51
|
Ji X, Sun W, Lv C, Huang J, Zhang H. Circular RNAs Regulate Glucose Metabolism in Cancer Cells. Onco Targets Ther 2021; 14:4005-4021. [PMID: 34239306 PMCID: PMC8259938 DOI: 10.2147/ott.s316597] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Circular RNAs (circRNAs) were originally thought to result from RNA splicing errors. However, it has been shown that circRNAs can regulate cancer onset and progression in various ways. They can regulate cancer cell proliferation, differentiation, invasion, and metastasis. Moreover, they modulate glucose metabolism in cancer cells through different mechanisms such as directly regulating glycolytic enzymes and glucose transporter (GLUT) or indirectly regulating signal transduction pathways. In this review, we elucidate on the role of circRNAs in regulating glucose metabolism in cancer cells, which partly explains the pathogenesis of malignant tumors, and provides new therapeutic targets or new diagnostic and prognostic markers for human cancers.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Wei Sun
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Chengzhou Lv
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Jiapeng Huang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, People's Republic of China
| |
Collapse
|
52
|
Kim H, Woo J, Dan K, Lee KM, Jin MS, Park IA, Ryu HS, Han D. Quantitative Proteomics Reveals Knockdown of CD44 Promotes Proliferation and Migration in Claudin-Low MDA-MB-231 and Hs 578T Breast Cancer Cell Lines. J Proteome Res 2021; 20:3720-3733. [PMID: 34075748 DOI: 10.1021/acs.jproteome.1c00293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD44 is a transmembrane glycoprotein that can regulate the oncogenic process. This is known to be a marker of the claudin-low subtype of breast cancer, as well as a cancer stem cell marker. However, its functional regulatory roles are poorly understood in claudin-low breast cancer. To gain comprehensive insight into the function of CD44, we performed an in-depth tandem mass tag-based proteomic analysis of two claudin-low breast cancer cell lines (MDA-MB-231 and Hs 578T) transfected with CD44 siRNA. As a result, we observed that 2736 proteins were upregulated and 2172 proteins were downregulated in CD44-knockdown MDA-MB-231 cells. For Hs 578T CD44-knockdown cells, 412 proteins were upregulated and 443 were downregulated. Gene ontology and network analyses demonstrated that the suppression of this marker mediates significant functional alterations related to oncogenic cellular processes, including proliferation, metabolism, adhesion, and gene expression regulation. A functional study confirmed that CD44 knockdown inhibited proliferation by regulating the expression of genes related to cell cycle, translation, and transcription. Moreover, this promoted the expression of multiple cell adhesion-associated proteins and attenuated cancer cell migration. Finally, our proteomic study defines the landscape of the CD44-regulated proteome of claudin-low breast cancer cells, revealing changes that mediate cell proliferation and migration. Our proteomics data set has been deposited to the ProteomeXchange Consortium via the PRIDE repository with the data set identifier PXD015171.
Collapse
Affiliation(s)
- Hyeyoon Kim
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea.,Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Jongmin Woo
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United States
| | - Kisoon Dan
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Kyung-Min Lee
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Min-Sun Jin
- Department of Pathology, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Gyeonggi-do 14647, Korea
| | - In Ae Park
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea.,Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| |
Collapse
|
53
|
Zhang Y, Sun Y, Ding L, Shi W, Ding K, Zhu Y. Long Non-Coding RNA LINC00467 Correlates to Poor Prognosis and Aggressiveness of Breast Cancer. Front Oncol 2021; 11:643394. [PMID: 33996559 PMCID: PMC8113855 DOI: 10.3389/fonc.2021.643394] [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: 12/18/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022] Open
Abstract
Breast cancer remains the leading cause of female cancer-related mortalities worldwide. Long non-coding RNAs (LncRNAs) have been increasingly reported to play pivotal roles in tumorigenesis and cancer progression. Herein, we focused on LINC00467, which has never been studied in breast cancer. Silence of LINC00467 suppressed proliferation, migration, invasion and epithelial-to-mesenchymal transition (EMT) of breast cancer cells in vitro, whereas forced expression of LINC00467 exhibited the opposite effects. Furthermore, we demonstrated overexpression of LINC00467 promoted tumor growth, while knockdown of LINC00467 inhibited pulmonary metastasis in vivo. Mechanistically, LINC00467 down-regulated miR-138-5p by acting as a miRNA “sponge”. Besides, LINC00467 also up-regulated the protein level of lin-28 homolog B (LIN28B) via a direct interaction. A higher expression level of LINC00467 was observed in breast cancer tissues as compared to the adjacent normal counterparts and elevated LINC00467 predicted poor overall survival. Our findings suggest LINC00467 promotes progression of breast cancer through interacting with miR-138-5p and LIN28B directly. LINC00467 may serve as a potential candidate for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Oncology of the First Affiliated Hospital, Division of Life Science and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Yi Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Lin Ding
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Wenjing Shi
- Department of Naval Medicine, Naval Medical University, Shanghai, China
| | - Keshuo Ding
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Yong Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.,Department of Oncology of the First Affiliated Hospital, Division of Life Science and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| |
Collapse
|
54
|
The New Serum-Free OptiPASS ® Medium in Cold and Oxygen-Free Conditions: An Innovative Conservation Method for the Preservation of MDA-MB-231 Triple Negative Breast Cancer Spheroids. Cancers (Basel) 2021; 13:cancers13081945. [PMID: 33919619 PMCID: PMC8073891 DOI: 10.3390/cancers13081945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cancer spheroids are reproducible and relevant multicellular in vitro preclinical models. Thus, their use is required more and more for drug development processes in oncology in order to improve the prediction of anticancer drugs responses. Moreover, spheroid models allow for the reduction in animal experimentation, in accordance with the rule of Reduce, Refine, Replace (3Rs). In order to optimize and extend the use of these spheroid models, this works was focused on the development of an original methodology to keep these cancer spheroids in the long term. This innovative concept is based on a cold storage for up to 7 days of Triple-Negative Breast Cancer (TNBC) spheroids cultured in the synthetic serum-free OptiPASS® culture medium. Major spheroid characteristics could be preserved with this new conservation method, allowing their use in high throughput screening tests. Abstract Cancer spheroids are very effective preclinical models to improve anticancer drug screening. In order to optimize and extend the use of spheroid models, these works were focused on the development of a new storage concept to maintain these models in the longer term using the Triple-Negative Breast Cancer MDA-MB-231 spheroid models. The results highlight that the combination of a temperature of 4 °C and oxygen-free conditions allowed the spheroid characteristics of OptiPASS® serum-free culture medium to preserve the spheroid characteristics during 3-, 5- or 7-day-long storage. Indeed, after storage they were returned to normal culture conditions, with recovered spheroids presenting similar growth rates (recovery = 96.2%), viability (Live/Dead® profiles) and metabolic activities (recovery = 90.4%) compared to nonstored control spheroids. Likewise, both recovered spheroids (after storage) and nonstored controls presented the same response profiles as two conventional drugs, i.e., epirubicin and cisplatin, and two anti-PARP1 targeted drugs—i.e., olaparib and veliparib. This new original storage concept seems to induce a temporary stop in spheroid growth while maintaining their principal characteristics for further use. In this way, this innovative and simple storage concept may instigate future biological sample preservation strategies.
Collapse
|
55
|
Zare ME, Kansestani AN, Hemmati S, Mansouri K, Vaisi-Raygani A. The rate of aerobic glycolysis is a pivotal regulator of tumor progression. J Diabetes Metab Disord 2021; 20:523-531. [PMID: 34178852 DOI: 10.1007/s40200-021-00774-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/18/2021] [Indexed: 12/30/2022]
Abstract
Purpose Cancer cells depend on glucose metabolism via exclusive glycolysis pathway is named Aerobic glycolysis or Warburg effect. The aim of this study was investigation of different glucose accessibility conditions on the rate of Warburg effect and its impact on Hypoxia inducible factors-1 α (HIF-1 α)/vascular endothelium growth factor (VEGF) pathway in breast cancer cells lines. Methods MDA-MB-231 (Warburg phenomenon) and MCF-7 (oxidative) cell lines were cultured in DMEM and exposed to three different glucose accessibility medium for 48 h (5.5 mM as normal glucose (NG), 25 mM as high glucose (HG) and 2-Deoxyglucose (2-DG) as restricted glucose accessibility). Glucose uptake, intra/extracellular lactate and pyruvate, HIF-1α accumulation and vascular endothelium growth factor (VEGF) expression were evaluated by standard methods. Results Our results showed in NG condition both of cell lines produce lactate, but it was higher in MDA-MB-231. HG condition increased extracellular lactate in both cell lines especially in MCF-7 cells whereas intracellular lactate and pyruvate raised only in MCF-7. 2-DG decreased extracellular and intracellular lactate and pyruvate in both cell lines especially in MDA-MB-231. HIF-1α accumulation was detectable in NG condition in both cell lines. HG condition increased HIF-1α accumulation in MCF-7 cells but not in MDA-MB-231 and 2-DG decreased it in both call lines, especially in MDA-MB-231. Expression of VEGF had similar pattern with HIF-1α in different conditions. Conclusions Our findings revealed the rate of Warburg effect is an important indicator for tumor promotion and invasion due to its impacts on important transcription factors like HIF-1α.
Collapse
Affiliation(s)
- Mohammad Erfan Zare
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Atefeh Nasir Kansestani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shahrooz Hemmati
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Asad Vaisi-Raygani
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Daneshgah Avenue, Kermanshah, 67148-69914 Iran
| |
Collapse
|
56
|
Tan YQ, Zhang X, Zhang S, Zhu T, Garg M, Lobie PE, Pandey V. Mitochondria: The metabolic switch of cellular oncogenic transformation. Biochim Biophys Acta Rev Cancer 2021; 1876:188534. [PMID: 33794332 DOI: 10.1016/j.bbcan.2021.188534] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/25/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria, well recognized as the "powerhouse" of cells, are maternally inherited organelles with bacterial ancestry that play essential roles in a myriad of cellular functions. It has become profoundly evident that mitochondria regulate a wide array of cellular and metabolic functions, including biosynthetic metabolism, cell signaling, redox homeostasis, and cell survival. Correspondingly, defects in normal mitochondrial functioning have been implicated in various human malignancies. Cancer development involves the activation of oncogenes, inactivation of tumor suppressor genes, and impairment of apoptotic programs in cells. Mitochondria have been recognized as the site of key metabolic switches for normal cells to acquire a malignant phenotype. This review outlines the role of mitochondria in human malignancies and highlights potential aspects of mitochondrial metabolism that could be targeted for therapeutic development.
Collapse
Affiliation(s)
- Yan Qin Tan
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, Guangdong, PR China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China
| | - Xi Zhang
- Shenzhen Bay Laboratory, Shenzhen 518055, Guangdong, PR China
| | - Shuwei Zhang
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, Guangdong, PR China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei 230000, Anhui, PR China; The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230000, Anhui, PR China
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Sector-125, Noida 201313, India
| | - Peter E Lobie
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, Guangdong, PR China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China; Shenzhen Bay Laboratory, Shenzhen 518055, Guangdong, PR China.
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, Guangdong, PR China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, PR China.
| |
Collapse
|
57
|
Pérez-Treviño P, Aguayo-Millán CD, Santuario-Facio SK, Vela-Guajardo JE, Salazar E, Camacho-Morales A, Ortiz R, García N. Metastatic TNBC is closely associated with a fused mitochondrial morphology and a glycolytic and lipogenic metabolism. Biochem Cell Biol 2020; 99:447-456. [PMID: 33342359 DOI: 10.1139/bcb-2020-0439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Mitochondria modify their function and morphology to satisfy the bioenergetic demand of the cells. Cancer cells take advantage of these features to sustain their metabolic, proliferative, metastatic, and survival necessities. Understanding the morphological changes to mitochondria in the different grades of triple-negative breast cancer (TNBC) could help to design new treatments. Consequently, this research explored mitochondrial morphology and the gene expression of some proteins related to mitochondrial dynamics, as well as proteins associated with oxidative and non-oxidative metabolism in metastatic and non-metastatic TNBC. We found that mitochondrial morphology and metabolism are different in metastatic and non-metastatic TNBC. In metastatic TNBC, there is overexpression of genes related to mitochondrial dynamics, fatty-acid metabolism, and glycolysis. These features are accompanied by a fused mitochondrial morphology. By comparison, in non-metastatic TNBC, there is a stress-associated mitochondrial morphology with hyperfragmented mitochondria, accompanied by the upregulated expression of genes associated with the biogenesis of mitochondria; both of which are characteristics related to the higher production of reactive oxygen species observed in this cell line. These differences between metastatic and non-metastatic TNBC should provide a better understanding of metastasis and contribute to the development of improved specific and personalized therapies for TNBC.
Collapse
Affiliation(s)
- Perla Pérez-Treviño
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Medicina Cardiovascular y Metabólica, Nuevo Leon, Mexico
| | - Claudia D Aguayo-Millán
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Investigación en Cáncer, Nuevo Leon, Mexico
| | - Sandra K Santuario-Facio
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Investigación en Cáncer, Nuevo Leon, Mexico
| | - Jorge E Vela-Guajardo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Medicina Cardiovascular y Metabólica, Nuevo Leon, Mexico
| | - Esteban Salazar
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Medicina Cardiovascular y Metabólica, Nuevo Leon, Mexico
| | - Alberto Camacho-Morales
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Mexico.,Unidad de Neurometabolismo, Centro de Investigación y Desarrollo en Ciencias de la Salud, Universidad Autonoma de Nuevo Leon, Nuevo Leon, Mexico
| | - Rocío Ortiz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Investigación en Cáncer, Nuevo Leon, Mexico
| | - Noemí García
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, GIEE Medicina Cardiovascular y Metabólica, Nuevo Leon, Mexico
| |
Collapse
|
58
|
Hu Y, Xu W, Zeng H, He Z, Lu X, Zuo D, Qin G, Chen W. OXPHOS-dependent metabolic reprogramming prompts metastatic potential of breast cancer cells under osteogenic differentiation. Br J Cancer 2020; 123:1644-1655. [PMID: 32934344 PMCID: PMC7686370 DOI: 10.1038/s41416-020-01040-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/22/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Microcalcification is one of the most reliable clinical features of the malignancy risk of breast cancer, and it is associated with enhanced tumour aggressiveness and poor prognosis. However, its underlying molecular mechanism remains unclear. METHODS Clinical data were retrieved to analyse the association between calcification and bone metastasis in patients with breast cancer. Using multiple human breast cancer cell lines, the osteogenic cocktail model was established in vitro to demonstrate calcification-exacerbated metastasis. Migration and invasion characteristics were determined by wound healing and transwell migration. mRNA and protein expression were identified by quantitative PCR and western blotting. Metabolic alterations in breast cancer cells were evaluated using Seahorse Analyser. RESULTS The osteogenic differentiation of human breast cancer cells activated the classical TGF-β/Smad signalling pathway and the non-canonical MAPK pathway, which, in turn, exacerbated the progression of epithelial-mesenchymal transition (EMT). The metabolic programme switched to enhancing mitochondrial oxidative phosphorylation (OXPHOS) upon osteogenic differentiation. Rotenone was used to inhibit the OXPHOS complex during osteogenesis to block mitochondrial function, consequently reversing the EMT phenotype. CONCLUSIONS This study provides important insights into the mechanisms involved in breast cancer bone metastasis, and outlines a possible strategy to intervene in OXPHOS for the treatment of breast tumours.
Collapse
Affiliation(s)
- Yangling Hu
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Weimin Xu
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Hui Zeng
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Zilong He
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Xiao Lu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - Daming Zuo
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, 510515, Guangzhou, China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Weiguo Chen
- Department of Radiology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
59
|
Guerra ÂR, Soares BIG, Freire CSR, Silvestre AJD, Duarte MF, Duarte IF. Metabolic Effects of a Eucalyptus Bark Lipophilic Extract on Triple Negative Breast Cancer and Nontumor Breast Epithelial Cells. J Proteome Res 2020; 20:565-575. [PMID: 32975121 DOI: 10.1021/acs.jproteome.0c00559] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this work, untargeted metabolomics was used to unveil the impact of a Eucalyptus (Eucalyptus nitens) outer bark lipophilic extract on the metabolism of triple negative breast cancer (TNBC) and nontumor breast cells. Integrative analysis of culture medium, intracellular polar metabolites, and cellular lipids provided a comprehensive picture of cell metabolic adaptations, which enabled several hypotheses about the metabolic targets and pathways affected to be proposed. One of the most marked effects in MDA-MB-231 breast cancer cells, upon 48 h incubation with the E. nitens extract (15 μg/mL), was the enhancement of the NAD+/NADH ratio, likely reflecting a shift to mitochondrial respiration, which appeared to be fueled by amino acids and fatty acids resulting from hydrolysis of neutral lipids (triglycerides and cholesteryl esters). Contrastingly, in MCF-10A breast epithelial cells, the E. nitens extract appeared to intensify glycolysis and the tricarboxylic acid cycle (resulting in a decreased NAD+/NADH ratio), while having no effect on the cell lipid composition. This knowledge improves the current understanding of the biological activity of E. nitens bark extracts and is potentially useful to promote their development in the field of TNBC anticancer therapy.
Collapse
Affiliation(s)
- Ângela R Guerra
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal.,Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL), Instituto Politécnico de Beja (IPBeja), Apartado 6158, 7801-908 Beja, Portugal
| | - Belinda I G Soares
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carmen S R Freire
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Armando J D Silvestre
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Maria F Duarte
- Centro de Biotecnologia Agrícola e Agro-Alimentar do Alentejo (CEBAL), Instituto Politécnico de Beja (IPBeja), Apartado 6158, 7801-908 Beja, Portugal.,MED-Mediterranean Institute for Agriculture, Environment and Development, CEBAL, 7801-908 Beja, Portugal
| | - Iola F Duarte
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
60
|
Medina MÁ. Metabolic Reprogramming is a Hallmark of Metabolism Itself. Bioessays 2020; 42:e2000058. [PMID: 32939776 DOI: 10.1002/bies.202000058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/13/2020] [Indexed: 12/16/2022]
Abstract
The reprogramming of metabolism has been identified as one of the hallmarks of cancer. It is becoming more and more frequent to connect other diseases with metabolic reprogramming. This article aims to argue that metabolic reprogramming is not driven by disease but instead is the main hallmark of metabolism, based on its dynamic behavior that allows it to continuously adapt to changes in the internal and external conditions.
Collapse
Affiliation(s)
- Miguel Ángel Medina
- Andalucía Tech, Facultad de Ciencias, Departamento de Biología Molecular y Bioquímica, and IBIMA (Biomedical Research Institute of Málaga), Universidad de Málaga, Málaga, E-29071, Spain.,CIBER de Enfermedades Raras (CIBERER), Málaga, E-29071, Spain
| |
Collapse
|
61
|
Folic Acid Induces Intake-Related Changes in the Mammary Tissue Transcriptome of C57BL/6 Mice. Nutrients 2020; 12:nu12092821. [PMID: 32942660 PMCID: PMC7551343 DOI: 10.3390/nu12092821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 12/17/2022] Open
Abstract
Folic acid (FA) intake has been associated with increased breast cancer risk in some studies. Although underlying mechanisms are unknown, epigenetic modifications that persistently alter transcription have been suggested. We tested the hypothesis that high FA (HFA) intake alters the adult mammary transcriptome in a manner consistent with increased potential for carcinogenesis, detectable beyond the period of intake. C57BL/6 mice were fed control FA (CFA) (1 mg/kg diet) or HFA (5 mg/kg diet) diets for 4 weeks, followed by AIN93M maintenance diet for 4 weeks. Plasma 5-methyltetrahydrofolate, p-aminobenzoylglutamate and unmetabolised FA concentrations were greater (1.62, 1.56, 5.80-fold, respectively) in HFA compared to CFA mice. RNA sequencing of the mammary transcriptome (~20 million reads) showed 222 transcripts (191 upregulated) differentially expressed between groups. Gene Set Enrichment showed upregulated genes significantly enriched in Epithelial Mesenchymal Transition, Myogenesis and Apical Junction and downregulated genes in E2F targets, MYC targets and G2M checkpoint. Cancer was the most altered Disease and Disorder pathway, with Metastasis, Mammary Tumour and Growth of Tumour the most upregulated pathways. ChIP-seq enrichment analysis showed that targets of histone methyltransferase EZH2 were enriched in HFA mice. This study demonstrates HFA intake during adulthood induces mammary transcriptome changes, consistent with greater tumorigenic potential.
Collapse
|
62
|
Metabolic Constrains Rule Metastasis Progression. Cells 2020; 9:cells9092081. [PMID: 32932943 PMCID: PMC7563739 DOI: 10.3390/cells9092081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Metastasis formation accounts for the majority of tumor-associated deaths and consists of different steps, each of them being characterized by a distinctive adaptive phenotype of the cancer cells. Metabolic reprogramming represents one of the main adaptive phenotypes exploited by cancer cells during all the main steps of tumor and metastatic progression. In particular, the metabolism of cancer cells evolves profoundly through all the main phases of metastasis formation, namely the metastatic dissemination, the metastatic colonization of distant organs, the metastatic dormancy, and ultimately the outgrowth into macroscopic lesions. However, the metabolic reprogramming of metastasizing cancer cells has only recently become the subject of intense study. From a clinical point of view, the latter steps of the metastatic process are very important, because patients often undergo surgical removal of the primary tumor when cancer cells have already left the primary tumor site, even though distant metastases are not clinically detectable yet. In this scenario, to precisely elucidate if and how metabolic reprogramming drives acquisition of cancer-specific adaptive phenotypes might pave the way to new therapeutic strategies by combining chemotherapy with metabolic drugs for better cancer eradication. In this review we discuss the latest evidence that claim the importance of metabolic adaptation for cancer progression.
Collapse
|
63
|
Breast Cancer Subtypes Underlying EMT-Mediated Catabolic Metabolism. Cells 2020; 9:cells9092064. [PMID: 32927665 PMCID: PMC7563728 DOI: 10.3390/cells9092064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Efficient catabolic metabolism of adenosine triphosphate (ATP) and reduced nicotinamide adenine dinucleotide phosphate (NADPH) is essentially required for cancer cell survival, especially in metastatic cancer progression. Epithelial–mesenchymal transition (EMT) plays an important role in metabolic rewiring of cancer cells as well as in phenotypic conversion and therapeutic resistance. Snail (SNAI1), a well-known inducer of cancer EMT, is critical in providing ATP and NADPH via suppression of several gatekeeper genes involving catabolic metabolism, such as phosphofructokinase 1 (PFK1), fructose-1,6-bisphosphatase 1 (FBP1), and acetyl-CoA carboxylase 2 (ACC2). Paradoxically, PFK1 and FBP1 are counter-opposing and rate-limiting reaction enzymes of glycolysis and gluconeogenesis, respectively. In this study, we report a distinct metabolic circuit of catabolic metabolism in breast cancer subtypes. Interestingly, PFKP and FBP1 are inversely correlated in clinical samples, indicating different metabolic subsets of breast cancer. The luminal types of breast cancer consist of the pentose phosphate pathway (PPP) subset by suppression of PFKP while the basal-like subtype (also known as triple negative breast cancer, TNBC) mainly utilizes glycolysis and mitochondrial fatty acid oxidation (FAO) by loss of FBP1 and ACC2. Notably, PPP remains active via upregulation of TIGAR in the FBP1-loss basal-like subset, indicating the importance of PPP in catabolic cancer metabolism. These results indicate different catabolic metabolic circuits and thus therapeutic strategies in breast cancer subsets.
Collapse
|
64
|
Drosophila melanogaster Mitochondrial Carriers: Similarities and Differences with the Human Carriers. Int J Mol Sci 2020; 21:ijms21176052. [PMID: 32842667 PMCID: PMC7504413 DOI: 10.3390/ijms21176052] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial carriers are a family of structurally related proteins responsible for the exchange of metabolites, cofactors and nucleotides between the cytoplasm and mitochondrial matrix. The in silico analysis of the Drosophila melanogaster genome has highlighted the presence of 48 genes encoding putative mitochondrial carriers, but only 20 have been functionally characterized. Despite most Drosophila mitochondrial carrier genes having human homologs and sharing with them 50% or higher sequence identity, D. melanogaster genes display peculiar differences from their human counterparts: (1) in the fruit fly, many genes encode more transcript isoforms or are duplicated, resulting in the presence of numerous subfamilies in the genome; (2) the expression of the energy-producing genes in D. melanogaster is coordinated from a motif known as Nuclear Respiratory Gene (NRG), a palindromic 8-bp sequence; (3) fruit-fly duplicated genes encoding mitochondrial carriers show a testis-biased expression pattern, probably in order to keep a duplicate copy in the genome. Here, we review the main features, biological activities and role in the metabolism of the D. melanogaster mitochondrial carriers characterized to date, highlighting similarities and differences with their human counterparts. Such knowledge is very important for obtaining an integrated view of mitochondrial function in D. melanogaster metabolism.
Collapse
|
65
|
Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative Stress in Cancer. Cancer Cell 2020; 38:167-197. [PMID: 32649885 PMCID: PMC7439808 DOI: 10.1016/j.ccell.2020.06.001] [Citation(s) in RCA: 1465] [Impact Index Per Article: 293.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/29/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022]
Abstract
Contingent upon concentration, reactive oxygen species (ROS) influence cancer evolution in apparently contradictory ways, either initiating/stimulating tumorigenesis and supporting transformation/proliferation of cancer cells or causing cell death. To accommodate high ROS levels, tumor cells modify sulfur-based metabolism, NADPH generation, and the activity of antioxidant transcription factors. During initiation, genetic changes enable cell survival under high ROS levels by activating antioxidant transcription factors or increasing NADPH via the pentose phosphate pathway (PPP). During progression and metastasis, tumor cells adapt to oxidative stress by increasing NADPH in various ways, including activation of AMPK, the PPP, and reductive glutamine and folate metabolism.
Collapse
Affiliation(s)
- John D Hayes
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland.
| | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, Jacqui Wood Cancer Centre, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, UK, Scotland; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth D Tew
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
66
|
An insight on type I collagen from horse tendon for the manufacture of implantable devices. Int J Biol Macromol 2020; 154:291-306. [DOI: 10.1016/j.ijbiomac.2020.03.082] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/11/2020] [Indexed: 12/14/2022]
|
67
|
Yu D, Liu C, Guo L. Mitochondrial metabolism and cancer metastasis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:904. [PMID: 32793748 DOI: 10.21037/atm.2020.03.42] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Metastasis is regarded as the most important cause of cancer-related deaths around the world. During the complicated metastatic cascade, altered mitochondrial metabolism adapts to serve distinct conditions and microenvironments. In this review, we discuss how cells regulate their mitochondria metabolism to adapt to environmental cues during the metastasis, as well as how cancer cells and their tumor micro-environment (TME) are metabolically coupled during the metastatic cascade. We place a strong emphasis on how mitochondrial proline metabolism and extracellular matrix (ECM) are coupled.
Collapse
Affiliation(s)
- Dandan Yu
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies, and Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China.,Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Chang Liu
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies, and Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China.,Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Ling Guo
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Academy for Advanced Interdisciplinary Studies, and Department of Biology, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
68
|
Bajor M, Graczyk-Jarzynka A, Marhelava K, Kurkowiak M, Rahman A, Aura C, Russell N, Zych AO, Firczuk M, Winiarska M, Gallagher WM, Zagozdzon R. Triple Combination of Ascorbate, Menadione and the Inhibition of Peroxiredoxin-1 Produces Synergistic Cytotoxic Effects in Triple-Negative Breast Cancer Cells. Antioxidants (Basel) 2020; 9:antiox9040320. [PMID: 32316111 PMCID: PMC7222372 DOI: 10.3390/antiox9040320] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of mammary malignancy currently without satisfactory systemic treatment options. Agents generating reactive oxygen species (ROS), such as ascorbate (Asc) and menadione (Men), especially applied in combination, have been proposed as an alternative anticancer modality. However, their effectiveness can be hampered by the cytoprotective effects of elevated antioxidant enzymes (e.g., peroxiredoxins, PRDX) in cancer. In this study, PRDX1 mRNA and protein expression were assessed in TNBC tissues by analysis of the online RNA-seq datasets and immunohistochemical staining of tissue microarray, respectively. We demonstrated that PRDX1 mRNA expression was markedly elevated in primary TNBC tumors as compared to non-malignant controls, with PRDX1 protein staining intensity correlating with favorable survival parameters. Subsequently, PRDX1 functionality in TNBC cell lines or non-malignant mammary cells was targeted by genetic silencing or chemically by auranofin (AUR). The PRDX1-knockdown or AUR treatment resulted in inhibition of the growth of TNBC cells in vitro. These cytotoxic effects were further synergistically potentiated by the incubation with a combination of the prooxidant agents, Asc and Men. In conclusion, we report that the PRDX1-related antioxidant system is essential for maintaining redox homeostasis in TNBC cells and can be an attractive therapeutic target in combination with ROS-generating agents.
Collapse
Affiliation(s)
- Malgorzata Bajor
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.B.); (K.M.)
| | - Agnieszka Graczyk-Jarzynka
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - Katsiaryna Marhelava
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.B.); (K.M.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Trojdena 2a, 02-091 Warsaw, Poland
| | - Malgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdansk, Wita Stwosza 63, 80-308 Gdansk, Poland;
| | - Arman Rahman
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
| | - Claudia Aura
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
| | - Niamh Russell
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
| | - Agata O. Zych
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - Magdalena Winiarska
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5, 02-097 Warsaw, Poland; (A.G.-J.); (A.O.Z.); (M.F.); (M.W.)
| | - William M. Gallagher
- Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, D04 Dublin 4, Ireland; (A.R.); (C.A.); (N.R.); (W.M.G.)
- OncoMark Ltd., Nova UCD, D04 Dublin 4, Ireland
| | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland; (M.B.); (K.M.)
- Department of Immunology, Transplantology, and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Poland
- Correspondence:
| |
Collapse
|
69
|
Pan Z, Cai J, Lin J, Zhou H, Peng J, Liang J, Xia L, Yin Q, Zou B, Zheng J, Qiao L, Zhang L. A novel protein encoded by circFNDC3B inhibits tumor progression and EMT through regulating Snail in colon cancer. Mol Cancer 2020; 19:71. [PMID: 32241279 PMCID: PMC7114813 DOI: 10.1186/s12943-020-01179-5] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/05/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Colon cancer (CC) is a common malignant cancer. Recently, circFNDC3B was found to exert biological function in multiple cancers. However, it was unclear whether the potential protein encoded by circFNDC3B is involved in carcinogenesis of CC. METHODS We used Sanger sequence and RNase R digestion assay to confirm the existence of circFNDC3B, and quantitative real-time PCR was used to evaluate the circRNA's expression. Then fluorescence in situ hybridization (FISH) was performed to study location of circFNDC3B. The identification of protein encoded by circFNDC3B was performed using LC-MS/MS. The function of circFNDC3B-218aa on proliferation, invasion and migration were assessed by CCK8 assays, colony formation assays, transwell assays, wound-healing assays and animal experiments. RNA-sequencing and western blot were used to identify the gene regulated by circFNDC3B-218aa. Finally, glucose metabolism-related assays were performed to further investigate function of circFNDC3B-218aa. RESULTS CircFNDC3B was localized mostly in the cytoplasm, and was decreased in CC cell lines and tissues. The patients with low circFNDC3B expression had a shorter OS (P = 0.0014) than patients with high expression. Moreover, circFNDC3B inhibited the proliferation, invasion and migration of CC cells. Next, we identified that circFNDC3B could encode a novel protein circFNDC3B-218aa. Furthermore, circFNDC3B-218aa, not circFNDC3B, inhibited the proliferation, invasion and migration of CC. Additionally, the in vivo experiments implied that up-regulated circFNDC3B-218aa exhibited an inhibitory effect on CC progression. By RNA-sequencing, western blot and glucose metabolism-related assays, we found that circFNDC3B-218aa inhibited the expression of Snail, and subsequently promoted the tumor-suppressive effect of FBP1 in CC. CONCLUSIONS The novel circFNDC3B-218aa may serve as a tumor suppressive factor and potential biomarker which may supply the potential therapeutic target for CC.
Collapse
Affiliation(s)
- Zihao Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianye Cai
- Department of Hepatic Surgery and Liver Transplantation Center, Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Jiatong Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huinian Zhou
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Jingwen Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinliang Liang
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Long Xia
- Department of Hepatic Surgery and Liver Transplantation Center, Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Qi Yin
- CookGen Biosciences Center, Guangzhou, China
| | - Baojia Zou
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center, Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
- Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine of Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney at Westmead Hospital, Westmead, NSW 2145 Australia
| | - Lei Zhang
- Department of Biliary-Pancreatic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630 China
| |
Collapse
|
70
|
Casadei-Gardini A, Del Coco L, Marisi G, Conti F, Rovesti G, Ulivi P, Canale M, Frassineti GL, Foschi FG, Longo S, Fanizzi FP, Giudetti AM. 1H-NMR Based Serum Metabolomics Highlights Different Specific Biomarkers between Early and Advanced Hepatocellular Carcinoma Stages. Cancers (Basel) 2020; 12:cancers12010241. [PMID: 31963766 PMCID: PMC7016798 DOI: 10.3390/cancers12010241] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/10/2020] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
The application of non-targeted serum metabolomics profiling represents a noninvasive tool to identify new clinical biomarkers and to provide early diagnostic differentiation, and insight into the pathological mechanisms underlying hepatocellular carcinoma (HCC) progression. In this study, we used proton Nuclear Magnetic Resonance (1H-NMR) Spectroscopy and multivariate data analysis to profile the serum metabolome of 64 HCC patients, in early (n = 28) and advanced (n = 36) disease stages. We found that 1H-NMR metabolomics profiling could discriminate early from advanced HCC patients with a cross-validated accuracy close to 100%. Orthogonal partial least squares discriminant analysis (OPLS-DA) showed significant changes in serum glucose, lactate, lipids and some amino acids, such as alanine, glutamine, 1-methylhistidine, lysine and valine levels between advanced and early HCC patients. Moreover, in early HCC patients, Kaplan-Meier analysis highlighted the serum tyrosine level as a predictor for overall survival (OS). Overall, our analysis identified a set of metabolites with possible clinical and biological implication in HCC pathophysiology.
Collapse
Affiliation(s)
- Andrea Casadei-Gardini
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, 41125 Modena, Italy; (A.C.-G.); (G.R.)
| | - Laura Del Coco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.D.C.); (S.L.); (A.M.G.)
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.M.); (P.U.); (M.C.)
| | - Fabio Conti
- Department of Internal Medicine, Degli Infermi Hospital, 48018 Faenza, Italy; (F.C.); (F.G.F.)
| | - Giulia Rovesti
- Division of Medical Oncology, Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, 41125 Modena, Italy; (A.C.-G.); (G.R.)
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.M.); (P.U.); (M.C.)
| | - Matteo Canale
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy; (G.M.); (P.U.); (M.C.)
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | | | - Serena Longo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.D.C.); (S.L.); (A.M.G.)
| | - Francesco Paolo Fanizzi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.D.C.); (S.L.); (A.M.G.)
- Correspondence: ; Tel.: +39-0832-299265
| | - Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (L.D.C.); (S.L.); (A.M.G.)
| |
Collapse
|
71
|
Sola-Penna M, Paixão LP, Branco JR, Ochioni AC, Albanese JM, Mundim DM, Baptista-de-Souza D, Figueiredo CP, Coelho WS, Marcondes MC, Zancan P. Serotonin activates glycolysis and mitochondria biogenesis in human breast cancer cells through activation of the Jak1/STAT3/ERK1/2 and adenylate cyclase/PKA, respectively. Br J Cancer 2020; 122:194-208. [PMID: 31819176 PMCID: PMC7052254 DOI: 10.1038/s41416-019-0640-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/01/2019] [Accepted: 10/28/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Although produced by several types of tumours, the role of serotonin on cancer biology is yet to be understood. METHODS The effects of serotonin (5-HT) on human breast cancer cells proliferation, signalling pathways and metabolic profile were evaluated by cytometry, western blotting, qPCR, enzymology and confocal microscopy. RESULTS Our results revealed that incubation of MCF-7 cells with 10 µM 5-HT increased cell growth rate by 28%, an effect that was prevented by the 5-HTR2A/C antagonist, ketanserin. Conversely, increasing concentrations of 5-HT promoted glucose consumption and lactate production by MCF-7 cells. We also showed that increased glucose metabolism is provoked by the upregulation of pyruvate kinase M2 (PKM2) isoform through 5-HTR2A/C-triggered activation of Jak1/STAT3 and ERK1/2 subcellular pathways. However, we noticed a decrease in the rate of produced lactate per consumed glucose as a function of the hormone concentration, suggesting a disruption of the Warburg effect. The latter effect is due to 5-HTR2A/C-dependent mitochondrial biogenesis and metabolism, which is triggered by adenylyl cyclase/PKA, enhancing the oxidation of lactate within these cells. CONCLUSIONS We showed that serotonin, through 5-HTR2A/C, interferes with breast cancer cells proliferation and metabolism by triggering two distinct signalling pathways: Jak1/STAT3 that boosts glycolysis through upregulation of PKM2, and adenylyl cyclase/PKA that enhances mitochondrial biogenesis.
Collapse
Affiliation(s)
- Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Larissa P Paixão
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Jessica R Branco
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Alan C Ochioni
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Jamille M Albanese
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Davi M Mundim
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | | | - Claudia P Figueiredo
- Nucleo de Neurociências da Faculdade de Farmácia, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Wagner S Coelho
- Laboratório de Enzimologia e Controle do Metabolismo, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
- Universidade Estadual da Zona Oeste, 23070-200, Rio de Janeiro, RJ, Brazil
| | - Mariah C Marcondes
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Câncer José Alencar Gomes da Silva, 20230-130, Rio de Janeiro, RJ, Brazil
| | - Patricia Zancan
- Laboratório de Oncobiologia Molecular, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
72
|
Raut GK, Chakrabarti M, Pamarthy D, Bhadra MP. Glucose starvation-induced oxidative stress causes mitochondrial dysfunction and apoptosis via Prohibitin 1 upregulation in human breast cancer cells. Free Radic Biol Med 2019; 145:428-441. [PMID: 31614178 DOI: 10.1016/j.freeradbiomed.2019.09.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/12/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022]
Abstract
In recent years there has been an upsurge in research focusing on reprogramming cancer cells through understanding of their metabolic signatures. Alterations in mitochondrial bioenergetics and impaired mitochondrial function may serve as effective targeting strategies especially in triple-negative breast cancers (TNBCs) where hormone receptors and endocrine therapy are absent. Glucose starvation (GS) of MDA-MB-231 and MCF-7 breast cancer cells showed decrease in mitochondrial Oxygen Consumption Rate (OCR), which was rescuable to control level through addition of exogenous antioxidant N-Acetyl Cysteine (NAC). Mechanistically, GS led to increase in mitochondrial ROS and upregulation of the pleiotropic protein, Prohibitin 1 (PHB1), leading to its dissociation from Dynamin-related protein 1 (DRP1), perturbance of mitochondrial membrane potential (MMP) and triggering of the apoptosis cascade. PHB1 also reduced the invasive and migratory potential of both cell lines. We emphasize that glucose starvation remarkably sensitized the highly glycolytic metastatic TNBC cell line, MDA-MB-231 to apoptosis and decreased its migratory potential. Based on our findings, additional TNBC cell lines can be evaluated and a nutritional paradigm be proposed for anticancer therapy.
Collapse
Affiliation(s)
- Ganesh Kumar Raut
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, 600 113, India
| | - Moumita Chakrabarti
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, 600 113, India
| | - Deepika Pamarthy
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, 600 113, India
| | - Manika Pal Bhadra
- Applied Biology Department, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, Telangana State, India; Academy of Scientific and Innovative Research (AcSIR), Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai, 600 113, India.
| |
Collapse
|
73
|
Crosstalk between mitochondrial metabolism and oxidoreductive homeostasis: a new perspective for understanding the effects of bioactive dietary compounds. Nutr Res Rev 2019; 33:90-101. [DOI: 10.1017/s0954422419000210] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
AbstractMitochondria play an important role in a number of fundamental cellular processes, including energy production, biosynthetic pathways and cellular oxidoreductive homeostasis (redox status), and their dysfunction can lead to numerous pathophysiological consequences. As the biochemical mechanisms orchestrating mitochondrial metabolism and redox homeostasis are functionally linked, mitochondria have been identified as a potential therapeutic target. Consequently, considerable effort has been made to evaluate the efficacy of natural compounds that modulate mitochondrial function. Molecules produced by plants (for example, polyphenols and isothiocyanates) have been shown to modulate mitochondrial metabolism/biogenesis and redox status; however, despite the existence of a functional link, few studies have considered the combined efficacy of these mitochondrial functions. The present review provides a complete overview of the molecular pathways involved in modulating mitochondrial metabolism/biogenesis and redox status. Crosstalk between these critical mechanisms is also discussed, whilst major data from the literature regarding their antioxidant abilities are described and critically analysed. We also provide a summary of recent evidence regarding the ability of several plant-derived compounds to target these mitochondrial functions. An in-depth understanding of the functional link between mitochondrial metabolism/biogenesis and redox status could facilitate the analysis of the biological effects of natural compounds as well as the development of new therapeutic approaches.
Collapse
|
74
|
Kang H, Kim H, Lee S, Youn H, Youn B. Role of Metabolic Reprogramming in Epithelial⁻Mesenchymal Transition (EMT). Int J Mol Sci 2019; 20:ijms20082042. [PMID: 31027222 PMCID: PMC6514888 DOI: 10.3390/ijms20082042] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Activation of epithelial–mesenchymal transition (EMT) is thought to be an essential step for cancer metastasis. Tumor cells undergo EMT in response to a diverse range of extra- and intracellular stimulants. Recently, it was reported that metabolic shifts control EMT progression and induce tumor aggressiveness. In this review, we summarize the involvement of altered glucose, lipid, and amino acid metabolic enzyme expression and the underlying molecular mechanisms in EMT induction in tumor cells. Moreover, we propose that metabolic regulation through gene-specific or pharmacological inhibition may suppress EMT and this treatment strategy may be applied to prevent tumor progression and improve anti-tumor therapeutic efficacy. This review presents evidence for the importance of metabolic changes in tumor progression and emphasizes the need for further studies to better understand tumor metabolism.
Collapse
Affiliation(s)
- Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea.
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea.
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea.
| |
Collapse
|
75
|
Avagliano A, Ruocco MR, Aliotta F, Belviso I, Accurso A, Masone S, Montagnani S, Arcucci A. Power in nursing: a collaborative approach. Nurs Outlook 1984; 8:cells8050401. [PMID: 31052256 PMCID: PMC6562467 DOI: 10.3390/cells8050401] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 01/09/2023]
Abstract
Breast cancers are very heterogeneous tissues with several cell types and metabolic pathways together sustaining the initiation and progression of disease and contributing to evasion from cancer therapies. Furthermore, breast cancer cells have an impressive metabolic plasticity that is regulated by the heterogeneous tumour microenvironment through bidirectional interactions. The structure and accessibility of nutrients within this unstable microenvironment influence the metabolism of cancer cells that shift between glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) to produce adenosine triphosphate (ATP). In this scenario, the mitochondrial energetic pathways of cancer cells can be reprogrammed to modulate breast cancer’s progression and aggressiveness. Moreover, mitochondrial alterations can lead to crosstalk between the mitochondria and the nucleus, and subsequently affect cancer tissue properties. This article reviewed the metabolic plasticity of breast cancer cells, focussing mainly on breast cancer mitochondrial metabolic reprogramming and the mitochondrial alterations influencing nuclear pathways. Finally, the therapeutic strategies targeting molecules and pathways regulating cancer mitochondrial alterations are highlighted.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy.
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| | - Federica Aliotta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy.
| | - Immacolata Belviso
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy.
| | - Antonello Accurso
- Department of General, Oncological, Bariatric and Endocrine-Metabolic Surgery, University of Naples Federico II, 80131 Naples, Italy.
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy.
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy.
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy.
| |
Collapse
|