1
|
Alfandari A, Moskovich D, Weisz A, Katzav A, Kidron D, Beiner M, Josephy D, Asali A, Hants Y, Yagur Y, Weitzner O, Ellis M, Itchaki G, Ashur‐Fabian O. The selenoenzyme type I iodothyronine deiodinase: a new tumor suppressor in ovarian cancer. Mol Oncol 2024; 18:2298-2313. [PMID: 38429887 PMCID: PMC11467794 DOI: 10.1002/1878-0261.13612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/26/2023] [Accepted: 02/05/2024] [Indexed: 03/03/2024] Open
Abstract
The selenoenzyme type I iodothyronine deiodinase (DIO1) catalyzes removal of iodine atoms from thyroid hormones. Although DIO1 action is reported to be disturbed in several malignancies, no work has been conducted in high-grade serous ovarian carcinoma (HGSOC), the most lethal gynecologic cancer. We studied DIO1 expression in HGSOC patients [The Cancer Genome Atlas (TCGA) data and tumor tissues], human cell lines (ES-2 and Kuramochi), normal Chinese hamster ovarian cells (CHO-K1), and normal human fallopian tube cells (FT282 and FT109). To study its functional role, DIO1 was overexpressed, inhibited [by propylthiouracil (PTU)], or knocked down (KD), and cell count, proliferation, apoptosis, cell viability, and proteomics analysis were performed. Lower DIO1 levels were observed in HGSOC compared to normal cells and tissues. TCGA analyses confirmed that low DIO1 mRNA expression correlated with worse survival and therapy resistance in patients. Silencing or inhibiting the enzyme led to enhanced ovarian cancer proliferation, while an opposite effect was shown following DIO1 ectopic expression. Proteomics analysis in DIO1-KD cells revealed global changes in proteins that facilitate tumor metabolism and progression. In conclusion, DIO1 expression and ovarian cancer progression are inversely correlated, highlighting a tumor suppressive role for this enzyme and its potential use as a biomarker in this disease.
Collapse
Affiliation(s)
- Adi Alfandari
- Translational Oncology LaboratoryHematology Institute, Meir Medical CenterKfar SabaIsrael
- Department of Human Molecular Genetics and Biochemistry, Faculty of MedicineTel Aviv UniversityIsrael
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| | - Dotan Moskovich
- Translational Oncology LaboratoryHematology Institute, Meir Medical CenterKfar SabaIsrael
- Department of Human Molecular Genetics and Biochemistry, Faculty of MedicineTel Aviv UniversityIsrael
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| | - Avivit Weisz
- Department of PathologyMeir Medical CenterKfar SabaIsrael
| | - Aviva Katzav
- Department of PathologyMeir Medical CenterKfar SabaIsrael
| | - Debora Kidron
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
- Department of PathologyMeir Medical CenterKfar SabaIsrael
| | - Mario Beiner
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
- Division of Gynecologic OncologyMeir Medical CenterKfar SabaIsrael
| | - Dana Josephy
- Division of Gynecologic OncologyMeir Medical CenterKfar SabaIsrael
| | - Aula Asali
- Division of Gynecologic OncologyMeir Medical CenterKfar SabaIsrael
| | - Yael Hants
- Division of Gynecologic OncologyMeir Medical CenterKfar SabaIsrael
| | - Yael Yagur
- Department of Obstetrics and GynecologyMeir Medical CenterKfar SabaIsrael
| | - Omer Weitzner
- Department of Obstetrics and GynecologyMeir Medical CenterKfar SabaIsrael
| | - Martin Ellis
- Translational Oncology LaboratoryHematology Institute, Meir Medical CenterKfar SabaIsrael
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| | - Gilad Itchaki
- Translational Oncology LaboratoryHematology Institute, Meir Medical CenterKfar SabaIsrael
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| | - Osnat Ashur‐Fabian
- Translational Oncology LaboratoryHematology Institute, Meir Medical CenterKfar SabaIsrael
- Department of Human Molecular Genetics and Biochemistry, Faculty of MedicineTel Aviv UniversityIsrael
- School of Medicine, Faculty of Medical and Health SciencesTel Aviv UniversityIsrael
| |
Collapse
|
2
|
Xu Y, Ni F, Sun D, Peng Y, Zhao Y, Wu X, Li S, Qi X, He X, Li M, Zhou Y, Zhang C, Yan M, Yao C, Zhu S, Yang Y, An B, Yang C, Zhang G, Jiang W, Mi J, Chen X, Wei P, Tian G, Zhang Y. Glucagon Enhances Chemotherapy Efficacy By Inhibition of Tumor Vessels in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307271. [PMID: 38072640 PMCID: PMC10853751 DOI: 10.1002/advs.202307271] [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] [Received: 09/30/2023] [Revised: 11/19/2023] [Indexed: 02/10/2024]
Abstract
Chemotherapy is widely used to treat colorectal cancer (CRC). Despite its substantial benefits, the development of drug resistance and adverse effects remain challenging. This study aimed to elucidate a novel role of glucagon in anti-cancer therapy. In a series of in vitro experiments, glucagon inhibited cell migration and tube formation in both endothelial and tumor cells. In vivo studies demonstrated decreased tumor blood vessels and fewer pseudo-vessels in mice treated with glucagon. The combination of glucagon and chemotherapy exhibited enhanced tumor inhibition. Mechanistic studies demonstrated that glucagon increased the permeability of blood vessels, leading to a pronounced disruption of vessel morphology. Signaling pathway analysis identified a VEGF/VEGFR-dependent mechanism whereby glucagon attenuated angiogenesis through its receptor. Clinical data analysis revealed a positive correlation between elevated glucagon expression and chemotherapy response. This is the first study to reveal a role for glucagon in inhibiting angiogenesis and vascular mimicry. Additionally, the delivery of glucagon-encapsulated PEGylated liposomes to tumor-bearing mice amplified the inhibition of angiogenesis and vascular mimicry, consequently reinforcing chemotherapy efficacy. Collectively, the findings demonstrate the role of glucagon in inhibiting tumor vessel network and suggest the potential utility of glucagon as a promising predictive marker for patients with CRC receiving chemotherapy.
Collapse
|
3
|
Xing Z, Gao Y, Shi Y, Gao Z, Liu C. Inhibition of PI3K/Akt/mTOR Signaling Pathway Suppresses 5-Fluorouracil Resistance in Gastric Cancer. Mol Biotechnol 2023:10.1007/s12033-023-00966-x. [PMID: 37999920 DOI: 10.1007/s12033-023-00966-x] [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: 09/08/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND At present, 5-Fluorouracil (5-FU) is a crucial anti-cancer drug and is widely used for the treatment of various carcinomas, including gastric cancer (GC). The resistance of GC cells to 5-FU is still a matter of great concern. OBJECTIVE To illustrate the role of PI3K/Akt/mTOR signaling in regulating the cell cycle progression and migration of 5-FU-resistant GC cells. MATERIAL AND METHODS After the establishment of drug-resistant GC cell lines, the effects of 5-FU and/or BEZ235 (the dual inhibitor of PI3K and mTOR) on the activity of parental or drug-resistant GC cells were explored. The viability and localization of GC cells (MKN-45 and MKN-74) and their drug-resistant cells (MKN-45/R and MKN-74/R) were assessed using MTT assays and immunofluorescence staining. The impacts of 5-FU and/or BEZ235 on GC cell cycle progression and cell migration were assessed via flow cytometry analyses and wound healing assays, respectively. GC tissues were collected from patients with GC sensitive or refractory to 5-FU chemotherapy. RT-qPCR and western blot were conducted to measure PI3K, AKT, and mTOR levels in GC cells or tissues. RESULTS After 5-FU treatment, GC cells displayed 5-FU resistance and the viability of drug-resistant cells (MKN-45/R and MKN-74/R) was higher than that of parental cells (MKN-45 and MKN-74). The IC50 values for MKN-45 and MKN-45/R were 8.93 ug/ml and 140 ug/ml, and the values for MKN-74 and MKN-74/R were 3.93 ug/ml and 114.29 ug/ml. Additionally, the PI3K/Akt/mTOR signaling pathway was activated in drug-resistant GC cells and tumor tissues of patients refractory to 5-FU chemotherapy, as evidenced by high PI3K, Akt, and mTOR levels in MKN-45/R, MKN-74/R, and GC tissues resistant to 5-FU. BEZ235 promoted cell cycle arrest and suppressed the migration of GC cells. Moreover, the combination of BEZ235 and 5-FU led to more effective suppressive influence on cell cycle progression and cell migration relative to the single 5-FU or BEZ235 treatment. CONCLUSIONS Silencing of the PI3K/Akt/mTOR signaling pathway suppressed the 5-FU resistance of GC cells.
Collapse
Affiliation(s)
- Zhiwei Xing
- Department of Oncology, Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Road, Huimin District, Hohhot, 010010, Inner Mongolia Autonomous Region, China
| | - Yanan Gao
- Graduate School of Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia Autonomous Region, China
| | - Yaxuan Shi
- Graduate School of Inner Mongolia Medical University, Hohhot, 010010, Inner Mongolia Autonomous Region, China
| | - Ziyu Gao
- Department of Oncology, Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Road, Huimin District, Hohhot, 010010, Inner Mongolia Autonomous Region, China
| | - Caixia Liu
- Department of Oncology, Affiliated Hospital of Inner Mongolia Medical University, No.1, Tongdao North Road, Huimin District, Hohhot, 010010, Inner Mongolia Autonomous Region, China.
| |
Collapse
|
4
|
Stange K, Schumacher T, Miersch C, Whelan R, Klünemann M, Röntgen M. Methionine Sources Differently Affect Production of Reactive Oxygen Species, Mitochondrial Bioenergetics, and Growth of Murine and Quail Myoblasts In Vitro. Curr Issues Mol Biol 2023; 45:2661-2680. [PMID: 37185698 PMCID: PMC10136669 DOI: 10.3390/cimb45040174] [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: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023] Open
Abstract
An optimal supply of L-methionine (L-Met) improves muscle growth, whereas over-supplementation exerts adverse effects. To understand the underlying mechanisms, this study aims at exploring effects on the growth, viability, ROS production, and mitochondrial bioenergetics of C2C12 (mouse) and QM7 (quail) myoblasts additionally supplemented (100 or 1000 µM) with L-Met, DL-methionine (DL-Met), or DL-2-hydroxy-4-(methylthio)butanoic acid (DL-HMTBA). In both cell lines, all the supplements stimulated cell growth. However, in contrast to DL-Met, 1000 µM of L-Met (C2C12 cells only) or DL-HMTBA started to retard growth. This negative effect was stronger with DL-HMTBA and was accompanied by significantly elevated levels of extracellular H2O2, an indicator for OS, in both cell types. In addition, oversupplementation with DL-HMTBA (1000 µM) induced adaptive responses in mitochondrial bioenergetics, including reductions in basal (C2C12 and QM7) and ATP-synthase-linked (C2C12) oxygen consumption, maximal respiration rate, and reserve capacity (QM7). Only QM7 cells switched to nonmitochondrial aerobic glycolysis to reduce ROS production. In conclusion, we found a general negative effect of methionine oversupplementation on cell proliferation. However, only DL-HMTBA-induced growth retardation was associated with OS and adaptive, species-specific alterations in mitochondrial functionality. OS could be better compensated by quail cells, highlighting the role of species differences in the ability to cope with methionine oversupplementation.
Collapse
Affiliation(s)
- Katja Stange
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Toni Schumacher
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | - Claudia Miersch
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
- Nutritional Physiology and Dietetics, International University of Applied Sciences (IU), Juri-Gagarin-Ring 152, 99084 Erfurt, Germany
| | - Rose Whelan
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany
| | - Martina Klünemann
- Evonik Operations GmbH, Rodenbacher Chaussee 4, 63457 Hanau, Germany
| | - Monika Röntgen
- Institute of Muscle Biology and Growth, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| |
Collapse
|
5
|
Singh V. F 1F o adenosine triphosphate (ATP) synthase is a potential drug target in non-communicable diseases. Mol Biol Rep 2023; 50:3849-3862. [PMID: 36715790 DOI: 10.1007/s11033-023-08299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023]
Abstract
F1Fo adenosine triphosphate (ATP) synthase, also known as the complex V, is the central ATP-producing unit in the cells arranged in the mitochondrial and plasma membranes. F1Fo ATP synthase also regulates the central metabolic processes in the human body driven by proton motive force (Δp). Numerous studies have immensely contributed toward highlighting its regulation in improving energy homeostasis and maintaining mitochondrial integrity, which otherwise gets compromised in illnesses. Yet, its role in the implication of non-communicable diseases remains unknown. F1Fo ATP synthase dysregulation at gene level leads to reduced activity and delocalization in the cristae and plasma membranes, which is directly associated with non-communicable diseases: cardiovascular diseases, diabetes, neurodegenerative disorders, cancer, and renal diseases. Individual subunits of the F1Fo ATP synthase target ligand-based competitive or non-competitive inhibition. After performing a systematic literature review to understand its specific functions and its novel drug targets, the present article focuses on the central role of F1Fo ATP synthase in primary non-communicable diseases. Next, it discusses its involvement through various pathways and the effects of multiple inhibitors, activators, and modulators specific to non-communicable diseases with a futuristic outlook.
Collapse
Affiliation(s)
- Varsha Singh
- Centre for Life Sciences, Chitkara School of Health Sciences, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
6
|
Lee H, Kang H, Kim C, Ku JL, Nam S, Lee EK. Long Non-Coding RNA GAS5 Promotes BAX Expression by Competing with microRNA-128-3p in Response to 5-Fluorouracil. Biomedicines 2022; 11:biomedicines11010058. [PMID: 36672566 PMCID: PMC9856034 DOI: 10.3390/biomedicines11010058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
The acquisition of drug resistance is a major hurdle for effective cancer treatment. Although several efforts have been made to overcome drug resistance, the underlying mechanisms have not been fully elucidated. This study investigated the role of long non-coding RNA (lncRNA) growth arrest-specific 5 (GAS5) in drug resistance. GAS5 was found to be downregulated in colon cancer cell lines that are resistant to 5-fluorouracil (5-FU). Downregulation of GAS5 decreased the viability of HCT116 cells and the level of the pro-apoptotic BAX protein, while GAS5 overexpression promoted cell death in response to 5-FU. The interaction between GAS5 and BAX mRNA was investigated using MS2-tagged RNA affinity purification (MS2-trap) followed by RT-qPCR, and the results showed that GAS5 bound to the 3'-untranslated region of BAX mRNA and enhanced its expression by interfering with the inhibitory effect of microRNA-128-3p, a negative regulator of BAX. In addition, ectopic expression of GAS5 increased the sensitivity of resistant cells in response to anti-cancer drugs. These results suggest that GAS5 promoted cell death by interfering with miR-128-3p-mediated BAX downregulation. Therefore, GAS5 overexpression in chemo-resistant cancer cells may be a potential strategy to improve the anti-cancer efficacy of drugs.
Collapse
Affiliation(s)
- Heejin Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hoin Kang
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chongtae Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Ja-Lok Ku
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Sukwoo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Correspondence: ; Tel.: +82-2-3147-8335; Fax: +82-2-596-4435
| |
Collapse
|
7
|
Peng J, Chen Z, Liang H, Yang J. Proteomics analyses of Xiaopi granules in MNNG-induced gastric epithelial dysplasia rat model by LC-MS. Biomed Chromatogr 2022; 36:e5414. [PMID: 35599573 DOI: 10.1002/bmc.5414] [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: 02/28/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Xiaopi granules have been shown to ameliorate gastric epithelial dysplasia in patients. However, the therapeutic mechanism is unclear. Herein, the proteomics method was applied to identify the differentially expressed proteins and related pathways. METHODS Sixty male Sprague-Dawley (SD) rats were randomly divided into four groups: control (C group, n=10), model (M group), Xiaopi granules (X group), and vitacoenzyme (V group). The rat gastric epithelial dysplasia model was established by intragastrically administering N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) and ranitidine, and drinking 0.05% ammonia solution. After 12 weeks, the stomach tissue was analyzed by H&E staining and proteomics analyses. Western blot analysis was applied to further validate the proteomics results. RESULTS Compared to the M group, levels of 326 and 350 proteins were altered significantly in the X and V groups (1.5-fold, P<0.05), which were significantly enriched in digestion, metabolism, coagulation, and cell apoptosis. CELA2A, GHRL, NDUFB9, and PGC were significantly upregulated (P<0.0001), while CLCA1, PLG, and DAC2 were downregulated (P<0.001 or P<0.0001) in the M group vs. the C group. The change in the above proteins could be reversed after the treatment of Xiaopi granules or vitacoenzyme tablets. CONCLUSION Xiaopi granules improve ameliorated gastric epithelial dysplasia by intervening in digestion, metabolism, blood coagulation, cell apoptosis, and other related pathways.
Collapse
Affiliation(s)
- Jisheng Peng
- Department of traditional Chinese medicine, Peking University Shougang Hospital, Beijing, China
| | - Zehui Chen
- Beijing University of Chinese Medicine, Beijing, China
| | - Huazheng Liang
- Clinical Research Center for Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jinxiang Yang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| |
Collapse
|
8
|
Zhang G, Zhong J, Lin L, Liu Z. Loss of ATP5A1 enhances proliferation and predicts poor prognosis of colon adenocarcinoma. Pathol Res Pract 2022; 230:153679. [PMID: 35007851 DOI: 10.1016/j.prp.2021.153679] [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: 06/16/2021] [Revised: 09/27/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND ATP Synthase F1 Subunit Alpha (ATP5F1A), also named as ATP5A1, is a subunit of mitochondrial ATP synthase. Dysregulated expression of ATP5A1 has been reported in several malignancies, nevertheless it showed either oncogenic or tumor-suppressing roles in different cancer types. Here we aimed to initially investigate the expression and role of ATP5A1 in colon adenocarcinoma. METHODS We firstly evaluated the transcription and mRNA levels of ATP5A1 using data from The Cancer Genome Atlas (TCGA). Besides, we tested its mRNA and protein expression in our enrolled retrospective cohort (n = 115). Univariate and multivariate analyzes were conducted to assess its prognostic value. Cellular experiments and xenografts in mice model were performed to validate the role of ATP5A1 in colon cancer. RESULTS ATP5A1 showed a significant lower level in colon adenocarcinoma than in adjacent nontumorous tissue. Advanced tumor stage was characterized with lower ATP5A1 level. Lower ATP5A1 was associated with poor prognosis in both TCGA dataset (P = 0.041) and our cohort (P = 0.001). Furthermore, Cox regression analysis demonstrated that ATP5A1 was a novel independent prognostic factor for colon cancer patients (HR=0.43, P = 0.018). Finally, cellular and xenografts data confirmed that overexpressing ATP5A1 can remarkably attenuate colon cancer growth. CONCLUSION Low expression of ATP5A1 may be a potential molecular marker for poor prognosis in colon cancer. DATA AVAILABILITY Data will be available upon request.
Collapse
Affiliation(s)
- Guifeng Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Jiangming Zhong
- Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Li Lin
- Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Zhenhua Liu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China.
| |
Collapse
|
9
|
Fu J, Pan J, Yang X, Zhang Y, Shao F, Chen J, Huang K, Wang Y. Mechanistic study of lncRNA UCA1 promoting growth and cisplatin resistance in lung adenocarcinoma. Cancer Cell Int 2021; 21:505. [PMID: 34544452 PMCID: PMC8454127 DOI: 10.1186/s12935-021-02207-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023] Open
Abstract
Aim This study aimed to explore the mechanism of LncRNA urothelial carcinoma-associated 1 (UCA1) promoting cisplatin resistance in lung adenocarcinoma (LUAD). Method The UCA1 expression level in LUAD cell lines was detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). We overexpressed UCA1 in A549 cells and downregulated UCA1 in A549/DDP cells by the lentivirus‑mediated technique. Subsequently, in vitro, and in vivo functional experiments were performed to investigate the functional roles of UCA1 in the growth and metastasis of LUAD cell lines. Furthermore, RNA pulldown, mass spectrometry, and RNA immunoprecipitation technique were performed to analyze various downstream target factors regulated by UCA1. Results The results revealed a higher UCA1 expression level in A549/DDP cells and LUAD tissues than in A549 cells and adjacent cancer tissues. UCA1 expression was significantly associated with distant metastasis, clinical stage, and survival time of patients with LUAD. UCA1 overexpression significantly increased the proliferation, invasion, clone formation, and cisplatin resistance ability and enhanced the expression levels of proliferating cell nuclear antigen and excision repair cross-complementing gene 1 in A549 cells. However, these trends were mostly reversed after the knockdown of UCA1 in A549/DDP cells. Tumorigenic assays in nude mice showed that UCA1 knockdown significantly inhibited tumor growth and reduced cisplatin resistance. Enolase 1 was the RNA-binding protein (RBP) of UCA1. Conclusion Based on the results, we concluded that UCA1 promoted LUAD progression and cisplatin resistance and hence could be a potential diagnostic marker and therapeutic target in patients with LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02207-0.
Collapse
Affiliation(s)
- Jiali Fu
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingjing Pan
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiang Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Fanggui Shao
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jie Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Kate Huang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yumin Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
10
|
Sosnowska M, Kutwin M, Strojny B, Wierzbicki M, Cysewski D, Szczepaniak J, Ficek M, Koczoń P, Jaworski S, Chwalibog A, Sawosz E. Diamond Nanofilm Normalizes Proliferation and Metabolism in Liver Cancer Cells. Nanotechnol Sci Appl 2021; 14:115-137. [PMID: 34511890 PMCID: PMC8420805 DOI: 10.2147/nsa.s322766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023] Open
Abstract
Purpose Surgical resection of hepatocellular carcinoma can be associated with recurrence resulting from the degeneration of residual volume of the liver. The objective was to assess the possibility of using a biocompatible nanofilm, made of a colloid of diamond nanoparticles (nfND), to fill the side after tumour resection and optimize its contact with proliferating liver cells, minimizing their cancerous transformation. Methods HepG2 and C3A liver cancer cells and HS-5 non-cancer cells were used. An aqueous colloid of diamond nanoparticles, which covered the cell culture plate, was used to create the nanofilm. The roughness of the resulting nanofilm was measured by atomic force microscopy. Mitochondrial activity and cell proliferation were measured by XTT and BrdU assays. Cell morphology and a scratch test were used to evaluate the invasiveness of cells. Flow cytometry determined the number of cells within the cell cycle. Protein expression in was measured by mass spectrometry. Results The nfND created a surface with increased roughness and exposed oxygen groups compared with a standard plate. All cell lines were prone to settling on the nanofilm, but cancer cells formed more relaxed clusters. The surface compatibility was dependent on the cell type and decreased in the order C3A >HepG2 >HS-5. The invasion was reduced in cancer lines with the greatest effect on the C3A line, reducing proliferation and increasing the G2/M cell population. Among the proteins with altered expression, membrane and nuclear proteins dominated. Conclusion In vitro studies demonstrated the antiproliferative properties of nfND against C3A liver cancer cells. At the same time, the need to personalize potential therapy was indicated due to the differential protein synthetic responses in C3A vs HepG2 cells. We documented that nfND is a source of signals capable of normalizing the expression of many intracellular proteins involved in the transformation to non-cancerous cells.
Collapse
Affiliation(s)
- Malwina Sosnowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Strojny
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Mateusz Wierzbicki
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Dominik Cysewski
- Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Science, Warsaw, Poland
| | - Jarosław Szczepaniak
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Mateusz Ficek
- Department of Metrology and Optoelectronics, Gdansk University of Technology, Gdansk, Poland
| | - Piotr Koczoń
- Department of Chemistry, Institute of Food Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - André Chwalibog
- Department of Veterinary and Animal, Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ewa Sawosz
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
11
|
Antiproliferative Activity of the Cyclization Products of 1-(2-Oxocyclohexyl)Ethane-1,1,2,2-Tetracarbonitrile and α,β-Unsaturated Aldehydes. Pharm Chem J 2021. [DOI: 10.1007/s11094-021-02367-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Kim MS, Gernapudi R, Cedeño YC, Polster BM, Martinez R, Shapiro P, Kesari S, Nurmemmedov E, Passaniti A. Targeting breast cancer metabolism with a novel inhibitor of mitochondrial ATP synthesis. Oncotarget 2020; 11:3863-3885. [PMID: 33196708 PMCID: PMC7597410 DOI: 10.18632/oncotarget.27743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 01/17/2023] Open
Abstract
Inhibitors of mitochondrial respiration and ATP synthesis may promote the selective killing of respiration-competent cancer cells that are critical for tumor progression. We previously reported that CADD522, a small molecule inhibitor of the RUNX2 transcription factor, has potential for breast cancer treatment. In the current study, we show that CADD522 inhibits mitochondrial oxidative phosphorylation by decreasing the mitochondrial oxygen consumption rate (OCR) and ATP production in human breast cancer cells in a RUNX2-independent manner. The enzyme activity of mitochondrial ATP synthase was inhibited by CADD522 treatment. Importantly, results from cellular thermal shift assays that detect drug-induced protein stabilization revealed that CADD522 interacts with both α and β subunits of the F1-ATP synthase complex. Differential scanning fluorimetry also demonstrated interaction of α subunits of the F1-ATP synthase to CADD522. These results suggest that CADD522 might target the enzymatic F1 subunits in the ATP synthase complex. CADD522 increased the levels of intracellular reactive oxygen species (ROS), which was prevented by MitoQ, a mitochondria-targeted antioxidant, suggesting that cancer cells exposed to CADD522 may elevate ROS from mitochondria. CADD522-increased mitochondrial ROS levels were enhanced by exogenously added pro-oxidants such as hydrogen peroxide or tert-butyl hydroperoxide. Conversely, CADD522-mediated cell growth inhibition was blocked by N-acetyl-l-cysteine, a general ROS scavenger. Therefore, CADD522 may exert its antitumor activity by increasing mitochondrial driven cellular ROS levels. Collectively, our data suggest in vitro proof-of-concept that supports inhibition of mitochondrial ATP synthase and ROS generation as contributors to the effectiveness of CADD522 in suppression of tumor growth.
Collapse
Affiliation(s)
- Myoung Sook Kim
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Ramkishore Gernapudi
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | | | - Brian M. Polster
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Research Health Scientist, The Veteran's Health Administration Research & Development Service (VAMHCS), Baltimore, MD, USA
| | - Ramon Martinez
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Santosh Kesari
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Elmar Nurmemmedov
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Antonino Passaniti
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- Research Health Scientist, The Veteran's Health Administration Research & Development Service (VAMHCS), Baltimore, MD, USA
| |
Collapse
|
13
|
Galber C, Acosta MJ, Minervini G, Giorgio V. The role of mitochondrial ATP synthase in cancer. Biol Chem 2020; 401:1199-1214. [PMID: 32769215 DOI: 10.1515/hsz-2020-0157] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/27/2020] [Indexed: 12/19/2022]
Abstract
The mitochondrial ATP synthase is a multi-subunit enzyme complex located in the inner mitochondrial membrane which is essential for oxidative phosphorylation under physiological conditions. In this review, we analyse the enzyme functions involved in cancer progression by dissecting specific conditions in which ATP synthase contributes to cancer development or metastasis. Moreover, we propose the role of ATP synthase in the formation of the permeability transition pore (PTP) as an additional mechanism which controls tumour cell death. We further describe transcriptional and translational modifications of the enzyme subunits and of the inhibitor protein IF1 that may promote adaptations leading to cancer metabolism. Finally, we outline ATP synthase gene mutations and epigenetic modifications associated with cancer development or drug resistance, with the aim of highlighting this enzyme complex as a potential novel target for future anti-cancer therapy.
Collapse
Affiliation(s)
- Chiara Galber
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Manuel Jesus Acosta
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Giovanni Minervini
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| | - Valentina Giorgio
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, V.le G. Colombo 3, I-35121, Padova, Italy
- Department of Biomedical Sciences, University of Padova, I-35121, Padova, Italy
| |
Collapse
|
14
|
Chakraborty M, Mahmud NU, Muzahid ANM, Rabby SMF, Islam T. Oligomycins inhibit Magnaporthe oryzae Triticum and suppress wheat blast disease. PLoS One 2020; 15:e0233665. [PMID: 32804955 PMCID: PMC7430738 DOI: 10.1371/journal.pone.0233665] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/29/2020] [Indexed: 02/05/2023] Open
Abstract
Oligomycins are macrolide antibiotics, produced by Streptomyces spp. that show antagonistic effects against several microorganisms such as bacteria, fungi, nematodes and the oomycete Plasmopara viticola. Conidiogenesis, germination of conidia and formation of appressoria are determining factors pertaining to pathogenicity and successful diseases cycles of filamentous fungal phytopathogens. The goal of this research was to evaluate the in vitro suppressive effects of two oligomycins, oligomycin B and F along with a commercial fungicide Nativo® 75WG on hyphal growth, conidiogenesis, conidial germination, and appressorial formation of the wheat blast fungus, Magnaporthe oryzae Triticum (MoT) pathotype. We also determined the efficacy of these two oligomycins and the fungicide product in vivo in suppressing wheat blast with a detached leaf assay. Both oligomycins suppressed the growth of MoT mycelium in a dose dependent manner. Between the two natural products, oligomycin F provided higher inhibition of MoT hyphal growth compared to oligomycin B with a minimum inhibitory concentration of 0.005 and 0.05 μg/disk, respectively. The application of the compounds completely halted conidial formation of the MoT mycelium in agar medium. Further bioassays showed that these compounds significantly inhibited MoT conidia germination and induced lysis. The compounds also caused abnormal germ tube formation and suppressed appressorial formation of germinated spores. Interestingly, the application of these macrolides significantly inhibited wheat blast on detached leaves of wheat. This is the first report on the inhibition of mycelial growth, conidiogenesis, germination of conidia, deleterious morphological changes in germinated conidia, and suppression of blast disease of wheat by oligomycins from Streptomyces spp. Further study is needed to unravel the precise mode of action of these natural compounds and consider them as biopesticides for controlling wheat blast.
Collapse
Affiliation(s)
- Moutoshi Chakraborty
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Nur Uddin Mahmud
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Abu Naim Md. Muzahid
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - S. M. Fajle Rabby
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Tofazzal Islam
- Institute of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
- * E-mail:
| |
Collapse
|
15
|
Blondy S, David V, Verdier M, Mathonnet M, Perraud A, Christou N. 5-Fluorouracil resistance mechanisms in colorectal cancer: From classical pathways to promising processes. Cancer Sci 2020; 111:3142-3154. [PMID: 32536012 PMCID: PMC7469786 DOI: 10.1111/cas.14532] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is a public health problem. It is the third most common cancer in the world, with nearly 1.8 million new cases diagnosed in 2018. The only curative treatment is surgery, especially for early tumor stages. When there is locoregional or distant invasion, chemotherapy can be introduced, in particular 5-fluorouracil (5-FU). However, the disease can become tolerant to these pharmaceutical treatments: resistance emerges, leading to early tumor recurrence. Different mechanisms can explain this 5-FU resistance. Some are disease-specific, whereas others, such as drug efflux, are evolutionarily conserved. These mechanisms are numerous and complex and can occur simultaneously in cells exposed to 5-FU. In this review, we construct a global outline of different mechanisms from disruption of 5-FU-metabolic enzymes and classic cellular processes (apoptosis, autophagy, glucose metabolism, oxidative stress, respiration, and cell cycle perturbation) to drug transporters and epithelial-mesenchymal transition induction. Particular interest is directed to tumor microenvironment function as well as epigenetic alterations and miRNA dysregulation, which are the more promising processes that will be the subject of much research in the future.
Collapse
Affiliation(s)
- Sabrina Blondy
- Faculty of Medicine, Laboratoire EA3842 CAPTuR "Control of cell activation, Tumor progression and Therapeutic resistance", Limoges cedex, France
| | - Valentin David
- Faculty of Medicine, Laboratoire EA3842 CAPTuR "Control of cell activation, Tumor progression and Therapeutic resistance", Limoges cedex, France.,Department of pharmacy, University Hospital of Limoges, Limoges, France
| | - Mireille Verdier
- Faculty of Medicine, Laboratoire EA3842 CAPTuR "Control of cell activation, Tumor progression and Therapeutic resistance", Limoges cedex, France
| | - Muriel Mathonnet
- Faculty of Medicine, Laboratoire EA3842 CAPTuR "Control of cell activation, Tumor progression and Therapeutic resistance", Limoges cedex, France.,Service de Chirurgie Digestive, Department of Digestive, General and Endocrine Surgery, University Hospital of Limoges, Limoges, France
| | - Aurélie Perraud
- Faculty of Medicine, Laboratoire EA3842 CAPTuR "Control of cell activation, Tumor progression and Therapeutic resistance", Limoges cedex, France.,Service de Chirurgie Digestive, Department of Digestive, General and Endocrine Surgery, University Hospital of Limoges, Limoges, France
| | - Niki Christou
- Faculty of Medicine, Laboratoire EA3842 CAPTuR "Control of cell activation, Tumor progression and Therapeutic resistance", Limoges cedex, France.,Service de Chirurgie Digestive, Department of Digestive, General and Endocrine Surgery, University Hospital of Limoges, Limoges, France
| |
Collapse
|
16
|
Hlozkova K, Pecinova A, Alquezar-Artieda N, Pajuelo-Reguera D, Simcikova M, Hovorkova L, Rejlova K, Zaliova M, Mracek T, Kolenova A, Stary J, Trka J, Starkova J. Metabolic profile of leukemia cells influences treatment efficacy of L-asparaginase. BMC Cancer 2020; 20:526. [PMID: 32503472 PMCID: PMC7275298 DOI: 10.1186/s12885-020-07020-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/28/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Effectiveness of L-asparaginase administration in acute lymphoblastic leukemia treatment is mirrored in the overall outcome of patients. Generally, leukemia patients differ in their sensitivity to L-asparaginase; however, the mechanism underlying their inter-individual differences is still not fully understood. We have previously shown that L-asparaginase rewires the biosynthetic and bioenergetic pathways of leukemia cells to activate both anti-leukemic and pro-survival processes. Herein, we investigated the relationship between the metabolic profile of leukemia cells and their sensitivity to currently used cytostatic drugs. METHODS Altogether, 19 leukemia cell lines, primary leukemia cells from 26 patients and 2 healthy controls were used. Glycolytic function and mitochondrial respiration were measured using Seahorse Bioanalyzer. Sensitivity to cytostatics was measured using MTS assay and/or absolute count and flow cytometry. Mitochondrial membrane potential was determined as TMRE fluorescence. RESULTS Using cell lines and primary patient samples we characterized the basal metabolic state of cells derived from different leukemia subtypes and assessed their sensitivity to cytostatic drugs. We found that leukemia cells cluster into distinct groups according to their metabolic profile. Lymphoid leukemia cell lines and patients sensitive to L-asparaginase clustered into the low glycolytic cluster. While lymphoid leukemia cells with lower sensitivity to L-asparaginase together with resistant normal mononuclear blood cells gathered into the high glycolytic cluster. Furthermore, we observed a correlation of specific metabolic parameters with the sensitivity to L-asparaginase. Greater ATP-linked respiration and lower basal mitochondrial membrane potential in cells significantly correlated with higher sensitivity to L-asparaginase. No such correlation was found in the other cytostatic drugs tested by us. CONCLUSIONS These data support that cell metabolism plays a prominent role in the treatment effect of L-asparaginase. Based on these findings, leukemia patients with lower sensitivity to L-asparaginase with no specific genetic characterization could be identified by their metabolic profile.
Collapse
Affiliation(s)
- Katerina Hlozkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alena Pecinova
- Department of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Natividad Alquezar-Artieda
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Pajuelo-Reguera
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Simcikova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Hovorkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katerina Rejlova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Marketa Zaliova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Tomas Mracek
- Department of Bioenergetics, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alexandra Kolenova
- Department of Pediatric Hematology and Oncology, National Institute of Children's Diseases and Medical Faculty, Comenius University, Bratislava, Slovakia
| | - Jan Stary
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Jan Trka
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Julia Starkova
- CLIP - Childhood Leukaemia Investigation Prague, Prague, Czech Republic.
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
17
|
Lin HC, Chu LJ, Huang PJ, Cheng WH, Zheng YH, Huang CY, Hong SW, Chen LC, Lin HA, Wang JY, Chen RM, Lin WN, Tang P, Huang KY. Proteomic signatures of metronidazole-resistant Trichomonas vaginalis reveal novel proteins associated with drug resistance. Parasit Vectors 2020; 13:274. [PMID: 32487244 PMCID: PMC7268490 DOI: 10.1186/s13071-020-04148-5] [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: 01/30/2020] [Accepted: 05/25/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Trichomoniasis is the most common non-viral sexually transmitted disease caused by the protozoan parasite Trichomonas vaginalis. Metronidazole (MTZ) is a widely used drug for the treatment of trichomoniasis; however, increased resistance of the parasite to MTZ has emerged as a highly problematic public health issue. METHODS We conducted iTRAQ-based analysis to profile the proteomes of MTZ-sensitive (MTZ-S) and MTZ-resistant (MTZ-R) parasites. STRING and gene set enrichment analysis (GESA) were utilized to explore the protein-protein interaction networks and enriched pathways of the differentially expressed proteins, respectively. Proteins potentially related to MTZ resistance were selected for functional validation. RESULTS A total of 3123 proteins were identified from the MTZ-S and MTZ-R proteomes in response to drug treatment. Among the identified proteins, 304 proteins were differentially expressed in the MTZ-R proteome, including 228 upregulated and 76 downregulated proteins. GSEA showed that the amino acid-related metabolism, including arginine, proline, alanine, aspartate, and glutamate are the most upregulated pathways in the MTZ-R proteome, whereas oxidative phosphorylation is the most downregulated pathway. Ten proteins categorized into the gene set of oxidative phosphorylation were ATP synthase subunit-related proteins. Drug resistance was further examined in MTZ-S parasites pretreated with the ATP synthase inhibitors oligomycin and bafilomycin A1, showing enhanced MTZ resistance and potential roles of ATP synthase in drug susceptibility. CONCLUSIONS We provide novel insights into previously unidentified proteins associated with MTZ resistance, paving the way for future development of new drugs against MTZ-refractory trichomoniasis.
Collapse
Affiliation(s)
- Hsin-Chung Lin
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan
| | - Lichieh Julie Chu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan City, 333, Taiwan.,Liver Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan City, 333, Taiwan
| | - Po-Jung Huang
- Department of Biomedical Sciences, Chang Gung University, Taoyuan City, 333, Taiwan.,Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taoyuan City, 333, Taiwan
| | - Wei-Hung Cheng
- Molecular Regulation and Bioinformatics Laboratory, Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan City, 333, Taiwan
| | - Yu-Hsing Zheng
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, 114, Taiwan
| | - Ching-Yun Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, 114, Taiwan
| | - Shu-Wen Hong
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, 114, Taiwan
| | - Lih-Chyang Chen
- Department of Medicine, Mackay Medical College, New Taipei City, 252, Taiwan
| | - Hsin-An Lin
- Division of Infection, Department of Medicine, Tri-Service General Hospital SongShan Branch, Taipei City, 105, Taiwan
| | - Jui-Yang Wang
- Division of Family Medicine, Tri-Service General Hospital Songshan Branch, Taipei City, 105, Taiwan
| | - Ruei-Min Chen
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei City, 242, Taiwan
| | - Petrus Tang
- Molecular Regulation and Bioinformatics Laboratory, Department of Parasitology, College of Medicine, Chang Gung University, Taoyuan City, 333, Taiwan
| | - Kuo-Yang Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei City, 114, Taiwan.
| |
Collapse
|
18
|
Wang SF, Chen S, Tseng LM, Lee HC. Role of the mitochondrial stress response in human cancer progression. Exp Biol Med (Maywood) 2020; 245:861-878. [PMID: 32326760 PMCID: PMC7268930 DOI: 10.1177/1535370220920558] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPACT STATEMENT Dysregulated mitochondria often occurred in cancers. Mitochondrial dysfunction might contribute to cancer progression. We reviewed several mitochondrial stresses in cancers. Mitochondrial stress responses might contribute to cancer progression. Several mitochondrion-derived molecules (ROS, Ca2+, oncometabolites, exported mtDNA, mitochondrial double-stranded RNA, humanin, and MOTS-c), integrated stress response, and mitochondrial unfolded protein response act as retrograde signaling pathways and might be critical in the development and progression of cancer. Targeting these mitochondrial stress responses may be an important strategy for cancer treatment.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, 112 Taipei
- School of Pharmacy, Taipei Medical University, 110 Taipei
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, 112 Taipei
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, CA 91010, USA
| | - Ling-Ming Tseng
- Division of General Surgery, Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, 112 Taipei
- Department of Surgery, School of Medicine, National Yang-Ming University, 112 Taipei
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, 112 Taipei
| |
Collapse
|
19
|
Kim EK, Song MJ, Jung Y, Lee WS, Jang HH. Proteomic Analysis of Primary Colon Cancer and Synchronous Solitary Liver Metastasis. Cancer Genomics Proteomics 2020; 16:583-592. [PMID: 31659112 DOI: 10.21873/cgp.20161] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIM Colon cancer is prone to distant metastases to other sites and the risk of recurrence is relatively high. Therefore, the identification of liver metastasis-related factors is important for the diagnosis or treatment of colon cancer. The aim of this study was to identify the metastasis-related factors that are differentially expressed in synchronous solitary liver metastasis compared to primary colon cancer. MATERIALS AND METHODS Tissues of primary colon cancer and associated with liver metastases of five patients were used for mass spectrometry. Identified proteins were validated by western blotting. The in silico analysis was performed using the STRING database and GeneMANIA. RESULTS We identified 58 differentially expressed proteins (DEPs), including 51 under-expressed and 7 over-expressed proteins among a total of 164 identified proteins. Major hubs of protein-protein networks were ACTC1, PRDX6, TPI1, and ALDH1A1. DEPs were located in the extracellular region and cytoplasm and were involved in the regulation of enzymatic activity. The metabolic process was significantly enriched in biological processes and an involvement in the KEGG pathway. CONCLUSION These DEPs can potentially be used as biomarkers for the diagnosis of liver metastasis and they may provide a new strategy for developing anti-metastatic liver drugs in colon cancer patients.
Collapse
Affiliation(s)
- Eun-Kyung Kim
- Department of Biochemistry, College of Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Min-Jeong Song
- Department of Biochemistry, College of Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Yunjae Jung
- Department of Microbiology, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Won-Suk Lee
- Department of Surgery, Gil Medical Center, Gachon University, Incheon, Republic of Korea
| | - Ho Hee Jang
- Department of Biochemistry, College of Medicine, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
20
|
Siska PJ, Singer K, Evert K, Renner K, Kreutz M. The immunological Warburg effect: Can a metabolic-tumor-stroma score (MeTS) guide cancer immunotherapy? Immunol Rev 2020; 295:187-202. [PMID: 32157706 DOI: 10.1111/imr.12846] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
The "glycolytic switch" also known as the "Warburg effect" is a key feature of tumor cells and leads to the accumulation of lactate and protons in the tumor environment. Intriguingly, non-malignant lymphocytes or stromal cells such as tumor-associated macrophages and cancer-associated fibroblasts contribute to the lactate accumulation in the tumor environment, a phenomenon described as the "Reverse Warburg effect." Localized lactic acidosis has a strong immunosuppressive effect and mediates an immune escape of tumors. However, some tumors do not display the Warburg phenotype and either rely on respiration or appear as a mosaic of cells with different metabolic properties. Based on these findings and on the knowledge that T cell infiltration is predictive for patient outcome, we suggest a metabolic-tumor-stroma score to determine the likelihood of a successful anti-tumor immune response: (a) a respiring tumor with high T cell infiltration ("hot"); (b) a reverse Warburg type with respiring tumor cells but glycolytic stromal cells; (c) a mixed type with glycolytic and respiring compartments; and (d) a glycolytic (Warburg) tumor with low T cell infiltration ("cold"). Here, we provide evidence that these types can be independent of the organ of origin, prognostically relevant and might help select the appropriate immunotherapy approach.
Collapse
Affiliation(s)
- Peter J Siska
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Katrin Singer
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Kathrin Renner
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.,Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
| | - Marina Kreutz
- Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.,Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
| |
Collapse
|
21
|
Oh SJ, Lee J, Kim Y, Song KH, Cho E, Kim M, Jung H, Kim TW. Far Beyond Cancer Immunotherapy: Reversion of Multi-Malignant Phenotypes of Immunotherapeutic-Resistant Cancer by Targeting the NANOG Signaling Axis. Immune Netw 2020; 20:e7. [PMID: 32158595 PMCID: PMC7049583 DOI: 10.4110/in.2020.20.e7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer immunotherapy, in the form of vaccination, adoptive cellular transfer, or immune checkpoint inhibitors, has emerged as a promising practice within the field of oncology. However, despite the developing field's potential to revolutionize cancer treatment, the presence of immunotherapeutic-resistant tumor cells in many patients present a challenge and limitation to these immunotherapies. These cells not only indicate immunotherapeutic resistance, but also show multi-modal resistance to conventional therapies, abnormal metabolism, stemness, and metastasis. How can immunotherapeutic-resistant tumor cells render multi-malignant phenotypes? We reasoned that the immune-refractory phenotype could be associated with multi-malignant phenotypes and that these phenotypes are linked together by a factor that acts as the master regulator. In this review, we discussed the role of the embryonic transcription factor NANOG as a crucial master regulator we named “common factor” in multi-malignant phenotypes and presented strategies to overcome multi-malignancy in immunotherapeutic-resistant cancer by restraining the NANOG-mediated multi-malignant signaling axis. Strategies that blunt the NANOG axis could improve the clinical management of therapy-refractory cancer.
Collapse
Affiliation(s)
- Se Jin Oh
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Jaeyoon Lee
- College of Science, College of Social Sciences and Humanities, Northeastern University, Boston, MA 02115, USA
| | - Yukang Kim
- Korea University College of Medicine, Seoul 02841, Korea
| | - Kwon-Ho Song
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Eunho Cho
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| | - Minsung Kim
- Korea University College of Medicine, Seoul 02841, Korea
| | - Heejae Jung
- Korea University College of Medicine, Seoul 02841, Korea
| | - Tae Woo Kim
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul 02841, Korea.,Department of Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea.,Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
22
|
Pawłowski P, Szczęsny P, Rempoła B, Poznańska A, Poznański J. Combined in silico and 19F NMR analysis of 5-fluorouracil metabolism in yeast at low ATP conditions. Biosci Rep 2019; 39:BSR20192847. [PMID: 31742586 PMCID: PMC6904775 DOI: 10.1042/bsr20192847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 11/27/2022] Open
Abstract
The cytotoxic effect of 5-fluorouracil (5-FU) on yeast cells is thought to be mainly via a misincorporation of fluoropyrimidines into both RNA and DNA, not only DNA damage via inhibition of thymidylate synthase (TYMS) by fluorodeoxyuridine monophosphate (FdUMP). However, some studies on Saccharomyces cerevisiae show a drastic decrease in ATP concentration under oxidative stress, together with a decrease in concentration of other tri- and diphosphates. This raises a question if hydrolysis of 5-fluoro-2-deoxyuridine diphosphate (FdUDP) under oxidative stress could not lead to the presence of FdUMP and the activation of so-called 'thymine-less death' route. We attempted to answer this question with in silico modeling of 5-FU metabolic pathways, based on new experimental results, where the stages of intracellular metabolism of 5-FU in Saccharomyces cerevisiae were tracked by a combination of 19F and 31P NMR spectroscopic study. We have identified 5-FU, its nucleosides and nucleotides, and subsequent di- and/or triphosphates. Additionally, another wide 19F signal, assigned to fluorinated unstructured short RNA, has been also identified in the spectra. The concentration of individual metabolites was found to vary substantially within hours, however, the initial steady-state was preserved only for an hour, until the ATP concentration dropped by a half, which was monitored independently via 31P NMR spectra. After that, the catabolic process leading from triphosphates through monophosphates and nucleosides back to 5-FU was observed. These results imply careful design and interpretation of studies in 5-FU metabolism in yeast.
Collapse
Affiliation(s)
- Piotr H. Pawłowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Paweł Szczęsny
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Institute of Experimental Plant Biology and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Bożenna Rempoła
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Poznańska
- National Institute of Public Health-National Institute of Hygiene, Department of Population Health Monitoring and Analysis, Warsaw, Poland
| | - Jarosław Poznański
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
23
|
Yang Y, Pan C, Yu L, Ruan H, Chang L, Yang J, Zheng Z, Zheng F, Liu T. SSBP1 Upregulation In Colorectal Cancer Regulates Mitochondrial Mass. Cancer Manag Res 2019; 11:10093-10106. [PMID: 31819642 PMCID: PMC6896925 DOI: 10.2147/cmar.s211292] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/28/2019] [Indexed: 12/27/2022] Open
Abstract
Background Colorectal cancers (CRC) are one of the most common forms of cancer seen worldwide, and also remain difficult to treat despite recent advances in chemotherapy. Although significant progress has been made in recent years towards precision medicine and mutation-guided therapy, common mechanisms that underlie tumor growth and progression remain incompletely understood. Methods Tumor tissue and nearby unaffected tissue were collected from >15 patients at each stage of CRC, from which we generated representative proteomics profiles of three stages. Bioinformatics analysis was performed to discover common differences that may be shared between the representative profiles and across larger cohorts. Flow cytometry was then used to identify functional consequences of SSBP1 depletion in cell lines, since its expression level was consistently increased in tumor cells across all of the datasets analyzed. Results Direct comparison of CRC tumor and unaffected tissue at each stage demonstrated that a number of proteins involved in mitochondrial function displayed significantly altered expression patterns. Depletion of SSBP1 in colon cancer cell lines was able to trigger loss of mitochondrial mass and an increase in tumor cell death, and this effect that was further accentuated in the presence of the common chemotherapy drug cisplatin. Conclusion Mitochondrial biogenesis and maintenance may play an important part in tumor cell survival during CRC progression, and may be a useful target for directed inhibition or adjuvant targeting in the cases of cisplatin resistance.
Collapse
Affiliation(s)
- Yongping Yang
- First Bethune Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Chenxi Pan
- Dalian Key Laboratory of Immune and Metabolic Kidney Diseases, Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Lingyun Yu
- First Bethune Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Hongxia Ruan
- Dalian Key Laboratory of Immune and Metabolic Kidney Diseases, Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Ling Chang
- Dalian Key Laboratory of Immune and Metabolic Kidney Diseases, Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of Nephrology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Jingbo Yang
- Second Bethune Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| | - Zihan Zheng
- Dalian Key Laboratory of Immune and Metabolic Kidney Diseases, Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Feng Zheng
- Dalian Key Laboratory of Immune and Metabolic Kidney Diseases, Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, Liaoning, People's Republic of China.,Department of Nephrology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Tongjun Liu
- First Bethune Hospital, Jilin University, Changchun, Jilin, People's Republic of China.,Second Bethune Hospital, Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
24
|
Huang CY, Huang CY, Pai YC, Lin BR, Lee TC, Liang PH, Yu LCH. Glucose Metabolites Exert Opposing Roles in Tumor Chemoresistance. Front Oncol 2019; 9:1282. [PMID: 31824857 PMCID: PMC6881467 DOI: 10.3389/fonc.2019.01282] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/05/2019] [Indexed: 01/01/2023] Open
Abstract
Reprogrammed glucose metabolism and increased glycolysis have been implicated in tumor chemoresistance. The aim was to investigate the distinct roles of the glucose metabolites pyruvate and ATP in chemoresistance mechanisms, including cell death and proliferation. Our data showed higher glucose transporters in colorectal cancer (CRC) from non-responsive patients than those responsive to chemotherapy. Human CRC cell lines exposed to 5-fluorouracil (5-FU) displayed elevated cell viability and larger tumors in xenograft mouse models if cultured in high-glucose medium. Glucose conferred resistance to 5-FU-induced necroptosis via pyruvate scavenging of mitochondrial free radicals, whereas ATP replenishment had no effect on cell death. Glucose attenuated the 5-FU-induced G0/G1 shift but not the S phase arrest. Opposing effects were observed by glucose metabolites; ATP increased while pyruvate decreased the G0/G1 shift. Lastly, 5-FU-induced tumor spheroid destruction was prevented by glucose and pyruvate, but not by ATP. Our finding argues against ATP as the main effector for glucose-mediated chemoresistance and supports a key role of glycolytic pyruvate as an antioxidant for dual modes of action: necroptosis reduction and a cell cycle shift to a quiescent state.
Collapse
Affiliation(s)
- Chung-Yen Huang
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,School of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Ying Huang
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Food Science and Biotechnology, National Chung-Hsing University, Taichung City, Taiwan
| | - Yu-Chen Pai
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Been-Ren Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Tsung-Chun Lee
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Pi-Hui Liang
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
25
|
Redox-Mediated Mechanism of Chemoresistance in Cancer Cells. Antioxidants (Basel) 2019; 8:antiox8100471. [PMID: 31658599 PMCID: PMC6826977 DOI: 10.3390/antiox8100471] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Cellular reactive oxygen species (ROS) status is stabilized by a balance of ROS generation and elimination called redox homeostasis. ROS is increased by activation of endoplasmic reticulum stress, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family members and adenosine triphosphate (ATP) synthesis of mitochondria. Increased ROS is detoxified by superoxide dismutase, catalase, and peroxiredoxins. ROS has a role as a secondary messenger in signal transduction. Cancer cells induce fluctuations of redox homeostasis by variation of ROS regulated machinery, leading to increased tumorigenesis and chemoresistance. Redox-mediated mechanisms of chemoresistance include endoplasmic reticulum stress-mediated autophagy, increased cell cycle progression, and increased conversion to metastasis or cancer stem-like cells. This review discusses changes of the redox state in tumorigenesis and redox-mediated mechanisms involved in tolerance to chemotherapeutic drugs in cancer.
Collapse
|
26
|
Bokil A, Sancho P. Mitochondrial determinants of chemoresistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:634-646. [PMID: 35582564 PMCID: PMC8992520 DOI: 10.20517/cdr.2019.46] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/10/2019] [Accepted: 07/17/2019] [Indexed: 12/17/2022]
Abstract
Chemoresistance constitute nowadays the major contributor to therapy failure in most cancers. There are main factors that mitigate cell response to therapy, such as target organ, inherent sensitivity to the administered compound, its metabolism, drug efflux and influx or alterations on specific cellular targets, among others. We now know that intrinsic properties of cancer cells, including metabolic features, substantially contribute to chemoresistance. In fact, during the last years, numerous reports indicate that cancer cells resistant to chemotherapy demonstrate significant alterations in mitochondrial metabolism, membrane polarization and mass. Metabolic activity and expression of several mitochondrial proteins are modulated under treatment to cope with stress, making these organelles central players in the development of resistance to therapies. Here, we review the role of mitochondria in chemoresistant cells in terms of metabolic rewiring and function of key mitochondria-related proteins.
Collapse
Affiliation(s)
- Ansooya Bokil
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| | - Patricia Sancho
- IIS Aragon, Hospital Universitario Miguel Servet, Zaragoza 50009, Spain
| |
Collapse
|
27
|
ATP Synthase Subunit Epsilon Overexpression Promotes Metastasis by Modulating AMPK Signaling to Induce Epithelial-to-Mesenchymal Transition and Is a Poor Prognostic Marker in Colorectal Cancer Patients. J Clin Med 2019; 8:jcm8071070. [PMID: 31330880 PMCID: PMC6678251 DOI: 10.3390/jcm8071070] [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: 06/07/2019] [Revised: 07/14/2019] [Accepted: 07/16/2019] [Indexed: 01/04/2023] Open
Abstract
Metastasis remains the major cause of death from colon cancer. We intend to identify differentially expressed genes that are associated with the metastatic process and prognosis in colon cancer. ATP synthase epsilon subunit (ATP5E) gene was found to encode the mitochondrial F0F1 ATP synthase subunit epsilon that was overexpressed in tumor cells compared to their normal counterparts, while other genes encoding the ATP synthase subunit were repressed in public microarray datasets. CRC cells in which ATP5E was silenced showed markedly reduced invasive and migratory abilities. ATP5E inhibition significantly reduced the incidence of distant metastasis in a mouse xenograft model. Mechanistically, increased ATP5E expression resulted in a prominent reduction in E-cadherin and an increase in Snail expression. Our data also showed that an elevated ATP5E level in metastatic colon cancer samples was significantly associated with the AMPK-AKT-hypoxia-inducible factor-1α (HIF1α) signaling axis; silencing ATP5E led to the degradation of HIF1α under hypoxia through AMPK-AKT signaling. Our findings suggest that elevated ATP5E expression could serve as a marker of distant metastasis and a poor prognosis in colon cancer, and ATP5E functions via modulating AMPK-AKT-HIF1α signaling.
Collapse
|
28
|
Nájera L, Alonso‐Juarranz M, Garrido M, Ballestín C, Moya L, Martínez‐Díaz M, Carrillo R, Juarranz A, Rojo F, Cuezva J, Rodríguez‐Peralto J. Prognostic implications of markers of the metabolic phenotype in human cutaneous melanoma. Br J Dermatol 2019; 181:114-127. [DOI: 10.1111/bjd.17513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2018] [Indexed: 12/12/2022]
Affiliation(s)
- L. Nájera
- Servicio de Anatomía Patológica Hospital Universitario Puerta de Hierro Majadahonda, MadridSpain
| | | | - M. Garrido
- Instituto de Investigación Hospital 12 de Octubre Universidad Complutense de Madrid MadridSpain
| | - C. Ballestín
- IIS‐Fundación Jiménez Diaz C/Reyes Católicos 2 28049 MadridSpain
| | - L. Moya
- Servicio de Anatomía Patológica Hospital Universitario Ramón y Cajal MadridSpain
| | - M. Martínez‐Díaz
- Departamento de Biología Molecular Centro de Biología Molecular Severo Ochoa CSIC‐UAM MadridSpain
| | - R. Carrillo
- Servicio de Anatomía Patológica Hospital Universitario Ramón y Cajal MadridSpain
| | - A. Juarranz
- Departamento de Biología Facultad de Ciencias Universidad Autónoma de Madrid C/Darwin, 2 28049 MadridSpain
- Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS) MadridSpain
| | - F. Rojo
- IIS‐Fundación Jiménez Diaz C/Reyes Católicos 2 28049 MadridSpain
| | - J.M. Cuezva
- Instituto de Investigación Hospital 12 de Octubre Universidad Complutense de Madrid MadridSpain
- Departamento de Biología Molecular Centro de Biología Molecular Severo Ochoa CSIC‐UAM MadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII MadridSpain
| | - J.L. Rodríguez‐Peralto
- Instituto de Investigación Hospital 12 de Octubre Universidad Complutense de Madrid MadridSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) ISCIII Madrid Spain
| |
Collapse
|
29
|
Xie D, Zhou Y, Luo X. Amorphous silica nanoparticles induce tumorigenesis via regulating ATP5H/SOD1-related oxidative stress, oxidative phosphorylation and EIF4G2/PABPC1-associated translational initiation. PeerJ 2019; 7:e6455. [PMID: 30863671 PMCID: PMC6404658 DOI: 10.7717/peerj.6455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/16/2019] [Indexed: 12/23/2022] Open
Abstract
Background Recent studies indicate amorphous silica nanoparticles (SiNPs), one of the widely applied nanomaterials, have potential toxicity in humans and induces cell malignant transformation. However, its carcinogenic mechanisms remain poorly understood. This study’s purpose was to investigate the underlying toxic mechanisms of amorphous SiNPs on human lung epithelial cells model by using microarray data. Methods Microarray dataset GSE82062 was collected from Gene Expression Omnibus database, including three repeats of Beas-2B exposed to amorphous SiNPs for 40 passages and three repeats of passage-matched control Beas-2B cells. Differentially expressed genes (DEGs) were identified using linear models for microarray data method. Protein–protein interaction (PPI) network was constructed using data from the STRING database followed by module analysis. The miRwalk2 database was used to predict the underlying target genes of differentially miRNAs. Function enrichment analysis was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool. Results A total of 323 genes were identified as DEGs, including 280 downregulated (containing 12 pre-miRNAs) and 43 upregulated genes (containing 29 pre-miRNAs). Function enrichment indicated these genes were involved in translational initiation (i.e., eukaryotic translation initiation factor 4 gamma 2 (EIF4G2), poly (A) binding protein cytoplasmic 1 (PABPC1)), response to reactive oxygen species (i.e., superoxide dismutase 1 (SOD1)) and oxidative phosphorylation (i.e., ATP5H). PABPC1 (degree = 15), ATP5H (degree = 11) and SOD1 (degree = 8)] were proved to be hub genes after PPI-module analyses. ATP5H/SOD1 and EIF4G2/PABPC1 were overlapped with the target genes of differentially expressed pre-miR-3648/572/661 and pre-miR-4521. Conclusions Amorphous SiNPs may induce tumorigenesis via influencing ATP5H/SOD1-related oxidative stress, oxidative phosphorylation and EIF4G2/PABPC1-associated translational initiation which may be regulated by miR-3648/572/661 and miR-4521, respectively.
Collapse
Affiliation(s)
- Dongli Xie
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| | - Yang Zhou
- School of Textile Science and Engineering/National Engineering Laboratory for Advanced Yarn and Clean Production, Wuhan Textile University, Wuhan, China
| | - Xiaogang Luo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, China
| |
Collapse
|
30
|
Koyama FC, Lopes Ramos CM, Ledesma F, Alves VAF, Fernandes JM, Vailati BB, São Julião GP, Habr-Gama A, Gama-Rodrigues J, Perez RO, Camargo AA. Effect of Akt activation and experimental pharmacological inhibition on responses to neoadjuvant chemoradiotherapy in rectal cancer. Br J Surg 2018; 105:e192-e203. [PMID: 29341150 DOI: 10.1002/bjs.10695] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (CRT) is one of the preferred initial treatment strategies for locally advanced rectal cancer. Responses are variable, and most patients still require surgery. The aim of this study was to identify molecular mechanisms determining poor response to CRT. METHODS Global gene expression and pathway enrichment were assessed in pretreatment biopsies from patients with non-metastatic cT2-4 N0-2 rectal cancer within 7 cm of the anal verge. Downstream Akt activation was assessed in an independent set of pretreatment biopsies and in colorectal cancer cell lines using immunohistochemistry and western blot respectively. The radiosensitizing effects of the Akt inhibitor MK2206 were assessed using clonogenic assays and xenografts in immunodeficient mice. RESULTS A total of 350 differentially expressed genes were identified, of which 123 were upregulated and 199 downregulated in tumours from poor responders. Mitochondrial oxidative phosphorylation (P < 0·001) and phosphatidylinositol signalling pathways (P < 0·050) were identified as significantly enriched pathways among the set of differentially expressed genes. Deregulation of both pathways is known to result in Akt activation, and high immunoexpression of phosphorylated Akt S473 was observed among patients with a poor histological response (tumour regression grade 0-2) to CRT (75 per cent versus 48 per cent in those with a good or complete response; P = 0·016). Akt activation was also confirmed in the radioresistant cell line SW480, and a 50 per cent improvement in sensitivity to CRT was observed in vitro and in vivo when SW480 cells were exposed to the Akt inhibitor MK2206 in combination with radiation and 5-fluorouracil. CONCLUSION Akt activation is a key event in the response to CRT. Pharmacological inhibition of Akt activation may enhance the effects of CRT. Surgical relevance Organ preservation is an attractive alternative in rectal cancer management following neoadjuvant chemoradiotherapy (CRT) to avoid the morbidity of radical surgery. Molecular steps associated with tumour response to CRT may provide a useful tool for the identification of patients who are candidates for no immediate surgery. In this study, tumours resistant to CRT were more likely to have activation of specific genetic pathways that result in phosphorylated Akt (pAkt) activation. Pretreatment biopsy tissues with high immunoexpression of pAkt were more likely to exhibit a poor histological response to CRT. In addition, the introduction of a pAkt inhibitor to cancer cell lines in vitro and in vivo led to a significant improvement in sensitivity to CRT. Identification of pAkt-activated tumours may thus allow the identification of poor responders to CRT. In addition, the concomitant use of pAkt inhibitors to increase sensitivity to CRT in patients with rectal cancer may constitute an interesting strategy for increasing the chance of a complete response to treatment and organ preservation.
Collapse
Affiliation(s)
- F C Koyama
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil.,Ludwig Institute for Cancer Research, São Paulo, Brazil
| | - C M Lopes Ramos
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil
| | - F Ledesma
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - V A F Alves
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - J M Fernandes
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil
| | - B B Vailati
- Instituto Angelita and Joaquim Gama, São Paulo, Brazil
| | | | - A Habr-Gama
- Instituto Angelita and Joaquim Gama, São Paulo, Brazil.,Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - J Gama-Rodrigues
- Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - R O Perez
- Ludwig Institute for Cancer Research, São Paulo, Brazil.,Instituto Angelita and Joaquim Gama, São Paulo, Brazil.,Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.,Digestive Surgical Oncology Division, BP - A Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | - A A Camargo
- Molecular Oncology Centre, Hospital Sírio Libanês, São Paulo, Brazil.,Ludwig Institute for Cancer Research, São Paulo, Brazil
| |
Collapse
|
31
|
Song KH, Kim JH, Lee YH, Bae HC, Lee HJ, Woo SR, Oh SJ, Lee KM, Yee C, Kim BW, Cho H, Chung EJ, Chung JY, Hewitt SM, Chung TW, Ha KT, Bae YK, Mao CP, Yang A, Wu T, Kim TW. Mitochondrial reprogramming via ATP5H loss promotes multimodal cancer therapy resistance. J Clin Invest 2018; 128:4098-4114. [PMID: 30124467 PMCID: PMC6118592 DOI: 10.1172/jci96804] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 06/28/2018] [Indexed: 01/12/2023] Open
Abstract
The host immune system plays a pivotal role in the emergence of tumor cells that are refractory to multiple clinical interventions including immunotherapy, chemotherapy, and radiotherapy. Here, we examined the molecular mechanisms by which the immune system triggers cross-resistance to these interventions. By examining the biological changes in murine and tumor cells subjected to sequential rounds of in vitro or in vivo immune selection via cognate cytotoxic T lymphocytes, we found that multimodality resistance arises through a core metabolic reprogramming pathway instigated by epigenetic loss of the ATP synthase subunit ATP5H, which leads to ROS accumulation and HIF-1α stabilization under normoxia. Furthermore, this pathway confers to tumor cells a stem-like and invasive phenotype. In vivo delivery of antioxidants reverses these phenotypic changes and resensitizes tumor cells to therapy. ATP5H loss in the tumor is strongly linked to failure of therapy, disease progression, and poor survival in patients with cancer. Collectively, our results reveal a mechanism underlying immune-driven multimodality resistance to cancer therapy and demonstrate that rational targeting of mitochondrial metabolic reprogramming in tumor cells may overcome this resistance. We believe these results hold important implications for the clinical management of cancer.
Collapse
Affiliation(s)
- Kwon-Ho Song
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
- Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Young-Ho Lee
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
- Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Hyun Cheol Bae
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Hyo-Jung Lee
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
- Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Seon Rang Woo
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
- Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Se Jin Oh
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
- Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul, South Korea
| | - Kyung-Mi Lee
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
| | - Cassian Yee
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bo Wook Kim
- Department of Obstetrics and Gynecology, International St. Mary’s Hospital, Catholic Kwandong University College of Medicine, Incheon, Seoul, South Korea
| | - Hanbyoul Cho
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Stephen M. Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Tae-Wook Chung
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, South Korea
| | - Ki-Tae Ha
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan, South Korea
| | - Young-Ki Bae
- Comparative Biomedicine Research Branch, Research Institute, National Cancer Center, Goyang, South Korea
| | - Chih-Ping Mao
- MD-PhD Program
- Immunology Training Program
- Department of Pathology
| | | | - T.C. Wu
- Department of Pathology
- Department of Oncology
- Department of Obstetrics and Gynecology, and
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Tae Woo Kim
- Department of Biochemistry and Molecular Biology
- Department of Biomedical Science, College of Medicine, and
- Translational Research Institute for Incurable Diseases, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Esparza-Moltó PB, Cuezva JM. The Role of Mitochondrial H +-ATP Synthase in Cancer. Front Oncol 2018; 8:53. [PMID: 29564224 PMCID: PMC5845864 DOI: 10.3389/fonc.2018.00053] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/20/2018] [Indexed: 01/23/2023] Open
Abstract
Cancer cells reprogram energy metabolism by boosting aerobic glycolysis as a main pathway for the provision of metabolic energy and of precursors for anabolic purposes. Accordingly, the relative expression of the catalytic subunit of the mitochondrial H+-ATP synthase—the core hub of oxidative phosphorylation—is downregulated in human carcinomas when compared with its expression in normal tissues. Moreover, some prevalent carcinomas also upregulate the ATPase inhibitory factor 1 (IF1), which is the physiological inhibitor of the H+-ATP synthase. IF1 overexpression, both in cells in culture and in tissue-specific mouse models, is sufficient to reprogram energy metabolism to an enhanced glycolysis by limiting ATP production by the H+-ATP synthase. Furthermore, the IF1-mediated inhibition of the H+-ATP synthase promotes the production of mitochondrial ROS (mtROS). mtROS modulate signaling pathways favoring cellular proliferation and invasion, the activation of antioxidant defenses, resistance to cell death, and modulation of the tissue immune response, favoring the acquisition of several cancer traits. Consistently, IF1 expression is an independent marker of cancer prognosis. By contrast, inhibition of the H+-ATP synthase by α-ketoglutarate and the oncometabolite 2-hydroxyglutarate, reduces mTOR signaling, suppresses cancer cell growth, and contributes to lifespan extension in several model organisms. Hence, the H+-ATP synthase appears as a conserved hub in mitochondria-to-nucleus signaling controlling cell fate. Unraveling the molecular mechanisms responsible for IF1 upregulation in cancer and the signaling cascades that are modulated by the H+-ATP synthase are of utmost interest to decipher the metabolic and redox circuits contributing to cancer origin and progression.
Collapse
Affiliation(s)
- Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre (i+12), Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre (i+12), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
33
|
Bai B, Xie B, Pan Z, Shan L, Zhao J, Zhu H. Identification of candidate genes and long non-coding RNAs associated with the effect of ATP5J in colorectal cancer. Int J Oncol 2018; 52:1129-1138. [PMID: 29484395 PMCID: PMC5843394 DOI: 10.3892/ijo.2018.4281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/15/2018] [Indexed: 12/27/2022] Open
Abstract
The incidence and development of colorectal cancer (CRC) is a process with multiple gene interactions. We have previously demonstrated that ATP synthase-coupling factor 6, mitochondrial (ATP5J) is associated with CRC migration and 5-fluorouracil resistance; nevertheless, the exact molecular mechanism remains unclear. The following study uses microarray and bioinformatics methods to identify candidate genes and long non-coding RNAs (lncRNAs) in CRC cells (two pairs) with upregulated and downregulated ATP5J. Briefly, a total of 2,190 differentially expressed mRNAs (DEmRNAs) were sorted. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed for 4 DEmRNAs to validate the results of microarray analysis. Functional annotation and pathway enrichment were analyzed for DEmRNAs using the Database for Annotation, Visualization and Integrated Discovery. Significantly enriched pathways included the regulation of gene expression and cell growth. The protein-protein interaction network was constructed, and AKT serine/threonine kinase 2 (AKT2) was considered as one of the hub genes. For further analysis, 51 DEmRNAs and 30 DElncRNAs were selected that were positively or negatively associated with the expression of ATP5J in the two cell pairs. X-inactive specific transcript (XIST), premature ovarian failure 1B (POF1B) and calmin (CLMN) were identified in the DEmRNA-DElncRNA co-expression network. The expression of AKT2 and XIST in CRC cells was confirmed by RT-qPCR. To sum up, the candidate genes and lncRNAs, as well as potential signaling pathways, which were identified using integrated bioinformatics analysis, could improve the understanding of molecular events involved in the function of ATP5J in CRC.
Collapse
Affiliation(s)
- Bingjun Bai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang 310016, P.R. China
| | - Binbin Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang 310016, P.R. China
| | - Zongyou Pan
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Lina Shan
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang 310016, P.R. China
| | - Jianpei Zhao
- Department of Colorectal Surgery, No. 2 Hospital of Ningbo, Ningbo, Zhejiang 315010, P.R. China
| | - Hongbo Zhu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Zhejiang 310016, P.R. China
| |
Collapse
|
34
|
Yoo BC, Yeo SG. Identification of CEA-interacting proteins in colon cancer cells and their changes in expression after irradiation. Radiat Oncol J 2017; 35:281-288. [PMID: 28881503 PMCID: PMC5654139 DOI: 10.3857/roj.2017.00255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/16/2017] [Accepted: 07/31/2017] [Indexed: 01/16/2023] Open
Abstract
Purpose The serum carcinoembryonic antigen (CEA) level has been recognized as a prognostic factor in colorectal cancer, and associated with response of rectal cancer to radiotherapy. This study aimed to identify CEA-interacting proteins in colon cancer cells and observe post-irradiation changes in their expression. Materials and Methods CEA expression in colon cancer cells was examined by Western blot analysis. Using an anti-CEA antibody or IgG as a negative control, immunoprecipitation was performed in colon cancer cell lysates. CEA and IgG immunoprecipitates were used for liquid chromatography–tandem mass spectrometry (LC-MS/MS) analysis. Proteins identified in the CEA immunoprecipitates but not in the IgG immunoprecipitates were selected as CEA-interacting proteins. After radiation treatment, changes in expression of CEA-interacting proteins were monitored by Western blot analysis. Results CEA expression was higher in SNU-81 cells compared with LoVo cells. The membrane localization of CEA limited the immunoprecipitation results and thus the number of CEA-interacting proteins identified. Only the Ras-related protein Rab-6B and lysozyme C were identified as CEA-interacting proteins in LoVo and SNU-81 cells, respectively. Lysozyme C was detected only in SNU-81, and CEA expression was differently regulated in two cell lines; it was down-regulated in LoVo but up-regulated in SNU-81 in radiation dosage-dependent manner. Conclusion CEA-mediated radiation response appears to vary, depending on the characteristics of individual cancer cells. The lysozyme C and Rab subfamily proteins may play a role in the link between CEA and tumor response to radiation, although further studies are needed to clarify functional roles of the identified proteins.
Collapse
Affiliation(s)
- Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang, Korea
| | - Seung-Gu Yeo
- Department of Radiation Oncology, Soonchunhyang University College of Medicine, Soonchunhyang University Hospital, Cheonan, Korea
| |
Collapse
|
35
|
Metformin increases chemo-sensitivity via gene downregulation encoding DNA replication proteins in 5-Fu resistant colorectal cancer cells. Oncotarget 2017; 8:56546-56557. [PMID: 28915611 PMCID: PMC5593582 DOI: 10.18632/oncotarget.17798] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/27/2017] [Indexed: 12/16/2022] Open
Abstract
Metformin is most widely prescribed for type 2 diabetes. Recently, evidences have shown that metformin has anticancer effects on pancreatic-, colorectal-, ovarian-, and other cancers. Because metformin has less adverse effects and is inexpensive, it could be a useful chemo-therapeutic agent with anticancer effects. In this study, we demonstrated metformin inhibited by cell proliferation, cell migration ability, clonogenic ability, and cancer stem cell population. Metformin also induced cell cycle arrest in parental-(SNU-C5), and 5-Fu resistant-colorectal cancer cell line (SNU-C5_5FuR). Moreover, a treatment that combines 5-Fu and metformin was found to have a synergistic effect on the cell proliferation rate, especially in SNU-C5_5FuR, which was mediated by the activation of AMPK pathway and NF-ƙB pathway, well-known metformin mechanisms. In this study, we suggested novel anticancer mechanism of metformin that inhibited DNA replication machinery, such as the MCM family in SNU-C5_5FuR. In conclusion, we provided that how metformin acts as not only a chemo-sensitizer, but also as a synergistic effector of 5-Fu in the 5-Fu resistant-cell line. We speculate that metformin used for adjuvant therapy is effective on 5-Fu resistant cancer cells.
Collapse
|
36
|
He J, Pei L, Jiang H, Yang W, Chen J, Liang H. Chemoresistance of colorectal cancer to 5-fluorouracil is associated with silencing of the BNIP3 gene through aberrant methylation. J Cancer 2017; 8:1187-1196. [PMID: 28607593 PMCID: PMC5463433 DOI: 10.7150/jca.18171] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 01/12/2017] [Indexed: 12/27/2022] Open
Abstract
Purpose To investigate the correlation between chemoresistance of colorectal cancer to 5-fluorouracil and BNIP3 and the underlying mechanism. Methods BNIP3 protein in specimens was evaluated using immunohistochemistry. Semi-quantitative reverse transcription PCR and Western blot was employed to assay gene expression. The promoter methylation status of BNIP3 was examined by methylation-specific PCR. Drug sensitivity was assayed using MTT assay. Results Specimens from 81 patients with colorectal cancer receiving 5-fluorouracil-based chemotherapy were analyzed. BNIP3 expression was negative in 42 cancer samples. The mean score of BNIP3 in cancer was 1.8±0.2 and it was 3.7±0.5 in adjacent colorectum (p<0.05). The response rate of the BNIP3 positive group was 63.6% and that of the negative group was 36.4% (p=0.021). The median PFS of the BNIP3 positive group was 9.25 months and that of the BNIP3 negative group was 6.5 months (p=0.011). BNIP3 mRNA was not detectable in 4 of 8 colorectal cell lines and all these 4 cell lines displayed BNIP3 methylated allele only. Other 4 cell lines what expressed detectable BNIP3 displayed BNIP3 unmethylated allele only or both unmethylated and methylated alleles. 5-Aza dramatically increased BNIP3 expression. Knockdown of DNMT1 increased BNIP3. Knockdown of DNMT3B alone did not detectably change BNIP3 expression while knockdown of both DNMT1 and DNMT3B increased BNIP3 expression more than knockdown of DNMT1 alone. Knockdown of BNIP3 decreased chemosensitivity to 5-fluorouracil and increasing BNIP3 through demethylation increased chemosensitivity. Conclusion Chemoresistance of colorectal cancer to 5-fluorouracil is associated with silencing of the BNIP3 gene through aberrant methylation via DNMT1/DNMT3B.
Collapse
Affiliation(s)
- Jianming He
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China.,Department Of Oncology, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang 050011, China
| | - Li Pei
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Heng Jiang
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Weiwen Yang
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Jianfang Chen
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| | - Houjie Liang
- Department Of Oncology And Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038 China
| |
Collapse
|
37
|
Dumas JF, Brisson L, Chevalier S, Mahéo K, Fromont G, Moussata D, Besson P, Roger S. Metabolic reprogramming in cancer cells, consequences on pH and tumour progression: Integrated therapeutic perspectives with dietary lipids as adjuvant to anticancer treatment. Semin Cancer Biol 2017; 43:90-110. [DOI: 10.1016/j.semcancer.2017.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 02/07/2023]
|
38
|
Xiaoyun X, Chaofei H, Weiqi Z, Chen C, Lixia L, Queping L, Cong P, Shuang Z, Juan S, Xiang C. Possible Involvement of F1F0-ATP synthase and Intracellular ATP in Keratinocyte Differentiation in normal skin and skin lesions. Sci Rep 2017; 7:42672. [PMID: 28209970 PMCID: PMC5314331 DOI: 10.1038/srep42672] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 01/11/2017] [Indexed: 12/15/2022] Open
Abstract
The F1F0-ATP synthase, an enzyme complex, is mainly located on the mitochondrial inner membrane or sometimes cytomembrane to generate or hydrolyze ATP, play a role in cell proliferation. This study focused on the role of F1F0-ATP synthase in keratinocyte differentiation, and its relationship with intracellular and extracellular ATP (InATP and ExATP). The F1F0-ATP synthase β subunit (ATP5B) expression in various skin tissues and confluence-dependent HaCaT differentiation models was detected. ATP5B expression increased with keratinocyte and HaCaT cell differentiation in normal skin, some epidermis hyper-proliferative diseases, squamous cell carcinoma, and the HaCaT cell differentiation model. The impact of InATP and ExATP content on HaCaT differentiation was reflected by the expression of the differentiation marker involucrin. Inhibition of F1F0-ATP synthase blocked HaCaT cell differentiation, which was associated with a decrease of InATP content, but not with changes of ExATP. Our results revealed that F1F0-ATP synthase expression is associated with the process of keratinocyte differentiation which may possibly be related to InATP synthesis.
Collapse
Affiliation(s)
- Xie Xiaoyun
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Department of Rheumatology and Immunology, XiangYa Hospital, Central South University, Changsha, China
| | - Han Chaofei
- Department of Plastic and Reconstructive Surgery, The Third XiangYa Hospital, Central South University, Changsha, China
| | - Zeng Weiqi
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| | - Chen Chen
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Department of Nephrology, XiangYa Hospital, Central South University, Changsha, China
| | - Lu Lixia
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| | - Liu Queping
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| | - Peng Cong
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| | - Zhao Shuang
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| | - Su Juan
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| | - Chen Xiang
- Department of Dermatology, XiangYa Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Skin Cancer and Psoriasis, XiangYa Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Wen R, Dhar S. Turn up the cellular power generator with vitamin E analogue formulation. Chem Sci 2016; 7:5559-5567. [PMID: 30034696 PMCID: PMC6022097 DOI: 10.1039/c6sc00481d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 05/07/2016] [Indexed: 01/22/2023] Open
Abstract
The down regulation of the cellular power generator, adenosine triphosphate (ATP) synthase, in various cancer cells plays an obstructive role in mitochondria-mediated cell death. Cancer cells up-regulate ATPase inhibitory factor 1 (IF1) and down-regulate β-F1-ATPase of ATP synthase to enhance aerobic glycolysis for tumor growth via inhibiting total ATP synthase activity in the oxidative phosphorylation (OXPHOS) pathway. Alpha-tocopheryl succinate (α-TOS), one of the most bioactive derivatives of vitamin E, can selectively induce apoptosis in numerous cancer cells. The cancer cell selective apoptosis inducing property of α-TOS is correlated to: mitochondrial destabilization, inhibition of anti-apoptotic B cell lymphoma 2 (Bcl2) and protein kinase C (PKC), caspase 3 activation, production of mitochondrial reactive oxygen species (ROS), and inhibition of succinate dehydrogenase activity of mitochondrial complex II, and interaction with complex I to some extent. There is no report which elucidates the effects of α-TOS on the cellular power generator, complex V or ATP synthase. Here, we report the activation of mitochondrial ATP synthase using a suitably designed chemical formulation of α-TOS for the first time. A mitochondria targeted α-TOS nanoparticle formulation demonstrated enhanced cytotoxicity and mitochondrial activities in cancer cells by inhibiting Bcl2 protein and activating ATP synthase. The modulation of ATP synthase in cancer cells by the engineered formulation of α-TOS can be promising for solid cancers with compromised ATP synthase.
Collapse
Affiliation(s)
- Ru Wen
- NanoTherapeutics Research Laboratory , Department of Chemistry , University of Georgia , Room 679 , Athens , GA 30602 , USA . ; ; Tel: +1-706-542-1012 ; http://shanta.uga.edu/
| | - Shanta Dhar
- NanoTherapeutics Research Laboratory , Department of Chemistry , University of Georgia , Room 679 , Athens , GA 30602 , USA . ; ; Tel: +1-706-542-1012 ; http://shanta.uga.edu/
| |
Collapse
|
40
|
Song K, Li M, Xu X, Xuan LI, Huang G, Liu Q. Resistance to chemotherapy is associated with altered glucose metabolism in acute myeloid leukemia. Oncol Lett 2016; 12:334-342. [PMID: 27347147 DOI: 10.3892/ol.2016.4600] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/08/2016] [Indexed: 12/18/2022] Open
Abstract
Altered glucose metabolism has been described as a cause of chemoresistance in multiple tumor types. The present study aimed to identify the expression profile of glucose metabolism in drug-resistant acute myeloid leukemia (AML) cells and provide potential strategies for the treatment of drug-resistant AML. Bone marrow and serum samples were obtained from patients with AML that were newly diagnosed or had relapsed. The messenger RNA expression of hypoxia inducible factor (HIF)-1α, glucose transporter (GLUT)1, and hexokinase-II was measured by quantitative polymerase chain reaction. The levels of LDH and β subunit of human F1-F0 adenosine triphosphate synthase (β-F1-ATPase) were detected by enzyme-linked immunosorbent and western blot assays. The HL-60 and HL-60/ADR cell lines were used to evaluate glycolytic activity and effect of glycolysis inhibition on cellular proliferation and apoptosis. Drug-resistant HL-60/ADR cells exhibited a significantly increased level of glycolysis compared with the drug-sensitive HL-60 cell line. The expression of HIF-1α, hexokinase-II, GLUT1 and LDH were increased in AML patients with no remission (NR), compared to healthy control individuals and patients with complete remission (CR) and partial remission. The expression of β-F1-ATPase in patients with NR was decreased compared with the expression in the CR group. Treatment of HL-60/ADR cells with 2-deoxy-D-glucose or 3-bromopyruvate increased in vitro sensitivity to Adriamycin (ADR), while treatment of HL-60 cells did not affect drug cytotoxicity. Subsequent to treatment for 24 h, apoptosis in these two cell lines showed no significant difference. However, glycolytic inhibitors in combination with ADR increased cellular necrosis. These findings indicate that increased glycolysis and low efficiency of oxidative phosphorylation may contribute to drug resistance. Targeting glycolysis is a viable strategy for modulating chemoresistance in AML.
Collapse
Affiliation(s)
- Kui Song
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, P.R. China; Department of Hematology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528400, P.R. China
| | - Min Li
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, P.R. China
| | - Xiaojun Xu
- Department of Hematology, Zhongshan City People's Hospital, Zhongshan, Guangdong 528400, P.R. China; Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - L I Xuan
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guinian Huang
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
41
|
Hsu CC, Tseng LM, Lee HC. Role of mitochondrial dysfunction in cancer progression. Exp Biol Med (Maywood) 2016; 241:1281-95. [PMID: 27022139 DOI: 10.1177/1535370216641787] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Deregulated cellular energetics was one of the cancer hallmarks. Several underlying mechanisms of deregulated cellular energetics are associated with mitochondrial dysfunction caused by mitochondrial DNA mutations, mitochondrial enzyme defects, or altered oncogenes/tumor suppressors. In this review, we summarize the current understanding about the role of mitochondrial dysfunction in cancer progression. Point mutations and copy number changes are the two most common mitochondrial DNA alterations in cancers, and mitochondrial dysfunction induced by chemical depletion of mitochondrial DNA or impairment of mitochondrial respiratory chain in cancer cells promotes cancer progression to a chemoresistance or invasive phenotype. Moreover, defects in mitochondrial enzymes, such as succinate dehydrogenase, fumarate hydratase, and isocitrate dehydrogenase, are associated with both familial and sporadic forms of cancer. Deregulated mitochondrial deacetylase sirtuin 3 might modulate cancer progression by regulating cellular metabolism and oxidative stress. These mitochondrial defects during oncogenesis and tumor progression activate cytosolic signaling pathways that ultimately alter nuclear gene expression, a process called retrograde signaling. Changes in the intracellular level of reactive oxygen species, Ca(2+), or oncometabolites are important in the mitochondrial retrograde signaling for neoplastic transformation and cancer progression. In addition, altered oncogenes/tumor suppressors including hypoxia-inducible factor 1 and tumor suppressor p53 regulate mitochondrial respiration and cellular metabolism by modulating the expression of their target genes. We thus suggest that mitochondrial dysfunction plays a critical role in cancer progression and that targeting mitochondrial alterations and mitochondrial retrograde signaling might be a promising strategy for the development of selective anticancer therapy.
Collapse
Affiliation(s)
- Chia-Chi Hsu
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan Department of Surgery, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan Taipei-Veterans General Hospital Comprehensive Breast Health Center, Taipei 112, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| |
Collapse
|
42
|
Inside the biochemical pathways of thymidylate synthase perturbed by anticancer drugs: Novel strategies to overcome cancer chemoresistance. Drug Resist Updat 2015; 23:20-54. [PMID: 26690339 DOI: 10.1016/j.drup.2015.10.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 10/08/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
Abstract
Our current understanding of the mechanisms of action of antitumor agents and the precise mechanisms underlying drug resistance is that these two processes are directly linked. Moreover, it is often possible to delineate chemoresistance mechanisms based on the specific mechanism of action of a given anticancer drug. A more holistic approach to the chemoresistance problem suggests that entire metabolic pathways, rather than single enzyme targets may better explain and educate us about the complexity of the cellular responses upon cytotoxic drug administration. Drugs, which target thymidylate synthase and folate-dependent enzymes, represent an important therapeutic arm in the treatment of various human malignancies. However, prolonged patient treatment often provokes drug resistance phenomena that render the chemotherapeutic treatment highly ineffective. Hence, strategies to overcome drug resistance are primarily designed to achieve either enhanced intracellular drug accumulation, to avoid the upregulation of folate-dependent enzymes, and to circumvent the impairment of DNA repair enzymes which are also responsible for cross-resistance to various anticancer drugs. The current clinical practice based on drug combination therapeutic regimens represents the most effective approach to counteract drug resistance. In the current paper, we review the molecular aspects of the activity of TS-targeting drugs and describe how such mechanisms are related to the emergence of clinical drug resistance. We also discuss the current possibilities to overcome drug resistance by using a molecular mechanistic approach based on medicinal chemistry methods focusing on rational structural modifications of novel antitumor agents. This paper also focuses on the importance of the modulation of metabolic pathways upon drug administration, their analysis and the assessment of their putative roles in the networks involved using a meta-analysis approach. The present review describes the main pathways that are modulated by TS-targeting anticancer drugs starting from the description of the normal functioning of the folate metabolic pathway, through the protein modulation occurring upon drug delivery to cultured tumor cells as well as cancer patients, finally describing how the pathways are modulated by drug resistance development. The data collected are then analyzed using network/netwire connecting methods in order to provide a wider view of the pathways involved and of the importance of such information in identifying additional proteins that could serve as novel druggable targets for efficacious cancer therapy.
Collapse
|
43
|
TAOKA Y, MATSUMOTO K, OHASHI K, MINAMIDA S, HAGIWARA M, NAGI S, SAITO T, KODERA Y, IWAMURA M. Protein expression profile related to cisplatin resistance in bladder cancer cell lines detected by two-dimensional gel electrophoresis . Biomed Res 2015; 36:253-61. [DOI: 10.2220/biomedres.36.253] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Yoshinori TAOKA
- Department of Urology, Kitasato University School of Medicine
| | | | - Kazuya OHASHI
- Department of Physics, Kitasato University School of Science
| | - Satoru MINAMIDA
- Department of Urology, Kitasato University School of Medicine
| | | | - Shoji NAGI
- Department of Urology, Kitasato University School of Medicine
| | - Tatsuya SAITO
- Department of Physics, Kitasato University School of Science
| | - Yoshio KODERA
- Department of Physics, Kitasato University School of Science
| | | |
Collapse
|
44
|
Shang J, Yang F, Wang Y, Wang Y, Xue G, Mei Q, Wang F, Sun S. MicroRNA-23a antisense enhances 5-fluorouracil chemosensitivity through APAF-1/caspase-9 apoptotic pathway in colorectal cancer cells. J Cell Biochem 2014; 115:772-84. [PMID: 24249161 DOI: 10.1002/jcb.24721] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/14/2013] [Indexed: 12/18/2022]
Abstract
Current literature provided information that alteration in microRNA expression impacted sensitivity or resistance of certain tumor types to anticancer treatment, including the possible intracellular pathways. The microRNA-23a (miR-23a)-regulated apoptosis in response to the 5-fluorouracil (5-FU)-induced mitochondria-mediated apoptotic pathway was determined in this study. The miR-23a expression in 5-FU-treated and untreated colon cancer cells and tissues was assessed using real-time PCR analysis. To determine the function of miR-23a in the regulation of 5-FU-induced apoptosis, cell-proliferation, cytotoxicity, and apoptosis analyses were performed. Dual luciferase reporter assay was used to identify the apoptosis-related target gene for miR-23a. The activity of caspases-3, -7, and -9 were also assessed in miR-23a antisense and 5-FU treated tumor cells. A xenograft tumor model was established to evaluate the biological relevance of altered miR-23a expression to the 5-FU-based chemotherapy in vivo. We found that the expression of miR-23a was increased and the level of apoptosis-activating factor-1 (APAF-1) was decreased in 5-FU-treated colon cancer cells compared to untreated cells. The activation of the caspases-3 and 7 was increased in miR-23a antisense and 5-FU-treated colon cancer cells compared to negative control. APAF-1, as a target gene of miR-23a, was identified and miR-23a antisense-induced increase in the activation of caspase-9 was observed. The overexpression of miR-23a antisense up-regulated the 5-FU induced apoptosis in colon cancer cells. However, the miR-23a knockdown did not increase the antitumor effect of 5-FU in xenograft model of colon cancer. This study shows that miR-23a antisense enhanced 5-FU-induced apoptosis in colorectal cancer cells through the APAF-1/caspase-9 apoptotic pathway.
Collapse
Affiliation(s)
- Jingli Shang
- Department of Medical Genetics, Institute of Genetics, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Martínez-Reyes I, Cuezva JM. The H+-ATP synthase: A gate to ROS-mediated cell death or cell survival. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2014; 1837:1099-112. [DOI: 10.1016/j.bbabio.2014.03.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/03/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
|
46
|
Lee H, Kim C, Ku JL, Kim W, Yoon SK, Kuh HJ, Lee JH, Nam SW, Lee EK. A long non-coding RNA snaR contributes to 5-fluorouracil resistance in human colon cancer cells. Mol Cells 2014; 37:540-6. [PMID: 25078450 PMCID: PMC4132306 DOI: 10.14348/molcells.2014.0151] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 12/11/2022] Open
Abstract
Several types of genetic and epigenetic regulation have been implicated in the development of drug resistance, one significant challenge for cancer therapy. Although changes in the expression of non-coding RNA are also responsible for drug resistance, the specific identities and roles of them remain to be elucidated. Long non-coding RNAs (lncRNAs) are a type of ncRNA (> 200 nt) that influence the regulation of gene expression in various ways. In this study, we aimed to identify differentially expressed lncRNAs in 5-fluorouracil-resistant colon cancer cells. Using two pairs of 5-FU-resistant cells derived from the human colon cancer cell lines SNU-C4 and SNU-C5, we analyzed the expression of 90 lncRNAs by qPCR-based profiling and found that 19 and 23 lncRNAs were differentially expressed in SNU-C4R and SNU-C5R cells, respectively. We confirmed that snaR and BACE1AS were downregulated in resistant cells. To further investigate the effects of snaR on cell growth, cell viability and cell cycle were analyzed after transfection of siRNAs targeting snaR. Down-regulation of snaR decreased cell death after 5-FU treatment, which indicates that snaR loss decreases in vitro sensitivity to 5-FU. Our results provide an important insight into the involvement of lncRNAs in 5-FU resistance in colon cancer cells.
Collapse
Affiliation(s)
- Heejin Lee
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Chongtae Kim
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Ja-Lok Ku
- Cancer Research Institute and Cancer Research Center, Seoul National University, Seoul 110-744, Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 443-749, Korea
| | - Sungjoo Kim Yoon
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
- Department of Biomedical Science, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Hyo-Jeong Kuh
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
- Department of Biomedical Science, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Jeong-Hwa Lee
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
| | - Suk Woo Nam
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
- Department of Pathology, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, Catholic University of Korea, Seoul 137-701, Korea
- Cancer Evolution Research Center, Catholic University of Korea, Seoul 137-701, Korea
| |
Collapse
|
47
|
Short-term exposure of nontumorigenic human bronchial epithelial cells to carcinogenic chromium(VI) compromises their respiratory capacity and alters their bioenergetic signature. FEBS Open Bio 2014; 4:594-601. [PMID: 25161867 PMCID: PMC4141194 DOI: 10.1016/j.fob.2014.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 12/24/2022] Open
Abstract
Chromium(VI) impaired respiration and increased glycolytic flux in BEAS-2B cells. Cr(VI)-exposed cells shifted to a more fermentative metabolism. This metabolic shift was in line with a decreased β-F1-ATPase/GAPDH protein ratio. Increased oxidative stress levels suggest impairment of antioxidant defenses.
Previous studies on the impact of hexavalent chromium [Cr(VI)] on mammalian cell energetics revealed alterations suggestive of a shift to a more fermentative metabolism. Aiming at a more defined understanding of the metabolic effects of Cr(VI) and of their molecular basis, we assessed the impact of a mild Cr(VI) exposure on critical bioenergetic parameters (lactate production, oxygen consumption and intracellular ATP levels). Cells derived from normal human bronchial epithelium (BEAS-2B cell line), the main in vivo target of Cr(VI) carcinogenicity, were subjected for 48 h to 1 μM Cr(VI). We could confirm a shift to a more fermentative metabolism, resulting from the simultaneous inhibition of respiration and stimulation of glycolysis. This shift was accompanied by a decrease in the protein levels of the catalytic subunit (subunit β) of the mitochondrial H+-ATP synthase (β-F1-ATPase) and a concomitant marked increase in those of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). The corresponding alteration in the β-F1-ATPase/GAPDH protein ratio (viewed as a bioenergetic signature) upon Cr(VI) exposure was in agreement with the observed attenuation of cellular respiration and enhancement of glycolytic flux. Altogether, these results constitute a novel finding in terms of the molecular mechanisms of Cr(VI) effects.
Collapse
Key Words
- 2,4-DNP, 2,4-dinitrophenol
- 2-DG, 2-deoxyglucose
- Aerobic glycolysis
- Cellular bioenergetic index
- Cellular energy status
- Cellular respiration
- Chromate lung cancer
- Cr(III), trivalent chromium
- Cr(IV), tetravalent chromium
- Cr(V), pentavalent chromium
- Cr(VI), hexavalent chromium
- DCF, 2′,7′-dichlorofluorescein
- EDTA, ethylenediaminetetracetic acid
- ETC, mitochondrial electron transport chain
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- IARC, International Agency for Research on Cancer
- OCR, oxygen consumption rate
- OXPHOS, oxidative phosphorylation
- PBS, phosphate-buffered saline
- PI, propidium iodide
- ROS, reactive oxygen species
- TCA, tricarboxylic acid
- Warburg effect
- β-F1-ATPase, catalytic subunit (subunit β) of the mitochondrial H+-ATP synthase
Collapse
|
48
|
Lin LL, Huang HC, Juan HF. Deciphering molecular determinants of chemotherapy in gastrointestinal malignancy using systems biology approaches. Drug Discov Today 2014; 19:1402-9. [PMID: 24793142 DOI: 10.1016/j.drudis.2014.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/15/2014] [Accepted: 04/24/2014] [Indexed: 12/26/2022]
Abstract
Gastrointestinal cancers are asymptomatic in early tumor development, leading to high mortality rates. Peri- or postoperative chemotherapy is a common strategy used to prolong the life expectancy of patients with these diseases. Understanding the molecular mechanisms by which anticancer drugs exert their effect is crucial to the development of anticancer therapies, especially when drug resistance occurs and an alternative drug is needed. By integrating high-throughput techniques and computational modeling to explore biological systems at different levels, from gene expressions to networks, systems biology approaches have been successfully applied in various fields of cancer research. In this review, we highlight chemotherapy studies that reveal potential signatures using microarray analysis, next-generation sequencing (NGS), proteomic and metabolomic approaches for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Li-Ling Lin
- Institute of Molecular and Cellular Biology, Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan.
| | - Hsueh-Fen Juan
- Institute of Molecular and Cellular Biology, Department of Life Science, National Taiwan University, Taipei, Taiwan; Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan; Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan.
| | | |
Collapse
|
49
|
Formentini L, Pereira MP, Sánchez-Cenizo L, Santacatterina F, Lucas JJ, Navarro C, Martínez-Serrano A, Cuezva JM. In vivo inhibition of the mitochondrial H+-ATP synthase in neurons promotes metabolic preconditioning. EMBO J 2014; 33:762-78. [PMID: 24521670 PMCID: PMC4000092 DOI: 10.1002/embj.201386392] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 01/08/2014] [Accepted: 01/13/2014] [Indexed: 12/25/2022] Open
Abstract
A key transducer in energy conservation and signaling cell death is the mitochondrial H(+)-ATP synthase. The expression of the ATPase inhibitory factor 1 (IF1) is a strategy used by cancer cells to inhibit the activity of the H(+)-ATP synthase to generate a ROS signal that switches on cellular programs of survival. We have generated a mouse model expressing a mutant of human IF1 in brain neurons to assess the role of the H(+)-ATP synthase in cell death in vivo. The expression of hIF1 inhibits the activity of oxidative phosphorylation and mediates the shift of neurons to an enhanced aerobic glycolysis. Metabolic reprogramming induces brain preconditioning affording protection against quinolinic acid-induced excitotoxicity. Mechanistically, preconditioning involves the activation of the Akt/p70S6K and PARP repair pathways and Bcl-xL protection from cell death. Overall, our findings provide the first in vivo evidence highlighting the H(+)-ATP synthase as a target to prevent neuronal cell death.
Collapse
Affiliation(s)
- Laura Formentini
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| | - Marta P Pereira
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
| | - Laura Sánchez-Cenizo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| | - Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| | - José J Lucas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadrid, Spain
| | - Carmen Navarro
- Departamento de Patología y Neuropatología, Instituto de Investigación Biomédica de Vigo (IBIV)Vigo, Spain
| | - Alberto Martínez-Serrano
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM)Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIIIMadrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de MadridMadrid, Spain
| |
Collapse
|
50
|
Park JW, Kim SC, Kim WK, Hong JP, Kim KH, Yeo HY, Lee JY, Kim MS, Kim JH, Yang SY, Kim DY, Oh JH, Cho JY, Yoo BC. Expression of phosphoenolpyruvate carboxykinase linked to chemoradiation susceptibility of human colon cancer cells. BMC Cancer 2014; 14:160. [PMID: 24602180 PMCID: PMC4016284 DOI: 10.1186/1471-2407-14-160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 02/28/2014] [Indexed: 01/13/2023] Open
Abstract
Background Resistance to 5-fluorouracil (5-FU) in patients with colorectal cancer prevents effective treatment and leads to unnecessary and burdensome chemotherapy. Therefore, prediction of 5-FU resistance is imperative. Methods To identify the proteins linked to 5-FU resistance, two-dimensional gel electrophoresis-based proteomics was performed using the human colon cancer cell line SNU-C4R with induced 5-FU resistance. Proteins showing altered expression in SNU-C4R were identified by matrix-associated laser desorption/ionization–time-of-flight analysis, and their roles in susceptibility to 5-FU or radiation were evaluated in various cell lines by transfection of specific siRNA or creation of overexpression constructs. Changes in cellular signaling and expression of mitochondrial apoptotic factors were investigated by Western Blot analysis. A mitochondrial membrane potential probe (JC-1 dye) and a flow cytometry system were employed to determine the mitochondrial membrane potential. Finally, protein levels were determined by Western Blot analysis in tissues from 122 patients with rectal cancer to clarify whether each identified protein is a useful predictor of a chemoradiation response. Results We identified mitochondrial phosphoenolpyruvate carboxykinase (mPEPCK) as a candidate predictor of 5-FU resistance. PEPCK was downregulated in SNU-C4R compared with its parent cell line SNU-C4. Overexpression of mPEPCK did not significantly alter the susceptibility to either 5-FU or radiation. Suppression of mPEPCK led to a decrease in both the cellular level of phosphoenolpyruvate and the susceptibility to 5-FU and radiation. Furthermore, the cellular levels of phosphoenolpyruvate (an end product of PEPCK and a substrate of pyruvate kinase), phosphorylated AKT, and phosphorylated 4EBP1 were decreased significantly secondary to the mPEPCK suppression in SNU-C4. However, mPEPCK siRNA transfection induced changes in neither the mitochondrial membrane potential nor the expression levels of mitochondrial apoptotic factors such as Bax, Bcl-2, and Bad. Downregulation of total PEPCK was observed in tissues from patients with rectal cancer who displayed poor responses to preoperative 5-FU-based radiation therapy. Conclusion Our overall results demonstrate that mPEPCK is a useful predictor of a response to chemoradiotherapy in patients with rectal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Byong Chul Yoo
- Colorectal Cancer Branch, Research Institute, National Cancer Center, Goyang, Gyeonggi, Republic of Korea.
| |
Collapse
|