1
|
Zhou X, Ye W, Xu J, Luo Q, Huang Y, Li J, Zhu Q, Liu G. The role of di-(2-ethylhexyl) phthalate in cancer initiation and progression: Mechanisms and health implications. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 959:178285. [PMID: 39756301 DOI: 10.1016/j.scitotenv.2024.178285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
The increasing prevalence of cancer has been linked to various environmental factors associated with modern industrial and societal advancements. Di-(2-ethylhexyl) phthalate (DEHP), a commonly used plasticizer, is one such environmental contaminant with potential carcinogenic effects. While epidemiological studies have suggested a positive association between DEHP exposure and cancer risk, the specific role of DEHP in cancer initiation and progression requires further clarification. This review systematically examines the relationship between DEHP exposure and cancer, highlighting key mechanisms involved in tumorigenesis. DEHP has been found to influence several critical aspects of cancer biology, including cell proliferation, apoptosis, metastasis, invasion, epithelial-mesenchymal transition, angiogenesis, drug resistance, immune regulation, and cancer stem cell maintenance. These carcinogenic effects are mediated through multiple pathways, such as the PI3K/AKT signaling pathway, estrogen receptor activation, epigenetic modifications, oxidative stress, and inflammation. By elucidating the molecular mechanisms underlying DEHP's role in cancer, this review aims to contribute to the development of targeted prevention and intervention strategies to mitigate the cancer risks associated with DEHP exposure.
Collapse
Affiliation(s)
- Xinrui Zhou
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; School of Pharmaceutical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Wei Ye
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; School of Pharmaceutical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Jiapeng Xu
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; School of Pharmaceutical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Qiting Luo
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China; School of Pharmaceutical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Yuanyuan Huang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Jieyu Li
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Qinchang Zhu
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Ge Liu
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| |
Collapse
|
2
|
Sfeir N, Kajdan M, Jalaguier S, Bonnet S, Teyssier C, Pyrdziak S, Yuan R, Bousquet E, Maraver A, Bernex F, Pirot N, Boissière‐Michot F, Castet‐Nicolas A, Lapierre M, Cavaillès V. RIP140 regulates transcription factor HES1 oscillatory expression and mitogenic activity in colon cancer cells. Mol Oncol 2024; 18:1510-1530. [PMID: 38459621 PMCID: PMC11161732 DOI: 10.1002/1878-0261.13626] [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: 06/28/2023] [Revised: 01/17/2024] [Accepted: 02/23/2024] [Indexed: 03/10/2024] Open
Abstract
The transcription factor receptor-interacting protein 140 (RIP140) regulates intestinal homeostasis and tumorigenesis through Wnt signaling. In this study, we investigated its effect on the Notch/HES1 signaling pathway. In colorectal cancer (CRC) cell lines, RIP140 positively regulated HES1 gene expression at the transcriptional level via a recombining binding protein suppressor of hairless (RBPJ)/neurogenic locus notch homolog protein 1 (NICD)-mediated mechanism. In support of these in vitro data, RIP140 and HES1 expression significantly correlated in mouse intestine and in a cohort of CRC samples, thus supporting the positive regulation of HES1 gene expression by RIP140. Interestingly, when the Notch pathway is fully activated, RIP140 exerted a strong inhibition of HES1 gene transcription controlled by the level of HES1 itself. Moreover, RIP140 directly interacts with HES1 and reversed its mitogenic activity in human CRC cells. In line with this observation, HES1 levels were associated with a better patient survival only when tumors expressed high levels of RIP140. Our data identify RIP140 as a key regulator of the Notch/HES1 signaling pathway, with a dual effect on HES1 gene expression at the transcriptional level and a strong impact on colon cancer cell proliferation.
Collapse
Affiliation(s)
- Nour Sfeir
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Marilyn Kajdan
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Stéphan Jalaguier
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Sandrine Bonnet
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Catherine Teyssier
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Samuel Pyrdziak
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Rong Yuan
- Department of Medical Microbiology, Immunology and Cell Biology, School of MedicineSouthern Illinois UniversitySpringfieldILUSA
| | - Emilie Bousquet
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Antonio Maraver
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Florence Bernex
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Nelly Pirot
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Florence Boissière‐Michot
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
- Translational Research UnitMontpellier Cancer Institute Val d'AurelleFrance
| | - Audrey Castet‐Nicolas
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Marion Lapierre
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| | - Vincent Cavaillès
- IRCM, Institut de Recherche en Cancérologie de MontpellierFrance
- INSERM, U1194France
- Université de MontpellierFrance
- Institut régional du Cancer de MontpellierFrance
| |
Collapse
|
3
|
Xing M, Ma X, Wang X, Wang H, Xie M, Zhang Z, Zhou J. Emodin disrupts the Notch1/Nrf2/GPX4 antioxidant system and promotes renal cell ferroptosis. J Appl Toxicol 2023; 43:1702-1718. [PMID: 37393915 DOI: 10.1002/jat.4509] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 07/04/2023]
Abstract
Emodin has been demonstrated to possess multiple pharmacological activities. However, emodin has also been reported to induce nephrotoxicity at high doses and with long-term use, and the underlying mechanism has not been fully disclosed. The current study aimed to investigate the roles of oxidative stress and ferroptosis in emodin-induced kidney toxicity. Mice were intraperitoneally treated with emodin, and NRK-52E cells were exposed to emodin in the presence or absence of treatment with Jagged1, SC79, or t-BHQ. Emodin significantly upregulated the levels of blood urea nitrogen, serum creatinine, malondialdehyde, and Fe2+ , reduced the levels of superoxide dismutase and glutathione, and induced pathological changes in the kidneys in vivo. Moreover, the viability of NRK-52E cells treated with emodin was reduced, and emodin induced iron accumulation, excessive reactive oxygen species production, and lipid peroxidation and depolarized the mitochondrial membrane potential (ΔΨm). In addition, emodin treatment downregulated the activity of neurogenic locus notch homolog protein 1 (Notch1), reduced the nuclear translocation of nuclear factor erythroid-2 related factor 2 (Nrf2), and decreased glutathione peroxidase 4 protein levels. However, Notch1 activation by Jagged1 pretreatment, Akt activation by SC79 pretreatment, or Nrf2 activation by t-BHQ pretreatment attenuated the toxic effects of emodin in NRK-52E cells. Taken together, these results revealed that emodin-induced ferroptosis triggered kidney toxicity through inhibition of the Notch1/Nrf2/glutathione peroxidase 4 axis.
Collapse
Affiliation(s)
- Miao Xing
- School of Medicine, Yichun University, Yichun, China
| | - Xiaoyu Ma
- School of Medicine, Yichun University, Yichun, China
| | - Xi Wang
- School of Medicine, Yichun University, Yichun, China
| | - Haoze Wang
- School of Medicine, Yichun University, Yichun, China
| | - Minjuan Xie
- School of Medicine, Yichun University, Yichun, China
| | - Ziwen Zhang
- School of Medicine, Yichun University, Yichun, China
| | - Jie Zhou
- School of Medicine, Yichun University, Yichun, China
| |
Collapse
|
4
|
Liu J, Pan Y, Liu Y, Wei W, Hu X, Xin W, Chen N. The regulation of PTEN: Novel insights into functions as cancer biomarkers and therapeutic targets. J Cell Physiol 2023; 238:1693-1715. [PMID: 37334436 DOI: 10.1002/jcp.31053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
This review summarizes the implications of the primary tumor suppressor protein phosphatase and tensin homolog (PTEN) in aggressive cancer development. PTEN interacts with other cellular proteins or factors suggesting the existence of an intricate molecular network that regulates their oncogenic function. Accumulating evidence has shown that PTEN exists and plays a role in the cytoplasmic organelles and in the nucleus. PTEN blocks phosphoinositide 3-kinases (PI3K)-protein kinase B-mammalian target of rapamycin signaling pathway by dephosphorylating phosphatidylinositol (PI)-3,4,5-triphosphate to PI-4,5-bisphosphate thus counteracting PI3K function. Studies have shown that PTEN expression is tightly regulated at transcriptional, posttranscriptional, and posttranslational levels (including protein-protein interactions and posttranslational modifications). Despite recent advances in PTEN research, the regulation and function of the PTEN gene remain largely unknown. How mutation or loss of specific exons in the PTEN gene occurs and involves in cancer development is not clear. This review illustrates the regulatory mechanisms of PTEN expression and discusses how PTEN participates in tumor development and/or suppression. Future prospects for the clinical applications are also highlighted.
Collapse
Affiliation(s)
- Jie Liu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yongli Pan
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Yuheng Liu
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoping Hu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Nan Chen
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
5
|
Dutta S, Shah RB, Singhal S, Dutta SB, Bansal S, Sinha S, Haque M. Metformin: A Review of Potential Mechanism and Therapeutic Utility Beyond Diabetes. Drug Des Devel Ther 2023; 17:1907-1932. [PMID: 37397787 PMCID: PMC10312383 DOI: 10.2147/dddt.s409373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/10/2023] [Indexed: 07/04/2023] Open
Abstract
Metformin has been designated as one of the most crucial first-line therapeutic agents in the management of type 2 diabetes mellitus. Primarily being an antihyperglycemic agent, metformin also has a plethora of pleiotropic effects on various systems and processes. It acts majorly by activating AMPK (Adenosine Monophosphate-Activated Protein Kinase) in the cells and reducing glucose output from the liver. It also decreases advanced glycation end products and reactive oxygen species production in the endothelium apart from regulating the glucose and lipid metabolism in the cardiomyocytes, hence minimizing the cardiovascular risks. Its anticancer, antiproliferative and apoptosis-inducing effects on malignant cells might prove instrumental in the malignancy of organs like the breast, kidney, brain, ovary, lung, and endometrium. Preclinical studies have also shown some evidence of metformin's neuroprotective role in Parkinson's disease, Alzheimer's disease, multiple sclerosis and Huntington's disease. Metformin exerts its pleiotropic effects through varied pathways of intracellular signalling and exact mechanism in the majority of them remains yet to be clearly defined. This article has extensively reviewed the therapeutic benefits of metformin and the details of its mechanism for a molecule of boon in various conditions like diabetes, prediabetes, obesity, polycystic ovarian disease, metabolic derangement in HIV, various cancers and aging.
Collapse
Affiliation(s)
- Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Rima B Shah
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Shubha Singhal
- Department of Pharmacology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Sudeshna Banerjee Dutta
- Department of Medical Surgical Nursing, Shri Anand Institute of Nursing, Rajkot, Gujarat, 360005, India
| | - Sumit Bansal
- Department of Anaesthesiology, All India Institute of Medical Sciences, Rajkot, Gujarat, India
| | - Susmita Sinha
- Department of Physiology, Khulna City Medical College and Hospital, Khulna, Bangladesh
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur, 57000, Malaysia
| |
Collapse
|
6
|
Lei Y, Shen HF, Li QW, Yang S, Xie HT, Li XF, Chen ML, Xia JW, Wang SC, Dai GQ, Zhou Y, Li YC, Huang SH, He DH, Zhou ZH, Cong JG, Lin XL, Lin TY, Wu AB, Xiao D, Xiao SJ, Zhang XK, Jia JS. Hairy gene homolog increases nasopharyngeal carcinoma cell stemness by upregulating Bmi-1. Aging (Albany NY) 2023; 15:204742. [PMID: 37219449 DOI: 10.18632/aging.204742] [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: 11/24/2022] [Accepted: 05/09/2023] [Indexed: 05/24/2023]
Abstract
B-cell-specific Moloney murine leukemia virus integration site 1 (Bmi-1) is overexpressed in various cancer types. We found that Bmi-1 mRNA levels were elevated in nasopharyngeal carcinoma (NPC) cell lines. In immunohistochemical analyses, high Bmi-1 levels were observed in not only 5 of 38 non-cancerous nasopharyngeal squamous epithelial biopsies, but also in 66 of 98 NPC specimens (67.3%). High Bmi-1 levels were detected more frequently in T3-T4, N2-N3 and stage III-IV NPC biopsies than in T1-T2, N0-N1 and stage I-II NPC samples, indicating that Bmi-1 is upregulated in advanced NPC. In 5-8F and SUNE1 NPC cells, stable depletion of Bmi-1 using lentiviral RNA interference greatly suppressed cell proliferation, induced G1-phase cell cycle arrest, reduced cell stemness and suppressed cell migration and invasion. Likewise, knocking down Bmi-1 inhibited NPC cell growth in nude mice. Both chromatin immunoprecipitation and Western blotting assays demonstrated that Hairy gene homolog (HRY) upregulated Bmi-1 by binding to its promoter, thereby increasing the stemness of NPC cells. Immunohistochemistry and quantitative real-time PCR analyses revealed that HRY expression correlated positively with Bmi-1 expression in a cohort of NPC biopsies. These findings suggested that HRY promotes NPC cell stemness by upregulating Bmi-1, and that silencing Bmi-1 can suppress NPC progression.
Collapse
Affiliation(s)
- Ye Lei
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
- Guangzhou Southern Medical Laboratory Animal Sci and Tech Co. Ltd., Guangzhou 510515, China
| | - Hong-Fen Shen
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qi-Wen Li
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng Yang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hong-Ting Xie
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Xu-Feng Li
- School of Basic Medical Sciences, Guangxi Medical University, Nanning 530000, China
| | - Mei-Ling Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Jia-Wei Xia
- The Third People’s Hospital of Kunming (The Sixth Affiliated Hospital of Dali University), Kunming 650041, China
| | - Sheng-Chun Wang
- Department of Pathology, School of Basic Medicine, Guangdong Medical University, Dongguan 523808, China
| | - Guan-Qi Dai
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ying Zhou
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ying-Chun Li
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shi-Hao Huang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Dan-Hua He
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhi-Hao Zhou
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jin-Ge Cong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
- Guangzhou Southern Medical Laboratory Animal Sci and Tech Co. Ltd., Guangzhou 510515, China
| | - Xiao-Lin Lin
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Tao-Yan Lin
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ai-Bing Wu
- Central People’s Hospital of Zhanjiang, Zhanjiang 524000, China
| | - Dong Xiao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
- Guangzhou Southern Medical Laboratory Animal Sci and Tech Co. Ltd., Guangzhou 510515, China
- National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng-Jun Xiao
- Department of Pathology, The Second Affiliated Hospital, Guilin Medical University, Guilin 541199, China
| | - Xin-Ke Zhang
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jun-Shuang Jia
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
7
|
Song E, Ghil S. Crosstalk between cannabinoid receptor 2 and lysophosphatidic acid receptor 5. Biochem Biophys Res Commun 2023; 666:154-161. [PMID: 37187093 DOI: 10.1016/j.bbrc.2023.04.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/17/2023]
Abstract
Cannabinoid receptor 2 (CB2) and lysophosphatidic acid receptor 5 (LPA5) are both classified as G-protein coupled receptors (GPCRs) activated by bioactive lipids and are highly expressed in colon cancer cells. However, crosstalk between two receptors and its potential effects on cancer cell physiology have not been fully elucidated. In the present study, the results of bioluminescence resonance energy transfer analysis showed that, among the LPA receptors, CB2 strongly and specifically interacted with LPA5. Both receptors were co-localized in the plasma membrane in the absence of agonists, and the receptors were co-internalized upon activation of either receptor alone or both receptors together. We further investigated the effects of expression of both receptors on cell proliferation and migration, and the molecular mechanisms underlying these effects in HCT116 colon cancer cells. Co-expression of receptors significantly increased cell proliferation and migration by increasing Akt phosphorylation and tumor progression-related gene expression, whereas no such effect was seen upon expression of either receptor alone. These results suggest the possibility of physical and functional crosstalk between CB2 and LPA5.
Collapse
Affiliation(s)
- Eunju Song
- Department of Life Science, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Sungho Ghil
- Department of Life Science, Kyonggi University, Suwon, 16227, Republic of Korea.
| |
Collapse
|
8
|
Emam O, Wasfey EF, Hamdy NM. Notch-associated lncRNAs profiling circuiting epigenetic modification in colorectal cancer. Cancer Cell Int 2022; 22:316. [PMID: 36229883 PMCID: PMC9558410 DOI: 10.1186/s12935-022-02736-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/28/2022] [Indexed: 11/15/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most prevalent digestive cancers, ranking the 2nd cause of cancer-related fatality worldwide. The worldwide burden of CRC is predicted to rise by 60% by 2030. Environmental factors drive, first, inflammation and hence, cancer incidence increase. Main The Notch-signaling system is an evolutionarily conserved cascade, has role in the biological normal developmental processes as well as malignancies. Long non-coding RNAs (LncRNAs) have become major contributors in the advancement of cancer by serving as signal pathways regulators. They can control gene expression through post-translational changes, interactions with micro-RNAs or down-stream effector proteins. Recent emerging evidence has emphasized the role of lncRNAs in controlling Notch-signaling activity, regulating development of several cancers including CRC. Conclusion Notch-associated lncRNAs might be useful prognostic biomarkers or promising potential therapeutic targets for CRC treatment. Therefore, here-in we will focus on the role of “Notch-associated lncRNAs in CRC” highlighting “the impact of Notch-associated lncRNAs as player for cancer induction and/or progression.” Graphical Abstract ![]()
Collapse
Affiliation(s)
| | - Eman F Wasfey
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
9
|
Kurtović M, Piteša N, Bartoniček N, Ozretić P, Musani V, Čonkaš J, Petrić T, King C, Sabol M. RNA-seq and ChIP-seq Identification of Unique and Overlapping Targets of GLI Transcription Factors in Melanoma Cell Lines. Cancers (Basel) 2022; 14:4540. [PMID: 36139698 PMCID: PMC9497141 DOI: 10.3390/cancers14184540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite significant progress in therapy, melanoma still has a rising incidence worldwide, and novel treatment strategies are needed. Recently, researchers have recognized the involvement of the Hedgehog-GLI (HH-GLI) signaling pathway in melanoma and its consistent crosstalk with the MAPK pathway. In order to further investigate the link between the two pathways and to find new target genes that could be considered for combination therapy, we set out to find transcriptional targets of all three GLI proteins in melanoma. METHODS We performed RNA sequencing on three melanoma cell lines (CHL-1, A375, and MEL224) with overexpressed GLI1, GLI2, and GLI3 and combined them with the results of ChIP-sequencing on endogenous GLI1, GLI2, and GLI3 proteins. After combining these results, 21 targets were selected for validation by qPCR. RESULTS RNA-seq revealed a total of 808 differentially expressed genes (DEGs) for GLI1, 941 DEGs for GLI2, and 58 DEGs for GLI3. ChIP-seq identified 527 genes that contained GLI1 binding sites in their promoters, 1103 for GLI2 and 553 for GLI3. A total of 15 of these targets were validated in the tested cell lines, 6 of which were detected by both RNA-seq and ChIP-seq. CONCLUSIONS Our study provides insight into the unique and overlapping transcriptional output of the GLI proteins in melanoma. We suggest that our findings could provide new potential targets to consider while designing melanoma-targeted therapy.
Collapse
Affiliation(s)
- Matea Kurtović
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Nikolina Piteša
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Nenad Bartoniček
- The Garvan Institute of Medical Research, 384 Victoria St., Darlinghurst, NSW 2010, Australia
- The Kinghorn Centre for Clinical Genomics, 370 Victoria St., Darlinghurst, NSW 2010, Australia
| | - Petar Ozretić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Vesna Musani
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Josipa Čonkaš
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Tina Petrić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| | - Cecile King
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maja Sabol
- Division of Molecular Medicine, Ruđer Bošković Institute, 10 000 Zagreb, Croatia
| |
Collapse
|
10
|
Xu J, Li L, Shi P, Cui H, Yang L. The Crucial Roles of Bmi-1 in Cancer: Implications in Pathogenesis, Metastasis, Drug Resistance, and Targeted Therapies. Int J Mol Sci 2022; 23:ijms23158231. [PMID: 35897796 PMCID: PMC9367737 DOI: 10.3390/ijms23158231] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/01/2022] Open
Abstract
B-cell-specific Moloney murine leukemia virus integration region 1 (Bmi-1, also known as RNF51 or PCGF4) is one of the important members of the PcG gene family, and is involved in regulating cell proliferation, differentiation and senescence, and maintaining the self-renewal of stem cells. Many studies in recent years have emphasized the role of Bmi-1 in the occurrence and development of tumors. In fact, Bmi-1 has multiple functions in cancer biology and is closely related to many classical molecules, including Akt, c-MYC, Pten, etc. This review summarizes the regulatory mechanisms of Bmi-1 in multiple pathways, and the interaction of Bmi-1 with noncoding RNAs. In particular, we focus on the pathological processes of Bmi-1 in cancer, and explore the clinical relevance of Bmi-1 in cancer biomarkers and prognosis, as well as its implications for chemoresistance and radioresistance. In conclusion, we summarize the role of Bmi-1 in tumor progression, reveal the pathophysiological process and molecular mechanism of Bmi-1 in tumors, and provide useful information for tumor diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
| | - Pengfei Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| |
Collapse
|
11
|
Zhang Q, Zhu Z, Guan J, Hu Y, Zhou W, Ye W, Lin B, Weng S, Chen Y, Zheng C. Hes1 Controls Proliferation and Apoptosis in Chronic Lymphoblastic Leukemia Cells by Modulating PTEN Expression. Mol Biotechnol 2022; 64:1419-1430. [PMID: 35704163 DOI: 10.1007/s12033-022-00476-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/02/2022] [Indexed: 12/26/2022]
Abstract
Hairy and enhancer of split homolog-1 (HES1), regulated by the Notch, has been reported to play important roles in the immune response and cancers, such as leukemia. In this study, we aim to explore the effect of HES1-mediated Notch1 signaling pathway in chronic lymphocytic leukemia (CLL). Reverse transcription quantitative polymerase chain reaction and Western blot assay were conducted to determine the expression of HES1, Notch1, and PTEN in B lymphocytes of peripheral blood samples of 60 CLL patients. We used lentivirus-mediated overexpression or silencing of HES1 and the Notch1 signaling pathway inhibitor, MW167, to detect the interaction among HES1, Notch1, and PTEN in CLL MEC1 and HG3 cells. MTT assay and flow cytometry were employed for detection of biological behaviors of CLL cells. HES1 and Notch1 showed high expression, but PTEN displayed low expression in B lymphocytes of peripheral blood samples of patients with CLL in association with poor prognosis. HES1 bound to the promoter region of PTEN and reduced PTEN expression. Overexpression of HES1 activated the Notch1 signaling pathway, thus promoting the proliferation of CLL cells, increasing the proportion of cells arrested at the S phase and limiting the apoptosis of CLL cells. Collectively, HES1 can promote activation of the Notch1 signaling pathway to cause PTEN transcription inhibition and the subsequent expression reduction, thereby promoting the proliferation and inhibiting the apoptosis of CLL cells.
Collapse
Affiliation(s)
- Qikai Zhang
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Zongsi Zhu
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jiaqiang Guan
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yingying Hu
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wenjin Zhou
- Department of Chemotherapy, Cancer Hospital of The University of Chinese Academy of Science, Wenzhou Campus, Wenzhou, 325000, People's Republic of China
| | - Wanchun Ye
- Department of Chemotherapy, Cancer Hospital of The University of Chinese Academy of Science, Wenzhou Campus, Wenzhou, 325000, People's Republic of China
| | - Bijing Lin
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Shanshan Weng
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yuemiao Chen
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Cuiping Zheng
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
12
|
Components of NOTCH Signaling for Uterine Cancer Patients’ Prognosis. JOURNAL OF ONCOLOGY 2022; 2022:8199306. [PMID: 35136410 PMCID: PMC8818413 DOI: 10.1155/2022/8199306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022]
Abstract
New molecular biomarkers that could have an independent prognostic value in endometrial cancer are currently under investigation. Recently, it was suggested that genetic changes in the Notch signaling pathway could be associated with the development of endometrial carcinoma. This study aimed to determine the expression of the Notch signaling pathway components in tumour and adjacent normal uterine tissue and to evaluate their importance for the survival of uterine cancer patients. The present study was performed on uterine body samples collected from 109 patients and paired adjacent noncancerous endometrial tissue samples. Kaplan–Meier curves and Cox regression were used for survival analyses. Expression alterations of NOTCH2, NOTCH3, NOTCH4, JAG2, and HES1 were evaluated as independent and significant prognostic factors for uterine cancer patients.
Collapse
|
13
|
Basiri P, Afshar S, Amini R, Soltanian AR, Saidijam M, Mahdavinezhad A. Evaluation of miR-330-3p and BMI1 Expression in Colorectal Cancer Patients, Healthy Adjacent Tissues, and Polypoid Adenomatous Lesions. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2022; 11:334-345. [PMID: 37727645 PMCID: PMC10506674 DOI: 10.22088/ijmcm.bums.11.4.334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 05/17/2023] [Accepted: 07/15/2023] [Indexed: 09/21/2023]
Abstract
MicroRNAs (miRNAs) have emerged as essential gene expression regulators associated with human diseases such as colorectal cancer (CRC). The purpose of this study was to evaluate the expression of miR-330-3p and its target gene BMI1 in tissue samples of patients with CRC, polyp, and healthy adjacent tissue samples and their association with clinicopathological and demographic factors such as age, tumor stage, grade, and lymph node invasion of the tumor. Following the extraction of total RNA from approximately 50 mg of colon and rectum tissue of 82 patients with CRC, 13 polypoid lesions, and 26 marginal healthy tissues using RiboEx reagent, cDNA synthesis was performed, and then quantitative real-time PCR was used to detect the expression levels of miR-330-3p and BMI1. Alterations in the gene expression were assessed using the 2(-∆∆ CT) method. The expression of miR-330-3p in all of the CRC samples was significantly lower than in adjacent healthy tissues and polyp (P<0.001). BMI1 was up-regulated in 97.9% of CRC tissue compared to healthy adjacent tissues and polyps (P<0.001). A negative reverse correlation between the miR-330-3p and BMI1 gene was observed in the CRC samples (r= -0.882, P<0.001). Down-regulation of miR-330-3p and BMI1 overexpression strongly correlates with higher tumor stage and lymph node invasion. The AUC for miR-330-3p and BMI1expression was 0.982 (sensitivity, 98.5%; specificity, 78.8%), and 0.971 (sensitivity, 97.6%; specificity, 84.6%) (P<0.001), respectively. Our results indicated that miR-330-3p and BMI1 expression probably could be considered potential diagnostic or prognostic biomarkers for CRC patient.
Collapse
Affiliation(s)
- Parviz Basiri
- School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Saeid Afshar
- Research Center for Molecular Medicine, Department of Molecular Medicine and Genetics, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Razieh Amini
- Research Center for Molecular Medicine, Department of Molecular Medicine and Genetics, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Ali Reza Soltanian
- Modeling of Non-Communicable Diseases Research Center, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Massoud Saidijam
- Research Center for Molecular Medicine, Department of Molecular Medicine and Genetics, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Ali Mahdavinezhad
- Research Center for Molecular Medicine, Department of Molecular Medicine and Genetics, Medical School, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
14
|
Li Q, Li S, Niu L, Yang S, Niu H, Cheng C. Long noncoding RNA MGC27382 inhibits proliferation and metastasis of non-small cell lung cancer cells via down-regulating AKT/GSK3β pathway. Clin Transl Oncol 2021; 23:2548-2559. [PMID: 34224057 DOI: 10.1007/s12094-021-02658-x] [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: 01/25/2021] [Accepted: 06/02/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Persistent abnormal proliferation and long distant metastasis of tumors contribute to high mortality rate in non-small cell lung cancer (NSCLC) patients. Strategies that prevent NSCLC proliferation and/or metastasis have been studied but still need to be further explored. Numerous studies have proved the diversity functions of long noncoding RNAs (lncRNAs) exerted in cancer, including NSCLC. In this study, we aim to identify and investigate the role of novel lncRNAs in NSCLC progression. METHODS RNA sequence data were retrieved from the Cancer Genome Atlas (TCGA), differentially expressed lncRNAs (DElncRNAs) were screened out based on the R language, then real-time PCR experiment was introduced to detect the DElncRNA expression levels. A series of experiments including MTT, cell cycle, transwell, and wound healing assays were employed to explore the effect of DElncRNA MGC27382 on cell proliferation and invasion ability. RESULTS We detected that DElncRNA MGC27382 is down-regulated in NSCLC tissues and cells. Overexpression of MGC27382 prevented NSCLC cell proliferation via down-regulating cyclin D1 and cyclin E. Moreover, wound healing and transwell assays indicated that the ability of cell invasion and migration could be impaired when cells were treated with MGC27382 overexpression. Further studies demonstrated that MGC27382-mediated inhibition on NSCLC progression can be impaired by LY294002, which is a frequently used inhibitor of AKT/GSK3β pathway. CONCLUSION MGC27382 is down-regulated in NSCLC. It exerts an inhibitory role in NSCLC development through suppressing the AKT/GSK3β pathway. Our results indicate that the lncRNA MGC27382 might be a tumor-suppressor gene in NSCLC. Overexpression of MGC27382 is thought to be a potential strategy for overcoming NSCLC progression.
Collapse
Affiliation(s)
- Q Li
- Department of Respiratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - S Li
- Department of Gastroenterology, People's Hospital of Weihaiwei, Weihai, China
| | - L Niu
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - S Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - H Niu
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - C Cheng
- Department of Thoracic Surgery, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan, China.
| |
Collapse
|
15
|
Corrado A, Aceto R, Silvestri R, Dell'Anno I, Ricci B, Miglietta S, Romei C, Giovannoni R, Poliseno L, Evangelista M, Vitiello M, Cipollini M, Garritano S, Giusti L, Zallocco L, Elisei R, Landi S, Gemignani F. Pro64His (rs4644) Polymorphism Within Galectin-3 Is a Risk Factor of Differentiated Thyroid Carcinoma and Affects the Transcriptome of Thyrocytes Engineered via CRISPR/Cas9 System. Thyroid 2021; 31:1056-1066. [PMID: 33308024 DOI: 10.1089/thy.2020.0366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Galectin-3 (LGALS3) is an important glycoprotein involved in the malignant transformation of thyrocytes acting in the extracellular matrix, cytoplasm, and nucleus where it regulates TTF-1 and TCF4 transcription factors. Within LGALS3 gene, a common single-nucleotide polymorphism (SNP) (c.191C>A, p.Pro64His; rs4644) encoding for the variant Proline to Histidine at codon 64 has been extensively studied. However, data on rs4644 in the context of thyroid cancer are lacking. Thus, the aim of the present work was to evaluate the role of the rs4644 SNP as risk factor for differentiated thyroid cancer (DTC) and to determine the effect on the transcriptome in thyrocytes. Methods: A case/control association study in 1223 controls and 1142 unrelated consecutive DTC patients was carried out to evaluate the association between rs4644-P64H and the risk of DTC. We used the nonmalignant cell line Nthy-Ori (rs4644-C/A) and the CRISPR/Cas9 technique to generate isogenic cells carrying either the rs4644-A/A or rs4644-C/C homozygosis. Then, the transcriptome of the derivative and unmodified parental cells was analyzed by RNA-seq. Genes differentially expressed were validated by quantitative reverse transcription PCR and further tested in the parental Nthy-Ori cells after LGALS3 gene silencing, to investigate whether the expression of target genes was dependent on galectin-3 levels. Results: rs4644 AA genotype was associated with a reduced risk of DTC (compared with CC, ORadj = 0.66; 95% confidence interval = 0.46-0.93; Pass = 0.02). We found that rs4644 affects galectin-3 as a transcriptional coregulator. Among 34 genes affected by rs4644, HES1, HSPA6, SPC24, and NHS were of particular interest since their expression was rs4644-dependent (CC>AA for the first and AA>CC for the others), also in 574 thyroid tissues of Genotype-Tissue Expression (GTEx) biobank. Moreover, the expression of these genes was regulated by LGALS3-silencing. Using the proximity ligation assay in Nthy-Ori cells, we found that the TTF-1 interaction was genotype dependent. Conclusions: Our data show that in thyroid, rs4644 is a trans-expression quantitative trait locus that can modify the transcriptional expression of downstream genes, through the modulation of TTF-1.
Collapse
Affiliation(s)
- Alda Corrado
- Genetic Unit, Department of Biology, University of Pisa, Pisa, Italy
| | - Romina Aceto
- Genetic Unit, Department of Biology, University of Pisa, Pisa, Italy
- Humanitas Clinical and Research Centre-IRCCS, Milan, Italy
| | - Roberto Silvestri
- Genetic Unit, Department of Biology, University of Pisa, Pisa, Italy
| | - Irene Dell'Anno
- Genetic Unit, Department of Biology, University of Pisa, Pisa, Italy
| | - Benedetta Ricci
- Fondazione I.R.C.C.S., Istituto Neurologico Carlo Besta, Milan, Italy
| | - Simona Miglietta
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Romei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Laura Poliseno
- Institute of Clinical Physiology (IFC), CNR, Pisa, Italy
| | | | | | - Monica Cipollini
- Genetic Unit, Department of Biology, University of Pisa, Pisa, Italy
| | - Sonia Garritano
- Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Laura Giusti
- School of Pharmacy, University of Camerino, Camerino, Italy
| | - Lorenzo Zallocco
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Landi
- Genetic Unit, Department of Biology, University of Pisa, Pisa, Italy
| | | |
Collapse
|
16
|
Zhu J, Zhang X, Wen B, Chen J, Lu Y, Xu W. Diethylhexyl phthalate (DEHP) regulates the proliferation and chemosensitivity of esophageal squamous cell carcinoma cells via regulation of PTEN. Hum Cell 2021; 34:1153-1162. [PMID: 33721218 DOI: 10.1007/s13577-021-00519-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/09/2021] [Indexed: 11/30/2022]
Abstract
As one of the most prevalent and deadly cancers worldwide, esophageal squamous cell carcinoma (ESCC) can be directly exposed to endocrine-disrupting chemical (EDC). As a potential EDC, diethylhexyl phthalate (DEHP) can trigger the development of various cancers, while the potential effect of DEHP on the ESCC progression was not clear. Our present study revealed that DEHP can trigger the proliferation of ESCC cells and decrease the cisplatin (CDDP) and fluorouracil (5-FU) sensitivity. Mechanistical studies indicated that DEHP can decrease the transcription of PTEN, a well-characterized tumor suppressor, in ESCC cells. Over expression of PTEN can reverse DEHP-regulated ESCC cell proliferation and chemosensitivity. Further, DEHP can increase the expression of HES-1, which can bind with the promoter of PTEN to inhibit its transcription. Collectively, DEHP can increase proliferation while decrease chemosensitivity of ESCC cells via regulation of HES-1/PTEN axis. Further, daily expression of DEHP may be a potent risk factor for ESCC development.
Collapse
Affiliation(s)
- Jian Zhu
- Department of Gastroenterology, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, People's Republic of China
| | - Xuesong Zhang
- Department of Central Laboratory, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, People's Republic of China
| | - Bin Wen
- Department of Oncology, Jingjiang Chinese Medicine Hospital, Jingjiang, Jiangsu, 214500, People's Republic of China
| | - Jing Chen
- Department of Pathology, Jingjiang People's Hospital, Jingjiang, Jiangsu, 214500, People's Republic of China
| | - Yuntian Lu
- Department of Bioinformatics, Nantong University Medical School, Nantong, Jiangsu, 226001, People's Republic of China
| | - Weijun Xu
- Department of Gastroenterology, East Theater General Hospital of PLA, Nanjing, Jiangsu, 210000, People's Republic of China.
| |
Collapse
|
17
|
Li X, Li Y, Du X, Wang X, Guan S, Cao Y, Jin F, Li F. HES1 promotes breast cancer stem cells by elevating Slug in triple-negative breast cancer. Int J Biol Sci 2021; 17:247-258. [PMID: 33390847 PMCID: PMC7757037 DOI: 10.7150/ijbs.53477] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. TNBC is enriched with breast cancer stem cells (BCSCs), which are responsible for cancer initiation, cancer progression and worse prognosis. Our previous study found that HES1 was overexpressed and promoted invasion in TNBC. However, the role of HES1 in modulating BCSC stemness of TNBC remains unclear. Here, we found that HES1 upregulates Slug both in transcriptional level and in protein level. HES1 also has a positive correlation with Slug expression in 150 TNBC patient samples. TNBC patients with high HES1 and Slug levels show worse prognosis in both progression-free survival and overall survival analyses. Survival analyses indicate that the effects of HES1 on survival prognosis may depend on Slug. Furthermore, we reveal that HES1 is a novel transcriptional activator for Slug through acting directly on its promoter. Meanwhile, HES1 knockdown reduces BCSC self-renewal, BCSC population, and cancer cell proliferation in TNBC, whereas overexpression of Slug restores the oncogenic function of HES1, both in vitro and in vivo, suggesting that HES1 performs its oncogenic role through upregulating Slug. Taken together, HES1 promotes BCSC stemness properties via targeting Slug, highlighting that HES1 might be a novel candidate for BCSC stemness regulation in TNBC and providing new clues for identifying promising prognostic biomarkers and therapeutic targets of TNBC.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, 110122 Shenyang, Liaoning, China
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, 110001 Shenyang, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, 110122 Shenyang, Liaoning, China
| | - Xianqiang Du
- Department of Breast Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Anji Road, Quanzhou, China
| | - Xu Wang
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, 110001 Shenyang, China
| | - Shu Guan
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, 110001 Shenyang, China
| | - Yu Cao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, 110001 Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, 155 Nanjing Road, 110001 Shenyang, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, No. 77, Puhe Road, Shenyang North New Area, 110122 Shenyang, Liaoning, China
| |
Collapse
|
18
|
Pourjafar M, Samadi P, Karami M, Najafi R. Assessment of clinicopathological and prognostic relevance of BMI-1 in patients with colorectal cancer: A meta-analysis. Biotechnol Appl Biochem 2020; 68:1313-1322. [PMID: 33086431 DOI: 10.1002/bab.2053] [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: 02/28/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
B-cell-specific Moloney leukemia virus insertion site 1 (BMI-1) is one of the stemness markers. The prognostic and clinicopathological effects of BMI-1 expression in colorectal cancer (CRC) have been in dispute with different studies. Eligible studies were retrieved from international databases up to December 2019. Studies with a relationship between the clinicopathological and prognostic value of CRC patients with BMI-1 expression were selected. The correlations in the random-effect model were evaluated using the hazard ratios, odds ratio, and 95% confidence intervals (CIs). A total of nine studies comprising Asian cases (seven studies) and European cases (two studies) covering 1,294 samples of CRC were included for this meta-analysis. The analysis suggested that in Asian cases, increased expression of BMI-1 was associated with poor overall survival (OS) and death-free survival, whereas in European populations, high expression of BMI-1 was associated with better OS. Also, overexpression of BMI-1 in the Asian population was associated with the tumor size, distant metastasis, and patient's gender and age. Results suggested that high expression of BMI-1 can be involved in the progression and invasion of CRC, and so its inhibitor-based therapies could be used to prevent the progression of CRC.
Collapse
Affiliation(s)
- Mona Pourjafar
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Manoochehr Karami
- Research Center for Health Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
19
|
Wang Z, Li MX, Xu CZ, Zhang Y, Deng Q, Sun R, Hu QY, Zhang SP, Zhang JW, Liang H. Comprehensive study of altered proteomic landscape in proximal renal tubular epithelial cells in response to calcium oxalate monohydrate crystals. BMC Urol 2020; 20:136. [PMID: 32867742 PMCID: PMC7461262 DOI: 10.1186/s12894-020-00709-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 08/26/2020] [Indexed: 12/18/2022] Open
Abstract
Background Calcium oxalate monohydrate (COM), the major crystalline composition of most kidney stones, induces inflammatory infiltration and injures in renal tubular cells. However, the mechanism of COM-induced toxic effects in renal tubular cells remain ambiguous. The present study aimed to investigate the potential changes in proteomic landscape of proximal renal tubular cells in response to the stimulation of COM crystals. Methods Clinical kidney stone samples were collected and characterized by a stone component analyzer. Three COM-enriched samples were applied to treat human proximal tubular epithelial cells HK-2. The proteomic landscape of COM-crystal treated HK-2 cells was screened by TMT-labeled quantitative proteomics analysis. The differentially expressed proteins (DEPs) were identified by pair-wise analysis. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of DEPs were performed. Protein interaction networks were identified by STRING database. Results The data of TMT-labeled quantitative proteomic analysis showed that a total of 1141 proteins were differentially expressed in HK-2 cells, of which 699 were up-regulated and 442 were down-regulated. Functional characterization by KEGG, along with GO enrichments, suggests that the DEPs are mainly involved in cellular components and cellular processes, including regulation of actin cytoskeleton, tight junction and focal adhesion. 3 high-degree hub nodes, CFL1, ACTN and MYH9 were identified by STRING analysis. Conclusion These results suggested that calcium oxalate crystal has a significant effect on protein expression profile in human proximal renal tubular epithelial cells.
Collapse
Affiliation(s)
- Zhu Wang
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China.
| | - Ming-Xing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chang-Zhi Xu
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Ying Zhang
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China
| | - Qiong Deng
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China
| | - Rui Sun
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China
| | - Qi-Yi Hu
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China
| | - Sheng-Ping Zhang
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China
| | - Jian-Wen Zhang
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China
| | - Hui Liang
- Department of Urology, People's Hospital of Longhua, Southern Medical University, Shenzhen, 518109, Guangdong, China.
| |
Collapse
|
20
|
Kim JW, Jun SY, Ylaya K, Chang HK, Oh YH, Hong SM, Chung JY, Hewitt SM. Loss of HES-1 Expression Predicts a Poor Prognosis for Small Intestinal Adenocarcinoma Patients. Front Oncol 2020; 10:1427. [PMID: 32974155 PMCID: PMC7466551 DOI: 10.3389/fonc.2020.01427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Objective: Hairy and enhancer of split-1 (HES-1), which is a downstream target of the Notch signaling pathway, has been linked to KRAS mutations. HES-1 has been proposed as harboring oncogenic activity in colorectal cancer but has not been investigated in adenocarcinoma of the small intestine, where the drivers of oncogenesis are not as well-understood. Materials and Methods: To investigate the clinicopathologic and prognostic implications of HES-1, HES-1 immunohistochemical expression was analyzed in digital images along with clinicopathological variables, including survival and KRAS genotype, in 185 small intestinal adenocarcinomas. Results: The loss of HES-1 expression (HES-1Loss) was observed in 38.4% (71/185) of the patients, and was associated with higher pT category (P = 0.018), pancreatic invasion (P = 0.005), high grade (P = 0.043), and non-tubular histology (P = 0.004). Specifically, in tumors with mutant KRAS (KRAS MT), HES-1Loss was related to proximal location (P = 0.024), high T and N categories (P = 0.005 and 0.047, respectively), and pancreatic invasion (P = 0.004). Patients with HES-1Loss showed worse overall survival compared to those with intact HES-1 (HES-1Intact) (P = 0.013). Patients with HES-1Loss/KRAS MT (median, 17.3 months) had significantly worse outcomes than those with HES-1Intact/KRAS WT (39.9 months), HES-1Intact/KRAS MT (47.6 month), and HES-1Loss/KRAS WT (36.2 months; P = 0.010). By multivariate analysis, HES-1Loss (hazard ratio = 1.55, 95% confidence interval (CI), 1.07-2.26; P = 0.022) remained an independent prognostic factor. Conclusion: HES-1expression can be used as a potential prognostic marker and may aid in the management of patients with small intestinal adenocarcinomas.
Collapse
Affiliation(s)
- Jeong Won Kim
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Sun-Young Jun
- Department of Pathology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Kris Ylaya
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hee-Kyung Chang
- Department of Pathology, Kosin University College of Medicine, Busan, South Korea
| | - Young-Ha Oh
- Department of Pathology, Hanyang University College of Medicine, Seoul, South Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Joon-Yong Chung
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Zaczek A, Jóźwiak P, Ciesielski P, Forma E, Wójcik-Krowiranda K, Cwonda Ł, Bieńkiewicz A, Bryś M, Krześlak A. Relationship between polycomb-group protein BMI-1 and phosphatases regulating AKT phosphorylation level in endometrial cancer. J Cell Mol Med 2019; 24:1300-1310. [PMID: 31863623 PMCID: PMC6991679 DOI: 10.1111/jcmm.14782] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 08/24/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022] Open
Abstract
The PI3K/AKT pathway is frequently activated in endometrial carcinoma. BMI‐1 (B‐lymphoma Mo‐MLV insertion region 1) protein affects expression of PTEN (phosphatase and tensin homolog) in some cancers, but its significance for endometrial tumorigenesis is not known. The objective of this study was to determine the relationship between BMI‐1 and expression of factors affecting AKT (protein kinase B) phosphorylation level in endometrial cancer. The expression of proteins and mRNAs was investigated in endometrial cancer specimens and samples of non‐neoplastic endometrial tissue by Western blot and RT‐PCR, respectively. The impact of BMI‐1 down‐regulation on AKT phosphorylation and expression of genes coding for several phosphatases were studied in HEC1A cells. The results showed that BMI‐1 depletion caused increase in PHLPP1 and PHLPP2 (PH domain and leucine‐rich repeat protein phosphatases 1/2) expression and decrease in phospho‐AKT (pAKT) level. In more advanced tumours with higher metastatic potential, the expression of BMI‐1 was lower compared to tumours less advanced and without lymph node metastasis. There were significant inverse correlations between BMI‐1 and PHLPPs, especially PHLPP1 in normal endometrial samples. The inverse correlation between BMI‐1 and PHLPP1/PHLPP2 expression was observed in PTEN positive but not PTEN negative cancers. Low PHLPP2 expression in tumours predicted poorer overall survival. BMI‐1 impacts on AKT phosphorylation level in endometrial cells by regulation of PHLPP expression.
Collapse
Affiliation(s)
- Agnieszka Zaczek
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Paweł Jóźwiak
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Piotr Ciesielski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Ewa Forma
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | | | - Łukasz Cwonda
- Clinical Division of Gynecological Oncology, Medical University of Lodz, Lodz, Poland
| | - Andrzej Bieńkiewicz
- Clinical Division of Gynecological Oncology, Medical University of Lodz, Lodz, Poland
| | - Magdalena Bryś
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Anna Krześlak
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| |
Collapse
|
22
|
Zhang Y, Zheng L, Lao X, Wen M, Qian Z, Liu X, Tang H, Gao F. Hes1 is associated with long non-coding RNAs in colorectal cancer. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:459. [PMID: 31700895 DOI: 10.21037/atm.2019.08.11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Long noncoding RNAs (lncRNAs) play important roles in the development and pathophysiology of colorectal cancer (CRC). Our previous study showed that Hes1 was involved in the self-renewal and tumorigenicity of stem-like cancer cells in CRC. Methods ArrayStar Human LncRNA/mRNA Expression Microarray Version 3.0 was used to detect lncRNA expression in CRC tissues compared with their matched non-tumoral tissues. RNA-binding protein immunoprecipitation and sequencing (RIP-seq) assay was used to detect lncRNAs binding to Hes1. Real-time qPCR was used to detect expression of specific lncRNAs in CRC tissues. Results We found significantly up-regulated as well as down-regulated lncRNAs in CRC tissues compared with their matched non-tumoral tissues. We also screened a number of lncRNAs interacting with Hes1 in CRC cells. Interestingly, we found several lncRNAs binding to Hes1 (such as, GNAS-AS1, RP11-89K10.1, and RP11-465L10.10) were up-regulated in CRC tissues showed by the tissue microarray. Next, we confirmed that Hes1 directly interacted with these lncRNAs using RIP-qPCR and RNA pulldown assay. Finally, we verified the expression of these lncRNAs in 32 CRC samples as well as the adjacent non-tumoral tissues using real-time qPCR. Conclusions Based on these, we speculate that Hes1 interacts with one or more lncRNAs which contribute to the development and progression of CRC.
Collapse
Affiliation(s)
- Yuqin Zhang
- Laboratory of Digestive Disease and Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.,Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.,Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Zheng
- Department of Pathology, Nanfang Hospital and School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuejun Lao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Mingbo Wen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhipeng Qian
- Laboratory of Digestive Disease and Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Xin Liu
- Laboratory of Digestive Disease and Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hui Tang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Fei Gao
- Laboratory of Digestive Disease and Oncology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.,Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.,Department of Physiology and Biomedical Engineering and Gastroenterology Research Unit, Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Mori M, Mori T, Yamamoto A, Takagi S, Ueda M. Proliferation of poorly differentiated endometrial cancer cells through autocrine activation of FGF receptor and HES1 expression. Hum Cell 2019; 32:367-378. [PMID: 30963412 DOI: 10.1007/s13577-019-00249-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Patients with poorly differentiated endometrial cancer show poor prognosis, and effective molecular target-based therapies are needed. Endometrial cancer cells proliferate depending on the activation of HES1 (hairy and enhancer of split-1), which is induced by several pathways, such as the Notch and fibroblast growth factor receptor (FGFR) signaling pathways. In addition, aberrant, ligand-free activation of the FGFR signaling pathway resulting from mutations in FGFR2 was also reported in endometrial cancer. However, a clinical trial showed that there was no difference in the effectiveness of FGFR inhibitors between patients with and without the FGFR2 mutation, suggesting a presence of another signaling pathway for the FGFR activation. Here, we investigated the signaling pathway regulating the expression of HES1 and proliferation of poorly and well-differentiated endometrial cancer cell lines Ishikawa and HEC-50B, respectively. Whereas Ishikawa cells proliferated and expressed HES1 in a Notch signaling-dependent manner, Notch signaling was not involved in HES1 and proliferation of HEC-50B cells. The FGFR inhibitor, NVP-BGJ398, decreased HES1 expression and proliferation of HEC-50B cells; however, HEC50B cells had no mutations in the FGFR2 gene. Instead, HEC-50B cells highly expressed ligands for FGFR2, suggesting that FGFR2 is activated by an autocrine manner, not by ligand-free activation. This autocrine pathway activated Akt downstream of FGFR for cell proliferation. Our findings suggest the usefulness of HES1 as a marker for the proliferation signaling and that FGFR inhibitor may be effective for poorly differentiated endometrial cancers that harbor wild-type FGFR.
Collapse
Affiliation(s)
- Michihiro Mori
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan. .,Kake Institute of Cytopathology, Okayama, Japan.
| | - Toshinori Mori
- Department of Clinical Laboratory, Mihara Medical Associations Hospital, Hiroshima, Japan.,Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Aina Yamamoto
- Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Shoji Takagi
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan.,Kake Institute of Cytopathology, Okayama, Japan
| | - Masatsugu Ueda
- Faculty of Health Sciences, Kio University, Nara, Japan.,Graduate School of Health Sciences, Kio University, Nara, Japan
| |
Collapse
|
24
|
Lu H, Tan Y, Chen L. A clinical study on the expression of PTEN in renal cell carcinoma in children. Oncol Lett 2019; 17:69-72. [PMID: 30655739 PMCID: PMC6313102 DOI: 10.3892/ol.2018.9571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 09/27/2018] [Indexed: 12/13/2022] Open
Abstract
The expression pattern of tumor suppressor gene phosphatase and tensin homolog deleted on chromosome ten (PTEN) and phosphatase and tensin homolog deleted on chromosome ten/phosphatidylinositol3-kinase/protein kinase B (PTEN/PI3K/AKT) cell signaling pathway in renal cell carcinoma (RCC) were investigated in children. A total of 5 cases of RCC (observation group) in children and 10 cases of benign kidney tumor (control group) diagnosed by pathological examinations were included to obtain tumor samples. Expression of PTEN mRNA was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The protein expression of PTEN, PI3K and AKT was detected by western blotting; relationships between the expression level of PTEN mRNA and the clinical features of RCC were analyzed. It turned out that expression level of PTEN mRNA in the observation group was significantly lower than that in the control group. The protein expression levels of PTEN, PI3K and AKT were significantly lower in the observation group than in the control group (P<0.05). The expression level of PTEN mRNA decreased with the increased clinical stage of RCC (P<0.05), and was not related to sex, age and maximum tumor diameter (P>0.05). The results showed that downregulation of the tumor suppressor gene PTEN expression and the inhibition of PTEN/PI3K/AKT cell signaling pathway may be involved in the occurrence and development of RCC in children.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pediatrics, Zibo Maternal and Child Health Care Hospital, Zibo, Shandong 255029, P.R. China
| | - Yuxia Tan
- Department of Pediatrics, Zibo Maternal and Child Health Care Hospital, Zibo, Shandong 255029, P.R. China
| | - Liping Chen
- Department of Pediatrics, Zibo Maternal and Child Health Care Hospital, Zibo, Shandong 255029, P.R. China
| |
Collapse
|
25
|
F-box and leucine-rich repeat protein 5 promotes colon cancer progression by modulating PTEN/PI3K/AKT signaling pathway. Biomed Pharmacother 2018; 107:1712-1719. [PMID: 30257389 DOI: 10.1016/j.biopha.2018.08.119] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 12/16/2022] Open
Abstract
The hyper-activation of PI3K/AKT signaling is common in many kinds of malignant tumors and promotes cell growth. Moreover, FBXL5 is reported to play an important role in the progression of gastric cancer and cervical cancer. In this view, this study aims to explore the function of FBXL5 in the progression of colon cancer and determine if PI3K/AKT signaling pathway involves in this process. Western blotting, RT-PCR, and immunohistochemistry were used to detect the expression pattern of FBXL5 in colon cancer tissues and cell lines. Immunofluorescence, Duolink, and immunoprecipitation (IP) assays were performed to evaluate the interaction between FBXL5 and PI3K/AKT signaling. Results showed that FBXL5 was elevated in colon cancer tissues and cells, which had physical interaction with PTEN protein and negatively regulated its expression, whereas positively modulated PI3K, AKT and mTOR expression and their phosphorylation. Besides, FBXL5 promoted cell proliferation and tumorigenesis and inhibited apoptosis by modulating PTEN/PI3K/AKT signaling. In conclusion, this study demonstrated that FBXL5 functioned as an oncogene in the progression of colon cancer through regulating PTEN/PI3K/AKT signaling.
Collapse
|
26
|
BMI1 Roles in Cancer Stem Cells and Its Association with MicroRNAs Dysregulation in Cancer: Emphasis on Colorectal Cancer. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2018. [DOI: 10.5812/ijcm.82926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
27
|
Tian J, Yuan L. Sirtuin 6 inhibits colon cancer progression by modulating PTEN/AKT signaling. Biomed Pharmacother 2018; 106:109-116. [PMID: 29957460 DOI: 10.1016/j.biopha.2018.06.070] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 02/08/2023] Open
Abstract
Decreased expression of the tumor suppressor sirtuin 6 (SIRT6) protein plays a role in tumorigenesis. The aim of this study was to investigate the effects of SIRT6 and its underlying mechanism in colon cancer progression. As shown by immunohistochemistry, Western blotting, and the real-time polymerase chain reaction (RT-PCR), SIRT6 expression was down-regulated in colon cancer tissues and different colon cancer cell lines, and down-regulation of SIRT6 showed a negative correlation with the overall survival of colon cancer patients. To assess the effects of SIRT6 on cell proliferation, apoptosis, invasion, and migration, 3-(4,5-dimethyl-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry, transwell, and wound healing assays were carried out, respectively. Results demonstrated that over-expression of SIRT6 inhibited cell proliferation, invasion, and migration and enhanced cell apoptosis. Co-immunoprecipitation (Co-IP) and Western blotting showed that up-regulation of SIRT6 increased the combined quantity of PTEN with SIRT6 proteins, and promoted the expression of PTEN and PIP2, as well as the stability of PTEN. SIRT6 also reduced the ubiquitination of PTEN and decreased protein levels of AKT1, phosphatidylinositol (3,4,5)-trisphosphate (PIP3), mTOR, cyclin d1, and c-myc. In addition, compared with cells over-expressed SIRT6, cell apoptosis was repressed and cell proliferation and tumorigenesis were enhanced in cells with SIRT6 over-expression and PTEN knockdown. In conclusion, the present study confirms that SIRT6 functions as a tumor suppressor gene in colon cancer by modulating PTEN/AKT signaling, which may provide a novel target for the treatment of colon cancer.
Collapse
Affiliation(s)
- Junhong Tian
- Department of Colorectal and Anal Surgery, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Leilei Yuan
- Department of Oncology, Jining No.1 People's Hospital, Jining, Shandong, 272000, China. 13905370037.@163.com
| |
Collapse
|
28
|
Kukcinaviciute E, Jonusiene V, Sasnauskiene A, Dabkeviciene D, Eidenaite E, Laurinavicius A. Significance of Notch and Wnt signaling for chemoresistance of colorectal cancer cells HCT116. J Cell Biochem 2018; 119:5913-5920. [PMID: 29637602 DOI: 10.1002/jcb.26783] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 02/02/2018] [Indexed: 12/21/2022]
Abstract
5-fluorouracil (5-FU) and oxaliplatin (OxaPt) are the main chemotherapeutics for colorectal cancer (CRC). Chemotherapy response rates for advanced CRC remain low, primarily due to intrinsic or acquired chemoresistance. The importance of Notch and Wnt signaling for carcinogenesis of CRC as well as crosstalk of Notch and Wnt signaling with many oncogenic signaling pathways suggest that Notch and Wnt pathways could be responsible for chemoresistance. In this study, we compared changes in Notch and Wnt signaling after 5-FU and OxaPt treatment in CRC cells HCT116 and its chemoresistant sublines HCT116/FU and HCT116/OXA. The levels of Notch1 receptor intracellular domain NICD1 and non-phosphorylated β-catenin, the reporters of Notch and Wnt signaling, were upregulated in untreated chemoresistant HCT116/FU and HCT116/OXA cells. Our data suggest that Notch inhibitor RO4929097 (RO) and Wnt inhibitor XAV939 (XAV) enhance the survival potential of OxaPt-treated cells. The protein level of Notch target gene HES1 was significantly upregulated in chemoresistant HCT116/FU and HCT116/OXA cells, compared to HCT116. HES1 silencing increased viability of HCT116 and its chemoresistant sublines after 5-FU or OxaPt treatment. The results of HES1 downregulation coincide with RO and XAV effects on cell viability of OxaPt-treated cells.
Collapse
Affiliation(s)
- Egle Kukcinaviciute
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Violeta Jonusiene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ausra Sasnauskiene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daiva Dabkeviciene
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Eigile Eidenaite
- Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania.,National Center of Pathology, Affiliate of Vilnius University Hospital Santara Clinics, Vilnius, Lithuania
| |
Collapse
|
29
|
Li Y, Zhang Y, Liu X, Wang M, Wang P, Yang J, Zhang S. Lutein inhibits proliferation, invasion and migration of hypoxic breast cancer cells via downregulation of HES1. Int J Oncol 2018; 52:2119-2129. [PMID: 29620169 DOI: 10.3892/ijo.2018.4332] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/14/2018] [Indexed: 11/06/2022] Open
Abstract
An intratumoral hypoxic microenvironment is frequently observed in solid tumors, including breast cancer. Lutein, a plant-derived compound and non-vitamin A carotenoid, has been demonstrated to possess multiple protective properties including anti-inflammation, anti-oxidative stress and antitumor effects. The main objective of the present research was to elucidate the involvement of lutein in the production of reactive oxygen species (ROS) under hypoxia, the activation of hairy and enhancer of split 1 (HES1), and the proliferation, invasion and migration of breast cancer cells. The human breast cancer cell lines MDA‑MB‑157 and MCF‑7 were exposed to hypoxic conditions and various concentrations of lutein. An MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay was performed to examine cell proliferation, and Annexin V-fluorescein isothiocyanate/propidium iodide staining was performed to analyze the apoptosis ratio. The levels of hypoxia inducible factor-1α (HIF‑1α), NOTCH signaling molecules, HES1 and epithelial-mesenchymal transition (EMT)-associated factors were examined by reverse transcription-quantitative polymerase chain reaction and western blot analysis. Wound healing and Transwell invasion assays were used to detect the invasion and migration of breast cancer cells. Intracellular ROS levels were examined using 2,7-dichlorodihydrofluorescein-diacetate and flow cytometry. The results revealed that cell proliferation was inhibited by lutein in a dose-dependent manner, and the apoptosis ratio gradually increased with lutein treatment under hypoxia as evident from flow cytometry-based analysis. Exposure to lutein inhibited hypoxia-mediated activation of HIF‑1α, NOTCH signaling and HES1 expression, and suppressed the hypoxia-induced expression of EMT-associated factors. Lutein markedly inhibited the invasion and migration of breast cancer cells under hypoxia. Hypoxia-induced production of ROS was also decreased by lutein. Furthermore, the ROS scavenger N‑acetylcysteine also suppressed hypoxia inducible factor 1α and HES1 expression in breast cancer cells during hypoxia, but hydrogen peroxide (H2O2) levels were increased. Taken together, the results of the present study suggested that lutein may be a novel candidate for the chemoprevention of breast cancer. Furthermore, HES1 may be crucial in mediating the involvement of lutein in the suppression of hypoxia-driven ROS-induced breast cancer progression.
Collapse
Affiliation(s)
- Yuan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuekun Zhang
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaosa Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Mingchen Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Peng Wang
- Department of Basic Medicine, Nursing College, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jianbo Yang
- Medical Laboratories, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - Shanfeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
30
|
Li Z, Wang L, Luo N, Zhao Y, Li J, Chen Q, Tian Y. Metformin inhibits the proliferation and metastasis of osteosarcoma cells by suppressing the phosphorylation of Akt. Oncol Lett 2018; 15:7948-7954. [PMID: 29725482 DOI: 10.3892/ol.2018.8297] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/17/2017] [Indexed: 02/07/2023] Open
Abstract
Metformin (Met) is a therapeutic agent for the treatment of type 2 diabetes mellitus. There is evidence that Met may reduce the risk of cancer in patients with type 2 diabetes mellitus by inhibiting tumor cell growth, prolonging the overall survival time in patients with various types of malignancy. However, the function and mechanism of Met have not been fully elucidated in osteosarcoma (OS). The present study evaluated the anti-proliferative effect of Met on MG63 and U2OS OS cells, identifying that it acted in a dose- and time-dependent manner. Met also inhibited OS cell migration and invasion, potentially by regulating the epithelial-mesenchymal transition in OS cells. Mechanistically, Met was demonstrated to partly exert these functions through the suppression of Akt phosphorylation, which was associated with increased phosphatase and tensin (PTEN) expression. Silencing PTEN prevented the Met-induced inhibition of the growth and metastasis of OS cells. As Met has anti-proliferative and anti-metastatic effects on OS cells it is a potential candidate, in combination with other chemotherapeutic agents, for use in the treatment of OS.
Collapse
Affiliation(s)
- Zuohong Li
- Department of Orthopedics, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Lesheng Wang
- Department of Blood Transfusion, Chinese People's Liberation Army 210 Hospital, Dalian, Liaoning 116015, P.R. China
| | - Nan Luo
- Department of Infectious Diseases, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116027, P.R. China
| | - Yantao Zhao
- Department of Orthopedics, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Jiazhi Li
- Department of Pathology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Qiwei Chen
- Department of Pathology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yu Tian
- Department of Pathology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
31
|
Liu X, Zhang Y, Shi M, Wang Y, Zhang F, Yan R, Liu L, Xiao Y, Guo B. Notch1 regulates PTEN expression to exacerbate renal tubulointerstitial fibrosis in diabetic nephropathy by inhibiting autophagy via interactions with Hes1. Biochem Biophys Res Commun 2018; 497:1110-1116. [PMID: 29496446 DOI: 10.1016/j.bbrc.2018.02.187] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 02/25/2018] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN) is a serious clinical microvascular complication of diabetes mellitus. DN is characterized by the accumulation of extracellular matrix, resulting in progressive fibrosis leading to the loss of renal function. Notch1 and phosphatase and tensin homolog deleted on chromosome ten (PTEN) signaling have been associated with fibrosis. Autophagy serves as an essential regulator of tubular cellular homeostasis. However, how these molecules control the balance between fibrosis and autophagy, the main homeostatic mechanism regulating fibrosis, is not well understood. This association was confirmed using Notch1-siRNA in vitro, which prevented the increase in Hes1 and restored PTEN expression. In contrast, transfection with pHAGE-Hes1 repressed PTEN promoter-driven luciferase activity, implying a direct relationship between Hes1 and PTEN. The expression of Notch1 and Hes1 was increased in diabetic db/db mice by western blotting; in contrast, the expression of PTEN was decreased. Importantly, the dysregulation of these signaling molecules was associated with an increase in extracellular matrix proteins (Collagen-I and III) and the inhibition of autophagy. Similar results were evident in response to high glucose concentrations in vitro in the NRK-52e cells. Therefore, the high glucose concentrations present in diabetes promote fibrosis through the Notch1 pathway via Hes1, while inhibiting the PTEN and autophagy. In conclusion, the inhibition of PTEN by Notch1/Hes1 in response to high glucose concentration inhibits autophagy, which is associated with the progression of fibrosis. Therefore, these signaling molecules may represent novel therapeutic targets in diabetic nephropathy.
Collapse
Affiliation(s)
- XingMei Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - YingYing Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - MingJun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - YuanYuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Rui Yan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550002, China
| | - LingLing Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou, 550025, China; Laboratory of Pathogenesis Research, Drug Prevention and Treatment of Major Diseases, Guizhou Medical University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
32
|
Liu J, Liu K, Jiang X, Wang X, Chen Y, Cui X, Pang L, Li S, Liu C, Zou H, Yang L, Zhao J, Qi Y, Hu JM, Li F. Clinicopathological significance of Bmi-1 overexpression in esophageal cancer: a meta-analysis. Biomark Med 2017; 12:71-81. [PMID: 29240461 DOI: 10.2217/bmm-2017-0092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
AIM The clinicopathological effects of Bmi-1 expression in esophageal cancer remain widely disputed. Our aim was to clarify this relationship. METHODS Available studies were retrieved from diverse databases. Review Manager 5.3 and Stata 12.0 software were used to identify correlations between Bmi-1 expression and the clinicopathological features of esophageal cancer. RESULTS From 16 studies, 1523 esophageal cancer patients were analyzed. Meta-analysis demonstrated that Bmi-1 overexpression was associated with differentiation (p = 0.03), tumor/node/metastasis stage (p = 0.02), depth of invasion (p = 0.0006) and lymph node metastasis (p = 0.008). CONCLUSION The expression of Bmi-1 is associated with the progression and invasion of esophageal cancer.
Collapse
Affiliation(s)
- Jihong Liu
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Kai Liu
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Xianli Jiang
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Xueli Wang
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Yunzhao Chen
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Xiaobin Cui
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Lijuan Pang
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Shugang Li
- Department of Preventive Medicine, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Chunxia Liu
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Hong Zou
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Lan Yang
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Jin Zhao
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Yan Qi
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Jian Ming Hu
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China
| | - Feng Li
- Department of Pathology & Key Laboratory for Xinjiang Endemic & Ethnic Diseases, Shihezi University School of Medicine, Xinjiang 832002, PR China.,Department of Pathology, Beijing ChaoYang Hospital, Capital Medical University, Beijing 100020, PR China
| |
Collapse
|
33
|
Wu Y, Gong L, Xu J, Mou Y, Xu X, Qian Z. The clinicopathological significance of HES1 promoter hypomethylation in patients with colorectal cancer. Onco Targets Ther 2017; 10:5827-5834. [PMID: 29263679 PMCID: PMC5726367 DOI: 10.2147/ott.s151857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hairy/enhancer of split 1 (HES1) is a basic helix-loop-helix transcriptional repressor. Aberrant demethylation has been considered a common mechanism of tumor promoter gene activation. In the current study, we aimed to investigate the methylation status of the HES1 promoter and correlations with clinicopathological parameters and prognosis in colorectal cancer (CRC). The expression of HES1 in 50 paired CRC specimens and adjacent normal tissues was determined by using quantitative real-time polymerase chain reaction and immunohistochemical analysis. Moreover, DNA methylation status was evaluated through methylation-specific polymerase chain reaction and bisulfite sequencing. The correlation of methylation status with HES1 expression level and clinicopathological parameters was statistically analyzed in CRC patients. Our data showed that the methylation level of HES1 was significantly decreased and negatively correlated with HES1 expression in CRC tissues. Moreover, HES1 hypomethylation was associated with a poor histological grade, Dukes' classification, lymph node metastasis, and clinical stages (P<0.05). Furthermore, survival analyses revealed that a decreased methylation status of HES1 was linked to poor prognosis of CRC patients. In conclusion, promoter hypomethylation upregulates HES1 expression and plays a critical role in the progression and prognosis of CRC patients.
Collapse
Affiliation(s)
- Yinfang Wu
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| | - Lijie Gong
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
| | - Ji Xu
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Yiping Mou
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaowu Xu
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhenyuan Qian
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Zhejiang
- Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
34
|
Xiao HJ, Ji Q, Yang L, Li RT, Zhang C, Hou JM. In vivo and in vitro effects of microRNA-124 on human gastric cancer by targeting JAG1 through the Notch signaling pathway. J Cell Biochem 2017; 119:2520-2534. [PMID: 28941308 DOI: 10.1002/jcb.26413] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/22/2017] [Indexed: 12/11/2022]
Abstract
In this study, we aim to determine the function of miR-124 on gastric cancer (GC) cells and the underlying mechanism that involves jaddeg1 (JAG1) and the Notch signaling pathway. GC tissues and adjacent tissues from 100 patients suffering from GC were selected. GC SGC-7901 and AGS cells were selected and grouped into control, mimic-NC, miR-124 mimic, inhibitor-NC, miR-124 inhibitor, and miR-124 inhibitor + si-JAG1 groups. RT-qPCR and a Western blotting assay were conducted to detect the expression of miR-124, JAG1, and Notch signaling pathway-related proteins (NICD, HES1, and HES5). MTS, wound-healing, transwell assay and flow cytometry were performed to detect cell proliferation, migration, invasion, cell cycle distribution, and apoptosis, respectively. Compared with adjacent tissues, a lower miR-124 expression and higher JAG1 expression were found in GC tissues. JAG1 is a direct target gene of miR-124. Compared with the control group, the expression of JAG1, NICD, HES1, and HES5, cell invasion, migration, and proliferation in the miR-124 mimic group were decreased, while the apoptosis rate was increased and cells were arrested at the G0/G1 phase. Compared with the miR-124 inhibitor group, the expression of JAG1, NICD, HES1, and HES5, cell invasion, migration, and proliferation in the miR-124 inhibitor + si-JAG1 group were decreased, while the apoptosis rate and cell ratio at the G0/G1 phase were increased. The demonstration that miR-124 inhibits GC cell growth supports the concept that miR-124 functions as a tumor suppressor by a mechanism that involves translational repression of the JAG1 and the inhibition of Notch signaling pathway.
Collapse
Affiliation(s)
- Hai-Juan Xiao
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, P.R. China.,Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, P.R. China
| | - Qing Ji
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, P.R. China
| | - Ren-Ting Li
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, P.R. China
| | - Cheng Zhang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, P.R. China
| | - Jun-Ming Hou
- Department of Surgical Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P.R. China
| |
Collapse
|
35
|
Fry NJ, Law BA, Ilkayeva OR, Holley CL, Mansfield KD. N6-methyladenosine is required for the hypoxic stabilization of specific mRNAs. RNA (NEW YORK, N.Y.) 2017; 23:1444-1455. [PMID: 28611253 PMCID: PMC5558913 DOI: 10.1261/rna.061044.117] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/02/2017] [Indexed: 05/19/2023]
Abstract
Post-transcriptional regulation of mRNA during oxygen deprivation, or hypoxia, can affect the survivability of cells. Hypoxia has been shown to increase stability of a subset of ischemia-related mRNAs, including VEGF. RNA binding proteins and miRNAs have been identified as important for post-transcriptional regulation of individual mRNAs, but corresponding mechanisms that regulate global stability are not well understood. Recently, mRNA modification by N6-methyladenosine (m6A) has been shown to be involved in post-transcriptional regulation processes including mRNA stability and promotion of translation, but the role of m6A in the hypoxia response is unknown. In this study, we investigate the effect of hypoxia on RNA modifications including m6A. Our results show hypoxia increases m6A content of poly(A)+ messenger RNA (mRNA), but not in total or ribosomal RNA in HEK293T cells. Using m6A mRNA immunoprecipitation, we identify specific hypoxia-modified mRNAs, including glucose transporter 1 (Glut1) and c-Myc, which show increased m6A levels under hypoxic conditions. Many of these mRNAs also exhibit increased stability, which was blocked by knockdown of m6A-specific methyltransferases METTL3/14. However, the increase in mRNA stability did not correlate with a change in translational efficiency or the steady-state amount of their proteins. Knockdown of METTL3/14 did reveal that m6A is involved in recovery of translational efficiency after hypoxic stress. Therefore, our results suggest that an increase in m6A mRNA during hypoxic exposure leads to post-transcriptional stabilization of specific mRNAs and contributes to the recovery of translational efficiency after hypoxic stress.
Collapse
Affiliation(s)
- Nate J Fry
- Biochemistry and Molecular Biology Department, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834, USA
| | - Brittany A Law
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina 27701, USA
| | - Christopher L Holley
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Kyle D Mansfield
- Biochemistry and Molecular Biology Department, Brody School of Medicine, East Carolina University, Greenville, North Carolina 27834, USA
| |
Collapse
|
36
|
Overexpression of microRNA-132 enhances the radiosensitivity of cervical cancer cells by down-regulating Bmi-1. Oncotarget 2017; 8:80757-80769. [PMID: 29113342 PMCID: PMC5655237 DOI: 10.18632/oncotarget.20358] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/21/2017] [Indexed: 02/04/2023] Open
Abstract
We examined the effects of microRNA-132 (miR-132) on Bmi-1 expression and radiosensitivity in HeLa, SiHa, and C33A cervical cancer (CC) cells and 104 CC patients. MiR-132 expression was decreased and Bmi-1 expression was increased in tumor tissues compared to adjacent normal tissues and in radiotherapy-resistant patients compared to radiotherapy-sensitive patients. MiR-132 expression and Bmi-1 mRNA expression were also negatively correlated in tumor tissues. HeLa, SiHa, and C33A cells were divided into blank, miR-132 negative control (NC), miR-132 inhibitor, miR-132 mimics, siBmi-1, and miR-132 inhibitor + siBmi-1 groups, after which expression of miR-132 and Bmi-1, and the interaction between them and cell survival, proliferation, and apoptosis were examined. Bmi-1 was confirmed as a target of miRNA-132. Survival was higher and apoptosis lower in the miR-132 inhibitor group than the blank group after various doses of radiation. By contrast, survival was lower and apoptosis higher in the miRNA-132 mimics and siBmi-1 groups than in the blank group. Moreover, miR-132 expression increased and Bmi-1 mRNA expression decreased in each group at radiation doses of 6 and 8 Gy. Finally, co-administration of radiotherapy and exogenous miR-132 inhibited the growth of HeLa cell transplant-induced tumors in nude mice more effectively than radiotherapy alone. These results suggest overexpression of miR-132 enhances the radiosensitivity of CC cells by down-regulating Bmi-1 and that miR-132 may be a useful new target for the treatment of CC.
Collapse
|
37
|
NOTCH1 activates the Wnt/β-catenin signaling pathway in colon cancer. Oncotarget 2017; 8:60378-60389. [PMID: 28947978 PMCID: PMC5601146 DOI: 10.18632/oncotarget.19534] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
PURPOSE AND METHODS The translocation of β-catenin/CTNNB1 to the nucleus activates Wnt signaling and cell proliferation; however, the precise mechanism underlying this phenomenon remains unknown. Previous reports have provided evidence that NOTCH1 is involved in the Wnt signaling pathway. Therefore, we sought to determine the mechanism by which NOTCH1 influences the Wnt/β-catenin pathway. We constructed a vector expressing the NOTCH1 intracellular domain (NICD1) and transfected the vector into HCT116 which has low expression of NICD1. Furthermore, inhibition of NOTCH signal pathway in SW480 which has abundant NICD1 expression, was performed by transfection of siNICD1 or DAPT, gamma secretase inhibitor, treatment. In addition, we evaluated NICD1 and β-catenin localization in colon cancer cell lines and in 189 colon cancer tissue samples and analyzed the correlation between the nuclear localization of NICD1 and the clinicopathological features of colon cancer patients. RESULTS Immunohistochemical assays demonstrated that NICD1 and β-catenin exhibited a similar localization pattern in colon cancer tissues. In addition, we found that NICD1 induced the translocation of β-catenin to the nucleus and that NICD1 and β-catenin co-localized in the nucleus. Overexpression of NICD1 increased luciferase activity of Wnt signal pathway. On the other hand, reduction of NICD1 reduced luciferase activity of Wnt signaling pathway. In the 189 analyzed colon cancer cases, multivariate COX regression analysis demonstrated the independent prognostic impact of nuclear localization of NICD1(p=0.0376). CONCLUSION NOTCH1 plays a key role in the Wnt pathway and activation of NOTCH1 is associated with the translocation of β-catenin to the nucleus.
Collapse
|
38
|
Goto N, Ueo T, Fukuda A, Kawada K, Sakai Y, Miyoshi H, Taketo MM, Chiba T, Seno H. Distinct Roles of HES1 in Normal Stem Cells and Tumor Stem-like Cells of the Intestine. Cancer Res 2017; 77:3442-3454. [PMID: 28536281 DOI: 10.1158/0008-5472.can-16-3192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/17/2017] [Accepted: 05/08/2017] [Indexed: 11/16/2022]
|
39
|
Yan Y, Wang Y, Zhao P, Ma W, Hu Z, Zhang K. BMI-1 Promotes Self-Renewal of Radio- and Temozolomide (TMZ)-Resistant Breast Cancer Cells. Reprod Sci 2017; 24:1620-1629. [DOI: 10.1177/1933719117697255] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yanfang Yan
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ying Wang
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pengxin Zhao
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weiyuan Ma
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhigang Hu
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kaili Zhang
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
40
|
Gu S, Zhang R, Gu J, Li X, Lv L, Jiang J, Xu Z, Wang S, Shi C, Wang DP, Wu C. HES5 promotes cellular proliferation of non-small cell lung cancer through STAT3 signaling. Oncol Rep 2016; 37:474-482. [DOI: 10.3892/or.2016.5268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/31/2016] [Indexed: 11/05/2022] Open
|
41
|
Zhou L, Jiang F, Chen X, Liu Z, Ouyang Y, Zhao W, Yu D. Downregulation of miR-221/222 by a microRNA sponge promotes apoptosis in oral squamous cell carcinoma cells through upregulation of PTEN. Oncol Lett 2016; 12:4419-4426. [PMID: 28101204 PMCID: PMC5228168 DOI: 10.3892/ol.2016.5250] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/02/2016] [Indexed: 12/18/2022] Open
Abstract
MicroRNA-221 and microRNA-222 (miR-221/222) have been identified as oncogenes and confirmed to be overexpressed in various types of cancer. However, the regulation mechanism of miR-221/222 in oral squamous cell carcinoma (OSCC) remains to be fully elucidated. Previously, an miR-221/222 sponge was successfully constructed and its effect on the downregulation of miR-221/222 expression was investigated. In the present study, the dual luciferase reporter assay revealed a phosphatase and tensin homolog (PTEN) deletion on chromosome 10 to be a target gene of miR-221/222. It was also demonstrated that miR-221/222 suppression by transfection with an miR-221/222 sponge in vitro resulted in upregulation of PTEN. Notably, the proliferation and invasiveness of the miR-221/222 sponge-transfected cells was significantly inhibited, while apoptosis was promoted, when determined by Cell Counting Kit-8, Transwell assays and flow cytometry. The results of the present study prove that miR-221/222 may downregulate the expression of PTEN in OSCC cells and function as oncogenes, providing a novel insight into the underlying mechanism of OSCC tumorigenesis. The present study suggests that upregulating the expression of PTEN by downregulation of miR-221/222 may be a potential treatment for OSCC.
Collapse
Affiliation(s)
- Lijie Zhou
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China; Department of Stomatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Fangfang Jiang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Xijuan Chen
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Zifeng Liu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Ying Ouyang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Wei Zhao
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Dongsheng Yu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|
42
|
Luo Y, Ye GY, Qin SL, Mu YF, Zhang L, Qi Y, Qiu YE, Yu MH, Zhong M. High expression of Rab3D predicts poor prognosis and associates with tumor progression in colorectal cancer. Int J Biochem Cell Biol 2016; 75:53-62. [DOI: 10.1016/j.biocel.2016.03.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/04/2016] [Accepted: 03/28/2016] [Indexed: 12/18/2022]
|