1
|
Xie Z, Zhou Z, Chen S, Li Y, He X, Chen G. GLUT1 sensitizes tumor cells to EGFR-TKIs by binding with activated EGFR and regulating its downstream signaling pathways. Cell Commun Signal 2025; 23:247. [PMID: 40437580 PMCID: PMC12121033 DOI: 10.1186/s12964-025-02259-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/20/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND We have previously demonstrated that GLUT1 can interact with phosphorylated EGFR and has an oncogenic role in lung cancer. Here, we aim to investigate their binding region and its signaling pathways. METHODS The AlphaFold 3 prediction, Co-immunoprecipitation, and Western blot were used to uncover the interaction conditions of GLUT1 and EGFR. The RNA-seq data was analyzed to evaluate the difference in signaling pathways between wild-type EGFR and activated mutated EGFR. The xenograft tumor model was established to determine the therapy effect of the combination of GLUT1 inhibitor BAY-876 and EGFR TKI Osimertinib. RESULTS We found that the interaction ability of GLUT1 and EGFR depended on the activation of EGFR. GLUT1 interacted with EGFRvIII (loss 2-7 exons) but not with EGFRvI (loss 1-16 exons), so GLUT1 interacts with EGFR in the EGFR extracellular transmembrane region. GLUT1 regulated EGFR downstream signaling pathways. GLUT1 inhibitor BAY-876 can sensitize tumor cells to EGFR TKI Osimertinib. CONCLUSIONS GLUT1 participates in tumor progression by interacting with phosphor-EGFR, suggesting that inhibition of the GLUT1-EGFR axis may be a potential therapeutic strategy for lung cancer treatment.
Collapse
Affiliation(s)
- Zhangrong Xie
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine,Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Zhiqing Zhou
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine,Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Sijie Chen
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine,Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Yu Li
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine,Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Xiaoniu He
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Guoan Chen
- Department of Human Cell Biology and Genetics, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, SUSTech Homeostatic Medicine Institute, School of Medicine,Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
2
|
Sui X, Zhou H, Wang T. GLUT1 maintains esophageal cancer stem cell-like characteristics by inhibiting autophagy-dependent ferroptosis via EGFR. Exp Cell Res 2025; 449:114600. [PMID: 40368081 DOI: 10.1016/j.yexcr.2025.114600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025]
Abstract
Esophageal cancer, a highly malignant tumor with poor prognosis, is characterized by the presence of cancer stem cells (CSCs) that drive tumor initiation, metastasis, and recurrence. This study investigates the molecular mechanism by which glucose transporter 1 (GLUT1) maintains esophageal CSC-like properties through regulation of autophagy-dependent ferroptosis via epidermal growth factor receptor (EGFR). Using shRNA to knock down GLUT1 or EGFR and constructing a GLUT1 overexpression vector in KYSE520 cells, we employed western blotting, qRT-PCR, flow cytometry, sphere formation, Transwell assays, and xenograft models to assess stemness markers (NANOG, OCT4, SOX2), autophagic flux (LC3B, P62, Beclin1), and ferroptosis-related parameters (ROS, Fe2+, GSH, GPX4, COX2). Mechanistic analyses included co-immunoprecipitation to validate the GLUT1-EGFR interaction, chloroquine to inhibit autophagy, and cycloheximide/MG132 to evaluate EGFR protein stability. Results showed that GLUT1 depletion reduced CSC marker expression, increased ROS and Fe2+ levels, depleted GSH, and induced lipid peroxidation, sensitizing cells to ferroptosis while activating autophagy (elevated LC3 II/I, Beclin1; reduced P62); autophagy inhibition exacerbated cell death, indicating a protective role for autophagy in this context. GLUT1 directly bound to EGFR, stabilizing the receptor by blocking ubiquitin-proteasome-mediated degradation, whereas EGFR knockdown enhanced autophagic flux and reversed GLUT1-overexpression-induced ferroptosis resistance and stemness maintenance. In vivo, GLUT1 knockdown suppressed tumor growth and lung metastasis, and clinical samples revealed a positive correlation between GLUT1 and EGFR expression, linked to advanced TNM stages and poor survival. Collectively, these findings demonstrate that GLUT1 preserves esophageal CSC-like characteristics by stabilizing EGFR to inhibit autophagy-dependent ferroptosis, highlighting targeting GLUT1 as a potential therapeutic strategy to eliminate CSCs and combat esophageal cancer progression.
Collapse
Affiliation(s)
- Xin Sui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Haining Zhou
- Department of Digestive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Tingting Wang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| |
Collapse
|
3
|
Yang D, Yang C, Huang L, Guan M, Song C. Role of ubiquitination-driven metabolisms in oncogenesis and cancer therapy. Semin Cancer Biol 2025; 110:17-35. [PMID: 39929409 DOI: 10.1016/j.semcancer.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025]
Abstract
Ubiquitination represents one of the most critical post-translational modifications, comprising a multi-stage enzyme process that plays a pivotal role in a myriad of cellular biological activities. The deregulation of the processes of ubiquitination and deubiquitination is associated with the development of cancers and other diseases. This typescript reviews the impact of ubiquitination on metabolic processes, elucidating the regulatory functions of ubiquitination on pivotal enzymes within metabolic pathways in pathological contexts. It underscores the role of ubiquitination-driven metabolism disorders in the etiology of cancers, and oncogenesis, and highlights the potential therapeutic efficacy of targeting ubiquitination-driven enzymes in cancer metabolism, their combination with immune checkpoint inhibitors, and their clinical applications.
Collapse
Affiliation(s)
- Dongqin Yang
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China; Central Laboratory, Huashan Hospital, Fudan University, 12 Middle Urumuqi Road, Shanghai 200040, China
| | - Can Yang
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Linlin Huang
- Central Laboratory, Huashan Hospital, Fudan University, 12 Middle Urumuqi Road, Shanghai 200040, China
| | - Ming Guan
- Department of Laboratory Medicine of Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Chunhua Song
- Division of Hematology, The Ohio State University Wexner Medical Center, the James Cancer Hospital, Columbus, OH 43210, USA.
| |
Collapse
|
4
|
Hayashi M, Nakamura K, Harada S, Tanaka M, Kobayashi A, Saito H, Tsuji T, Yamamoto D, Moriyama H, Kinoshita J, Inaki N. GLUT1 inhibition by BAY-876 induces metabolic changes and cell death in human colorectal cancer cells. BMC Cancer 2025; 25:716. [PMID: 40247224 PMCID: PMC12004878 DOI: 10.1186/s12885-025-14141-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/11/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Glucose transporter 1 (GLUT1) is known to play a crucial role in glucose uptake in malignant tumors. GLUT1 inhibitors reportedly exhibit anti-tumor effects by suppressing cancer cell proliferation. BAY-876, a selective GLUT1 inhibitor, has been shown to inhibit tumor growth in ovarian and breast cancers. In this study, we investigated the anti-proliferative effects of BAY-876 treatment in human colorectal cancer (CRC) cell lines. METHODS We investigated the metabolic changes and effects on proliferation from BAY-876 treatment in HCT116, DLD1, COLO205, LoVo, and Caco-2 cells in vitro. Additionally, a mouse xenograft model was established using HCT116 cells to examine the tumor-inhibitory effects of BAY-876 treatment in vivo. RESULTS BAY-876 treatment inhibited cell proliferation in HCT116, DLD1, COLO205, and LoVo cells. Reduced GLUT1 protein expression levels were observed through western blot analysis. Flux analysis indicated enhanced mitochondrial respiration, accompanied by increased reactive oxygen species levels and apoptosis rates. Tumor-inhibitory effects were also observed in the xenograft model, with the BAY-876-treated groups showing GLUT1 suppression. CONCLUSIONS BAY-876 treatment induced metabolic changes and inhibited cell proliferation in human CRC cell lines. Using BAY-876 is a potential novel approach for treating CRC.
Collapse
Affiliation(s)
- Masato Hayashi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Keishi Nakamura
- Department of Surgery, Public Central Hospital of Matto Ishikawa, 3-8 Kuramitsu, Hakusan, Ishikawa, 924-8588, Japan.
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Mariko Tanaka
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Akiko Kobayashi
- Center for Biomedical Research and Education, School of Medicine, Kanazawa University, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroto Saito
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Toshikatsu Tsuji
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Daisuke Yamamoto
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hideki Moriyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| | - Noriyuki Inaki
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa, 920-8641, Japan
| |
Collapse
|
5
|
Karimpur-Zahmatkesh A, Khalaj-Kondori M. The perspective of targeting cancer cell metabolism: combination therapy approaches. Mol Biol Rep 2025; 52:375. [PMID: 40202553 DOI: 10.1007/s11033-025-10472-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Cancer cells are considered the most adaptable for their metabolic status, which supports growth, survival, rapid proliferation, invasiveness, and metastasis in a nutrient-deficient microenvironment. Since the discovery of altered glucose metabolism (aerobic glycolysis), which is generally known as a part of metabolic reprogramming and an innate trait of cancer cells, in 1930 via Dr. Otto Warburg, numerous studies have endeavored to recognize various aspects of cancer cell metabolism and find new methods for efficiently eradicating described cells by targeting their energy metabolism. In this way, the outcomes have mainly been promising. Accordingly, outlining the related results will indeed assist us in making a definitive path for developing targeted therapy strategies based on cancer cell-altered metabolism. The present study reviews the key features of cancer cell metabolism and treatment strategies based on them. It emphasizes the importance of targeting cancer cell dysregulated metabolic pathways that influence the cell energy supply and manage cancer cell growth and survival. This trial also introduces a multimodal therapeutic strategy hypothesis, a potential next-generation combination therapy approach, and suggests interdisciplinary research to recognize the complexities of cancer metabolism and exploit them for designing more efficacious cancer therapeutic strategies.
Collapse
Affiliation(s)
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
6
|
Zhang F, Pan Z, Wu J, Huang Y. Relationship between MRI features and HIF-1α, GLUT1 and Ki-67 expression in pituitary adenoma with cystic degeneration. BMC Med Imaging 2025; 25:76. [PMID: 40050749 PMCID: PMC11887064 DOI: 10.1186/s12880-025-01574-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Pituitary adenomas (PAs) are prevalent tumors that often exhibit ischemia, hypoxia, and cystic transformations, impacting their prognosis. The relationship between cystic degeneration in PAs and the expressions of hypoxia-inducible factor-1α (HIF-1α), glucose transporter 1 (GLUT1), and Ki-67 remains unclear. This study aims to analyze the correlation between MRI characteristics of cystic PA and the expression of these proteins. METHODS This is a retrospective analysis. A total of 74 patients with cystic PA and 30 PA patients without cystic degeneration were enrolled. Their MRI signs were analyzed. According to the T2WI signs of PA, they were divided into the fluid level group (n = 26), non-fluid level group (n = 48), and non-cyst group (n = 30). Immunohistochemistry was performed to evaluate the expression levels of HIF-lα, GLUT1, and Ki-67 protein. Univariate and multinomial logistic regression analyses were used to evaluate the factors affecting MRI signs of PA. Spearman correlation was also performed. RESULTS There was no significant difference in gender, age, and HIF-1α protein expression among the three groups. Significant differences were found in invasiveness (P = 0.008), GLUT1 (P < 0.001), and Ki-67 protein expression (P = 0.009) among the three groups. Pairwise comparisons revealed statistically significant differences in invasiveness, GLUT1, and Ki-67 protein expressions between the non-fluid level group and the non-cyst group. Furthermore, GLUT1 protein expression was significantly different between the non-fluid level group and the fluid level group. Notably, GLUT1 was identified as an independent factor for the non-fluid level cystic characteristics of PA. Additionally, GLUT1 was positively correlated with invasiveness and Ki-67. CONCLUSION The non-fluid level cystic PA has higher invasiveness and higher proliferation than fluid level cystic PA and non-cyst PA, which may be related to high glucose metabolism as indicated by GLUT1 expression.
Collapse
Affiliation(s)
- Fangfang Zhang
- Department of Endocrinology, Fuzhou First General Hospital Affiliated with Fujian Medical University, Fuzhou, China
| | - Zhenhong Pan
- Fujian University of Traditional Chinese Medicine Fuzhou General Hospital, Fuzhou, China
| | - Jianwu Wu
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, (The 900TH Hospital), Xi'erhuanbei Road, Fuzhou, 350025, Fujian, China.
- Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, China.
| | - Yinxing Huang
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, (The 900TH Hospital), Xi'erhuanbei Road, Fuzhou, 350025, Fujian, China.
- Fujian Provincial Clinical Medical Research Center for Minimally Invasive Diagnosis and Treatment of Neurovascular Diseases, Fuzhou, China.
| |
Collapse
|
7
|
Dong XM, Chen L, Xu YX, Wu P, Xie T, Liu ZQ. Exploring metabolic reprogramming in esophageal cancer: the role of key enzymes in glucose, amino acid, and nucleotide pathways and targeted therapies. Cancer Gene Ther 2025; 32:165-183. [PMID: 39794467 DOI: 10.1038/s41417-024-00858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 01/13/2025]
Abstract
Esophageal cancer (EC) is one of the most common malignancies worldwide with the character of poor prognosis and high mortality. Despite significant advancements have been achieved in elucidating the molecular mechanisms of EC, for example, in the discovery of new biomarkers and metabolic pathways, effective treatment options for patients with advanced EC are still limited. Metabolic heterogeneity in EC is a critical factor contributing to poor clinical outcomes. This heterogeneity arises from the complex interplay between the tumor microenvironment and genetic factors of tumor cells, which drives significant metabolic alterations in EC, a process known as metabolic reprogramming. Understanding the mechanisms of metabolic reprogramming is essential for developing new antitumor therapies and improving treatment outcomes. Targeting the distinct metabolic alterations in EC could enable more precise and effective therapies. In this review, we explore the complex metabolic changes in glucose, amino acid, and nucleotide metabolism during the progression of EC, and how these changes drive unique nutritional demands in cancer cells. We also evaluate potential therapies targeting key metabolic enzymes and their clinical applicability. Our work will contribute to enhancing knowledge of metabolic reprogramming in EC and provide new insights and approaches for the clinical treatment of EC.
Collapse
Affiliation(s)
- Xue-Man Dong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Pu Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.
| | - Zhao-Qian Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
| |
Collapse
|
8
|
Kuo CY, Hsu YC, Chen MJ, Lin CH, Li YS, Cheng SP. Glucose Transporter 1 Inhibitors Induce Autophagy and Synergize With Lenvatinib in Thyroid Cancer Cells. Head Neck 2025; 47:615-624. [PMID: 39360406 DOI: 10.1002/hed.27953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Less differentiated thyroid cancer may upregulate the expression of glucose transporter 1 (GLUT1) and increase glycolytic activity. However, it is uncertain whether GLUT1 can be used as a target for therapy. METHODS Thyroid cancer cell lines were treated with two different GLUT1 inhibitors, STF-31 and BAY-876. Functional assays were conducted to evaluate the effects of these inhibitors on cell biology. RESULTS GLUT1 inhibitors dose-dependently decreased cell growth and clonogenicity of thyroid cancer cells. Cell cycle analysis showed that these inhibitors caused G2/M arrest instead of apoptosis. Additionally, treatment with GLUT1 inhibitors led to the activation of autophagy. In both the Transwell and spheroid models, GLUT1 inhibitors significantly suppressed cell invasiveness. Moreover, GLUT1 inhibitors demonstrated synergistic interactions when combined with lenvatinib. CONCLUSIONS Treatment with GLUT1 inhibitors activates autophagy and provokes cell cycle arrest, accompanied by a decrease in colony formation and invasive capacity in thyroid cancer cells.
Collapse
Affiliation(s)
- Chi-Yu Kuo
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
- Center for Astronautical Physics and Engineering, National Central University, Taoyuan City, Taiwan
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chi-Hsin Lin
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan City, Taiwan
| | - Ying-Syuan Li
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, School of Medicine, MacKay Medical College, New Taipei City, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
9
|
Zhang J, Song S, Li Y, Gong A. Predicting Outcomes in Esophageal Squamous Cell Carcinoma Using scRNA-Seq and Bulk RNA-Seq: A Model Development and Validation Study. Cancer Med 2025; 14:e70617. [PMID: 39840762 PMCID: PMC11751878 DOI: 10.1002/cam4.70617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Altered glucose metabolism is a critical characteristic from the beginning stage of esophageal squamous cell carcinoma (ESCC), and the phenomenon is presented as a pink-color sign under endoscopy after iodine staining. Therefore, calculating the metabolic score based on the glucose metabolic gene sets may bring some novel insights, enabling the prediction of prognosis and the identification of treatment choices for ESCC. METHODS A total of 8, 99, and 140 individuals from The Gene Expression Omnibus database, The Cancer Genome Atlas database, and the Memorial Sloan Kettering Cancer Center, respectively, were encompassed in the investigation. Patients diagnosed with ESCC after surgery were enrolled for further validation. RESULTS A total of 13 kinds of cell clusters were screened, and the squamous epithelium was identified with the highest score. And 558 differential genes were selected from the single-cell RNA sequencing (scRNA-seq) dataset. Four glucose metabolism-related genes, namely, SERP1, CTSC, RAP2B, and SSR4, were identified as hub genes to develop a risk prognostic model. The model was validated in another external cohort. According to the risk score (RS) determined by the model, the patients were categorized into low- and high-risk groups (LRG and HRG). Compared with LRG, HRG indicated poor survival and decreased drug sensitivity. Additionally, the immune microenvironment and pathway enrichment were different between the two groups. Immunohistochemical staining revealed that hub genes were expressed differently in ESCC tissues, high- and low-grade intraepithelial neoplasia, and adjacent normal tissues. CONCLUSION Four hub genes (SERP1, CTSC, RAP2B, and SSR4) screened based on glucose metabolism developed a predictive model in ESCC patients. The RS was established as an independent risk factor for predicting prognosis. These findings may enhance understanding of ESCC's molecular profile and serve as a new prognostic tool for better patient stratification and treatment planning in clinical practice.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Digestive EndoscopyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| | - Shunzhe Song
- Department of Digestive EndoscopyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| | - Yuqing Li
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| | - Aixia Gong
- Department of Digestive EndoscopyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
- Department of GastroenterologyThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningPeople's Republic of China
| |
Collapse
|
10
|
Sui X, Wang W, Zhang D, Xu J, Li J, Jia Y, Qin Y. Integrated analysis of ferroptosis and stemness based on single-cell and bulk RNA-sequencing data provide insights into the prognosis and treatment of esophageal carcinoma. Gene 2024; 927:148701. [PMID: 38885819 DOI: 10.1016/j.gene.2024.148701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Cancer stem cells (CSCs) play a significant role in the recurrence and drug resistance of esophageal carcinoma (ESCA). Ferroptosis is a promising anticancer therapeutic strategy that effectively targets CSCs exhibiting high tumorigenicity and treatment resistance. However, there is a lack of research on the combined role of ferroptosis-related genes (FRGs) and stemness signature in the prognosis of ESCA. METHODS The cellular compositions were characterized using single-cell RNA sequencing (scRNA-seq) data from 18 untreated ESCA samples. 50 ferroptosis-related stemness genes (FRSGs) were identified by integrating FRGs with stemness-related genes (SRGs), and then the cells were grouped by AUCell analysis. Next, functional enrichment, intercellular communication, and trajectory analyses were performed to characterize the different groups of cells. Subsequently, the stem-ferr-index was calculated using machine learning algorithms based on the expression profiles of the identified risk genes. Additionally, therapeutic drugs were predicted by analyzing the GDSC2 database. Finally, the expression and functional roles of the identified marker genes were validated through in vitro experiments. RESULTS The analysis of scRNA-seq data demonstrates the diversity and cellular heterogeneity of ESCA. Then, we identified 50 FRSGs and classified cells into high or low ferroptosis score stemness cells accordingly. Functional enrichment analysis conducted on the differentially up-regulated genes between these groups revealed predominant enrichment in pathways associated with intercellular communication and cell differentiation. Subsequently, we identified 9 risk genes and developed a prognostic signature, termed stem_ferr_index, based on these identified risk genes. We found that the stem-ferr-index was correlated with the clinical characteristics of patients, and patients with high stem-ferr-index had poor prognosis. Furthermore, we identified four drugs (Navitoclax, Foretinib, Axitinib, and Talazoparib) with potential efficacy targeting patients with a high stem_ferr_index. Additionally, we delineated two marker genes (STMN1 and SLC2A1). Particularly noteworthy, SLC2A1 exhibited elevated expression levels in ESCA tissues and cells. We provided evidence suggesting that SLC2A1 could influence the migration, invasion, and stemness of ESCA cells, and it was associated with sensitivity to Foretinib. CONCLUSION This study constructed a novel ferroptosis-related stemness signature, identified two marker genes for ESCA, and provided valuable insights for developing more effective therapeutic targets targeting ESCA CSCs in the future.
Collapse
Affiliation(s)
- Xin Sui
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Wenjia Wang
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Daidi Zhang
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Jiayao Xu
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Jing Li
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Yongxu Jia
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China
| | - Yanru Qin
- Department of Clinical Oncology, The First Affiliated Hospital Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
11
|
Xu Q, Jin Z, Yuan Z, Yu Z, Gao J, Zhao R, Li H, Ren H, Cao B, Wei B, Jiang L. YAP Promotes Chemoresistance to 5-FU in Colorectal Cancer Through mTOR/GLUT3 Axis. J Cancer 2024; 15:6784-6797. [PMID: 39668819 PMCID: PMC11632981 DOI: 10.7150/jca.100179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/28/2024] [Indexed: 12/14/2024] Open
Abstract
Background: Although chemoresistance constitutes a significant barrier to the effectiveness of chemotherapy in colorectal cancer (CRC), its precise mechanisms remain unclear. YAP functions as an oncogene in various malignancies. However, the relationship between YAP and chemoresistance in CRC needs clarification. Methods: The expression level of YAP in CRC tissues was assessed through immunohistochemistry (IHC), and the impact of YAP on CRC cell chemoresistance was evaluated using the Cell Counting Kit-8, EdU, and flow cytometry assays. Meanwhile, tumor proliferation was assessed in vivo by analyzing the expression of PCNA and Ki-67 in subcutaneous tumors via IHC. In addition, the TUNEL assay was employed to evaluate tumor apoptosis levels and western blot was utilized to detect the mTOR/GLUT3 pathway-related protein expression to provide insights into the underlying mechanism. Results: YAP was highly expressed in CRC tissues and correlated with patient prognosis and clinicopathological features. Bioinformatic analysis based on the TCGA database revealed that YAP was associated with DNA replication, glycolysis, and the mTOR pathway. Meanwhile, YAP could enhance chemoresistance and glycolysis in CRC cells both in vitro and in vivo. Additional mechanistic experiments unveiled that YAP promoted CRC cell chemoresistance via the mTOR/GLUT3 axis. Conclusion: This study validated the role of YAP as an oncogene in CRC, as it promoted chemoresistance through the mTOR/GLUT3 axis. These results suggested YAP as a potential target for promoting the efficacy of chemotherapy in patients with CRC.
Collapse
Affiliation(s)
- Qixuan Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zhesi Jin
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu,China
| | - Zhen Yuan
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiyuan Yu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Jingwang Gao
- Department of General Surgery, Linfen Central Hospital, Linfen, Shanxi, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Huiguang Ren
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Linhua Jiang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
12
|
Li D, Xiong Y, Li M, Long L, Zhang Y, Yan H, Xiang H. STC2 knockdown inhibits cell proliferation and glycolysis in hepatocellular carcinoma through promoting autophagy by PI3K/Akt/mTOR pathway. Arch Biochem Biophys 2024; 761:110149. [PMID: 39271096 DOI: 10.1016/j.abb.2024.110149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/02/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The pathogenesis exploration and timely intervention of hepatocellular carcinoma (HCC) are crucial due to its global impact on human health. As a general tumor biomarker, stanniocalcin 2 (STC2), its role in HCC remains unclear. We aimed to analyze the effect and mechanism of STC2 on HCC. METHODS STC2 expressions in HCC tissues and cell lines were measured. si-STC2 and oe-STC2 transfections were utilized to analyze how STC2 affected cell functions. Functional enrichment analysis of STC2 was performed by Gene Set Enrichment Analysis (GSEA). The regulatory mechanism of STC2 on HCC was investigated using 2-DG, 3-MA, IGF-1, Rap, and LY294002. The impact of STC2 on HCC progression in vivo was evaluated by the tumor formation experiment. RESULTS Higher levels of STC2 expression were observed in HCC tissues and cell lines. Besides, STC2 knockdown reduced proliferation, migration, and invasion, while inducing cell apoptosis. Further analysis indicated a positive correlation between STC2 and glycolysis. STC2 knockdown inhibited glycolysis progression and down-regulated the expressions of PKM2, GLUT1, and HK2 in HCC cells. However, treatment with glycolysis inhibitor (2-DG) prevented oe-STC2 from promoting the growth of HCC cells. Additionally, STC2 knockdown up-regulated the levels of LC3II/LC3I and Beclin1 and reduced the phosphorylation of PI3K, AKT, and mTOR. Treatment with 3-MA, IGF-1, Rap, and LY294002 altered the function of STC2 on proliferation and glycolysis in HCC cells. Tumor formation experiment results revealed that STC2 knockdown inhibited HCC progression. CONCLUSIONS STC2 knockdown inhibited cell proliferation and glycolysis in HCC through the PI3K/Akt/mTOR pathway-mediated autophagy induction.
Collapse
Affiliation(s)
- Ding Li
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Yuanyuan Xiong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Muzi Li
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Lin Long
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Yongjin Zhang
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China
| | - Huifeng Yan
- The Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hua Xiang
- Department of Interventional Radiology and Vascular Surgery, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, 410005, Hunan, China.
| |
Collapse
|
13
|
Kooshan Z, Cárdenas-Piedra L, Clements J, Batra J. Glycolysis, the sweet appetite of the tumor microenvironment. Cancer Lett 2024; 600:217156. [PMID: 39127341 DOI: 10.1016/j.canlet.2024.217156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Cancer cells display an altered metabolic phenotype, characterised by increased glycolysis and lactate production, even in the presence of sufficient oxygen - a phenomenon known as the Warburg effect. This metabolic reprogramming is a crucial adaptation that enables cancer cells to meet their elevated energy and biosynthetic demands. Importantly, the tumor microenvironment plays a pivotal role in shaping and sustaining this metabolic shift in cancer cells. This review explores the intricate relationship between the tumor microenvironment and the Warburg effect, highlighting how communication within this niche regulates cancer cell metabolism and impacts tumor progression and therapeutic resistance. We discuss the potential of targeting the Warburg effect as a promising therapeutic strategy, with the aim of disrupting the metabolic advantage of cancer cells and enhancing our understanding of this complex interplay within the tumor microenvironment.
Collapse
Affiliation(s)
- Zeinab Kooshan
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Lilibeth Cárdenas-Piedra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell & Tissue Engineering Technologies, Brisbane, Australia
| | - Judith Clements
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Jyotsna Batra
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia; Center for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology, Brisbane, Australia; ARC Training Centre for Cell & Tissue Engineering Technologies, Brisbane, Australia.
| |
Collapse
|
14
|
Ni X, Lu CP, Xu GQ, Ma JJ. Transcriptional regulation and post-translational modifications in the glycolytic pathway for targeted cancer therapy. Acta Pharmacol Sin 2024; 45:1533-1555. [PMID: 38622288 PMCID: PMC11272797 DOI: 10.1038/s41401-024-01264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 03/08/2024] [Indexed: 04/17/2024]
Abstract
Cancer cells largely rely on aerobic glycolysis or the Warburg effect to generate essential biomolecules and energy for their rapid growth. The key modulators in glycolysis including glucose transporters and enzymes, e.g. hexokinase 2, enolase 1, pyruvate kinase M2, lactate dehydrogenase A, play indispensable roles in glucose uptake, glucose consumption, ATP generation, lactate production, etc. Transcriptional regulation and post-translational modifications (PTMs) of these critical modulators are important for signal transduction and metabolic reprogramming in the glycolytic pathway, which can provide energy advantages to cancer cell growth. In this review we recapitulate the recent advances in research on glycolytic modulators of cancer cells and analyze the strategies targeting these vital modulators including small-molecule inhibitors and microRNAs (miRNAs) for targeted cancer therapy. We focus on the regulation of the glycolytic pathway at the transcription level (e.g., hypoxia-inducible factor 1, c-MYC, p53, sine oculis homeobox homolog 1, N6-methyladenosine modification) and PTMs (including phosphorylation, methylation, acetylation, ubiquitination, etc.) of the key regulators in these processes. This review will provide a comprehensive understanding of the regulation of the key modulators in the glycolytic pathway and might shed light on the targeted cancer therapy at different molecular levels.
Collapse
Affiliation(s)
- Xuan Ni
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China
| | - Cheng-Piao Lu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, China
| | - Guo-Qiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, China.
- Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Jing-Jing Ma
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
15
|
Afonso J, Barbosa-Matos C, Silvestre R, Pereira-Vieira J, Gonçalves SM, Mendes-Alves C, Parpot P, Pinto J, Carapito Â, Guedes de Pinho P, Santos L, Longatto-Filho A, Baltazar F. Cisplatin-Resistant Urothelial Bladder Cancer Cells Undergo Metabolic Reprogramming beyond the Warburg Effect. Cancers (Basel) 2024; 16:1418. [PMID: 38611096 PMCID: PMC11010907 DOI: 10.3390/cancers16071418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Advanced urothelial bladder cancer (UBC) patients are tagged by a dismal prognosis and high mortality rates, mostly due to their poor response to standard-of-care platinum-based therapy. Mediators of chemoresistance are not fully elucidated. This work aimed to study the metabolic profile of advanced UBC, in the context of cisplatin resistance. Three isogenic pairs of parental cell lines (T24, HT1376 and KU1919) and the matching cisplatin-resistant (R) sublines were used. A set of functional assays was used to perform a metabolic screening on the cells. In comparison to the parental sublines, a tendency was observed towards an exacerbated glycolytic metabolism in the cisplatin-resistant T24 and HT1376 cells; this glycolytic phenotype was particularly evident for the HT1376/HT1376R pair, for which the cisplatin resistance ratio was higher. HT1376R cells showed decreased basal respiration and oxygen consumption associated with ATP production; in accordance, the extracellular acidification rate was also higher in the resistant subline. Glycolytic rate assay confirmed that these cells presented higher basal glycolysis, with an increase in proton efflux. While the results of real-time metabolomics seem to substantiate the manifestation of the Warburg phenotype in HT1376R cells, a shift towards distinct metabolic pathways involving lactate uptake, lipid biosynthesis and glutamate metabolism occurred with time. On the other hand, KU1919R cells seem to engage in a metabolic rewiring, recovering their preference for oxidative phosphorylation. In conclusion, cisplatin-resistant UBC cells seem to display deep metabolic alterations surpassing the Warburg effect, which likely depend on the molecular signature of each cell line.
Collapse
Affiliation(s)
- Julieta Afonso
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Catarina Barbosa-Matos
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Joana Pereira-Vieira
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Samuel Martins Gonçalves
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Camille Mendes-Alves
- CQUM, Centre of Chemistry, Chemistry Department, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.M.-A.); (P.P.)
| | - Pier Parpot
- CQUM, Centre of Chemistry, Chemistry Department, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.M.-A.); (P.P.)
- CEB—Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Joana Pinto
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (J.P.); (Â.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ângela Carapito
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (J.P.); (Â.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University of Porto, 4050-313 Porto, Portugal; (J.P.); (Â.C.); (P.G.d.P.)
- UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Lúcio Santos
- Experimental Pathology and Therapeutics Group, Research Center of the Portuguese Institute of Oncology (CI-IPOP), 4200-072 Porto, Portugal;
- Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Laboratory of Medical Investigation (LIM14), Faculty of Medicine, São Paulo State University, São Paulo 01049-010, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, São Paulo 14784-400, Brazil
| | - Fátima Baltazar
- Life and Health Sciences Research Institute (ICVS), Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (C.B.-M.); (R.S.); (J.P.-V.); (S.M.G.); (A.L.-F.); (F.B.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
16
|
Fujiyoshi S, Honda S, Ara M, Kondo T, Kobayashi N, Taketomi A. SGLT2 is upregulated to acquire cisplatin resistance and SGLT2 inhibition reduces cisplatin resistance in hepatoblastoma. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2024; 31:223-233. [PMID: 37927142 DOI: 10.1002/jhbp.1391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
BACKGROUND Cancer cells can alter glucose metabolism and regulate the expression of glucose transporters. Hepatoblastoma patients undergo cisplatin-based chemotherapy; however, 22.3% of patients develop cisplatin resistance and thus face a poor prognosis. We hypothesized that glucose transporters are associated with acquiring cisplatin resistance with increasing sugar intake inhibiting glucose transporters could reduce cisplatin resistance in hepatoblastoma patients. METHODS We established cisplatin-resistant HepG2 and HuH6 cells by continuous treatment with cisplatin. We evaluated the relationship between cisplatin resistance and glucose uptake. We used an expression array to select cisplatin-resistant associated glucose transporters and selected sodium-glucose cotransporter 2 (SGLT2). We used dapagliflozin as an SGLT2 inhibitor and evaluated glucose uptake and IC50 after dapagliflozin treatment in wild-type and resistant hepatoblastoma cells in vitro and in vivo. RESULTS We found a strong relationship between cisplatin resistance and glucose uptake. Additionally, SGLT2 was upregulated in resistant cells after cisplatin treatment. After dapagliflozin treatment, glucose uptake and cisplatin resistance decreased in resistant cells. CONCLUSIONS Cisplatin-resistant hepatoblastoma cells exhibited upregulated SGLT2 expression and activated glucose uptake to survive under cisplatin stress. SGLT2 inhibition decreased cellular resistance to cisplatin. SGLT2 inhibition with cisplatin therapy could be a novel therapeutic strategy for cisplatin-resistant hepatoblastoma patients.
Collapse
Affiliation(s)
- Sunao Fujiyoshi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Shohei Honda
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Momoko Ara
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Takafumi Kondo
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Nozomi Kobayashi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery 1, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
17
|
Gao L, Li L, Zhang D, Qiu J, Qian J, Liu H. TAPI-1 Exhibits Anti-tumor Efficacy in Human Esophageal Squamous Cell Carcinoma Cells via Suppression of NF-κB Signaling Pathway. Dig Dis Sci 2024; 69:81-94. [PMID: 38007701 PMCID: PMC10787672 DOI: 10.1007/s10620-023-08181-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 11/02/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND TNF-α processing inhibitor-1 (TAPI-1) is a known metalloproteinase inhibitor with potential anti-inflammatory effects. However, its anti-cancer effects on esophageal squamous cell carcinoma (ESCC) have not been uncovered. AIM In the present study, the effects of TAPI-1 on ESCC cell viability, migration, invasion, and cisplatin resistance and the underlying molecular mechanisms were investigated in TE-1 and Eca109 cells. METHODS To this end, TE-1 and Eca109 cells were exposed to TAPI-1 for indicated time intervals. Cell viability was assessed using cell counting kit-8 assay and apoptosis was evaluated using flow cytometry assay. Migration and invasion were assessed using Transwell assays. Gene expressions were analyzed using quantitative reverse transcription polymerase chain reaction. The activation of NF-κB signaling pathway was elucidated via Western blot and chromatin immunoprecipitation assay. RESULTS We observed that higher doses (10, 20 μM) of TAPI-1 inhibited ESCC cell viability, while a lower dose (5 μM) of TAPI-1 inhibited ESCC cell migration and invasion and enhanced the chemosensitivity of ESCC cells to cisplatin. Moreover, TAPI-1 suppressed the activation of NF-κB signaling and the target genes expression in the stage of transcription initiation. Furthermore, blocking NF-κB signaling in advance could abolish all the effects of TAPI-1 on ESCC cells. CONCLUSION Overall, these results indicated that TAPI-1 impairs ESCC cell viability, migration, and invasion and facilitates cisplatin-induced apoptosis via suppression of NF-κB signaling pathway. TAPI-1 may serve as a potential adjuvant agent with cisplatin for ESCC therapy.
Collapse
Affiliation(s)
- Lin Gao
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Li Li
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China
| | - Dongmei Zhang
- Medical Research Center, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Jianwei Qiu
- Department of Gastroenterology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Junbo Qian
- Department of Gastroenterology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, People's Republic of China
| | - Hongbin Liu
- Department of Pathology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Shengli Road No. 666, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Kouzu K, Tsujimoto H, Tamura K, Einama T, Kishi Y, Ishida J, Ueno H. Impact of retention index on the neoadjuvant chemotherapy effect and the prognosis in oesophageal cancer. Jpn J Clin Oncol 2023; 53:1130-1137. [PMID: 37626445 DOI: 10.1093/jjco/hyad115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVE The relationship between retention index calculated from dual-time point 18F-fluorodeoxyglucose positron emission tomography-computed tomography and oesophageal cancer prognosis remains unknown. This study aimed to determine usefulness of retention index as a predictor of long-term prognosis of oesophageal cancer and neoadjuvant chemotherapy efficacy. METHODS A total of 151 patients with oesophageal cancer who underwent esophagectomy were evaluated retrospectively in this study. We acquired positron emission tomography scans 60 and 120 min (SUVmax1 and SUVmax2, respectively) after the intravenous administration of 3.7 Mbq/kg 18F-fluorodeoxyglucose. The patients were divided into two groups: high-retention index (retention index ≥29%, 107 patients) and low-retention index (retention index <29%, 44 patients). Retention index was calculated as follows: retention index (%) = [(SUVmax2 - SUVmax1)/SUVmax1] × 100. RESULTS The overall survival and relapse-free survival rates in the high-retention index group were significantly lower than those in the low-retention index group (P < 0.001). Our multivariate analysis identified that the high-retention index group contained independent risk factors for overall survival (hazard ratio: 2.44, P = 0.009) and relapse-free survival (hazard ratio: 2.61, P = 0.002). The high-retention index group exhibited a lower partial response rate to neoadjuvant chemotherapy evaluated by computed tomography (P < 0.001) and a lower pathological therapeutic effect in the resected specimen (P = 0.019) than the low-retention index group. CONCLUSIONS The retention index was associated with neoadjuvant chemotherapy responses and long-term prognosis for oesophageal cancer.
Collapse
Affiliation(s)
- Keita Kouzu
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | | | - Takahiro Einama
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Jiro Ishida
- Eijinkai Seeds Clinic, Tokorozawa, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
19
|
Liu T, Ren Y, Wang Q, Wang Y, Li Z, Sun W, Fan D, Luan Y, Gao Y, Yan Z. Exploring the role of the disulfidptosis-related gene SLC7A11 in adrenocortical carcinoma: implications for prognosis, immune infiltration, and therapeutic strategies. Cancer Cell Int 2023; 23:259. [PMID: 37919768 PMCID: PMC10623781 DOI: 10.1186/s12935-023-03091-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Disulfidptosis and the disulfidptosis-related gene SLC7A11 have recently attracted significant attention for their role in tumorigenesis and tumour management. However, its association with adrenocortical carcinoma (ACC) is rarely discussed. METHODS Differential analysis, Cox regression analysis, and survival analysis were used to screen for the hub gene SLC7A11 in the TCGA and GTEx databases and disulfidptosis-related gene sets. Then, we performed an association analysis between SLC7A11 and clinically relevant factors in ACC patients. Univariate and multivariate Cox regression analyses were performed to evaluate the prognostic value of SLC7A11 and clinically relevant factors. Weighted gene coexpression analysis was used to find genes associated with SLC7A11. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses and the LinkedOmics database were used to analyse the functions of SLC7A11-associated genes. The CIBERSORT and Xcell algorithms were used to analyse the relationship between SLC7A11 and immune cell infiltration in ACC. The TISIDB database was applied to search for the correlation between SLC7A11 expression and immune chemokines. In addition, we performed a correlation analysis for SLC7A11 expression and tumour mutational burden and immune checkpoint-related genes and assessed drug sensitivity based on SLC7A11 expression. Immunohistochemistry and RT‒qPCR were used to validate the upregulation of SLC7A11 in the ACC. RESULTS SLC7A11 is highly expressed in multiple urological tumours, including ACC. SLC7A11 expression is strongly associated with clinically relevant factors (M-stage and MYL6 expression) in ACC. SLC7A11 and the constructed nomogram can accurately predict ACC patient outcomes. The functions of SLC7A11 and its closely related genes are tightly associated with the occurrence of disulfidptosis in ACC. SLC7A11 expression was tightly associated with various immune cell infiltration disorders in the ACC tumour microenvironment (TME). It was positively correlated with the expression of immune chemokines (CXCL8, CXCL3, and CCL20) and negatively correlated with the expression of immune chemokines (CXCL17 and CCL14). SLC7A11 expression was positively associated with the expression of immune checkpoint genes (NRP1, TNFSF4, TNFRSF9, and CD276) and tumour mutation burden. The expression level of SLC7A11 in ACC patients is closely associated withcthe drug sensitivity. CONCLUSION In ACC, high expression of SLC7A11 is associated with migration, invasion, drug sensitivity, immune infiltration disorders, and poor prognosis, and its induction of disulfidptosis is a promising target for the treatment of ACC.
Collapse
Affiliation(s)
- Tonghu Liu
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Yilin Ren
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Qixin Wang
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Department of Surgery, Nanyang Central Hospital, 473005, Nanyang, Henan, China
| | - Yu Wang
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Zhiyuan Li
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Weibo Sun
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
- Department of Radiation Oncology and Oncology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, 450003, Zhengzhou, Henan, China
| | - Dandan Fan
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China
| | - Yongkun Luan
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China.
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China.
| | - Yukui Gao
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, 241001, Wuhu, Anhui, China.
| | - Zechen Yan
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, 450001, Zhengzhou, Henan, China.
- Henan Engineering Research Center of Tumour Molecular Diagnosis and Treatment, 450001, Zhengzhou, Henan, China.
- Institute of Molecular Cancer Surgery of Zhengzhou University, 450001, Zhengzhou, Henan, China.
| |
Collapse
|
20
|
Liu Y, Zhang X, Cheng F, Cao W, Geng Y, Chen Z, Wei W, Zhang L. Xanthatin induce DDP-resistance lung cancer cells apoptosis through regulation of GLUT1 mediated ROS accumulation. Drug Dev Res 2023; 84:1266-1278. [PMID: 37260173 DOI: 10.1002/ddr.22084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
Chemoresistance to cisplatin (DDP) therapy is a major obstacle that needs to be overcome in treating lung cancer patients. Xanthatin has been reported to exhibit an antitumor effect on various cancers, but the function of xanthatin in DDP-resistance lung cancer remains unclear. The study aimed to explore the effect and mechanisms of xanthatin on proliferation, apoptosis, and migration in DDP-resistance lung cancer cells. In the present study, xanthatin suppresses the expression of glucose transporter 1 (GLUT1), attenuates the pentose phosphate pathway (PPP), and causes ROS accumulation and apoptosis, thereby mitigating the antioxidative capacity in DDP-resistance cells. Previous studies have shown that GLUT1 is associated with resistance to platinum drugs. We found that GLUT1 was significantly increased in the DDP-resistant lung cancer cell line compared to the parental cell line, and xanthatin significantly downregulated GLUT1 expression in DDP-resistant lung cancer cells. Notably, overexpression of GLUT1 significantly reduced the production of ROS and increased cellular NADPH/NADP+ and GSH/GSSG ratios. Thus, these results suggest that xanthatin induces DDP-resistance lung cancer cells apoptosis through regulation of GLUT1-mediated ROS accumulation. These findings might provide a possible strategy for the clinical treatment of DDP-resistant lung cancer.
Collapse
Affiliation(s)
- Yunxiao Liu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Xinge Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Fenting Cheng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Cao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yadi Geng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaolin Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Lei Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
21
|
Chu YD, Chen CW, Lai MW, Lim SN, Lin WR. Bioenergetic alteration in gastrointestinal cancers: The good, the bad and the ugly. World J Gastroenterol 2023; 29:4499-4527. [PMID: 37621758 PMCID: PMC10445009 DOI: 10.3748/wjg.v29.i29.4499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 08/02/2023] Open
Abstract
Cancer cells exhibit metabolic reprogramming and bioenergetic alteration, utilizing glucose fermentation for energy production, known as the Warburg effect. However, there are a lack of comprehensive reviews summarizing the metabolic reprogramming, bioenergetic alteration, and their oncogenetic links in gastrointestinal (GI) cancers. Furthermore, the efficacy and treatment potential of emerging anticancer drugs targeting these alterations in GI cancers require further evaluation. This review highlights the interplay between aerobic glycolysis, the tricarboxylic acid (TCA) cycle, and oxidative phosphorylation (OXPHOS) in cancer cells, as well as hypotheses on the molecular mechanisms that trigger this alteration. The role of hypoxia-inducible transcription factors, tumor suppressors, and the oncogenetic link between hypoxia-related enzymes, bioenergetic changes, and GI cancer are also discussed. This review emphasizes the potential of targeting bioenergetic regulators for anti-cancer therapy, particularly for GI cancers. Emphasizing the potential of targeting bioenergetic regulators for GI cancer therapy, the review categorizes these regulators into aerobic glycolysis/ lactate biosynthesis/transportation and TCA cycle/coupled OXPHOS. We also detail various anti-cancer drugs and strategies that have produced pre-clinical and/or clinical evidence in treating GI cancers, as well as the challenges posed by these drugs. Here we highlight that understanding dysregulated cancer cell bioenergetics is critical for effective treatments, although the diverse metabolic patterns present challenges for targeted therapies. Further research is needed to comprehend the specific mechanisms of inhibiting bioenergetic enzymes, address side effects, and leverage high-throughput multi-omics and spatial omics to gain insights into cancer cell heterogeneity for targeted bioenergetic therapies.
Collapse
Affiliation(s)
- Yu-De Chu
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chun-Wei Chen
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ming-Wei Lai
- Department of Pediatrics, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Siew-Na Lim
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Wey-Ran Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
22
|
Zhong J, Tian L, Gou Y, Zhao P, Dong X, Guo M, Zhao G, Li A, Hao A, He TC, Fan J. BMP4 upregulates glycogen synthesis through the SMAD/SLC2A1 (GLUT1) signaling axis in hepatocellular carcinoma (HCC) cells. Cancer Metab 2023; 11:9. [PMID: 37443106 DOI: 10.1186/s40170-023-00310-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Excessive hepatic glycogen accumulation benefits tumorigenesis and cancer cell survival. We previously reported that BMP4 has the strongest ability to promote glycogenesis among the 14 BMPs in hepatocytes and augmented hepatocellular carcinoma (HCC) cell survival under hypoxia and hypoglycemia conditions by promoting the glycolysis pathway. However, the mechanism underlying BMP4's effect on glycogenesis in HCC remains elusive. METHODS The expression of BMP4 and SLC2A1 were acquired by analyzing the TCGA-LIHC dataset, as well as by immunohistochemical analysis of the 40 pairs of human HCC samples and para-tumor tissues. Gene expressions were detected by qPCR, immunoflurorescence staining, and Western blotting. Overexpression and silencing of BMP4 were accomplished through adenoviruses Ad-B4 and Ad-siB4 infection. Hepatic glycogen was detected by PAS staining. SLC2A1 (GLUT1) function was blocked by the inhibitor BAY-876. ChIP assay was used to determine the binding of SMADs to the promoter region of SLC2A1 in HCC cells. Lastly, the in vivo effect of BMP4-regulated SLC2A1 on HCC tumor growth was assessed in a xenograft model of HCC. RESULTS The elevated expression of BMP4 in HCC tumor tissues was highly correlated with hepatic glycogen accumulation in clinical samples. SLC2A1 was highly expressed in HCC tumor tissue and correlated with clinical stage and prognosis. Exogenous BMP4 augmented glycogen accumulation and upregulated the expression of glycogen synthesis-related genes in Huh7 and HepG2 cells, both of which were effectively blunted by SLC2A1inhibitor BAY-876. In mechanism, BMP4 activated SMAD5 to regulate the promoter of SLC2A1to enhance its expression. The in vivo xenograft experiments revealed that BMP4 promoted glycogen accumulation and tumor growth, which were effectively diminished by BAY-876. CONCLUSION These results demonstrate that BMP4 upregulates glycogen synthesis through the SMAD/SLC2A1 (GLUT1) signaling axis in HCC cells, which may be exploited as novel therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL, 60637, USA
| | - Luyao Tian
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL, 60637, USA
| | - Piao Zhao
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL, 60637, USA
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
| | - Meichun Guo
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
| | - Guozhi Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL, 60637, USA
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL, 60637, USA.
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, Chongqing, China.
- Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, No. 1 Medical School Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
23
|
Bahremani M, Rashtchizadeh N, Sabzichi M, Vatankhah AM, Danaiyan S, Poursistany H, Mohammadian J, Ghorbanihaghjo A. Enhanced chemotherapeutic efficacy of docetaxel in human lung cancer cell line via GLUT1 inhibitor. J Biochem Mol Toxicol 2023; 37:e23348. [PMID: 36999407 DOI: 10.1002/jbt.23348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/01/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
The dose-dependent adverse effects of anticancer agents need new methods with lesser toxicity. The objective of the current research was to evaluate the efficacy of GLUT1 inhibitor, as an inhibitor of glucose consumption in cancer cells, in augmenting the efficiency of docetaxel with respect to cytotoxicity and apoptosis. Cell cytotoxicity was assessed by using methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. Annexin V/PI double staining was employed to evaluate apoptosis percentage. Quantitative real-time polymerase chain reaction (RT-PCR) analysis was accomplished to detect the expression of genes involved in the apoptosis pathway. The IC50 values for docetaxel and BAY-876 were 3.7 ± 0.81 and 34.1 ± 3.4 nM, respectively. The severity of synergistic mutual effects of these agents on each other was calculated by synergy finder application. It showed that the percentage of apoptotic cells following co-administration of docetaxel and BAY-876 increased to 48.1 ± 2.8%. In comparison without GLUT1 co-administration, the combined therapy decreased significantly the transcriptome levels of the Bcl-2 and Ki-67 and a remarkable increase in the level of the Bax as proapoptotic protein(p < 0.05). Co-treatment of BAY-876 and docetaxel depicted a synergistic effect which was calculated using the synergy finder highest single agent (HSA) method (HSA synergy score: 28.055). These findings recommend that the combination of GLUT-1 inhibitor and docetaxel can be considered as a promising therapeutic approach for the treatment of patients with lung cancer.
Collapse
Affiliation(s)
- Mona Bahremani
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadereh Rashtchizadeh
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sabzichi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sepideh Danaiyan
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haniyeh Poursistany
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jamal Mohammadian
- Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghorbanihaghjo
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
24
|
Chelakkot C, Chelakkot VS, Shin Y, Song K. Modulating Glycolysis to Improve Cancer Therapy. Int J Mol Sci 2023; 24:2606. [PMID: 36768924 PMCID: PMC9916680 DOI: 10.3390/ijms24032606] [Citation(s) in RCA: 147] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Cancer cells undergo metabolic reprogramming and switch to a 'glycolysis-dominant' metabolic profile to promote their survival and meet their requirements for energy and macromolecules. This phenomenon, also known as the 'Warburg effect,' provides a survival advantage to the cancer cells and make the tumor environment more pro-cancerous. Additionally, the increased glycolytic dependence also promotes chemo/radio resistance. A similar switch to a glycolytic metabolic profile is also shown by the immune cells in the tumor microenvironment, inducing a competition between the cancer cells and the tumor-infiltrating cells over nutrients. Several recent studies have shown that targeting the enhanced glycolysis in cancer cells is a promising strategy to make them more susceptible to treatment with other conventional treatment modalities, including chemotherapy, radiotherapy, hormonal therapy, immunotherapy, and photodynamic therapy. Although several targeting strategies have been developed and several of them are in different stages of pre-clinical and clinical evaluation, there is still a lack of effective strategies to specifically target cancer cell glycolysis to improve treatment efficacy. Herein, we have reviewed our current understanding of the role of metabolic reprogramming in cancer cells and how targeting this phenomenon could be a potential strategy to improve the efficacy of conventional cancer therapy.
Collapse
Affiliation(s)
| | - Vipin Shankar Chelakkot
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Youngkee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, Research Institute of Pharmaceutical Science, Department of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyoung Song
- College of Pharmacy, Duksung Women’s University, Seoul 01366, Republic of Korea
| |
Collapse
|
25
|
Li R, Yan L, Tian S, Zhao Y, Zhu Y, Wang X. Increased response to TPF chemotherapy promotes immune escape in hypopharyngeal squamous cell carcinoma. Front Pharmacol 2023; 13:1097197. [PMID: 36712687 PMCID: PMC9880322 DOI: 10.3389/fphar.2022.1097197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
Background: There is an urgent need to identify which patients would benefit from TPF chemotherapy in hypopharyngeal squamous cell carcinoma (HPSCC) and to explore new combinations to improve the treatment effect. Materials and methods: Gene-expression profiles in 15 TPF-sensitive patients were compared to 13 resistant patients. Immunohistochemistry (IHC) was performed to detect CD8+ T cells in 28 samples. Patient-Derived Tumor Xenograft (PDX) model and IHC were used to verify markers that optimize treatment for HPSCC. Results: Through RNA sequencing 188 genes were up-regulated in TPF chemotherapy-resistant (CR) tissues were involved in T cell activation, while 60 down-regulated genes were involved in glycolysis. Gene set enrichment analysis (GSEA) showed that chemotherapy-sensitive (CS) group upregulation of the pathways of glycolysis, while immune response was downregulated. CIBERSORT, MCP-counter, and IHC proved that most immune cells including CD8+ T cells in the CR significantly higher than that in CS group. Among the 16 up-regulated genes in CS had close associations, the most significant negative correlation between the gene level and CD8+ T cells existed in SEC61G. SEC61G was related to glycolysis, which was transcriptionally regulated by E2F1, and participated in antigen degradation through ubiquitin-dependent protein catabolic process. Palbociclib, combined with Cetuximab decreased the tumor burden and significantly suppressed the expression of E2F1 and SEC61G while activating MHC-I in PDX model. Conclusion: Enhanced glycolysis promoted immune escape, but increased response to TPF chemotherapy. SEC61G was the center of the molecular network and targeting the E2F1/SEC61G pathway increased the expression level of MHC-I.
Collapse
Affiliation(s)
| | | | - Shu Tian
- *Correspondence: Xiaoshen Wang, ; Yi Zhu, ; Shu Tian,
| | | | - Yi Zhu
- *Correspondence: Xiaoshen Wang, ; Yi Zhu, ; Shu Tian,
| | - Xiaoshen Wang
- *Correspondence: Xiaoshen Wang, ; Yi Zhu, ; Shu Tian,
| |
Collapse
|
26
|
Cao B, Zhao R, Li H, Xu X, Gao J, Chen L, Wei B. Inhibition of androgen receptor enhanced the anticancer effects of everolimus through targeting glucose transporter 12. Int J Biol Sci 2023; 19:104-119. [PMID: 36594084 PMCID: PMC9760431 DOI: 10.7150/ijbs.75106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/08/2022] [Indexed: 11/24/2022] Open
Abstract
Everolimus was designed as a mammalian target of rapamycin (mTOR) inhibitor. It has been proven as a targeted drug for gastric cancer (GC) therapy. However, long-term treatment with everolimus may cause severe side effects for recipients. Decreasing the dosage and attenuating the associated risks are feasible to promote clinical translation of everolimus. This study aimed to identify the underlying mechanisms of responses to everolimus and develop novel regimens for GC treatment. Our findings proved that there was a significant dose-dependent relationship of everolimus-induced GC cell apoptosis and glycolysis inhibition. Then, we found that a member of glucose transporter (GLUT12) family, GLUT12, was actively upregulated to counteract the anticancer effects of everolimus. GLUT12 might be overexpressed in GC. High expression of GLUT12 might be correlated with tumor progression and short survival time of GC patients. Bioinformatic analysis suggested that GLUT12 might be involved in regulating cancer development and metabolism. The experiments proved that GLUT12 significantly promoted GC growth, glycolysis and impaired the anticancer effects of everolimus. Androgen receptor (AR) is a classical oncogenic factor in many types of cancer. Everolimus elevated GLUT12 expression in an AR-dependent manner. Inhibition of AR activity abrogated the promotive effects on GLUT12 expression. Both in-vitro and in-vivo experiments demonstrated that GLUT12 knockdown augmented anticancer effects of everolimus. Enzalutamide, an AR inhibitor, or AR knockdown was comparable to GLUT12 suppression. This study identified the role of the AR/GLUT12 pathway in the development of poor responses to everolimus. Interference with AR/GLUT12 pathway may serve as a promising approach to promoting the translational application of everolimus in GC therapy.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Xingming Xu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Jingwang Gao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China
| | - Lin Chen
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China.,✉ Corresponding authors: Bo Wei, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938071; E-mail: ; Lin Chen, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938066; E-mail:
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Medical School of Chinese PLA, Beijing 100853, China.,✉ Corresponding authors: Bo Wei, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938071; E-mail: ; Lin Chen, MD, PhD, Chief Doctor, Professor, Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Tel: +86-10-66938066; E-mail:
| |
Collapse
|
27
|
The Effect of Oxidative Phosphorylation on Cancer Drug Resistance. Cancers (Basel) 2022; 15:cancers15010062. [PMID: 36612059 PMCID: PMC9817696 DOI: 10.3390/cancers15010062] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Recent studies have shown that oxidative phosphorylation (OXPHOS) is a target for the effective attenuation of cancer drug resistance. OXPHOS inhibitors can improve treatment responses to anticancer therapy in certain cancers, such as melanomas, lymphomas, colon cancers, leukemias and pancreatic ductal adenocarcinoma (PDAC). However, the effect of OXPHOS on cancer drug resistance is complex and associated with cell types in the tumor microenvironment (TME). Cancer cells universally promote OXPHOS activity through the activation of various signaling pathways, and this activity is required for resistance to cancer therapy. Resistant cancer cells are prevalent among cancer stem cells (CSCs), for which the main metabolic phenotype is increased OXPHOS. CSCs depend on OXPHOS to survive targeting by anticancer drugs and can be selectively eradicated by OXPHOS inhibitors. In contrast to that in cancer cells, mitochondrial OXPHOS is significantly downregulated in tumor-infiltrating T cells, impairing antitumor immunity. In this review, we summarize novel research showing the effect of OXPHOS on cancer drug resistance, thereby explaining how this metabolic process plays a dual role in cancer progression. We highlight the underlying mechanisms of metabolic reprogramming in cancer cells, as it is vital for discovering new drug targets.
Collapse
|
28
|
Temre MK, Kumar A, Singh SM. An appraisal of the current status of inhibition of glucose transporters as an emerging antineoplastic approach: Promising potential of new pan-GLUT inhibitors. Front Pharmacol 2022; 13:1035510. [PMID: 36386187 PMCID: PMC9663470 DOI: 10.3389/fphar.2022.1035510] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/18/2022] [Indexed: 07/23/2023] Open
Abstract
Neoplastic cells displayed altered metabolism with accelerated glycolysis. Therefore, these cells need a mammoth supply of glucose for which they display an upregulated expression of various glucose transporters (GLUT). Thus, novel antineoplastic strategies focus on inhibiting GLUT to intersect the glycolytic lifeline of cancer cells. This review focuses on the current status of various GLUT inhibition scenarios. The GLUT inhibitors belong to both natural and synthetic small inhibitory molecules category. As neoplastic cells express multiple GLUT isoforms, it is necessary to use pan-GLUT inhibitors. Nevertheless, it is also necessary that such pan-GLUT inhibitors exert their action at a low concentration so that normal healthy cells are left unharmed and minimal injury is caused to the other vital organs and systems of the body. Moreover, approaches are also emerging from combining GLUT inhibitors with other chemotherapeutic agents to potentiate the antineoplastic action. A new pan-GLUT inhibitor named glutor, a piperazine-one derivative, has shown a potent antineoplastic action owing to its inhibitory action exerted at nanomolar concentrations. The review discusses the merits and limitations of the existing GLUT inhibitory approach with possible future outcomes.
Collapse
Affiliation(s)
- Mithlesh Kumar Temre
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ajay Kumar
- Deparment of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sukh Mahendra Singh
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
29
|
Cui Y, Li C, Sang F, Cao W, Qin Z, Zhang P. Natural products targeting glycolytic signaling pathways-an updated review on anti-cancer therapy. Front Pharmacol 2022; 13:1035882. [PMID: 36339566 PMCID: PMC9631946 DOI: 10.3389/fphar.2022.1035882] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/30/2022] [Indexed: 11/30/2022] Open
Abstract
Glycolysis is a complex metabolic process that occurs to convert glucose into pyruvate to produce energy for living cells. Normal cells oxidized pyruvate into adenosine triphosphate and carbon dioxide in the presence of oxygen in mitochondria while cancer cells preferentially metabolize pyruvate to lactate even in the presence of oxygen in order to maintain a slightly acidic micro-environment of PH 6.5 and 6.9, which is beneficial for cancer cell growth and metastasis. Therefore targeting glycolytic signaling pathways provided new strategy for anti-cancer therapy. Natural products are important sources for the treatment of diseases with a variety of pharmacologic activities. Accumulated studies suggested that natural products exhibited remarkable anti-cancer properties both in vitro and in vivo. Plenty of studies suggested natural products like flavonoids, terpenoids and quinones played anti-cancer properties via inhibiting glucose metabolism targets in glycolytic pathways. This study provided an updated overview of natural products controlling glycolytic pathways, which also provide insight into druggable mediators discovery targeting cancer glucose metabolism.
Collapse
Affiliation(s)
- Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Chuang Li
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Feng Sang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Zhuo Qin
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong, China
- *Correspondence: Weiling Cao, ; Zhuo Qin, ; Peng Zhang,
| |
Collapse
|
30
|
Targeting Glucose Metabolism Enzymes in Cancer Treatment: Current and Emerging Strategies. Cancers (Basel) 2022; 14:cancers14194568. [PMID: 36230492 PMCID: PMC9559313 DOI: 10.3390/cancers14194568] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Reprogramming of glucose metabolism is a hallmark of cancer and can be targeted by therapeutic agents. Some metabolism regulators, such as ivosidenib and enasidenib, have been approved for cancer treatment. Currently, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Furthermore, some natural products have shown efficacy in killing tumor cells by regulating glucose metabolism, offering novel therapeutic opportunities in cancer. However, most of them have failed to be translated into clinical applications due to low selectivity, high toxicity, and side effects. Recent studies suggest that combining glucose metabolism modulators with chemotherapeutic drugs, immunotherapeutic drugs, and other conventional anticancer drugs may be a future direction for cancer treatment. Abstract Reprogramming of glucose metabolism provides sufficient energy and raw materials for the proliferation, metastasis, and immune escape of cancer cells, which is enabled by glucose metabolism-related enzymes that are abundantly expressed in a broad range of cancers. Therefore, targeting glucose metabolism enzymes has emerged as a promising strategy for anticancer drug development. Although several glucose metabolism modulators have been approved for cancer treatment in recent years, some limitations exist, such as a short half-life, poor solubility, and numerous adverse effects. With the rapid development of medicinal chemicals, more advanced and effective glucose metabolism enzyme-targeted anticancer drugs have been developed. Additionally, several studies have found that some natural products can suppress cancer progression by regulating glucose metabolism enzymes. In this review, we summarize the mechanisms underlying the reprogramming of glucose metabolism and present enzymes that could serve as therapeutic targets. In addition, we systematically review the existing drugs targeting glucose metabolism enzymes, including small-molecule modulators and natural products. Finally, the opportunities and challenges for glucose metabolism enzyme-targeted anticancer drugs are also discussed. In conclusion, combining glucose metabolism modulators with conventional anticancer drugs may be a promising cancer treatment strategy.
Collapse
|
31
|
Zhao J, Wang C, Zhang X, Li J, Liu Y, Pan X, Zhu L, Chen D, Xie T. Cell membrane coated electrochemical sensor for kinetic measurements of GLUT transport. Anal Chim Acta 2022; 1226:340263. [DOI: 10.1016/j.aca.2022.340263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 11/01/2022]
|
32
|
Tian X, Yan T, Liu F, Liu Q, Zhao J, Xiong H, Jiang S. Link of sorafenib resistance with the tumor microenvironment in hepatocellular carcinoma: Mechanistic insights. Front Pharmacol 2022; 13:991052. [PMID: 36071839 PMCID: PMC9441942 DOI: 10.3389/fphar.2022.991052] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022] Open
Abstract
Sorafenib, a multi-kinase inhibitor with antiangiogenic, antiproliferative, and proapoptotic properties, is the first-line treatment for patients with late-stage hepatocellular carcinoma (HCC). However, the therapeutic effect remains limited due to sorafenib resistance. Only about 30% of HCC patients respond well to the treatment, and the resistance almost inevitably happens within 6 months. Thus, it is critical to elucidate the underlying mechanisms and identify effective approaches to improve the therapeutic outcome. According to recent studies, tumor microenvironment (TME) and immune escape play critical roles in tumor occurrence, metastasis and anti-cancer drug resistance. The relevant mechanisms were focusing on hypoxia, tumor-associated immune-suppressive cells, and immunosuppressive molecules. In this review, we focus on sorafenib resistance and its relationship with liver cancer immune microenvironment, highlighting the importance of breaking sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Xinchen Tian
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tinghao Yan
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fen Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Jing Zhao
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Basic Medical School, Jining Medical University, Jining, China
- *Correspondence: Huabao Xiong, ; Shulong Jiang,
| | - Shulong Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Clinical Medical Laboratory Center, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Huabao Xiong, ; Shulong Jiang,
| |
Collapse
|
33
|
Endoplasmic Reticulum Stress-Related Four-Biomarker Risk Classifier for Survival Evaluation in Esophageal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:5860671. [PMID: 35342421 PMCID: PMC8956413 DOI: 10.1155/2022/5860671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/27/2022] [Accepted: 03/02/2022] [Indexed: 12/03/2022]
Abstract
Purpose Esophageal cancer (EC) is a lethal digestive tumor worldwide with a dismal clinical outcome. Endoplasmic reticulum (ER) stress poses essential implications for a variety of tumor malignant behaviors. Here, we set up an ER stress-based risk classifier for assessing patient outcome and exploiting robust targets for medical decision-making of EC cases. Methods 340 EC cases with transcriptome and survival data from two independent public datasets (TCGA and GEO) were recruited for this project. Cox regression analyses were employed to create a risk classifier based on ER stress-related genes (ERGs) which were strongly linked to EC cases' outcomes. Then, we detected and confirmed the predictive ability of our proposed classifier via a host of statistical methods, including survival analysis and ROC method. In addition, immune-associated algorithm was implemented to analyze the immune activity of EC samples. Results Four EGRs (BCAP31, HSPD1, PDHA1, and UBE2D1) were selected to build an EGR-related classifier (ERC). This classifier could distinguish the patients into different risky subgroups. The remarkable differences in patient outcome between the two groups were observed, and similar results were also confirmed in GEO cohort. In terms of the immune analysis, the ERC could forecast the infiltration level of immunocytes, such as Tregs and NK cells. Conclusion We created a four-ERG risk classifier which displays the powerful capability of survival evaluation for EC cases.
Collapse
|
34
|
Qiu BQ, Lin XH, Lai SQ, Lu F, Lin K, Long X, Zhu SQ, Zou HX, Xu JJ, Liu JC, Wu YB. ITGB1-DT/ARNTL2 axis may be a novel biomarker in lung adenocarcinoma: a bioinformatics analysis and experimental validation. Cancer Cell Int 2021; 21:665. [PMID: 34906142 PMCID: PMC8670189 DOI: 10.1186/s12935-021-02380-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Lung cancer is one of the most lethal malignant tumors that endangers human health. Lung adenocarcinoma (LUAD) has increased dramatically in recent decades, accounting for nearly 40% of all lung cancer cases. Increasing evidence points to the importance of the competitive endogenous RNA (ceRNA) intrinsic mechanism in various human cancers. However, behavioral characteristics of the ceRNA network in lung adenocarcinoma need further study. METHODS Groups based on SLC2A1 expression were used in this study to identify associated ceRNA networks and potential prognostic markers in lung adenocarcinoma. The Cancer Genome Atlas (TCGA) database was used to obtain the patients' lncRNA, miRNA, and mRNA expression profiles, as well as clinical data. Informatics techniques were used to investigate the effect of hub genes on prognosis. The Cox regression analyses were performed to evaluate the prognostic effect of hub genes. The methylation, GSEA, and immune infiltration analyses were utilized to explore the potential mechanisms of the hub gene. The CCK-8, transwell, and colony formation assays were performed to detect the proliferation and invasion of lung cancer cells. RESULTS We eventually identified the ITGB1-DT/ARNTL2 axis as an independent fact may promote lung adenocarcinoma progression. Furthermore, methylation analysis revealed that hypo-methylation may cause the dysregulated ITGB1-DT/ARNTL2 axis, and immune infiltration analysis revealed that the ITGB1-DT/ARNTL2 axis may affect the immune microenvironment and the progression of lung adenocarcinoma. The CCK-8, transwell, and colonu formation assays suggested that ITGB1-DT/ARNTL2 promotes the progression of lung adenocarcinoma. And hsa-miR-30b-3p reversed the ITGB1/ARNTL2-mediated oncogenic processes. CONCLUSION Our study identified the ITGB1-DT/ARNTL2 axis as a novel prognostic biomarker affects the prognosis of lung adenocarcinoma.
Collapse
Affiliation(s)
- Bai-Quan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xia-Hui Lin
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Song-Qing Lai
- Institute of Cardiovascular Disease, Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Feng Lu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kun Lin
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiang Long
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shu-Qiang Zhu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Hua-Xi Zou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jian-Jun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ji-Chun Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Yong-Bing Wu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
35
|
Wang L, Zhao X, Fu J, Xu W, Yuan J. The Role of Tumour Metabolism in Cisplatin Resistance. Front Mol Biosci 2021; 8:691795. [PMID: 34250022 PMCID: PMC8261055 DOI: 10.3389/fmolb.2021.691795] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022] Open
Abstract
Cisplatin is a chemotherapy drug commonly used in cancer treatment. Tumour cells are more sensitive to cisplatin than normal cells. Cisplatin exerts an antitumour effect by interfering with DNA replication and transcription processes. However, the drug-resistance properties of tumour cells often cause loss of cisplatin efficacy and failure of chemotherapy, leading to tumour progression. Owing to the large amounts of energy and compounds required by tumour cells, metabolic reprogramming plays an important part in the occurrence and development of tumours. The interplay between DNA damage repair and metabolism also has an effect on cisplatin resistance; the molecular changes to glucose metabolism, amino acid metabolism, lipid metabolism, and other metabolic pathways affect the cisplatin resistance of tumour cells. Here, we review the mechanism of action of cisplatin, the mechanism of resistance to cisplatin, the role of metabolic remodelling in tumorigenesis and development, and the effects of common metabolic pathways on cisplatin resistance.
Collapse
Affiliation(s)
- Lude Wang
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiaoya Zhao
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jianfei Fu
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Wenxia Xu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jianlie Yuan
- Department of Neurosurgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
36
|
Abstract
The GLUT is a key regulator of glucose metabolism and is widely expressed on the surface of most cells of the body. GLUT provides a variety of nutrients for the growth, proliferation and differentiation of cells. In recent years, the development of drugs affecting the energy intake of tumor cells has become a research hotspot. GLUT inhibitors are gaining increased attention because they can block the energy supply of malignant tumors. Herein, we elaborate on the structure and function of GLUT1, the structural and functional differences among GLUT1-4 transporters and the relationship between GLUT1 and tumor development, as well as GLUT1 transporter inhibitors, to provide a reference for the development of new GLUT1 inhibitors.
Collapse
|
37
|
van Gisbergen MW, Zwilling E, Dubois LJ. Metabolic Rewiring in Radiation Oncology Toward Improving the Therapeutic Ratio. Front Oncol 2021; 11:653621. [PMID: 34041023 PMCID: PMC8143268 DOI: 10.3389/fonc.2021.653621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
To meet the anabolic demands of the proliferative potential of tumor cells, malignant cells tend to rewire their metabolic pathways. Although different types of malignant cells share this phenomenon, there is a large intracellular variability how these metabolic patterns are altered. Fortunately, differences in metabolic patterns between normal tissue and malignant cells can be exploited to increase the therapeutic ratio. Modulation of cellular metabolism to improve treatment outcome is an emerging field proposing a variety of promising strategies in primary tumor and metastatic lesion treatment. These strategies, capable of either sensitizing or protecting tissues, target either tumor or normal tissue and are often focused on modulating of tissue oxygenation, hypoxia-inducible factor (HIF) stabilization, glucose metabolism, mitochondrial function and the redox balance. Several compounds or therapies are still in under (pre-)clinical development, while others are already used in clinical practice. Here, we describe different strategies from bench to bedside to optimize the therapeutic ratio through modulation of the cellular metabolism. This review gives an overview of the current state on development and the mechanism of action of modulators affecting cellular metabolism with the aim to improve the radiotherapy response on tumors or to protect the normal tissue and therefore contribute to an improved therapeutic ratio.
Collapse
Affiliation(s)
- Marike W van Gisbergen
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Dermatology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Emma Zwilling
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
38
|
lncRNA GAS6-AS1 inhibits progression and glucose metabolism reprogramming in LUAD via repressing E2F1-mediated transcription of GLUT1. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 25:11-24. [PMID: 34141461 PMCID: PMC8181633 DOI: 10.1016/j.omtn.2021.04.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
Glucose metabolism reprogramming is one of the hallmarks of cancer cells, although functional and regulatory mechanisms of long noncoding RNA (lncRNA) in the contribution of glucose metabolism in lung adenocarcinoma (LUAD) remain incompletely understood. The aim of this study was to uncover the role of GAS6-AS1 in the regulation of progression and glucose metabolism in LUAD. We discovered that overexpression of GAS6-AS1 suppressed tumor progression of LUAD both in vitro and in vivo. Metabolism-related assays revealed that GAS6-AS1 inhibited glucose metabolism reprogramming. Mechanically, GAS6-AS1 was found to repress the expression of glucose transporter GLUT1, a key regulator of glucose metabolism. Ectopic expression of GLUT1 restored the inhibition effect of GAS6-AS1 on cancer progression and glucose metabolism reprogramming. Further investigation identified that GAS6-AS1 directly interacted with transcription factor E2F1 and suppressed E2F1-mediated transcription of GLUT1, and GAS6-AS1 was downregulated in LUAD tissues and correlated with clinicopathological characteristics and survival of patients. Taken together, our results identified GAS6-AS1 as a novel tumor suppressor in LUAD and unraveled its underlying molecular mechanism in reprogramming glucose metabolism. GAS6-AS1 potentially may serve as a prognostic marker and therapeutic target in LUAD.
Collapse
|
39
|
Yu B, Zhang F, Liu L, Liang Y, Tang X, Peng Y, Cai F, Zeng D, Yuan X, Li J, Guo Y, Lv B, Wang M, Liao Q, Lv XB. The novel prognostic risk factor STC2 can regulate the occurrence and progression of osteosarcoma via the glycolytic pathway. Biochem Biophys Res Commun 2021; 554:25-32. [PMID: 33774276 DOI: 10.1016/j.bbrc.2021.03.067] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Osteosarcoma, a highly aggressive malignant tumor of the bone, usually occurs in children and young adults. However, although the considerable achievement in the clinical treatment of osteosarcoma recent years, the overall survival of osteosarcoma patients has not been obviously improved. Cancer cells preferentially use glycolysis instead of oxidative phosphorylation to meet their increased energetic and biosynthetic demands, a phenomenon known as the Warburg effect. Glycolysis is a driving factor in multiple cancers and is emerging as a new cancer target treatment. In the present study, we established a model to screen for glycolysis-associated genes in osteosarcoma. This risk score of the model were correlated with clinical characteristics osteosarcoma patients. Besides, a functional assay identified that STC2 enhanced the glycolysis of osteosarcoma cells. Modulation of STC2 changes glucose consumption and lactate production as well as GLUT1 expression in osteosarcoma. Furthermore, we identified that change in the expression levels of STC2 affected the proliferation, invasion, and migration of osteosarcoma cells. Our findings showed STC2 as a new tumor-promoting factor of osteosarcoma cells through enhancing glycolysis.
Collapse
Affiliation(s)
- Bo Yu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Feifei Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China
| | - Lang Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Yiping Liang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China
| | - Xiaofeng Tang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China
| | - Yuanxiang Peng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Feng Cai
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Duo Zeng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Xuhui Yuan
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Jiayu Li
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Yuhong Guo
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, 154 An Shan Road, Heping District, Tianjin, 300052, PR China
| | - Qi Liao
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China; Department of Orthopedics, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, Jiangxi, 330008, PR China.
| | - Xiao-Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, PR China.
| |
Collapse
|
40
|
Sun CX, Zhu F, Qi L. Demethylated miR-216a Regulates High Mobility Group Box 3 Promoting Growth of Esophageal Cancer Cells Through Wnt/β-Catenin Pathway. Front Oncol 2021; 11:622073. [PMID: 33842327 PMCID: PMC8025835 DOI: 10.3389/fonc.2021.622073] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background Esophageal cancer (EC) is the eighth most common cause of cancer-associated mortality in humans. Recent studies have revealed the important roles of microRNAs (miRs) in mediating tumor initiation and progression. miR-216a has been found to be involved in the progression of EC, but the underlying mechanisms remain largely unknown. The aim of this study is to explore the mechanism of miR-216a and the downstream molecules in esophageal cancer. Materials and Methods The degree of methylation of miR-216a promoter in EC tissues and cell lines was determined with methylation specific polymerase chain reaction (MSP). The levels of miR-216a and HMGB3 in EC cells were quantified by quantitative PCR (qPCR) and Western blot (WB). EC cell lines were treated with DNA methylation inhibitor 5-aza-2’-deoxycytidine (5-AZ), miR-216a mimics, and HMGB3 siRNA to explore the effects of miR-216a and HMGB3 on the proliferation, migration, invasion, and apoptosis of cells. Dual-luciferase reporter assay was employed to verify the binding of miR-216a to the 3’UTR of HMGB2 mRNA. Results The promoter of MiR-216a was hypermethylated and the expression of miR-216a was down-regulated in EC, while HMGB3 was up-regulated. Dual luciferase reporter assay confirmed the binding of miR-216a to the 3’UTR of HMGB3 mRNA. Demethylated miR-216a and miR-216a mimics elevated miR-216a expression and down-regulated HMGB3, as well as inhibited cell proliferation, migration, and invasion. Inhibiting the expression of HMGB3 played an important role in inducing apoptosis, suppressing cell expansion, and down-regulating the activity of Wnt/β-catenin pathway. Conclusions Hypermethylation in the promoter of miR-216a upregulated HMGB3 and the Wnt/β-catenin pathway, resulting in enhanced EC progression.
Collapse
Affiliation(s)
- Cheng-Xi Sun
- Department of Clinical Laboratory, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Feng Zhu
- Department of Thoracic Surgery, Shandong Provincial Chest Hospital, Jinan, China
| | - Lei Qi
- Department of Thoracic Surgery, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
41
|
Cotton S, Ferreira D, Soares J, Peixoto A, Relvas-Santos M, Azevedo R, Piairo P, Diéguez L, Palmeira C, Lima L, Silva AMN, Lara Santos L, Ferreira JA. Target Score-A Proteomics Data Selection Tool Applied to Esophageal Cancer Identifies GLUT1-Sialyl Tn Glycoforms as Biomarkers of Cancer Aggressiveness. Int J Mol Sci 2021; 22:ijms22041664. [PMID: 33562270 PMCID: PMC7915893 DOI: 10.3390/ijms22041664] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/28/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Esophageal cancer (EC) is a life-threatening disease, demanding the discovery of new biomarkers and molecular targets for precision oncology. Aberrantly glycosylated proteins hold tremendous potential towards this objective. In the current study, a series of esophageal squamous cell carcinomas (ESCC) and EC-derived circulating tumor cells (CTCs) were screened by immunoassays for the sialyl-Tn (STn) antigen, a glycan rarely expressed in healthy tissues and widely observed in aggressive gastrointestinal cancers. An ESCC cell model was glycoengineered to express STn and characterized in relation to cell proliferation and invasion in vitro. STn was found to be widely present in ESCC (70% of tumors) and in CTCs in 20% of patients, being associated with general recurrence and reduced survival. Furthermore, STn expression in ESCC cells increased invasion in vitro, while reducing cancer cells proliferation. In parallel, an ESCC mass spectrometry-based proteomics dataset, obtained from the PRIDE database, was comprehensively interrogated for abnormally glycosylated proteins. Data integration with the Target Score, an algorithm developed in-house, pinpointed the glucose transporter type 1 (GLUT1) as a biomarker of poor prognosis. GLUT1-STn glycoproteoforms were latter identified in tumor tissues in patients facing worst prognosis. Furthermore, healthy human tissues analysis suggested that STn glycosylation provided cancer specificity to GLUT1. In conclusion, STn is a biomarker of worst prognosis in EC and GLUT1-STn glycoforms may be used to increase its specificity on the stratification and targeting of aggressive ESCC forms.
Collapse
Affiliation(s)
- Sofia Cotton
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal
| | - Dylan Ferreira
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal
| | - Janine Soares
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- QOPNA/LAQV, Department of Chemistry, Campus Universitário de Santiago, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Andreia Peixoto
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal
| | - Marta Relvas-Santos
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute for Biomedical Engineering (INEB), 4200-135 Porto, Portugal
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal;
| | - Rita Azevedo
- Institute of Biomedicine, University of Turku, FI-20014 Turku, Finland;
| | - Paulina Piairo
- Medical Devices Research Group, International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal; (P.P.); (L.D.)
| | - Lorena Diéguez
- Medical Devices Research Group, International Iberian Nanotechnology Laboratory (INL), 4715-330 Braga, Portugal; (P.P.); (L.D.)
| | - Carlos Palmeira
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Department of Immunology, Portuguese Institute of Oncology of Porto, 4200-072 Porto, Portugal
- Health Science Faculty, University of Fernando Pessoa, 4249-004 Porto, Portugal
| | - Luís Lima
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal
| | - André M. N. Silva
- REQUIMTE-LAQV, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal;
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Health Science Faculty, University of Fernando Pessoa, 4249-004 Porto, Portugal
- Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal
- Department of Surgical Oncology, Portuguese Institute of Oncology, 4200-072 Porto, Portugal
| | - José Alexandre Ferreira
- Experimental Pathology and Therapeutics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute (IPO Porto), 4200-072 Porto, Portugal; (S.C.); (D.F.); (J.S.); (A.P.); (M.R.-S.); (C.P.); (L.L.); (L.L.S.)
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Porto Comprehensive Cancer Center (P.ccc), 4200-072 Porto, Portugal
- Correspondence: ; Tel.: +351-225-084-000 (ext. 5111)
| |
Collapse
|
42
|
Tu MJ, Duan Z, Liu Z, Zhang C, Bold RJ, Gonzalez FJ, Kim EJ, Yu AM. MicroRNA-1291-5p Sensitizes Pancreatic Carcinoma Cells to Arginine Deprivation and Chemotherapy through the Regulation of Arginolysis and Glycolysis. Mol Pharmacol 2020; 98:686-694. [PMID: 33051382 PMCID: PMC7673485 DOI: 10.1124/molpharm.120.000130] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cells are dysregulated and addicted to continuous supply and metabolism of nutritional glucose and amino acids (e.g., arginine) to drive the synthesis of critical macromolecules for uncontrolled growth. Recent studies have revealed that genome-derived microRNA (miRNA or miR)-1291-5p (miR-1291-5p or miR-1291) may modulate the expression of argininosuccinate synthase (ASS1) and glucose transporter protein type 1 (GLUT1). We also developed a novel approach to produce recombinant miR-1291 agents for research, which are distinguished from conventional chemo-engineered miRNA mimics. Herein, we firstly demonstrated that bioengineered miR-1291 agent was selectively processed to high levels of target miR-1291-5p in human pancreatic cancer (PC) cells. After the suppression of ASS1 protein levels, miR-1291 perturbed arginine homeostasis and preferably sensitized ASS1-abundant L3.3 cells to arginine deprivation therapy. In addition, miR-1291 treatment reduced the protein levels of GLUT1 in both AsPC-1 and PANC-1 cells, leading to a lower glucose uptake (deceased > 40%) and glycolysis capacity (reduced approximately 50%). As a result, miR-1291 largely improved cisplatin efficacy in the inhibition of PC cell viability. Our results demonstrated that miR-1291 was effective to sensitize PC cells to arginine deprivation treatment and chemotherapy through targeting ASS1- and GLUT1-mediated arginolysis and glycolysis, respectively, which may provide insights into understanding miRNA signaling underlying cancer cell metabolism and development of new strategies for the treatment of lethal PC. SIGNIFICANCE STATEMENT: Many anticancer drugs in clinical use and under investigation exert pharmacological effects or improve efficacy of coadministered medications by targeting cancer cell metabolism. Using new recombinant miR-1291 agent, we revealed that miR-1291 acts as a metabolism modulator in pancreatic carcinoma cells through the regulation of argininosuccinate synthase- and glucose transporter protein type 1-mediated arginolysis and glycolysis. Consequently, miR-1291 effectively enhanced the efficacy of arginine deprivation (pegylated arginine deiminase) and chemotherapy (cisplatin), offering new insights into development of rational combination therapies.
Collapse
Affiliation(s)
- Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Zhijian Duan
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Zhenzhen Liu
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Chao Zhang
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Richard J Bold
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Frank J Gonzalez
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Edward J Kim
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine (M.-J.T., Z.D., Z.L., C.Z., A.-M.Y.), Division of Surgical Oncology (R.J.B.), Division of Hematology and Oncology, Department of Internal Medicine (E.J.K.), University of California (UC) Davis School of Medicine, Sacramento, California; and Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.)
| |
Collapse
|
43
|
Tilekar K, Upadhyay N, Iancu CV, Pokrovsky V, Choe JY, Ramaa CS. Power of two: combination of therapeutic approaches involving glucose transporter (GLUT) inhibitors to combat cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188457. [PMID: 33096154 PMCID: PMC7704680 DOI: 10.1016/j.bbcan.2020.188457] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022]
Abstract
Cancer research of the Warburg effect, a hallmark metabolic alteration in tumors, focused attention on glucose metabolism whose targeting uncovered several agents with promising anticancer effects at the preclinical level. These agents' monotherapy points to their potential as adjuvant combination therapy to existing standard chemotherapy in human trials. Accordingly, several studies on combining glucose transporter (GLUT) inhibitors with chemotherapeutic agents, such as doxorubicin, paclitaxel, and cytarabine, showed synergistic or additive anticancer effects, reduced chemo-, radio-, and immuno-resistance, and reduced toxicity due to lowering the therapeutic doses required for desired chemotherapeutic effects, as compared with monotherapy. The combinations have been specifically effective in treating cancer glycolytic phenotypes, such as pancreatic and breast cancers. Even combining GLUT inhibitors with other glycolytic inhibitors and energy restriction mimetics seems worthwhile. Though combination clinical trials are in the early phase, initial results are intriguing. The various types of GLUTs, their role in cancer progression, GLUT inhibitors, and their anticancer mechanism of action have been reviewed several times. However, utilizing GLUT inhibitors as combination therapeutics has received little attention. We consider GLUT inhibitors agents that directly affect glucose transporters by binding to them or indirectly alter glucose transport by changing the transporters' expression level. This review mainly focuses on summarizing the effects of various combinations of GLUT inhibitors with other anticancer agents and providing a perspective on the current status.
Collapse
Affiliation(s)
- Kalpana Tilekar
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Neha Upadhyay
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| | - Cristina V. Iancu
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - Vadim Pokrovsky
- Laboratory of Combined Therapy, N.N. Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People’s Friendship University, Moscow, Russia
| | - Jun-yong Choe
- East Carolina Diabetes and Obesity Institute, Department of Chemistry, East Carolina University, Greenville, North Carolina, USA
| | - C. S. Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth’s College of Pharmacy, Navi Mumbai, Maharashtra, India
| |
Collapse
|
44
|
Zhang H, Qi J, Wang Y, Sun J, Li Z, Sui L, Fan J, Liu C, Shang Y, Kong L, Kong Y. Progesterone Regulates Glucose Metabolism Through Glucose Transporter 1 to Promote Endometrial Receptivity. Front Physiol 2020; 11:543148. [PMID: 33101047 PMCID: PMC7546771 DOI: 10.3389/fphys.2020.543148] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Successful embryo implantation requires receptive endometrium, which is conducive to the process of embryo recognition, adhesion, and invasion within a certain period of time and is inseparable from the dynamic interaction between 17β-estradiol (E2) and progesterone (P4). Proper glucose metabolism is critical for the profound physiological changes in the endometrium entering the receptive state. And glucose transporters (GLUTs) are responsible for intracellular uptake of glucose and are the first step in glucose metabolism. Prior literature has reported the presence of GLUTs in the endometrium. However, we still do not understand the specific mechanisms of this process. In this study, we identified the effect of P4 on glucose transporter 1 (GLUT1) using in vivo animal models and determined the regulation of glucose metabolism by P4 in cells. We highly suspect that this pregnancy failure may be due to reduced GLUT1-mediated glucose metabolism, resulting in a decrease in endometrial receptivity caused by an inadequate energy supply and synthesis of substrate. Here, we propose a possible mechanism to explain how embryo implantation is affected by P4 and glucose utilization under abnormal endometrial conditions.
Collapse
Affiliation(s)
- Hongshuo Zhang
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jia Qi
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yufei Wang
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jing Sun
- Department of Ultrasound, Xijing Hospital, Xi'an, China
| | - Zhen Li
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Linlin Sui
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jianhui Fan
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chao Liu
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Yuhong Shang
- Department of Gynecology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li Kong
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ying Kong
- Core Laboratory of Glycobiology and Glycoengineering, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
45
|
Chen H, Zheng B, Xue S, Chen C. Knockdown of miR-183 Enhances the Cisplatin-Induced Apoptosis in Esophageal Cancer Through Increase of FOXO1 Expression. Onco Targets Ther 2020; 13:8463-8474. [PMID: 32943877 PMCID: PMC7468590 DOI: 10.2147/ott.s258680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
Background As an important member of platinum-based chemotherapeutic drugs, cisplatin is effective and is commonly used in the treatment of esophageal cancer. However, repeated use of cisplatin usually causes severe side-effects on patients. Novel approaches should be explored to increase the sensitivity of cancer cells to cisplatin. Methods The expression level of miR-183 in esophageal cancer tissues and cell lines was measured by quantitative reverse transcriptase real-time PCR (qRT-PCR). The sensitivity of EC cell lines to cisplatin was evaluated by CCK-8 assay and flow cytometry. Luciferase reporter assay was used to confirm the association between miR-183 and FOXO1. The apoptosis pathway of EC cells was tested by Western blot assay. Results The expression level of miR-183 was increased in esophageal cancer patients' tumor tissues and esophageal cancer cell lines. However, knockdown of miR-183 was found to enhance the effect of cisplatin on inducing the apoptotic cell death of esophageal cancer cells. In the mechanism research, we proved that FOXO1 was the target of miR-183 in esophageal cancer cells. Inhibition of miR-183 increased the expression of FOXO1 to promote the expression of Bim and Noxa. As Bim and Noxa acted as key pro-apoptotic proteins in mitochondrial apoptosis, inhibition of miR-183 enhanced the cisplatin-induced apoptosis pathway in esophageal cancer. Conclusion Knockdown of miR-183 enhanced the cisplatin-induced apoptosis in esophageal cancer through an increase of FOXO1 expression.
Collapse
Affiliation(s)
- Hao Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou City, Fujian Province 350000, People's Republic of China
| | - Bin Zheng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou City, Fujian Province 350000, People's Republic of China
| | - Songtao Xue
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou City, Fujian Province 350000, People's Republic of China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou City, Fujian Province 350000, People's Republic of China
| |
Collapse
|
46
|
Varghese E, Samuel SM, Líšková A, Samec M, Kubatka P, Büsselberg D. Targeting Glucose Metabolism to Overcome Resistance to Anticancer Chemotherapy in Breast Cancer. Cancers (Basel) 2020; 12:E2252. [PMID: 32806533 PMCID: PMC7464784 DOI: 10.3390/cancers12082252] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023] Open
Abstract
Breast cancer (BC) is the most prevalent cancer in women. BC is heterogeneous, with distinct phenotypical and morphological characteristics. These are based on their gene expression profiles, which divide BC into different subtypes, among which the triple-negative breast cancer (TNBC) subtype is the most aggressive one. The growing interest in tumor metabolism emphasizes the role of altered glucose metabolism in driving cancer progression, response to cancer treatment, and its distinct role in therapy resistance. Alterations in glucose metabolism are characterized by increased uptake of glucose, hyperactivated glycolysis, decreased oxidative phosphorylation (OXPHOS) component, and the accumulation of lactate. These deviations are attributed to the upregulation of key glycolytic enzymes and transporters of the glucose metabolic pathway. Key glycolytic enzymes such as hexokinase, lactate dehydrogenase, and enolase are upregulated, thereby conferring resistance towards drugs such as cisplatin, paclitaxel, tamoxifen, and doxorubicin. Besides, drug efflux and detoxification are two energy-dependent mechanisms contributing to resistance. The emergence of resistance to chemotherapy can occur at an early or later stage of the treatment, thus limiting the success and outcome of the therapy. Therefore, understanding the aberrant glucose metabolism in tumors and its link in conferring therapy resistance is essential. Using combinatory treatment with metabolic inhibitors, for example, 2-deoxy-D-glucose (2-DG) and metformin, showed promising results in countering therapy resistance. Newer drug designs such as drugs conjugated to sugars or peptides that utilize the enhanced expression of tumor cell glucose transporters offer selective and efficient drug delivery to cancer cells with less toxicity to healthy cells. Last but not least, naturally occurring compounds of plants defined as phytochemicals manifest a promising approach for the eradication of cancer cells via suppression of essential enzymes or other compartments associated with glycolysis. Their benefits for human health open new opportunities in therapeutic intervention, either alone or in combination with chemotherapeutic drugs. Importantly, phytochemicals as efficacious instruments of anticancer therapy can suppress events leading to chemoresistance of cancer cells. Here, we review the current knowledge of altered glucose metabolism in contributing to resistance to classical anticancer drugs in BC treatment and various ways to target the aberrant metabolism that will serve as a promising strategy for chemosensitizing tumors and overcoming resistance in BC.
Collapse
Affiliation(s)
- Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (E.V.); (S.M.S.)
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (E.V.); (S.M.S.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (A.L.); (M.S.)
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (A.L.); (M.S.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar; (E.V.); (S.M.S.)
| |
Collapse
|
47
|
Shao QP, Wei C, Yang J, Zhang WZ. miR-3609 Decelerates the Clearance of Sorafenib in Hepatocellular Carcinoma Cells by Targeting EPAS-1 and Reducing the Activation of the Pregnane X Receptor Pathway. Onco Targets Ther 2020; 13:7213-7227. [PMID: 32801751 PMCID: PMC7394586 DOI: 10.2147/ott.s246471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Background The pregnane X receptor (PXR) not only plays an important role in cellular metabolism processes but also induces the resistance of hepatocellular carcinoma (HCC) cells to molecularly targeted drugs by mediating their metabolism and clearance by these cells. Endothelial PAS domain-containing protein 1 (EPAS-1) acts as a coactivator to regulate the transcription factor activity of PXR. In the present study, a microRNA that potentially targets EPAS-1, namely miR-3609, was identified using the miRDB tool. Methods The expression of miR-3609 and EPAS-1 was examined by qPCR. Lentiviral particles containing the full-length sequences of miR-3609 (pri-miR-3609) were prepared. The antitumor effect of antitumor agents was examined by the in vitro and in vivo assays. Results The expression of miR-3609 was negatively correlated with that of EPAS-1 in both HCC clinical specimens and paired non-tumor specimens, and the effect of miR-3609 on the expression of EPAS-1 was confirmed by Western blot experiments. Overexpression of miR-3609 decreased the expression of EPAS-1 and, in turn, repressed the activation of the PXR pathway. miR-3609 decreased the transcription factor activation of PXR, repressed its recruitment to its target gene promoter regions, and decreased the expression of its target genes CYP3A4 and P-GP. In addition, miR-3609 decelerated the metabolism and clearance of sorafenib in HCC cells and enhanced the antitumor effect of sorafenib in HCC cells. Conclusion Therefore, the results indicate that miR-3609 decreases the expression of EPAS-1 and enhances the sensitivity of HCC cells to sorafenib.
Collapse
Affiliation(s)
- Qing-Ping Shao
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province 450008, People's Republic of China
| | - Chen Wei
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, People's Republic of China
| | - Jie Yang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province 450052, People's Republic of China
| | - Wen-Zhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province 450008, People's Republic of China
| |
Collapse
|
48
|
Bednarz-Misa I, Fortuna P, Diakowska D, Jamrozik N, Krzystek-Korpacka M. Distinct Local and Systemic Molecular Signatures in the Esophageal and Gastric Cancers: Possible Therapy Targets and Biomarkers for Gastric Cancer. Int J Mol Sci 2020; 21:ijms21124509. [PMID: 32630408 PMCID: PMC7349922 DOI: 10.3390/ijms21124509] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric (GC) and esophageal (EC) cancers are highly lethal. Better understanding of molecular abnormalities is needed for new therapeutic targets and biomarkers to be found. Expression of 18 cancer-related genes in 31 paired normal-tumor samples was quantified by reversely-transcribed quantitative polymerase chain reaction (RTqPCR) and systemic concentration of 27 cytokines/chemokines/growth factors in 195 individuals was determined using Luminex xMAP technology. Only Ki67, CLDN2, and BCLxL were altered in GC while Ki67, CDKN1A, ODC1, SLC2A1, HIF1A, VEGFA, NOS2, CCL2, PTGS2, IL10, IL10Ra, and ACTA2 were changed in EC. The relatively unaltered molecular GC landscape resulted from high expression of BCLxL, CDKN1A, BCL2, Ki67, HIF1A, VEGFA, ACTA2, TJP1, CLDN2, IL7Ra, ODC1, PTGS2, and CCL2 in non-cancerous tissue. The NOS2 expression and IL-4, IL-9, FGF2, and RANTES secretion were higher in cardiac than non-cardiac GC. Four-cytokine panels (interleukin (IL)-1β/IL-1ra/IL-6/RANTES or IL-1β/IL-6/IL-4/IL-13) differentiated GC from benign conditions with 87–89% accuracy. Our results showed increased proliferative, survival, inflammatory and angiogenic capacity in gastric tumor-surrounding tissue, what might contribute to GC aggressiveness and facilitate cancer recurrence. Further studies are needed to determine the CLDN2 and NOS2 suitability as candidate molecular targets in GC and cardiac GC, respectively, and discern the role of CLDN2 or to verify IL-1β/IL-1ra/IL-6/RANTES or IL-1β/IL-6/IL-4/IL-13 usefulness as differential biomarkers.
Collapse
Affiliation(s)
- Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (N.J.)
| | - Paulina Fortuna
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (N.J.)
| | - Dorota Diakowska
- Department of Gastrointestinal and General Surgery, Wroclaw Medical University, 50-368 Wroclaw, Poland;
- Department of Nervous System Diseases, Wroclaw Medical University, 51-618 Wroclaw, Poland
| | - Natalia Jamrozik
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (N.J.)
| | - Małgorzata Krzystek-Korpacka
- Department of Medical Biochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (I.B.-M.); (P.F.); (N.J.)
- Correspondence:
| |
Collapse
|
49
|
Liu XS, Yuan LL, Gao Y, Zhou LM, Yang JW, Pei ZJ. Overexpression of METTL3 associated with the metabolic status on 18F-FDG PET/CT in patients with Esophageal Carcinoma. J Cancer 2020; 11:4851-4860. [PMID: 32626532 PMCID: PMC7330681 DOI: 10.7150/jca.44754] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background: To investigate the expression of methyltransferase 3 (METTL3) and its relationship with 18F-FDG uptake in patients with esophageal carcinoma (ESCA). Materials and methods: This study analyzed the expression of METTL3 in ESCA and its relationship with clinicopathological features by The Cancer Genome Atlas (TCGA) database. Immunohistochemical staining was performed on 57 tumor tissues of ESCA patients who underwent PET/CT scan before surgery to evaluate the expression of METTL3, glucose transporter 1 (GLUT1), and hexokinase 2 (HK2) in tumor tissues and peritumoral tissues. Analyze the relationship between SUVmax with METTL3, HK2, and GLUT1 expression. Results: The expression of METTL3, GLUT1, and HK2 was significantly increased in ESCA tissues compared with normal tissues (p < 0.001). The expression of METTL3 was correlated with tumor size and histological differentiation (p < 0.05), and there was no significant difference between age, sex, pathological types, tumor staging, or lymph node metastasis (p > 0.05). The SUVmax was significantly higher in tumors with high METTL3 expression (17.822±6.249) compared to low METTL3 expression (9.573±5.082) (p < 0.001). There was a positive correlation between the SUVmax and METTL3 expression in ESCA (r2 = 0.647, p < 0.001). Multivariate analysis confirmed the association between SUVmax and METTL3 expression (p < 0.05). GLUT1 and HK2 expression in ESCA was positively correlated with 18F-FDG uptake and METTL3 status (p < 0.001). Conclusions: The high expression of METTL3 is related to the high SUVmax in ESCA, and METTL3 may increase 18F-FDG uptake by regulating GLUT1 and HK2.
Collapse
Affiliation(s)
- Xu-Sheng Liu
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 44200, China
| | - Ling-Ling Yuan
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Yan Gao
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 44200, China
| | - Lu-Meng Zhou
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 44200, China
| | - Jian-Wei Yang
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 44200, China
| | - Zhi-Jun Pei
- Department of Nuclear Medicine and Institute of Anesthesiology and Pain, Taihe Hospital, Hubei University of Medicine, Shiyan, 44200, China.,Hubei Key Laboratory of WudangLocal Chinese Medicine Research, Shiyan, 442000, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, 442000, China
| |
Collapse
|
50
|
Choi SH, Jin CC, Do SK, Lee SY, Choi JE, Kang HG, Kim JH, Lee JH, Hong MJ, Lee WK, Jeong JY, Shin KM, Lee YH, Seo H, Yoo SS, Lee J, Cha SI, Kim CH, Park JY. Polymorphisms in Glycolysis-Related Genes Are Associated with Clinical Outcomes of Paclitaxel-Cisplatin Chemotherapy in Non-Small Cell Lung Cancer. Oncology 2020; 98:468-477. [PMID: 32252059 DOI: 10.1159/000504175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/03/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study was conducted to investigate whether polymorphisms in glycolysis-related genes are associated with clinical outcomes of patients with advanced-stage non-small cell lung cancer (NSCLC) undergoing chemotherapy. METHODS A total of 377 patients with NSCLC were enrolled. Sixty-five single-nucleotide polymorphisms in 26 genes involved in the glycolytic pathway were evaluated. The associations of the variants with the chemotherapy response and overall survival (OS) were analyzed. RESULTS Among the 65 variants investigated, PFKL rs2073436C>G and GPI rs7248411C>G significantly correlated with clinical outcomes after chemotherapy in multivariate analyses. PFKL rs2073436C>G was significantly associated with both a worse response to chemotherapy (adjusted odds ratio [aOR] = 0.64, 95% CI = 0.45-0.90, p = 0.01) and a worse OS (adjusted hazard ratio [aHR] = 1.35, 95% CI = 1.14-1.61, p = 0.001). GPI rs7248411C>G was significantly associated with both a better chemotherapy response (aOR = 1.58, 95% CI = 1.07-2.23, p = 0.02) and a better OS (aHR = 0.80, 95% CI = 0.66-0.98, p = 0.03). When stratified by tumor histology, PFKL rs2073436C>G was significantly associated with OS only in squamous cell carcinoma, whereas GPI rs7248411C>G exhibited a significant association with the chemotherapy response and OS only in adenocarcinoma. CONCLUSION This result suggests that the PFKL rs2073436C>G and GPI rs7248411C>G are useful for predicting the clinical outcome of first-line paclitaxel-cisplatin chemotherapy in NSCLC.
Collapse
Affiliation(s)
- Sun Ha Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Cheng Cheng Jin
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Sook Kyung Do
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Jin Eun Choi
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyo-Gyoung Kang
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Hyun Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Jang Hyuck Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Mi Jeong Hong
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Won Kee Lee
- Medical Research Collaboration Center in Kyungpook National University Hospital and School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung Min Shin
- Department of Radiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Hoon Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyewon Seo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Soo Yoo
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Jaehee Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Ick Cha
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Ho Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae Yong Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea, .,Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea, .,Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea, .,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea, .,Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea,
| |
Collapse
|