1
|
Xu K, Fu A, Li Z, Miao L, Lou Z, Jiang K, Lau C, Su T, Tong T, Bao J, Lyu A, Kwan HY. Elevated extracellular matrix protein 1 in circulating extracellular vesicles supports breast cancer progression under obesity conditions. Nat Commun 2024; 15:1685. [PMID: 38402239 PMCID: PMC10894219 DOI: 10.1038/s41467-024-45995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
The cargo content in small extracellular vesicles (sEVs) changes under pathological conditions. Our data shows that in obesity, extracellular matrix protein 1 (ECM1) protein levels are significantly increased in circulating sEVs, which is dependent on integrin-β2. Knockdown of integrin-β2 does not affect cellular ECM1 protein levels but significantly reduces ECM1 protein levels in the sEVs released by these cells. In breast cancer (BC), overexpressing ECM1 increases matrix metalloproteinase 3 (MMP3) and S100A/B protein levels. Interestingly, sEVs purified from high-fat diet-induced obesity mice (D-sEVs) deliver more ECM1 protein to BC cells compared to sEVs from control diet-fed mice. Consequently, BC cells secrete more ECM1 protein, which promotes cancer cell invasion and migration. D-sEVs treatment also significantly enhances ECM1-mediated BC metastasis and growth in mouse models, as evidenced by the elevated tumor levels of MMP3 and S100A/B. Our study reveals a mechanism and suggests sEV-based strategies for treating obesity-associated BC.
Collapse
Affiliation(s)
- Keyang Xu
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ai Fu
- Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaoyi Li
- Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liangbin Miao
- Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhonghan Lou
- Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keying Jiang
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Condon Lau
- Department of Physics, City University of Hong Kong, Hong Kong, China
| | - Tao Su
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tiejun Tong
- Department of Mathematics, Hong Kong Baptist University, Hong Kong, China
| | - Jianfeng Bao
- Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Aiping Lyu
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Systems Medicine and Health Sciences, Hong Kong Baptist University, Hong Kong, China.
| | - Hiu Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
- Institute of Systems Medicine and Health Sciences, Hong Kong Baptist University, Hong Kong, China.
- Institute of Research and Continuing Education, Hong Kong Baptist University, Shenzhen, China.
| |
Collapse
|
2
|
Gehris J, Ervin C, Hawkins C, Womack S, Churillo AM, Doyle J, Sinusas AJ, Spinale FG. Fibroblast activation protein: Pivoting cancer/chemotherapeutic insight towards heart failure. Biochem Pharmacol 2024; 219:115914. [PMID: 37956895 PMCID: PMC10824141 DOI: 10.1016/j.bcp.2023.115914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
An important mechanism for cancer progression is degradation of the extracellular matrix (ECM) which is accompanied by the emergence and proliferation of an activated fibroblast, termed the cancer associated fibroblast (CAF). More specifically, an enzyme pathway identified to be amplified with local cancer progression and proliferation of the CAF, is fibroblast activation protein (FAP). The development and progression of heart failure (HF) irrespective of the etiology is associated with left ventricular (LV) remodeling and changes in ECM structure and function. As with cancer, HF progression is associated with a change in LV myocardial fibroblast growth and function, and expresses a protein signature not dissimilar to the CAF. The overall goal of this review is to put forward the postulate that scientific discoveries regarding FAP in cancer as well as the development of specific chemotherapeutics could be pivoted to target the emergence of FAP in the activated fibroblast subtype and thus hold translationally relevant diagnostic and therapeutic targets in HF.
Collapse
Affiliation(s)
- John Gehris
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlie Ervin
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlotte Hawkins
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Sydney Womack
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Jonathan Doyle
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Albert J Sinusas
- Yale University Cardiovascular Imaging Center, New Haven CT, United States
| | - Francis G Spinale
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States.
| |
Collapse
|
3
|
Lv C, Ren C, Yu Y, Yin H, Huang C, Yang G, Hong Y. Wentilactone A Reverses the NF-κB/ECM1 Signaling-Induced Cisplatin Resistance through Inhibition of IKK/IκB in Ovarian Cancer Cells. Nutrients 2022; 14:nu14183790. [PMID: 36145166 PMCID: PMC9504226 DOI: 10.3390/nu14183790] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Wentilactone A (WA) is a tetranorditerpenoid isolated from marine algae. We previously found that WA inhibited cancer cell proliferation with little toxicity. In this study, we show that high expression of extracellular matrix protein-1 (ECM1) promotes cancer cell cisplatin resistance, and the secreted ECM1 activates normal fibroblasts (NFs) to transform cells with characteristics of cancer-associated fibroblasts (CAFs). Transcription of the ECM1 gene is regulated largely by NF-κB through EP881C/T-EP266C binding sites. WA supresses the phosphorylation of NF-κB through inhibition of the upstream IKK/IκB phoshorylation to block the expression of ECM1, which reverses the cisplatin-induced activation of NF-κB/ECM1. On the contrary, cisplatin facilitates phosphorylation of NF-κB to enhance the expression of ECM1. These results highlight ECM1 as a potential target for treatment of cisplatin-resistant cancers associated with the ECM1 activated signaling. In addition, WA reverses cisplatin resistance by targeting both tumor cells and the tumor microenvironment through IKK/IκB/NF-κB signaling to reduce the expression of the ECM1 protein.
Collapse
Affiliation(s)
- Cuiting Lv
- Central Laboratory, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Chunxia Ren
- Center for Reproductive Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yinjue Yu
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Huijing Yin
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Caiguo Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medical, Navy Medical University, Shanghai 200433, China
- Correspondence: (C.H.); (G.Y.); (Y.H.)
| | - Gong Yang
- Central Laboratory, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical School, Fudan University, Shanghai 200032, China
- Correspondence: (C.H.); (G.Y.); (Y.H.)
| | - Yang Hong
- Central Laboratory, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China
- Department of Orthopedics, The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai 200240, China
- Correspondence: (C.H.); (G.Y.); (Y.H.)
| |
Collapse
|
4
|
Liu YR, Song DD, Liang DM, Li YJ, Yan YF, Sun HF, Zhang ML, Hu JX, Zhao YL, Liang Y, Li YM, Yang Z, Wang RR, Zheng HF, Wang P, Xie SY. Oncogenic TRIB2 interacts with and regulates PKM2 to promote aerobic glycolysis and lung cancer cell procession. Cell Death Dis 2022; 8:306. [PMID: 35790734 PMCID: PMC9256704 DOI: 10.1038/s41420-022-01095-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022]
Abstract
PKM2 is an important regulator of the aerobic glycolysis that plays a vital role in cancer cell metabolic reprogramming. In general, Trib2 is considered as a “pseudokinase”, contributing to different kinds of cancer. However, the detailed roles of TRIB2 in regulating cancer metabolism by PKM2 remain unclear. This study demonstrated that TRIB2, not a “pseudokinase”, has the kinase activity to directly phosphorylate PKM2 at serine 37 in cancer cells. The elevated pSer37-PKM2 would subsequently promote the PKM2 dimers to enter into nucleus and increase the expression of LDHA, GLUT1, and PTBP1. The aerobic glycolysis is then elevated to promote cancer cell proliferation and migration in TRIB2- or PKM2-overexpressed cultures. The glucose uptake and lactate production increased, but the ATP content decreased in TRIB2- or PKM2-treated cultures. Experiments of TRIB2−/− mice further supported that TRIB2 could regulate aerobic glycolysis by PKM2. Thus, these results reveal the new kinase activity of TRIB2 and its mechanism in cancer metabolism may be related to regulating PKM2 to promote lung cancer cell proliferation in vitro and in vivo, suggesting promising therapeutic targets for cancer therapy by controlling cancer metabolism.
Collapse
|
5
|
Abstract
Extracellular matrix protein 1 (ECM1) is associated with a poor prognosis of breast cancers. However, the role of ECM1 with endocrine resistance in estrogen receptor-positive (ER+) breast cancers has not been elucidated yet. We show that ECM1 promotes endocrine resistance in ER+ breast cancers. ECM1 is overexpressed in luminal breast cancer patients compared to the basal type of breast cancer. Significantly, higher expression of ECM1 is associated with poor response to endocrine therapies in luminal B breast cancer patients. We found that ECM1 is upregulated in CAMA1 and MDA-MB-361 cells grown in long-term estrogen-deprived (LTED) conditions. Moreover, the ablation of ECM1 significantly inhibited the proliferation of CAMA1 LTED and MDA-MB-361 LTED cells. Finally, an interrogation of a dataset containing transcriptome and proteome of breast cancer cell lines revealed that the level of ECM1 mRNA is positively correlated with that of phosphorylated Src. Based on these findings, we strongly suggest that ECM1 significantly contributes to the acquisition of endocrine resistance in ER+ breast cancers by the activation of Src.
Collapse
Affiliation(s)
- Tae Won Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
| | - Kyung-min Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Natural Sciences, Hanyang University, Seoul, Republic of Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, Republic of Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Zhou WJ, Yang HL, Mei J, Chang KK, Lu H, Lai ZZ, Shi JW, Wang XH, Wu K, Zhang T, Wang J, Sun JS, Ye JF, Li DJ, Zhao JY, Jin LP, Li MQ. Fructose-1,6-bisphosphate prevents pregnancy loss by inducing decidual COX-2 + macrophage differentiation. SCIENCE ADVANCES 2022; 8:eabj2488. [PMID: 35196096 PMCID: PMC8865779 DOI: 10.1126/sciadv.abj2488] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 12/23/2021] [Indexed: 05/23/2023]
Abstract
Decidualization is an intricate biological process in which extensive remodeling of the endometrium occurs to support the development of an implanting blastocyst. However, the immunometabolic mechanisms underlying this process are still largely unknown. We found that the decidualization process is accompanied by the accumulation of fructose-1,6-bisphosphate (FBP). The combination of FBP with pyruvate kinase M stimulated IL-27 secretion by endometrial stromal cells in an ERK/c-FOS-dependent manner. IL-27 induced decidual COX-2+ M2-like macrophage differentiation, which promotes decidualization, trophoblast invasion, and maternal-fetal tolerance. Transfer of Ptgs2+/COX-2+ macrophages prevented fetal loss in Il27ra-deleted pregnant mice. FBP levels were low in plasma and decidual tissues of patients with unexplained recurrent spontaneous abortion. In therapeutic studies, FBP supplementation significantly improved embryo loss by up-regulation of IL-27-induced COX-2+ macrophage differentiation in a mouse model of spontaneous abortion. These findings collectively provide a scientific basis for a potential therapeutic strategy to prevent pregnancy loss.
Collapse
Affiliation(s)
- Wen-Jie Zhou
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People’s Republic of China
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Hui-Li Yang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jie Mei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medicine School, Nanjing 210000, People’s Republic of China
| | - Kai-Kai Chang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Han Lu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Zhen-Zhen Lai
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jia-Wei Shi
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Xiao-Hui Wang
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Ke Wu
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, People’s Republic of China
| | - Jian Wang
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jian-Song Sun
- National Research Centre for Carbohydrate Synthesis, Jiangxi Normal University, Nanchang, Jiangxi Province 330022, People’s Republic of China
| | - Jiang-Feng Ye
- Division of Obstetrics and Gynecology, KK Women’s and Children’s Hospital, Singapore 229899, Singapore
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| | - Jian-Yuan Zhao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, People’s Republic of China
| | - Li-Ping Jin
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, People’s Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People’s Republic of China
| |
Collapse
|
7
|
Liu F, Ma M, Gao A, Ma F, Ma G, Liu P, Jia C, Wang Y, Donahue K, Zhang S, Ong IM, Keles S, Li L, Xu W. PKM2-TMEM33 axis regulates lipid homeostasis in cancer cells by controlling SCAP stability. EMBO J 2021; 40:e108065. [PMID: 34487377 PMCID: PMC8591543 DOI: 10.15252/embj.2021108065] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 02/01/2023] Open
Abstract
The pyruvate kinase M2 isoform (PKM2) is preferentially expressed in cancer cells to regulate anabolic metabolism. Although PKM2 was recently reported to regulate lipid homeostasis, the molecular mechanism remains unclear. Herein, we discovered an ER transmembrane protein 33 (TMEM33) as a downstream effector of PKM2 that regulates activation of SREBPs and lipid metabolism. Loss of PKM2 leads to up-regulation of TMEM33, which recruits RNF5, an E3 ligase, to promote SREBP-cleavage activating protein (SCAP) degradation. TMEM33 is transcriptionally regulated by nuclear factor erythroid 2-like 1 (NRF1), whose cleavage and activation are controlled by PKM2 levels. Total plasma cholesterol levels are elevated by either treatment with PKM2 tetramer-promoting agent TEPP-46 or by global PKM2 knockout in mice, highlighting the essential function of PKM2 in lipid metabolism. Although depletion of PKM2 decreases cancer cell growth, global PKM2 knockout accelerates allografted tumor growth. Together, our findings reveal the cell-autonomous and systemic effects of PKM2 in lipid homeostasis and carcinogenesis, as well as TMEM33 as a bona fide regulator of lipid metabolism.
Collapse
Affiliation(s)
- Fabao Liu
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Min Ma
- School of PharmacyUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Ang Gao
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Fengfei Ma
- School of PharmacyUniversity of Wisconsin‐MadisonMadisonWIUSA
- Present address:
Protein Sciences, Discovery BiologicsMerck & Co., Inc.South San FranciscoCAUSA
| | - Gui Ma
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Peng Liu
- Department of Biostatistics and Medical InformaticsUniversity of Wisconsin‐MadisonMadisonWIUSA
- UW Carbone Cancer CenterSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Chenxi Jia
- School of PharmacyUniversity of Wisconsin‐MadisonMadisonWIUSA
- Present address:
State Key Laboratory of ProteomicsNational Center for Protein Sciences‐BeijingBeijing Proteome Research CenterBeijing Institute of Radiation MedicineBeijingChina
| | - Yidan Wang
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Kristine Donahue
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Shengjie Zhang
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Irene M Ong
- Department of Biostatistics and Medical InformaticsUniversity of Wisconsin‐MadisonMadisonWIUSA
- UW Carbone Cancer CenterSchool of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Sunduz Keles
- Department of StatisticsUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Lingjun Li
- School of PharmacyUniversity of Wisconsin‐MadisonMadisonWIUSA
- Department of ChemistryUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Wei Xu
- McArdle Laboratory for Cancer ResearchUniversity of Wisconsin‐MadisonMadisonWIUSA
| |
Collapse
|
8
|
Extracellular matrix protein-1 secretory isoform promotes ovarian cancer through increasing alternative mRNA splicing and stemness. Nat Commun 2021; 12:4230. [PMID: 34244494 PMCID: PMC8270969 DOI: 10.1038/s41467-021-24315-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 06/10/2021] [Indexed: 12/27/2022] Open
Abstract
Extracellular matrix protein-1 (ECM1) promotes tumorigenesis in multiple organs but the mechanisms associated to ECM1 isoform subtypes have yet to be clarified. We report in this study that the secretory ECM1a isoform induces tumorigenesis through the GPR motif binding to integrin αXβ2 and the activation of AKT/FAK/Rho/cytoskeleton signaling. The ATP binding cassette subfamily G member 1 (ABCG1) transduces the ECM1a-integrin αXβ2 interactive signaling to facilitate the phosphorylation of AKT/FAK/Rho/cytoskeletal molecules and to confer cancer cell cisplatin resistance through up-regulation of the CD326-mediated cell stemness. On the contrary, the non-secretory ECM1b isoform binds myosin and blocks its phosphorylation, impairing cytoskeleton-mediated signaling and tumorigenesis. Moreover, ECM1a induces the expression of the heterogeneous nuclear ribonucleoprotein L like (hnRNPLL) protein to favor the alternative mRNA splicing generating ECM1a. ECM1a, αXβ2, ABCG1 and hnRNPLL higher expression associates with poor survival, while ECM1b higher expression associates with good survival. These results highlight ECM1a, integrin αXβ2, hnRNPLL and ABCG1 as potential targets for treating cancers associated with ECM1-activated signaling. Extracellular matrix protein 1 (ECM1) has been associated with cancer but the underlying molecular mechanisms are not clear. Here, the authors show that while ECM1b isoform is a tumour suppressor, the secreted isoform ECM1a promotes tumourigenesis and chemoresistance through increasing stemness and alternative mRNA splicing in ovarian cancer.
Collapse
|
9
|
Abbaszadegan MR, Mojarrad M, Moghbeli M. Role of extra cellular proteins in gastric cancer progression and metastasis: an update. Genes Environ 2020; 42:18. [PMID: 32467737 PMCID: PMC7227337 DOI: 10.1186/s41021-020-00157-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background Gastric cancer (GC) is one of the most common cancers in the world with a high ratio of mortality. Regarding the late diagnosis, there is a high ratio of distant metastasis among GC cases. Despite the recent progresses in therapeutic modalities, there is not still an efficient therapeutic method to increase survival rate of metastatic GC cases. Main body Apart from the various intracellular signaling pathways which are involved in tumor cell migration and metastasis, the local microenvironment is also a critical regulator of tumor cell migration. Indeed, the intracellular signaling pathways also exert their final metastatic roles through regulation of extra cellular matrix (ECM). Therefore, it is required to assess the role of extra cellular components in biology of GC. Conclusion In the present review, we summarize 48 of the significant ECM components including 17 ECM modifying enzymes, seven extracellular angiogenic factors, 13 cell adhesion and cytoskeletal organizers, seven matricellular proteins and growth factors, and four proteoglycans and extra cellular glycoproteins. This review paves the way of determination of a specific extra cellular diagnostic and prognostic panel marker for the GC patients.
Collapse
Affiliation(s)
| | - Majid Mojarrad
- 2Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- 2Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Identification of Sca-1 +Abcg1 + bronchioalveolar epithelial cells as the origin of lung adenocarcinoma in Gprc5a-knockout mouse model through the interaction between lung progenitor AT2 and Lgr5 cells. Oncogene 2020; 39:3754-3773. [PMID: 32157214 PMCID: PMC7190569 DOI: 10.1038/s41388-020-1251-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/14/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
The reason for the reduced efficacy of lung cancer therapy is the existence of lung cancer stem cells (CSCs). Targeting CSCs results in evolved phenotypes with increased malignancy, leading to therapy failure. Here, we propose a new therapeutic strategy: investigating the “transitional” cells that represent the stage between normal lung stem cells and lung CSCs. Identifying and targeting the key molecule that drives carcinogenesis to inhibit or reverse this process would thus provide new perspectives for early diagnosis and intervention in lung cancer. We used Gprc5a-knockout (KO) mice, the first animal model of spontaneous lung adenocarcinoma established by the deletion of a single lung tumor suppressor gene. We investigated the interaction of lung progenitor cells AT2 with Lgr5 cells in the generation of CSCs and related signaling mechanism. In the present study, using Gprc5a-KO mice, we found the initiator Sca-1+Abcg1+ subset with a CSC-like phenotype within the lung progenitor AT2 cell population in mice that had not yet developed tumors. We confirmed the self-renewal and tumor initiation capacities of this subset in vitro, in vivo, and clinical samples. Mechanistically, we found that the generation of Sca-1+Abcg1+ cells was associated with an interaction between AT2 and Lgr5 cells and the subsequent activation of the ECM1-α6β4-ABCG1 axis. Importantly, Sca-1+Abcg1+ and SPA+ABCG1+ cells specifically existed in the small bronchioles of Gprc5a-KO mice and patients with pneumonia, respectively. Thus, the present study unveiled a new kind of lung cancer-initiating cells (LCICs) and provided potential markers for the early diagnosis of lung cancer.
Collapse
|
11
|
Wang Z, Zhou Q, Li A, Huang W, Cai Z, Chen W. Extracellular matrix protein 1 (ECM1) is associated with carcinogenesis potential of human bladder cancer. Onco Targets Ther 2019; 12:1423-1432. [PMID: 30863109 PMCID: PMC6389008 DOI: 10.2147/ott.s191321] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Bladder cancer (BCa) is a common urological malignant tumor worldwide, and recurrence and death still remain high. New therapeutic targets are needed to treat patients who are not sensitive to current therapy. Extracellular matrix protein 1 (ECM1) is a key player in multiple epithelial malignancies. However, the knowledge regarding the expression of ECM1 in BCa and the mechanisms by which ECM1 affects BCa tumor progression is unclear. Materials and methods ECM1 expression levels in BCa tissues and cells were detected by quantitative real-time PCR (qRT-PCR), immunohistochemistry and Western blot. ECM1 expression was suppressed by shRNAs. Cell Counting Kit-8 (CCK-8), luminescent cell viability assay and 5-ethynyl-2′-deoxyuridine (EdU) assay were used to detect cell proliferation. Flow cytometry and transwell assay were used to evaluate cell apoptosis and invasion, respectively. All statistical analyses were performed by using the GraphPad Prism 7 software package. Results In this study, the expression of ECM1 in BCa specimens and cell lines was examined and displayed a significant increase compared with noncancerous counterparts, while ECM1-knockdown affected not only cell proliferation and migration, but also cell invasion ability and apoptosis potential, corresponding to the finding that ECM1 overexpression in BCa patients was associated with a poor prognosis. Additionally, after suppression of ECM1, the expression of glucose transporter 1 (GLUT1), lactate dehydrogenase (LDHA) and hypoxia-inducible factor 1α (HIF-1α), genes involved in Warburg effect regulation, were significantly decreased, and the lactate production was also obviously reduced in ECM1-silenced cells. Conclusion Our investigations revealed that the expression of ECM1 was closely associated with tumor cell growth, migration and apoptosis at least in part through regulation of Warburg effect, defining ECM1 as an effective predictor in the carcinogenesis and postoperative recurrence of human BCa.
Collapse
Affiliation(s)
- Zhicai Wang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| | - Qun Zhou
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| | - Aolin Li
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| | - Weiren Huang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ; .,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China, .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China,
| | - Zhiming Cai
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ; .,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China, .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China,
| | - Wei Chen
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| |
Collapse
|
12
|
A critical review of the role of M 2PYK in the Warburg effect. Biochim Biophys Acta Rev Cancer 2019; 1871:225-239. [PMID: 30708038 PMCID: PMC6525063 DOI: 10.1016/j.bbcan.2019.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/14/2019] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
It is becoming generally accepted in recent literature that the Warburg effect in cancer depends on inhibition of M2PYK, the pyruvate kinase isozyme most commonly expressed in tumors. We remain skeptical. There continues to be a general lack of solid experimental evidence for the underlying idea that a bottle neck in aerobic glycolysis at the level of M2PYK results in an expanded pool of glycolytic intermediates (which are thought to serve as building blocks necessary for proliferation and growth of cancer cells). If a bottle neck at M2PYK exists, then the remarkable increase in lactate production by cancer cells is a paradox, particularly since a high percentage of the carbons of lactate originate from glucose. The finding that pyruvate kinase activity is invariantly increased rather than decreased in cancer undermines the logic of the M2PYK bottle neck, but is consistent with high lactate production. The "inactive" state of M2PYK in cancer is often described as a dimer (with reduced substrate affinity) that has dissociated from an active tetramer of M2PYK. Although M2PYK clearly dissociates easier than other isozymes of pyruvate kinase, it is not clear that dissociation of the tetramer occurs in vivo when ligands are present that promote tetramer formation. Furthermore, it is also not clear whether the dissociated dimer retains any activity at all. A number of non-canonical functions for M2PYK have been proposed, all of which can be challenged by the finding that not all cancer cell types are dependent on M2PYK expression. Additional in-depth studies of the Warburg effect and specifically of the possible regulatory role of M2PYK in the Warburg effect are needed.
Collapse
|
13
|
Hassani F, Oryan S, Eftekhari-Yazdi P, Bazrgar M, Moini A, Nasiri N, Sharifi-Zarchi A. Downregulation of Extracellular Matrix and Cell Adhesion Molecules in Cumulus Cells of Infertile Polycystic Ovary Syndrome Women with and without Insulin Resistance. CELL JOURNAL 2018; 21:35-42. [PMID: 30507086 PMCID: PMC6275428 DOI: 10.22074/cellj.2019.5576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/27/2018] [Indexed: 11/04/2022]
Abstract
Objective The extracellular matrix (ECM) of the cumulus oocyte complex (COC) is composed of several molecules that have different roles during follicle development. This study aims to explore gene expression profiles for ECM and cell adhesion molecules in the cumulus cells of polycystic ovary syndrome (PCOS) patients based on their insulin sensitivity following controlled ovarian stimulation (COS). Materials and Methods In this prospective case-control study enrolled 23 women less than 36 years of age who participated in an intracytoplasmic sperm injection (ICSI) program. Patients were subdivided into 3 groups: control (n=8, fertile women with male infertility history), insulin resistant (IR) PCOS (n=7), and insulin sensitive (IS) PCOS (n=8). We compared 84 ECM component and adhesion molecule gene expressions by quantitative real-time polymerase chain reaction array (qPCR-array) among the groups. Results We noted that 21 of the 84 studied genes differentially expressed among the groups, from which 18 of these genes downregulated. Overall, comparison of PCOS cases with controls showed downregulation of extracellular matrix protein 1 (ECM1); catenin (cadherin-associated protein), alpha 1 (CTNNA1); integrin, alpha 5 (ITGA5); laminin, alpha 3 (LAMA3); laminin, beta 1 (LAMB1); fibronectin 1 (FN1); and integrin, alpha 7 (ITGA7). In the IS group, there was upregulation of ADAM metallopeptidase with thrombospondin type 1 motif, 8 (ADAMTS8) and neural cell adhesion molecule 1 (NCAM1) compared with the controls (P<0.05). Conclusion Downregulation of ECM and cell adhesion molecules seem to be related to PCOS. Gene expression profile alterations in cumulus cells from both the IS and IR groups of PCOS patients seems to be involved in the composition and regulation of ECM during the ovulation process. This study highlights the association of ECM gene alteration as a viewpoint for additional understanding of the etiology of PCOS.
Collapse
Affiliation(s)
- Fatemeh Hassani
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.Electronic Address:
| | - Shahrbanoo Oryan
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Poopak Eftekhari-Yazdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran. Electronic Address:
| | - Masood Bazrgar
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Nasiri
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ali Sharifi-Zarchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
14
|
MiR-3662 suppresses hepatocellular carcinoma growth through inhibition of HIF-1α-mediated Warburg effect. Cell Death Dis 2018; 9:549. [PMID: 29748591 PMCID: PMC5945826 DOI: 10.1038/s41419-018-0616-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 02/07/2023]
Abstract
Glucose metabolic reprogramming from oxidative to aerobic glycolysis, referred as the Warburg effect, is a hallmark of tumor cells. Accumulating evidence suggests that a subset of microRNAs play pivotal roles in modulating such reprogramming of glucose metabolism in cancer cells. miR-3662 has been implicated previously in both pro-tumorigenic and anti-tumorigenic effects in several types of cancer. The expression level of miR-3662 is downregulated in acute myeloid leukemia, whereas increased miR-3662 expression is observed in lung adenocarcinoma. However, the roles and underlying mechanisms of miR-3662 in hepatocellular carcinoma (HCC) metabolic reprogramming remain unclear. Our present study revealed that miR-3662 was frequently downregulated in HCC tissues and cell lines. The low expression level of miR-3662 was associated with tumor size, tumor multiplicity, Edmondson grade, and tumor-node-metastasis stage. Gain-of-function and loss-of-function assays showed that miR-3662 dampened glycolysis by reducing lactate production, glucose consumption, cellular glucose-6-phosphate level, ATP generation, and extracellular acidification rate, and increasing oxygen consumption rate in HCC cells after treatment with the hypoxia mimetic CoCl2. Moreover, miR-3662 suppressed cell growth in vitro and in vivo, and induced G1/S cell cycle arrest. miR-3662 inhibited the activation of ERK and JNK signaling pathways in HCC. By combined computational and experimental approaches, hypoxia-inducible factor-1α (HIF-1α) was determined as a direct target of miR-3662. After treatment with the hypoxia mimetic CoCl2, miR-3662 regulated the Warburg effect and HCC progression via decreasing HIF-1α expression. Our findings uncover a mechanistic role for miR-3662/HIF-1α axis in HCC metabolic reprogramming, providing a potential therapeutic strategy in liver cancer.
Collapse
|
15
|
Li Y, He ZC, Liu Q, Zhou K, Shi Y, Yao XH, Zhang X, Kung HF, Ping YF, Bian XW. Large Intergenic Non-coding RNA-RoR Inhibits Aerobic Glycolysis of Glioblastoma Cells via Akt Pathway. J Cancer 2018; 9:880-889. [PMID: 29581766 PMCID: PMC5868152 DOI: 10.7150/jca.20869] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 01/29/2018] [Indexed: 12/21/2022] Open
Abstract
Reprogramming energy metabolism is a hallmark of malignant tumors, including glioblastoma (GBM). Aerobic glycolysis is often utilized by tumor cells to maintain survival and proliferation. However, the underlying mechanisms of aerobic glycolysis in GBM remain elusive. Herein, we demonstrated that large intergenic non-coding RNA-RoR (LincRNA-RoR) functioned as a critical suppressor to inhibit the aerobic glycolysis and viability of GBM cells. We found that LincRNA-RoR was markedly reduced in GBM tissues compared with adjacent non-tumor tissues from 10 cases of GBM patients. Consistently, LincRNA-RoR expression in GBM cells was significantly lower than that in normal glial cells. The aerobic glycolysis of GBM cells, as determined by the measurement of glucose uptake and lactate production, was impaired by LincRNA-RoR overexpression. Mechanistically, LincRNA-RoR inhibited the expression of Rictor, the key component of mTORC2 (mammalian target of rapamycin complex 2), to suppress the activity of Akt pathway and impair the expression of glycolytic effectors, including Glut1, HK2, PKM2 and LDHA. Finally, enforced expression of LincRNA-RoR reduced the proliferation of GBM cells in vitro, restrained tumor growth in vivo, and repressed the expression of glycolytic molecules in GBM xenografts. Collectively, our results underscore LincRNA-RoR as a new suppressor of GBM aerobic glycolysis with therapeutic potential.
Collapse
Affiliation(s)
- Yong Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Kai Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Hsiang-Fu Kung
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing 400038, China
| |
Collapse
|
16
|
Prakasam G, Iqbal MA, Bamezai RNK, Mazurek S. Posttranslational Modifications of Pyruvate Kinase M2: Tweaks that Benefit Cancer. Front Oncol 2018; 8:22. [PMID: 29468140 PMCID: PMC5808394 DOI: 10.3389/fonc.2018.00022] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/22/2018] [Indexed: 01/02/2023] Open
Abstract
Cancer cells rewire metabolism to meet biosynthetic and energetic demands. The characteristic increase in glycolysis, i.e., Warburg effect, now considered as a hallmark, supports cancer in various ways. To attain such metabolic reshuffle, cancer cells preferentially re-express the M2 isoform of pyruvate kinase (PKM2, M2-PK) and alter its quaternary structure to generate less-active PKM2 dimers. The relatively inactive dimers cause the accumulation of glycolytic intermediates that are redirected into anabolic pathways. In addition, dimeric PKM2 also benefits cancer cells through various non-glycolytic moonlight functions, such as gene transcription, protein kinase activity, and redox balance. A large body of data have shown that several distinct posttranslation modifications (PTMs) regulate PKM2 in a way that benefits cancer growth, e.g., formation of PKM2 dimers. This review discusses the recent advancements in our understanding of various PTMs and the benefits they impart to the sustenance of cancer. Understanding the PTMs in PKM2 is crucial to assess their therapeutic potential and to design novel anticancer strategies.
Collapse
Affiliation(s)
- Gopinath Prakasam
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mohammad Askandar Iqbal
- Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, India
| | | | - Sybille Mazurek
- Institute of Veterinary Physiology and Biochemistry, University of Giessen, Giessen, Germany
| |
Collapse
|
17
|
Regulation of Tumor Progression by Programmed Necrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3537471. [PMID: 29636841 PMCID: PMC5831895 DOI: 10.1155/2018/3537471] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022]
Abstract
Rapidly growing malignant tumors frequently encounter hypoxia and nutrient (e.g., glucose) deprivation, which occurs because of insufficient blood supply. This results in necrotic cell death in the core region of solid tumors. Necrotic cells release their cellular cytoplasmic contents into the extracellular space, such as high mobility group box 1 (HMGB1), which is a nonhistone nuclear protein, but acts as a proinflammatory and tumor-promoting cytokine when released by necrotic cells. These released molecules recruit immune and inflammatory cells, which exert tumor-promoting activity by inducing angiogenesis, proliferation, and invasion. Development of a necrotic core in cancer patients is also associated with poor prognosis. Conventionally, necrosis has been thought of as an unregulated process, unlike programmed cell death processes like apoptosis and autophagy. Recently, necrosis has been recognized as a programmed cell death, encompassing processes such as oncosis, necroptosis, and others. Metabolic stress-induced necrosis and its regulatory mechanisms have been poorly investigated until recently. Snail and Dlx-2, EMT-inducing transcription factors, are responsible for metabolic stress-induced necrosis in tumors. Snail and Dlx-2 contribute to tumor progression by promoting necrosis and inducing EMT and oncogenic metabolism. Oncogenic metabolism has been shown to play a role(s) in initiating necrosis. Here, we discuss the molecular mechanisms underlying metabolic stress-induced programmed necrosis that promote tumor progression and aggressiveness.
Collapse
|
18
|
Poma AM, Giannini R, Piaggi P, Ugolini C, Materazzi G, Miccoli P, Vitti P, Basolo F. A six-gene panel to label follicular adenoma, low- and high-risk follicular thyroid carcinoma. Endocr Connect 2018; 7:124-132. [PMID: 29298844 PMCID: PMC5754511 DOI: 10.1530/ec-17-0261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022]
Abstract
The distinction between follicular thyroid carcinomas (FTCs) and follicular-patterned benign lesions is almost impossible on fine-needle aspiration cytology. Furthermore, minimally invasive FTCs (MI-FTCs) with less than 4 vascular invasion foci generally have an excellent prognosis, but there are exceptions and, so far, no molecular marker appears able to identify them reliably. We aimed to distinguish benign lesions from low- and high-risk FTCs by a small-scale combination of genes. The expression analysis of 75 selected genes was performed on 18 follicular adenomas (FAs), 14 MI-FTCs and 6 widely invasive FTC (WI-FTCs). The mutational status of the RAS genes, TERT promoter and PAX8-PPARG rearrangements was also investigated. Seven samples were mutated, namely 3 MI-FTCs and 4 WI-FTCs. Twenty-five genes were differentially expressed (FDR <0.05) between FAs and WI-FTCs. Six of these (ECM1, RXRG, SDPR, SLC26A4, TIFF3, TIMP1) were also differently expressed among MI-FTCs and FAs or WI-FTCs and were considered to build a classification model, which was tested to classify samples according to their histological class. Hence, 31 out of 38 were correctly classified, and accuracy remained high after cross-validation (27/38). The 2 MI-FTCs incorrectly classified as WI-FTCs harbored both RAS and TERT promoter mutations. The capability of these six genes to stratify benign, low- and high-risk lesions appears to be promising in supporting the diagnosis of indeterminate thyroid nodules.
Collapse
Affiliation(s)
- Anello Marcello Poma
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Riccardo Giannini
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo Piaggi
- National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of Health, Phoenix, Arizona, USA
| | - Clara Ugolini
- Department of Laboratory MedicineSection of Pathology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Gabriele Materazzi
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo Miccoli
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo Vitti
- Department of Clinical and Experimental MedicineUniversity of Pisa, Pisa, Italy
| | - Fulvio Basolo
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
He Y, Gao M, Cao Y, Tang H, Liu S, Tao Y. Nuclear localization of metabolic enzymes in immunity and metastasis. Biochim Biophys Acta Rev Cancer 2017; 1868:359-371. [PMID: 28757126 DOI: 10.1016/j.bbcan.2017.07.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/19/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
Metabolism is essential to all living organisms that provide cells with energy, regulators, building blocks, enzyme cofactors and signaling molecules, and is in tune with nutritional conditions and the function of cells to make the appropriate developmental decisions or maintain homeostasis. As a fundamental biological process, metabolism state affects the production of multiple metabolites and the activation of various enzymes that participate in regulating gene expression, cell apoptosis, cancer progression and immunoreactions. Previous studies generally focus on the function played by the metabolic enzymes in the cytoplasm and mitochondrion. In this review, we conclude the role of them in the nucleus and their implications for cancer progression, immunity and metastasis.
Collapse
Affiliation(s)
- Yuchen He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Menghui Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiqu Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Liu
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China; Cancer Research Institute, School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, Hunan 410078, China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
20
|
Pravenec M, Saba LM, Zídek V, Landa V, Mlejnek P, Šilhavý J, Šimáková M, Strnad H, Trnovská J, Škop V, Hüttl M, Marková I, Oliyarnyk O, Malínská H, Kazdová L, Smith H, Tabakoff B. Systems genetic analysis of brown adipose tissue function. Physiol Genomics 2017; 50:52-66. [PMID: 29127223 DOI: 10.1152/physiolgenomics.00091.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Brown adipose tissue (BAT) has been suggested to play an important role in lipid and glucose metabolism in rodents and possibly also in humans. In the current study, we used genetic and correlation analyses in the BXH/HXB recombinant inbred (RI) strains, derived from Brown Norway (BN) and spontaneously hypertensive rats (SHR), to identify genetic determinants of BAT function. Linkage analyses revealed a quantitative trait locus (QTL) associated with interscapular BAT mass on chromosome 4 and two closely linked QTLs associated with glucose oxidation and glucose incorporation into BAT lipids on chromosome 2. Using weighted gene coexpression network analysis (WGCNA) we identified 1,147 gene coexpression modules in the BAT from BXH/HXB rats and mapped their module eigengene QTLs. Through an unsupervised analysis, we identified modules related to BAT relative mass and function. The Coral4.1 coexpression module is associated with BAT relative mass (includes Cd36 highly connected gene), and the Darkseagreen coexpression module is associated with glucose incorporation into BAT lipids (includes Hiat1, Fmo5, and Sort1 highly connected transcripts). Because multiple statistical criteria were used to identify candidate modules, significance thresholds for individual tests were not adjusted for multiple comparisons across modules. In summary, a systems genetic analysis using genomic and quantitative transcriptomic and physiological information has produced confirmation of several known genetic factors and significant insight into novel genetic components functioning in BAT and possibly contributing to traits characteristic of the metabolic syndrome.
Collapse
Affiliation(s)
- Michal Pravenec
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Laura M Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Václav Zídek
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Vladimír Landa
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Petr Mlejnek
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Jan Šilhavý
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Miroslava Šimáková
- Institute of Physiology of the Czech Academy of Sciences , Prague , Czech Republic
| | - Hynek Strnad
- Institute of Molecular Genetics of the Czech Academy of Sciences , Prague , Czech Republic
| | - Jaroslava Trnovská
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vojtěch Škop
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Martina Hüttl
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Irena Marková
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Olena Oliyarnyk
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Hana Malínská
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Ludmila Kazdová
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Harry Smith
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado.,Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| |
Collapse
|
21
|
Gan L, Meng J, Xu M, Liu M, Qi Y, Tan C, Wang Y, Zhang P, Weng W, Sheng W, Huang M, Wang Z. Extracellular matrix protein 1 promotes cell metastasis and glucose metabolism by inducing integrin β4/FAK/SOX2/HIF-1α signaling pathway in gastric cancer. Oncogene 2017; 37:744-755. [PMID: 29059156 DOI: 10.1038/onc.2017.363] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/03/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022]
Abstract
Extracellular matrix protein 1 (ECM1) is related to strong invasiveness and poor prognosis in major malignancies, but the underlying mechanism remains unknown. Here we aimed to elucidate the function of ECM1 on cell metastasis and glucose metabolism in gastric cancer (GC). The level of ECM1 in sera and tissues of patient with GC were positively correlated with tumor invasion and recurrence. Genetic manipulation of ECM1 expression affected cell metastasis and glucose metabolism in GC cell lines. Enhanced ECM1 expression facilitated gene expression levels associated with epithelial-mesenchymal transition (EMT) and glucose metabolism. Interestingly, our results indicated that ECM1 directly interacted with integrin β4 (ITGB4) and activated ITGB4/focal adhesion kinase (FAK)/glycogen synthase kinase 3β signaling pathway, which further induced the expression of transcription factor SOX2. Aberrant expression of SOX2 altered gene expression of EMT factors and glucose metabolism enzymes. Furthermore, SOX2 enhanced hypoxia-inducible factor α (HIF-1α) promoter activity to regulate glucose metabolism. The micro-positron emission tomography/computed tomography imaging of xenograft model showed that ECM1 substantially increased 18F-fluorodeoxyglucose uptake in xenograft tumors. Using in vivo mouse tail vein injection experiments, ECM1 was also found to increase in lung surface metastasis. These findings provide evidence that ECM1 regulates GC cell metastasis and glucose metabolism by inducing ITGB4/FAK/SOX2/HIF-1α signal pathway and have important implications for the development of therapeutic target to prevent tumor metastasis and recurrence.
Collapse
Affiliation(s)
- L Gan
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - J Meng
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - M Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - M Liu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Y Qi
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - C Tan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Y Wang
- Nanchang Medical College, Nanchang University, Nanchang, China
| | - P Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - W Weng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - W Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - M Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Z Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Pkm2 can enhance pluripotency in ESCs and promote somatic cell reprogramming to iPSCs. Oncotarget 2017; 8:84276-84284. [PMID: 29137422 PMCID: PMC5663594 DOI: 10.18632/oncotarget.20685] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/25/2017] [Indexed: 01/17/2023] Open
Abstract
Aerobic glycolysis is one of the most important common characteristics in both cancer cells and stem cells. Metabolism switch has been discovered as an important early event in the process of reprogramming somatic cells to induced pluripotent stem cells (iPSCs). As a rate limiting kinase in glycolysis, Pkm2 has been reported playing critical roles in many tumors, yet its role in stem cells and iPSCs induction is poorly defined. In the present study, we showed that Pkm2 is a predominant pyruvate kinase in embryonic stem cells (ESCs), and its expression increases many pluripotent genes. During somatic cell reprogramming, up-regulation of Pkm2 can be observed and over-expression of Pkm2 can facilitate iPSCs induction, while Pkm1 or a mutant form of Pkm2 (Pkm2K422R) showed no enhancement role in iPSCs induction. Therefore, our data demonstrated that Pkm2 enhances the pluripotency maintenance in ESCs and promotes the pluripotency acquisition during somatic cell reprogramming.
Collapse
|
23
|
Li Z, Li H, Lu Y, Yang P, Li Z. Berberine Inhibited the Proliferation of Cancer Cells by Suppressing the Activity of Tumor Pyruvate Kinase M2. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701200909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Berberine, an isoquinoline alkaloid extracted from coptis, exerts anti-proliferation and anticancer properties. Pyruvate kinase M2 (PKM2) is a key enzyme of aerobic glycolysis and considered as the potential anticancer target. However, the inhibition effects and interaction action between Berberine and PKM2 is not well known. In this study, berberine showed antitumor activity of HCT-116 and HeLa cells with the suppression of cell proliferation. Moreover, berberine inhibited the enzyme activity of PKM2 in cancer cells, but had no impact on PKM2 expression. Further research showed that the interaction between berberine and PKM2 was dynamic fluorescence quenching and the main intermolecular force was hydrogen bonding. These findings revealed that berberine may serve as a therapeutic drug for cancer chemotherapy.
Collapse
Affiliation(s)
- Zhichao Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Hanqing Li
- College of Life Science, Shanxi University, Taiyuan 030006, China
| | - Yangxu Lu
- College of Life Science, Shanxi University, Taiyuan 030006, China
| | - Peng Yang
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
- College of Life Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
24
|
Creaney J, Dick IM, Leon JS, Robinson BWS. A Proteomic Analysis of the Malignant Mesothelioma Secretome Using iTRAQ. Cancer Genomics Proteomics 2017; 14:103-117. [PMID: 28387650 DOI: 10.21873/cgp.20023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 12/30/2022] Open
Abstract
Backgound/Aim: Malignant mesothelioma (MM) is an aggressive and fatal pleural cancer. The cell secretome offers information allowing insight into the pathogenesis of MM while offering the possibility to identify potential therapeutic targets and biomarkers. In the present study the secretome protein profile of MM cell lines was compared to normal mesothelial cells. MATERIALS AND METHODS Six MM cell lines were compared against three primary mesothelial cell culture preparations using iTRAQ® mass spectrometry. RESULTS MM cell lines more abundantly secreted exosome-associated proteins than mesothelial cells. MM cell secretomes were enriched in proteins that are involved in response to stress, carbon metabolism, biosynthesis of amino acids, antigen processing and presentation and protein processing in the endoplasmic reticulum. CONCLUSION The MM cell secretome is enriched in proteins that are likely to enhance its growth and response to stress and help it inhibit an adaptive immune response. These are potential targets for therapeutic and biomarker discovery.
Collapse
Affiliation(s)
- Jenette Creaney
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia and Australian Mesothelioma Tissue Bank, Sir Charles Gairdner Hospital, Perth, Western Australia
| | - Ian M Dick
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia
| | - Justine S Leon
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia
| | - Bruce W S Robinson
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, University of Western Australia, Sir Charles Gairdner Hospital, Perth, Western Australia
| |
Collapse
|
25
|
Wei X, Li M, Ma M, Jia H, Zhang Y, Kang W, Wang T, Shi X. Induction of apoptosis by FFJ-5, a novel naphthoquinone compound, occurs via downregulation of PKM2 in A549 and HepG2 cells. Oncol Lett 2016; 13:791-799. [PMID: 28356960 PMCID: PMC5351257 DOI: 10.3892/ol.2016.5522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/10/2016] [Indexed: 12/12/2022] Open
Abstract
Pyruvate kinase isoenzyme M2 (PKM2) has previously been identified as a tumor biomarker and as a potential target for cancer therapy. In this study, F§FJ-5, a characterized naphthoquinone modifier of mollugin, was synthesized in order to investigate its anticancer activity and the potential mechanisms. It was observed that FFJ-5 inhibited the cell growth of human lung adenocarcinoma cells A549 and human hepatoma cells HepG2 by MTT assays. FFJ-5 arrested cell cycle at the G2/M phase. Further analyses demonstrated that FFJ-5 attenuated the expression of PKM2 and reduced the production of adenosine triphosphate (ATP). Reduced expression and activity of epidermal growth factor receptor (EGFR) and Akt were observed in A549 and HepG2 cells exposed to FFJ-5. FFJ-5 exposure also resulted in cell apoptosis, in association with decreased intracellular pH level and mitochondrial membrane potential. In addition, FFJ-5 activated the caspase-3 cascade. In conclusion, FFJ-5 inhibited cancer cell growth via the blocking the EGFR-Akt-PKM2 pathway or through the synergistic action of EGFR, Akt and PKM2 proteins, alongside a decrease in ATP production. In addition, FFJ-5 induced cancer cell apoptosis by decreasing the intracellular pH level and the mitochondrial apoptosis pathway. The present results suggest a potential role of FFJ-5 on the therapy of human cancer.
Collapse
Affiliation(s)
- Xiaoli Wei
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Ming Li
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Mingming Ma
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Huina Jia
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Yu Zhang
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Wenyi Kang
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Tianxiao Wang
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Xiaoyan Shi
- Institute of Traditional Chinese Medicine, College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| |
Collapse
|
26
|
Li Y, Li Y, Zhao J, Zheng X, Mao Q, Xia H. Development of a Sensitive Luciferase-Based Sandwich ELISA System for the Detection of Human Extracellular Matrix 1 Protein. Monoclon Antib Immunodiagn Immunother 2016; 35:273-279. [PMID: 27923104 DOI: 10.1089/mab.2016.0033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Enzyme-linked immunosorbent assay (ELISA) has been one of the main methods for detecting an antigen in an aqueous sample for more than four decades. Nowadays, one of the biggest concerns for ELISA is still how to improve the sensitivity of the assay, and the luciferase-luciferin reaction system has been noticed as a new detection method with high sensitivity. In this study, a luciferin-luciferase reaction system was used as the detection method for a sandwich ELISA system. It was shown that this new system led to an increase in the detection sensitivity of at least two times when compared with the traditional horseradish peroxidase (HRP) detection method. Lastly, the serum levels of the human extracellular matrix 1 protein of breast cancer patients were determined by the new system, which were overall similar to the HRP chemiluminescent system. Furthermore, this new luciferase reporter can be implemented into other ELISA systems for the purpose of increasing the assay sensitivity.
Collapse
Affiliation(s)
- Ya Li
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Yanqing Li
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Junli Zhao
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Xiaojing Zheng
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Qinwen Mao
- 2 Department of Pathology, Northwestern University Feinberg School of Medicine Chicago , Chicago, Illinois
| | - Haibin Xia
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| |
Collapse
|
27
|
Stålberg P, Westin G, Thirlwell C. Genetics and epigenetics in small intestinal neuroendocrine tumours. J Intern Med 2016; 280:584-594. [PMID: 27306880 DOI: 10.1111/joim.12526] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroendocrine tumour of the small intestine (SI-NET), formerly known as midgut carcinoid tumour, is the most common small intestinal malignancy. The incidence is rising, with recent reports of 0.67 per 100 000 in the USA and 1.12 per 100 000 in Sweden. SI-NETs often present a challenge in terms of diagnosis and treatment, as patients often have widespread disease and are beyond cure by surgery. Somatostatin analogues provide the mainstay of medical treatment to control hormonal excess and increase the time to progression. Despite overall favourable prognosis (5-year overall survival of 65%), there is a need to find markers to identify both patients with worse outcome and new targets for therapy. Loss on chromosome 18 has been reported in 60-90% of SI-NETs, but mutated genes on this chromosome have failed detection. Recently, a putative tumour suppressor role has been suggested for TCEB3C occurring at 18q21 (encoding elongin A3), which may undergo epigenetic repression. CDKN1B has recently been revealed as the only recurrently mutated gene in SI-NETs but, with a frequency as low as 8%, its role as a driver in SI-NET development may be questioned. Integrated genomewide analysis including exome and whole-genome sequencing, gene expression, DNA methylation and copy number analysis has identified three novel molecular subtypes of SI-NET with differing clinical outcome. DNA methylation analysis has demonstrated that SI-NETs have significant epigenetic dysregulation in 70-80% of tumours. In this review, we focus on understanding of the genetic, epigenetic and molecular events that lead to development and progression of SI-NETs.
Collapse
Affiliation(s)
- P Stålberg
- Department of Surgical Sciences, Uppsala University and University Hospital, Uppsala, Sweden
| | - G Westin
- Department of Surgical Sciences, Uppsala University and University Hospital, Uppsala, Sweden
| | - C Thirlwell
- Cancer Institute, University College London, London, UK
| |
Collapse
|
28
|
Slaninova V, Krafcikova M, Perez-Gomez R, Steffal P, Trantirek L, Bray SJ, Krejci A. Notch stimulates growth by direct regulation of genes involved in the control of glycolysis and the tricarboxylic acid cycle. Open Biol 2016; 6:150155. [PMID: 26887408 PMCID: PMC4772804 DOI: 10.1098/rsob.150155] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Glycolytic shift is a characteristic feature of rapidly proliferating cells, such as cells during development and during immune response or cancer cells, as well as of stem cells. It results in increased glycolysis uncoupled from mitochondrial respiration, also known as the Warburg effect. Notch signalling is active in contexts where cells undergo glycolytic shift. We decided to test whether metabolic genes are direct transcriptional targets of Notch signalling and whether upregulation of metabolic genes can help Notch to induce tissue growth under physiological conditions and in conditions of Notch-induced hyperplasia. We show that genes mediating cellular metabolic changes towards the Warburg effect are direct transcriptional targets of Notch signalling. They include genes encoding proteins involved in glucose uptake, glycolysis, lactate to pyruvate conversion and repression of the tricarboxylic acid cycle. The direct transcriptional upregulation of metabolic genes is PI3K/Akt independent and occurs not only in cells with overactivated Notch but also in cells with endogenous levels of Notch signalling and in vivo. Even a short pulse of Notch activity is able to elicit long-lasting metabolic changes resembling the Warburg effect. Loss of Notch signalling in Drosophila wing discs as well as in human microvascular cells leads to downregulation of glycolytic genes. Notch-driven tissue overgrowth can be rescued by downregulation of genes for glucose metabolism. Notch activity is able to support growth of wing during nutrient-deprivation conditions, independent of the growth of the rest of the body. Notch is active in situations that involve metabolic reprogramming, and the direct regulation of metabolic genes may be a common mechanism that helps Notch to exert its effects in target tissues.
Collapse
Affiliation(s)
- Vera Slaninova
- Faculty of Science, University of South Bohemia, Branisovska 31, 37005 Ceske Budejovice, Czech Republic Institute of Entomology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| | - Michaela Krafcikova
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Raquel Perez-Gomez
- Faculty of Science, University of South Bohemia, Branisovska 31, 37005 Ceske Budejovice, Czech Republic
| | - Pavel Steffal
- Faculty of Science, University of South Bohemia, Branisovska 31, 37005 Ceske Budejovice, Czech Republic
| | - Lukas Trantirek
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Sarah J Bray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Alena Krejci
- Faculty of Science, University of South Bohemia, Branisovska 31, 37005 Ceske Budejovice, Czech Republic Institute of Entomology, Biology Centre, Czech Academy of Sciences, 37005 Ceske Budejovice, Czech Republic
| |
Collapse
|
29
|
The impact of RNA binding motif protein 4-regulated splicing cascade on the progression and metabolism of colorectal cancer cells. Oncotarget 2016; 6:38046-60. [PMID: 26506517 PMCID: PMC4741983 DOI: 10.18632/oncotarget.5710] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/06/2015] [Indexed: 01/17/2023] Open
Abstract
Dysregulated splicing of pre-messenger (m)RNA is considered a molecular occasion of carcinogenesis. However, the underlying mechanism is complex and remains to be investigated. Herein, we report that the upregulated miR-92a reduced the RNA-binding motif 4 (RBM4) protein expression, leading to the imbalanced expression of the neuronal polypyrimidine tract-binding (nPTB) protein through alternative splicing-coupled nonsense mediated decay (NMD) mechanism. Increase in nPTB protein enhances the relative level of fibroblast growth factor receptor 2 IIIc (FGFR2) and pyruvate kinase M2 (PKM2) transcripts which contribute to the progression and metabolic signature of CRC cells. Expression profiles of RBM4 and downstream alternative splicing events are consistently observed in cancerous tissues compared to adjacent normal tissues. These results constitute a mechanistic understanding of RBM4 on repressing the carcinogenesis of colorectal cells.
Collapse
|
30
|
Littlejohn NK, Keen HL, Weidemann BJ, Claflin KE, Tobin KV, Markan KR, Park S, Naber MC, Gourronc FA, Pearson NA, Liu X, Morgan DA, Klingelhutz AJ, Potthoff MJ, Rahmouni K, Sigmund CD, Grobe JL. Suppression of Resting Metabolism by the Angiotensin AT2 Receptor. Cell Rep 2016; 16:1548-1560. [PMID: 27477281 DOI: 10.1016/j.celrep.2016.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/09/2016] [Accepted: 07/01/2016] [Indexed: 11/15/2022] Open
Abstract
Activation of the brain renin-angiotensin system (RAS) stimulates energy expenditure through increasing of the resting metabolic rate (RMR), and this effect requires simultaneous suppression of the circulating and/or adipose RAS. To identify the mechanism by which the peripheral RAS opposes RMR control by the brain RAS, we examined mice with transgenic activation of the brain RAS (sRA mice). sRA mice exhibit increased RMR through increased energy flux in the inguinal adipose tissue, and this effect is attenuated by angiotensin II type 2 receptor (AT2) activation. AT2 activation in inguinal adipocytes opposes norepinephrine-induced uncoupling protein-1 (UCP1) production and aspects of cellular respiration, but not lipolysis. AT2 activation also opposes inguinal adipocyte function and differentiation responses to epidermal growth factor (EGF). These results highlight a major, multifaceted role for AT2 within inguinal adipocytes in the control of RMR. The AT2 receptor may therefore contribute to body fat distribution and adipose depot-specific effects upon cardio-metabolic health.
Collapse
Affiliation(s)
| | - Henry L Keen
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Kristin E Claflin
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin V Tobin
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Kathleen R Markan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Sungmi Park
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Meghan C Naber
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Nicole A Pearson
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Xuebo Liu
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA
| | - Curt D Sigmund
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA.
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
31
|
Wu J, Hu L, Chen M, Cao W, Chen H, He T. Pyruvate kinase M2 overexpression and poor prognosis in solid tumors of digestive system: evidence from 16 cohort studies. Onco Targets Ther 2016; 9:4277-88. [PMID: 27478385 PMCID: PMC4951066 DOI: 10.2147/ott.s106508] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose The expression of pyruvate kinase M2 (PKM2) has been linked to tumor formation and invasion. Specifically, the relationship between high PKM2 expression and prognosis has been evaluated in solid tumors of digestive system. However, the prognostic value of PKM2 remains controversial. Methods A literature search of PubMed, Embase, and Cochrane databases was conducted until October 2015. The end point focused on overall survival (OS). The pooled hazard ratio (HR) or odds ratio and the 95% confidence intervals were calculated to correlate PKM2 overexpression with OS and clinicopathological characteristics by employing fixed- or random-effects models, depending on the heterogeneity of the included studies. Results We identified 18 cohorts in 16 studies involving 2,812 patients for this meta-analysis. Overall, the combined HR for OS in all tumor types was 1.74 (1.44–2.11; P<0.001). When stratified by tumor type, the influence of PKM2 expression on poor prognosis was also found in gastric cancer (HR =1.54 [1.08–2.21], P=0.018), esophageal squamous cell carcinoma (HR =1.71 [1.38–2.12], P<0.001), hepatocellular cancer (HR =1.92 [1.52–2.42], P<0.001), biliary cancer (HR =2.11 [1.50–2.95], P<0.001), and oral cancer (HR =3.49 [1.97–6.18], P<0.001), but not in pancreatic ductal adenocarcinoma (HR =1.03 [0.28–3.76], P=0.968). Furthermore, PKM2 overexpression had a negative effect on the late clinical stage of all tumor types except for pancreatic ductal adenocarcinoma. The high density of PKM2 overexpression was significantly associated with some clinical characteristics in different cancer types, such as tumor stage, modal metastasis, and tumor size. Conclusion Our findings revealed significant association of PKM2 overexpression with OS and certain clinicopathological features in solid tumors of digestive system, thereby suggesting that PKM2 might be an indicator of poor prognosis in digestive system cancers.
Collapse
Affiliation(s)
- Jiayuan Wu
- Nutritional Department, the Affiliated Hospital of Guangdong Medical University
| | - Liren Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Guangdong Medical University
| | - Manyu Chen
- Department of Oncology, the Affiliated Hospital of Guangdong Medical University
| | - Wenjun Cao
- School of Public Health, Guangdong Medical University
| | - Haicong Chen
- Department of Orthopedics, the Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, People's Republic of China
| | - Taiping He
- School of Public Health, Guangdong Medical University
| |
Collapse
|
32
|
Ye H, Yu X, Xia J, Tang X, Tang L, Chen F. MiR-486-3p targeting ECM1 represses cell proliferation and metastasis in cervical cancer. Biomed Pharmacother 2016; 80:109-114. [PMID: 27133046 DOI: 10.1016/j.biopha.2016.02.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/25/2016] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) regulate gene expression and are involved in cervical cancer. But the molecular mechanism is still unclear. Here, miRNA profile of cervical cancer was performed and demonstrated that miR-486-3p decreased in specimens of cervical cancer patients. In addition, our clinical data show that decreased miR-486-3p was associated with metastasis in cervical cancer patients. ECM1 was predicted and velified as a target gene of miR-486-3p. Overexpression of miR-486-3p inhibited cell growth and metastasis by targeting ECM1. In a conclusion, these findings suggest that miR-486-3p is a tumor suppressor miRNA and induction of miR-486-3p is a potential strategy to inhibit cervical cancer progression.
Collapse
Affiliation(s)
- Haiqiong Ye
- Department of Obstetrics and Gynecology, The Affiliated Hospital of SouthWest Medical University, Sichuan, China
| | - Xiaolan Yu
- Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of SouthWest Medical University, Sichuan, China
| | - Jiyi Xia
- The Institute of Cancer Research, SouthWest Medical University, Sichuan, China
| | - Xiaoping Tang
- Experimental Medicine Center, The Affiliated Hospital of SouthWest Medical University, Sichuan, China
| | - Li Tang
- Experimental Medicine Center, The Affiliated Hospital of SouthWest Medical University, Sichuan, China
| | - Feng Chen
- Experimental Medicine Center, The Affiliated Hospital of SouthWest Medical University, Sichuan, China.
| |
Collapse
|
33
|
Production and characterization of domain-specific monoclonal antibodies against human ECM1. Protein Expr Purif 2016; 121:103-11. [PMID: 26826312 DOI: 10.1016/j.pep.2016.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/13/2022]
Abstract
Human extracellular matrix protein-1 (hECM1), a secreted glycoprotein, is widely expressed in different tissues and organs. ECM1 has been implicated in multiple biological functions, which are potentially mediated by the interaction of different ECM1 domains with its ligands. However, the exact biological functions of ECM1 have not been elucidated yet, and the functional study of ECM1 has been partially hampered by the lack of sensitive and specific antibodies, especially those targeting different ECM1 domains. In this study, six strains of monoclonal antibody (MAb) against hECM1 were generated using purified, prokaryotically-expressed hECM1 as an immunogen. The MAbs were shown to be highly sensitive and specific, and suitable for western blot, immunoprecipitation assays and immunohistochemistry. Furthermore, the particular ECM1 domains recognized by different MAbs were identified. Lastly, the MAbs were found to have neutralizing activities, inhibiting the proliferation, migration and metastasis of MDA-MB-231 cells. In conclusion, the domain-specific anti-ECM1 MAbs produced in this study should provide a useful tool for investigating ECM1's biological functions, and cellular pathways in which it is involved.
Collapse
|
34
|
Secreted pyruvate kinase M2 facilitates cell migration via PI3K/Akt and Wnt/β-catenin pathway in colon cancer cells. Biochem Biophys Res Commun 2015; 459:327-332. [PMID: 25732087 DOI: 10.1016/j.bbrc.2015.02.112] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/17/2015] [Indexed: 02/02/2023]
Abstract
Pyruvate Kinase M2 (PKM2) is a key glycolytic enzyme, which highly expressed in tumor cells, and plays a pivotal role in the growth, survival and metabolism reprogramming of cancer cells. Besides the location of cytoplasm as a glycolytic enzyme and the location of nucleus as a protein kinase, extracellular PKM2 is present in serum and feces of tumor patients. However, little is known about the secretion of PKM2 and its significance in the progression of colon cancer. Here we demonstrated that PKM2 could be secreted from colon cancer cells, and purified PKM2 protein mimicing the secreted PKM2 was able to promote colon cancer cell migration. Moreover, PI3K/Akt and Wnt/β-catenin signaling were involved in secreted PKM2 induced colon cancer cell migration. The results reveal critical roles of secreted PKM2 in the progression of colon cancer, and indicate that PKM2 may be a therapeutic target for colon cancer.
Collapse
|