagnostic and therapeutic target for PC.
Collapse
Affiliation(s)
- Shi-Lei Liu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Xiang-Song Wu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Feng-Nan Li
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Wen-Yan Yao
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Zi-You Wu
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Ping Dong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Xue-Feng Wang
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang Road, Shanghai 200092, China
| | - Wei Gong
- Department of General Surgery, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang Road, Shanghai 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang Road, Shanghai 200092, China
| |
Collapse
|
20
|
Crevet L, Vanacker JM. Regulation of the expression of the estrogen related receptors (ERRs). Cell Mol Life Sci 2020; 77:4573-4579. [PMID: 32448995 PMCID: PMC11104921 DOI: 10.1007/s00018-020-03549-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 10/23/2019] [Accepted: 05/13/2020] [Indexed: 10/24/2022]
Abstract
Estrogen related receptors (ERRα, β and γ in mammals) are orphan members of the nuclear receptor superfamily acting as transcription factors. ERRs are expressed in several tissues and cells and they display various physiological and pathological functions, controlling, amongst others and depending on the receptor, bone homeostasis, energy metabolism, embryonic stem cell pluripotency, and cancer progression. In contrast to classical nuclear receptors, the activities of the ERRs are not controlled by a natural ligand. Regulation of their activities thus rely on other means such as post-translational modification or availability of transcriptional co-regulators. In addition, regulation of their mere expression under given physiological or pathological conditions is a particularly important level of control. Here we discuss the mechanisms involved in the regulation of ERRs expression and the reported means to impact on it using pharmacological approaches.
Collapse
Affiliation(s)
- Lucile Crevet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France.
| |
Collapse
|
21
|
Dai C, Shen L, Jin W, Lv B, Liu P, Wang X, Yin Y, Fu Y, Liang L, Ma Z, Zhang X, Wang Y, Xu D, Chen Z. Physapubescin B enhances the sensitivity of gastric cancer cells to trametinib by inhibiting the STAT3 signaling pathway. Toxicol Appl Pharmacol 2020; 408:115273. [PMID: 33035574 DOI: 10.1016/j.taap.2020.115273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/24/2020] [Accepted: 10/04/2020] [Indexed: 02/07/2023]
Abstract
Given the poor prognosis of unresectable advanced gastric cancer (GC), novel therapeutic strategies are needed. The mitogen-activated protein kinase (MAPK) signaling cascade, the most frequently activated pathway in GC, plays an important role in tumorigenesis and metastasis. The MAPK/extracellular signal-regulated kinase (ERK) pathway is an attractive therapeutic target for GC. In this study, trametinib, a mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) inhibitor, reduced the p-ERK level and significantly increased signal transducer and activator of transcription 3 (STAT3) phosphorylation in GC cells, resulting in reduced sensitivity to trametinib. Physapubescin B (PB), a steroidal compound extracted from the plant Physalis pubescens L., inhibited the proliferation and induced the apoptosis of GC cells by suppressing STAT3 phosphorylation. The combination of PB and trametinib suppressed the STAT3 phosphorylation induced by trametinib, and synergistically suppressed gastric tumor growth in vitro and in vivo. Together, these results indicate that inhibition of both MEK and STAT3 may be effective for patients with MAPK/ERK pathway-addicted GC.
Collapse
Affiliation(s)
- Chunyan Dai
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Li Shen
- Institute of Basic Theory of TCM, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Weiyang Jin
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310006, China
| | - Bing Lv
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Pei Liu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Yifei Yin
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Yufei Fu
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Liguo Liang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China
| | - Zhongjun Ma
- School of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, PR China
| | - Xiaojian Zhang
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Yiping Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China.
| | - Daogun Xu
- Department of Colorectal Surgery, Wenling Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Wenling, China.
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, the First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian Road, Hangzhou 310006, China.
| |
Collapse
|
22
|
Cai S, Wang J, Zeng W, Cheng X, Liu L, Li W. Lysine-specific histone demethylase 1B (LSD2/KDM1B) represses p53 expression to promote proliferation and inhibit apoptosis in colorectal cancer through LSD2-mediated H3K4me2 demethylation. Aging (Albany NY) 2020; 12:14990-15001. [PMID: 32726297 PMCID: PMC7425427 DOI: 10.18632/aging.103558] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/04/2020] [Indexed: 11/25/2022]
Abstract
Epigenetic alterations have been reported to play critical roles in the development of colorectal cancer (CRC). However, the biological function of the lysine-specific histone demethylase 1B (LSD2/KDM1B) in CRC is not well understood. Therefore, we investigated the characteristics of LSD2 in CRC. We observed significant upregulation of LSD2 in CRC tissue compared to that in normal colorectal tissue. LSD2 promotes CRC cell proliferation and inhibits cell apoptosis through cell cycle regulation, promoting CRC progression both in vitro and in vivo. We found that LSD2 performs these functions by inhibiting the p53-p21-Rb pathway. Finally, we found that LSD2 directly binds to p53 and represses p53 expression via H3K4me2 demethylation at the p53 promoter. Our results revealed that LSD2 acts as an oncogene by binding and inhibiting p53 activity in CRC. Thus, LSD2 may be a new molecular target for CRC treatment.
Collapse
Affiliation(s)
- Shaoxin Cai
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Jinsi Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Wei Zeng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Xuefei Cheng
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Lihang Liu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Weihua Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
- Department of Surgical Oncology, Fujian Provincial Hospital, Fuzhou 350001, China
| |
Collapse
|
23
|
The Flavonoid Metabolite 2,4,6-Trihydroxybenzoic Acid Is a CDK Inhibitor and an Anti-Proliferative Agent: A Potential Role in Cancer Prevention. Cancers (Basel) 2019; 11:cancers11030427. [PMID: 30917530 PMCID: PMC6468648 DOI: 10.3390/cancers11030427] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/16/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022] Open
Abstract
Flavonoids have emerged as promising compounds capable of preventing colorectal cancer (CRC) due to their anti-oxidant and anti-inflammatory properties. It is hypothesized that the metabolites of flavonoids are primarily responsible for the observed anti-cancer effects owing to the unstable nature of the parent compounds and their degradation by colonic microflora. In this study, we investigated the ability of one metabolite, 2,4,6-trihydroxybenzoic acid (2,4,6-THBA) to inhibit Cyclin Dependent Kinase (CDK) activity and cancer cell proliferation. Using in vitro kinase assays, we demonstrated that 2,4,6-THBA dose-dependently inhibited CDKs 1, 2 and 4 and in silico studies identified key amino acids involved in these interactions. Interestingly, no significant CDK inhibition was observed with the structurally related compounds 3,4,5-trihydroxybenzoic acid (3,4,5-THBA) and phloroglucinol, suggesting that orientation of the functional groups and specific amino acid interactions may play a role in inhibition. We showed that cellular uptake of 2,4,6-THBA required the expression of functional SLC5A8, a monocarboxylic acid transporter. Consistent with this, in cells expressing functional SLC5A8, 2,4,6-THBA induced CDK inhibitory proteins p21Cip1 and p27Kip1 and inhibited cell proliferation. These findings, for the first time, suggest that the flavonoid metabolite 2,4,6-THBA may mediate its effects through a CDK- and SLC5A8-dependent pathway contributing to the prevention of CRC.
Collapse
|
24
|
Dai X, Xia H, Zhou S, Tang Q, Bi F. Zoledronic acid enhances the efficacy of the MEK inhibitor trametinib in KRAS mutant cancers. Cancer Lett 2018; 442:202-212. [PMID: 30429107 DOI: 10.1016/j.canlet.2018.10.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/09/2018] [Accepted: 10/17/2018] [Indexed: 02/05/2023]
Abstract
KRAS mutation is the most common type of mutation in human cancers. However, the direct pharmacological inhibition of KRAS has not been clinically successful. Trametinib (GSK1120212, Tram), a newer MEK inhibitor, inhibits RAS signaling through mitogen-activated protein kinase (MAPK) cascade suppression. The effectiveness of Tram in clinical practice is limited in KRAS mutant tumors compared to that in BRAF mutant tumors. Here, we found that Tram treatment provoked feedback activation of upstream RAS, thus causing an induction of phosphorylated MEK (pMEK) and phosphorylated ERK (pERK) rebound in KRAS mutant tumors. This failure of persistent ERK inhibition led to drug resistance. Zoledronic acid (ZA), a nitrogen-containing bisphosphonate, disrupts the biological activity of RAS by inhibiting its isoprenylation. Surprisingly, ZA overcame Tram resistance, and augmented antitumor activity was observed in KRAS mutant tumors both in vitro and in vivo. Furthermore, ZA enhanced the effect of Tram partially through the mevalonate pathway. In summary, the combination of the two FDA-approved drugs Tram and ZA may represent a novel therapeutic strategy for the treatment of KRAS mutant cancers.
Collapse
Affiliation(s)
- Xinyu Dai
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, China
| | - Hongwei Xia
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, China.
| | - Sheng Zhou
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, China
| | - Qiulin Tang
- Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, China
| | - Feng Bi
- Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Laboratory of Molecular Targeted Therapy in Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, China.
| |
Collapse
|