51
|
Ganguly D, Sims M, Cai C, Fan M, Pfeffer LM. Chromatin Remodeling Factor BRG1 Regulates Stemness and Chemosensitivity of Glioma Initiating Cells. Stem Cells 2018; 36:1804-1815. [PMID: 30171737 DOI: 10.1002/stem.2909] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/23/2018] [Accepted: 08/18/2018] [Indexed: 12/13/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive and malignant brain tumor that is refractory to existing therapeutic regimens, which reflects the presence of stem-like cells, termed glioma-initiating cells (GICs). The complex interactions between different signaling pathways and epigenetic regulation of key genes may be critical in the maintaining GICs in their stem-like state. Although several signaling pathways have been identified as being dysregulated in GBM, the prognosis of GBM patients remains miserable despite improvements in targeted therapies. In this report, we identified that BRG1, the catalytic subunit of the SWI/SNF chromatin remodeling complex, plays a fundamental role in maintaining GICs in their stem-like state. In addition, we identified a novel mechanism by which BRG1 regulates glycolysis genes critical for GICs. BRG1 downregulates the expression of TXNIP, a negative regulator of glycolysis. BRG1 knockdown also triggered the STAT3 pathway, which led to TXNIP activation. We further identified that TXNIP is an STAT3-regulated gene. Moreover, BRG1 suppressed the expression of interferon-stimulated genes, which are negatively regulated by STAT3 and regulate tumorigenesis. We further demonstrate that BRG1 plays a critical role in the drug resistance of GICs and in GIC-induced tumorigenesis. By genetic and pharmacological means, we found that inhibiting BRG1 can sensitize GICs to chemotherapeutic drugs, temozolomide and carmustine. Our studies suggest that BRG1 may be a novel therapeutic target in GBM. The identification of the critical role that BRG1 plays in GIC stemness and chemosensitivity will inform the development of better targeted therapies in GBM and possibly other cancers. Stem Cells 2018;36:1806-12.
Collapse
Affiliation(s)
- Debolina Ganguly
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Michelle Sims
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Chun Cai
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Meiyun Fan
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
52
|
Klepinin A, Ounpuu L, Mado K, Truu L, Chekulayev V, Puurand M, Shevchuk I, Tepp K, Planken A, Kaambre T. The complexity of mitochondrial outer membrane permeability and VDAC regulation by associated proteins. J Bioenerg Biomembr 2018; 50:339-354. [PMID: 29998379 PMCID: PMC6209068 DOI: 10.1007/s10863-018-9765-9] [Citation(s) in RCA: 15] [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: 01/15/2018] [Accepted: 07/05/2018] [Indexed: 12/18/2022]
Abstract
Previous studies have shown that class II β-tubulin plays a key role in the regulation of oxidative phosphorylation (OXPHOS) in some highly differentiated cells, but its role in malignant cells has remained unclear. To clarify these aspects, we compared the bioenergetic properties of HL-1 murine sarcoma cells, murine neuroblastoma cells (uN2a) and retinoic acid - differentiated N2a cells (dN2a). We examined the expression and possible co-localization of mitochondrial voltage dependent anion channel (VDAC) with hexokinase-2 (HK-2) and βII-tubulin, the role of depolymerized βII-tubuline and the effect of both proteins in the regulation of mitochondrial outer membrane (MOM) permeability. Our data demonstrate that neuroblastoma and sarcoma cells are prone to aerobic glycolysis, which is partially mediated by the presence of VDAC bound HK-2. Microtubule destabilizing (colchicine) and stabilizing (taxol) agents do not affect the MOM permeability for ADP in N2a and HL-1 cells. The obtained results show that βII-tubulin does not regulate the MOM permeability for adenine nucleotides in these cells. HL-1 and NB cells display comparable rates of ADP-activated respiration. It was also found that differentiation enhances the involvement of OXPHOS in N2a cells due to the rise in their mitochondrial reserve capacity. Our data support the view that the alteration of mitochondrial affinity for ADNs is one of the characteristic features of cancer cells. It can be concluded that the binding sites for tubulin and hexokinase within the large intermembrane protein supercomplex Mitochondrial Interactosome, could be different between muscle and cancer cells.
Collapse
Affiliation(s)
- Aleksandr Klepinin
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Lyudmila Ounpuu
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Kati Mado
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Laura Truu
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Vladimir Chekulayev
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Marju Puurand
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Kersti Tepp
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia
| | - Anu Planken
- Oncology and Hematology Clinic at the North Estonia Medical Centre, Tallinn, Estonia
| | - Tuuli Kaambre
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618, Tallinn, Estonia.
| |
Collapse
|
53
|
Zhou K, Yao YL, He ZC, Chen C, Zhang XN, Yang KD, Liu YQ, Liu Q, Fu WJ, Chen YP, Niu Q, Ma QH, Zhou R, Yao XH, Zhang X, Cui YH, Bian XW, Shi Y, Ping YF. VDAC2 interacts with PFKP to regulate glucose metabolism and phenotypic reprogramming of glioma stem cells. Cell Death Dis 2018; 9:988. [PMID: 30250190 PMCID: PMC6155247 DOI: 10.1038/s41419-018-1015-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/18/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Plastic phenotype convention between glioma stem cells (GSCs) and non-stem tumor cells (NSTCs) significantly fuels glioblastoma heterogeneity that causes therapeutic failure. Recent progressions indicate that glucose metabolic reprogramming could drive cell fates. However, the metabolic pattern of GSCs and NSTCs and its association with tumor cell phenotypes remain largely unknown. Here we found that GSCs were more glycolytic than NSTCs, and voltage-dependent anion channel 2 (VDAC2), a mitochondrial membrane protein, was critical for metabolic switching between GSCs and NSTCs to affect their phenotypes. VDAC2 was highly expressed in NSTCs relative to GSCs and coupled a glycolytic rate-limiting enzyme platelet-type of phosphofructokinase (PFKP) on mitochondrion to inhibit PFKP-mediated glycolysis required for GSC maintenance. Disruption of VDAC2 induced dedifferentiation of NSTCs to acquire GSC features, including the enhanced self-renewal, preferential expression of GSC markers, and increased tumorigenicity. Inversely, enforced expression ofVDAC2 impaired the self-renewal and highly tumorigenic properties of GSCs. PFK inhibitor clotrimazole compromised the effect of VDAC2 disruption on glycolytic reprogramming and GSC phenotypic transition. Clinically, VDAC2 expression inversely correlated with glioma grades (Immunohistochemical staining scores of VDAC2 were 4.7 ± 2.8, 3.2 ± 1.9, and 1.9 ± 1.9 for grade II, grade III, and IV, respectively, p < 0.05 for all) and the patients with high expression of VDAC2 had longer overall survival than those with low expression of VDAC2 (p = 0.0008). In conclusion, we demonstrate that VDAC2 is a new glycolytic regulator controlling the phenotype transition between glioma stem cells and non-stem cells and may serves as a new prognostic indicator and a potential therapeutic target for glioma patients.
Collapse
Affiliation(s)
- Kai Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Yue-Liang Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Cong Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Xiao-Ning Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Kai-Di Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Yu-Qi Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Wen-Juan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Ya-Ping Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Qing-Hua Ma
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Rong Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Xiao-Hong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China.
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China.
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, 400038, China.
| |
Collapse
|
54
|
Chen G, Wang Q, Yang Q, Li Z, Du Z, Ren M, Zhao H, Song Y, Zhang G. Circular RNAs hsa_circ_0032462, hsa_circ_0028173, hsa_circ_0005909 are predicted to promote CADM1 expression by functioning as miRNAs sponge in human osteosarcoma. PLoS One 2018; 13:e0202896. [PMID: 30153287 PMCID: PMC6112665 DOI: 10.1371/journal.pone.0202896] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 08/07/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a primary malignant bone tumor with a high fatality rate. Many circRNAs have been proved to play important roles in the pathogenesis of some diseases. However, the occurrence of circRNAs in OS remains little known. METHODS The circular RNA (circRNA) expression file GSE96964 dataset, which included seven osteosarcoma cell lines and one control sample (osteoblast cell line), was downloaded from the Gene Expression Omnibus (GEO) database to explore the potential function of circRNAs in osteosarcoma by competing endogenous RNA (ceRNA) analysis. Three gene expression profiles of OS were downloaded from GEO database and then used for the pathway enrichment analysis, Venn analysis and protein-protein interaction (PPI) network analysis. Real-time qPCR validation and RNA interference were conducted to verify our prediction. RESULTS Differentially expressed circRNAs between OS and control, including 8 up-regulated and 102 down-regulated circRNAs, were generated and ceRNA analysis for 5 most up-regulated or 5 most down-regulated circRNAs in OS were then performed. The pathway enrichment analysis of gene expression profiles indicated differentially expressed genes (DEGs) of three gene profiles significantly enriched in cell cycle pathway, cell adhesion molecules (CAMs) pathway, oxidative phosphorylation pathway, cytokine-cytokine receptor interaction pathway, p53 signaling pathway and proteoglycans in cancer pathway, which were critical important pathways in the pathogenesis of OS. The Venn analysis showed that 2 (one is a pseudogene) up-regulated and 39 down-regulated DEGs were co-expressed in all three gene profiles. Then PPI networks of 41 co-expressed DEGs (up- and down-regulated DEGs) were constructed to predict their functions using the GeneMANIA. The expression levels of these related RNAs also matched our predictions really well. CONCLUSION Ultimately, we found cell adhesion molecule 1 (CADM1) gene was not only a co-expression mRNA of the three mRNA expression profiles of OS, but also are predicted to be regulated by hsa_circ_0032462, hsa_circ_0028173, hsa_circ_0005909 by functioning as miRNAs 'Sponge' in human osteosarcoma. These over-expressed circRNAs may result in the over expression of CADM1 which promote the development of OS. We envision this discovery of these important moleculars, incuding hsa_circ_0032462, hsa_circ_0028173, hsa_circ_0005909 and CADM1 may lead to further development of new concepts, thus allowing for more opportunities in diagnosis and therapy of OS.
Collapse
Affiliation(s)
- Gaoyang Chen
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Qingyu Wang
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Qiwei Yang
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Zhaoyan Li
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Zhenwu Du
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Ming Ren
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Haiyue Zhao
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
| | - Yang Song
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
- * E-mail: (GZ); (YS)
| | - Guizhen Zhang
- Department of Orthopedics of the Second Hospital of Jilin University, Changchun, Jilin, China
- Research Centre of the Second Hospital of Jilin University, Changchun, Jilin, China
- The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Changchun, Jilin, China
- * E-mail: (GZ); (YS)
| |
Collapse
|
55
|
Targeting cancer stem cells and their niche: perspectives for future therapeutic targets and strategies. Semin Cancer Biol 2018; 53:139-155. [PMID: 30081228 DOI: 10.1016/j.semcancer.2018.08.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
Abstract
A small subpopulation of cells within the bulk of tumors share features with somatic stem cells, in that, they are capable of self-renewal, they differentiate, and are highly resistant to conventional therapy. These cells have been referred to as cancer stem cells (CSCs). Recent reports support the central importance of a cancer stem cell-like niche that appears to help foster the generation and maintenance of CSCs. In response to signals provided by this microenvironment, CSCs express the tumorigenic characteristics that can drive tumor metastasis by the induction of epithelial-mesenchymal-transition (EMT) that in turn fosters the migration and recolonization of the cells as secondary tumors within metastatic niches. We summarize here recent advances in cancer stem cell research including the characterization of their genetic and epigenetic features, metabolic specialities, and crosstalk with aging-associated processes. Potential strategies for targeting CSCs, and their niche, by regulating CSCs plasticity, or therapeutic sensitivity is discussed. Finally, it is hoped that new strategies and related therapeutic approaches as outlined here may help prevent the formation of the metastatic niche, as well as counter tumor progression and metastatic growth.
Collapse
|
56
|
Shimamura M, Yamamoto K, Kurashige T, Nagayama Y. Intracellular redox status controls spherogenicity, an in vitro cancer stem cell marker, in thyroid cancer cell lines. Exp Cell Res 2018; 370:699-707. [PMID: 30053445 DOI: 10.1016/j.yexcr.2018.07.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/17/2018] [Accepted: 07/23/2018] [Indexed: 01/17/2023]
Abstract
Cancer stem cells (CSCs), a small fraction of a tumor mass, are proposed to be highly crucial for cancer initiation, recurrence and metastasis. We have recently found that aldehyde dehydrogenase (ALDH) 1A3 is a CSC marker in some thyroid cancer cell lines, whose functional activity is, however, not relevant for thyroid cancer stemness. Since previous studies on malignancies in other organs suggest that intracellular reactive oxygen species (ROS) might be a functional and targetable CSC marker, the present study was conducted to elucidate the significance of ROS as a functional CSC marker in thyroid cancer cell lines. We first found that ROS levels controlled spherogenicity; that is, ROSlow cells were more spherogenic than ROShigh cells. However, unlike typical CSCs in other cancers, CSC-like ROSlow cells in thyroid cancer cells were plastic and were not accompanied by de-differentiation status (i.e., expression of stemness markers/thyroid-specific transcription factors) or chemo-/radio-resistance. The lower levels of ROS were functionally critical because a forced increase in ROS levels by L-buthionine-S,R-sulfoximine, an inhibitor of glutathione (GSH) synthesis, and irradiation suppressed spherogenicity. ROS levels were also correlated with the number of double strand DNA breaks determined by 53BP1 staining. Lower ROS levels appear to be a result of decreased mitochondrial oxidative phosphorylation and elevated GSH contents. Given the importance of CSC-targeted therapy for achieving long-term disease eradication by exhausting self-renewal and growth potential of cancer tissues, ROS may be a good candidate for CSC-targeted therapy in thyroid cancer.
Collapse
Affiliation(s)
- Mika Shimamura
- Department of Molecular Medicine, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Kazuo Yamamoto
- Biomedical Research Support Center, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Tomomi Kurashige
- Department of Molecular Medicine, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yuji Nagayama
- Department of Molecular Medicine, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| |
Collapse
|
57
|
Snyder V, Reed-Newman TC, Arnold L, Thomas SM, Anant S. Cancer Stem Cell Metabolism and Potential Therapeutic Targets. Front Oncol 2018; 8:203. [PMID: 29922594 PMCID: PMC5996058 DOI: 10.3389/fonc.2018.00203] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022] Open
Abstract
Malignant tumors contain heterogeneous populations of cells in various states of proliferation and differentiation. The presence of cancer stem or initiating cells is a well-established concept wherein quiescent and poorly differentiated cells within a tumor mass contribute to drug resistance, and under permissive conditions, are responsible for tumor recurrence and metastasis. A number of studies have identified molecular markers that are characteristic of tissue-specific cancer stem cells (CSCs). Isolation of CSCs has enabled studies on the metabolic status of CSCs. As metabolic plasticity is a hallmark of cancer cell adaptation, the intricacies of CSC metabolism and their phenotypic behavior are critical areas of research. Unlike normal stem cells, which rely heavily on oxidative phosphorylation (OXPHOS) as their primary source of energy, or cancer cells, which are primarily glycolytic, CSCs demonstrate a unique metabolic flexibility. CSCs can switch between OXPHOS and glycolysis in the presence of oxygen to maintain homeostasis and, thereby, promote tumor growth. Here, we review key factors that impact CSC metabolic phenotype including heterogeneity of CSCs across different histologic tumor types, tissue-specific variations, tumor microenvironment, and CSC niche. Furthermore, we discuss how targeting key players of glycolytic and mitochondrial pathways has shown promising results in cancer eradication and attenuation of disease recurrence in preclinical models. In addition, we highlight studies on other potential therapeutic targets including complex interactions within the microenvironment and cellular communications in the CSC niche to interfere with CSC growth, resistance, and metastasis.
Collapse
Affiliation(s)
- Vusala Snyder
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Tamika C Reed-Newman
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Levi Arnold
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS, United States.,Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States.,Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Shrikant Anant
- Department of General Surgery, University of Kansas Medical Center, Kansas City, KS, United States.,Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
58
|
Cancer stem cells (CSCs): metabolic strategies for their identification and eradication. Biochem J 2018; 475:1611-1634. [PMID: 29743249 PMCID: PMC5941316 DOI: 10.1042/bcj20170164] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/12/2018] [Accepted: 04/12/2018] [Indexed: 02/08/2023]
Abstract
Phenotypic and functional heterogeneity is one of the most relevant features of cancer cells within different tumor types and is responsible for treatment failure. Cancer stem cells (CSCs) are a population of cells with stem cell-like properties that are considered to be the root cause of tumor heterogeneity, because of their ability to generate the full repertoire of cancer cell types. Moreover, CSCs have been invoked as the main drivers of metastatic dissemination and therapeutic resistance. As such, targeting CSCs may be a useful strategy to improve the effectiveness of classical anticancer therapies. Recently, metabolism has been considered as a relevant player in CSC biology, and indeed, oncogenic alterations trigger the metabolite-driven dissemination of CSCs. More interestingly, the action of metabolic pathways in CSC maintenance might not be merely a consequence of genomic alterations. Indeed, certain metabotypic phenotypes may play a causative role in maintaining the stem traits, acting as an orchestrator of stemness. Here, we review the current studies on the metabolic features of CSCs, focusing on the biochemical energy pathways involved in CSC maintenance and propagation. We provide a detailed overview of the plastic metabolic behavior of CSCs in response to microenvironment changes, genetic aberrations, and pharmacological stressors. In addition, we describe the potential of comprehensive metabolic approaches to identify and selectively eradicate CSCs, together with the possibility to 'force' CSCs within certain metabolic dependences, in order to effectively target such metabolic biochemical inflexibilities. Finally, we focus on targeting mitochondria to halt CSC dissemination and effectively eradicate cancer.
Collapse
|
59
|
Ayob AZ, Ramasamy TS. Cancer stem cells as key drivers of tumour progression. J Biomed Sci 2018; 25:20. [PMID: 29506506 PMCID: PMC5838954 DOI: 10.1186/s12929-018-0426-4] [Citation(s) in RCA: 563] [Impact Index Per Article: 93.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are subpopulations of cancer cells sharing similar characteristics as normal stem or progenitor cells such as self-renewal ability and multi-lineage differentiation to drive tumour growth and heterogeneity. Throughout the cancer progression, CSC can further be induced from differentiated cancer cells via the adaptation and cross-talks with the tumour microenvironment as well as a response from therapeutic pressures, therefore contributes to their heterogeneous phenotypes. Challengingly, conventional cancer treatments target the bulk of the tumour and are unable to target CSCs due to their highly resistance nature, leading to metastasis and tumour recurrence. MAIN BODY This review highlights the roles of CSCs in tumour initiation, progression and metastasis with a focus on the cellular and molecular regulators that influence their phenotypical changes and behaviours in the different stages of cancer progression. We delineate the cross-talks between CSCs with the tumour microenvironment that support their intrinsic properties including survival, stemness, quiescence and their cellular and molecular adaptation in response to therapeutic pressure. An insight into the distinct roles of CSCs in promoting angiogenesis and metastasis has been captured based on in vitro and in vivo evidences. CONCLUSION Given dynamic cellular events along the cancer progression and contributions of resistance nature by CSCs, understanding their molecular and cellular regulatory mechanism in a heterogeneous nature, provides significant cornerstone for the development of CSC-specific therapeutics.
Collapse
Affiliation(s)
- Ain Zubaidah Ayob
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Wilayah Persekutuan Kuala Lumpur, Malaysia
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Wilayah Persekutuan Kuala Lumpur, Malaysia
- Cell and Molecular Laboratory (CMBL), The Dean’s Office, Faculty of Medicine, University of Malaya, 50603 Wilayah Persekutuan Kuala Lumpur, Malaysia
| |
Collapse
|
60
|
Li Y, Li X, Kan Q, Zhang M, Li X, Xu R, Wang J, Yu D, Goscinski MA, Wen JG, Nesland JM, Suo Z. Mitochondrial pyruvate carrier function is negatively linked to Warburg phenotype in vitro and malignant features in esophageal squamous cell carcinomas. Oncotarget 2018; 8:1058-1073. [PMID: 27911865 PMCID: PMC5352034 DOI: 10.18632/oncotarget.13717] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/28/2016] [Indexed: 02/07/2023] Open
Abstract
Aerobic glycolysis is one of the emerging hallmarks of cancer cells. In this study, we investigated the relationship between blocking mitochondrial pyruvate carrier (MPC) with MPC blocker UK5099 and the metabolic alteration as well as aggressive features of esophageal squamous carcinoma. It was found that blocking pyruvate transportation into mitochondria attenuated mitochondrial oxidative phosphorylation (OXPHOS) and triggered aerobic glycolysis, a feature of Warburg effect. In addition, the HIF-1α expression and ROS production were also activated upon UK5099 application. It was further revealed that the UK5099-treated cells became significantly more resistant to chemotherapy and radiotherapy, and the UK5099-treated tumor cells also exhibited stronger invasive capacity compared to the parental cells. In contrast to esophageal squamous epithelium cells, decreased MPC protein expression was observed in a series of 157 human squamous cell carcinomas, and low/negative MPC1 expression predicted an unfavorable clinical outcome. All these results together revealed the potential connection of altered MPC expression/activity with the Warburg metabolic reprogramming and tumor aggressiveness in cell lines and clinical samples. Collectively, our findings highlighted a therapeutic strategy targeting Warburg reprogramming of human esophageal squamous cell carcinomas.
Collapse
Affiliation(s)
- Yaqing Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China.,Department of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway
| | - Xiaoran Li
- Department of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway.,Department of Pathology, the Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| | - Quancheng Kan
- Department of Clinical Pharmacology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Mingzhi Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Xiaoli Li
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China.,Department of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway
| | - Ruiping Xu
- Department of Oncology, the Anyang Tumor Hospital, Anyang, 455000, Henan Province, China
| | - Junsheng Wang
- Department of Oncology, the Anyang Tumor Hospital, Anyang, 455000, Henan Province, China
| | - Dandan Yu
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China.,Department of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway
| | - Mariusz Adam Goscinski
- Department of Surgery, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway
| | - Jian-Guo Wen
- Institute of Clinical Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Jahn M Nesland
- Department of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway.,Department of Pathology, the Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| | - Zhenhe Suo
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China.,Department of Pathology, the Norwegian Radium Hospital, Oslo University Hospital, University of Oslo, Oslo, 0379, Norway.,Department of Pathology, the Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| |
Collapse
|
61
|
Abstract
Glycolysis has long been considered as the major metabolic process for energy production and anabolic growth in cancer cells. Although such a view has been instrumental for the development of powerful imaging tools that are still used in the clinics, it is now clear that mitochondria play a key role in oncogenesis. Besides exerting central bioenergetic functions, mitochondria provide indeed building blocks for tumor anabolism, control redox and calcium homeostasis, participate in transcriptional regulation, and govern cell death. Thus, mitochondria constitute promising targets for the development of novel anticancer agents. However, tumors arise, progress, and respond to therapy in the context of an intimate crosstalk with the host immune system, and many immunological functions rely on intact mitochondrial metabolism. Here, we review the cancer cell-intrinsic and cell-extrinsic mechanisms through which mitochondria influence all steps of oncogenesis, with a focus on the therapeutic potential of targeting mitochondrial metabolism for cancer therapy.
Collapse
Affiliation(s)
- Paolo Ettore Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, 10124 Torino, Italy
| | - Nicoletta Filigheddu
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - José Manuel Bravo-San Pedro
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France
- Université Pierre et Marie Curie/Paris VI, 75006 Paris, France
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France
- INSERM, U1138, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France
- Université Pierre et Marie Curie/Paris VI, 75006 Paris, France
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France
- INSERM, U1138, 75006 Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, 75015 Paris, France
- Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, New York, NY 10065, USA
| |
Collapse
|
62
|
Ullmann P, Qureshi-Baig K, Rodriguez F, Ginolhac A, Nonnenmacher Y, Ternes D, Weiler J, Gäbler K, Bahlawane C, Hiller K, Haan S, Letellier E. Hypoxia-responsive miR-210 promotes self-renewal capacity of colon tumor-initiating cells by repressing ISCU and by inducing lactate production. Oncotarget 2018; 7:65454-65470. [PMID: 27589845 PMCID: PMC5323168 DOI: 10.18632/oncotarget.11772] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/25/2016] [Indexed: 01/01/2023] Open
Abstract
Low oxygen concentrations (hypoxia) are known to affect the cellular metabolism and have been suggested to regulate a subpopulation of cancer cells with tumorigenic properties, the so-called tumor-initiating cells (TICs). To better understand the mechanism of hypoxia-induced TIC activation, we set out to study the role of hypoxia-responsive miRNAs in recently established colon cancer patient-derived TICs. We were able to show that low oxygen concentrations consistently lead to the upregulation of miR-210 in different primary TIC-enriched cultures. Both stable overexpression of miR-210 and knockdown of its target gene ISCU resulted in enhanced TIC self-renewal. We could validate the tumorigenic properties of miR- 210 in in vivo experiments by showing that ectopic expression of miR-210 results in increased tumor incidence. Furthermore, enhanced miR-210 expression correlated with reduced TCA cycle activity and increased lactate levels. Importantly, by blocking lactate production via inhibition of LDHA, we could reverse the promoting effect of miR-210 on self-renewal capacity, thereby emphasizing the regulatory impact of the glycolytic phenotype on colon TIC properties. Finally, by assessing expression levels in patient tissue, we could demonstrate the clinical relevance of the miR-210/ISCU signaling axis for colorectal carcinoma. Taken together, our study highlights the importance of hypoxia-induced miR-210 in the regulation of colon cancer initiation.
Collapse
Affiliation(s)
- Pit Ullmann
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Komal Qureshi-Baig
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Fabien Rodriguez
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Aurélien Ginolhac
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | | | - Dominik Ternes
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Jil Weiler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karoline Gäbler
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Christelle Bahlawane
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Karsten Hiller
- Luxembourg Centre for Systems Biomedicine, L-4367 Belvaux, Luxembourg
| | - Serge Haan
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Elisabeth Letellier
- Life Sciences Research Unit, University of Luxembourg, L-4367 Belvaux, Luxembourg
| |
Collapse
|
63
|
Mancini R, Noto A, Pisanu ME, De Vitis C, Maugeri-Saccà M, Ciliberto G. Metabolic features of cancer stem cells: the emerging role of lipid metabolism. Oncogene 2018; 37:2367-2378. [PMID: 29445137 DOI: 10.1038/s41388-018-0141-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/10/2017] [Accepted: 12/12/2017] [Indexed: 12/14/2022]
Abstract
Cancer stem cells (CSCs) are an uncommon subset of tumor cells capable of self-renewal, differentiating, and recreating the parental tumor when transplanted into the murine background. Over the past two decades, efforts toward understanding CSC biology culminated into identifying a set of signaling pathways sustaining "stemness". Nevertheless, while metabolic rewiring is nowadays considered a hallmark of cancer, no consensus has been reached on the metabolic features underlying the plastic nature of CSCs, which are capable of residing in a dormant state, and able to rapidly proliferate when the need to repopulate the tumor mass arises. An emerging concept in the field of CSC metabolism is that these cells are extremely reliant on the activity of enzymes involved in lipid metabolism, such as stearoyl-CoA desaturase 1 (SCD1) and 3-hydroxy-3-methylglutharyl-coenzyme A reductase (HMG-CoAR). Indeed, SCD1 and HMG-CoAR have been described as key factors for the correct function of a number of concatenated pathways involved in CSC fate decision, such as Hippo and Wnt. In the present review, we describe metabolic futures of CSCs with a special focus on lipid metabolism, which until now represents an underappreciated force in maintaining CSCs and an attractive therapeutic target.
Collapse
Affiliation(s)
- Rita Mancini
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Alessia Noto
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Maria Elena Pisanu
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Gennaro Ciliberto
- Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy.
| |
Collapse
|
64
|
Zhang C, Yang L, Geng YD, An FL, Xia YZ, Guo C, Luo JG, Zhang LY, Guo QL, Kong LY. Icariside II, a natural mTOR inhibitor, disrupts aberrant energy homeostasis via suppressing mTORC1-4E-BP1 axis in sarcoma cells. Oncotarget 2017; 7:27819-37. [PMID: 27056897 PMCID: PMC5053690 DOI: 10.18632/oncotarget.8538] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 03/23/2016] [Indexed: 12/21/2022] Open
Abstract
The aberrant energy homeostasis that characterized by high rate of energy production (glycolysis) and energy consumption (mRNA translation) is associated with the development of cancer. As mammalian target of rapamycin (mTOR) is a critical regulator of aberrant energy homeostasis, it is an attractive target for anti-tumor intervention. The flavonoid compound Icariside II (IS) is a natural mTOR inhibitor derived from Epimedium. Koreanum. Herein, we evaluate the effect of IS on aberrant energy homeostasis. The reduction of glycolysis and mRNA translation in U2OS (osteosarcoma), S180 (fibrosarcoma) and SW1535 (chondrosarcoma) cells observed in our study, indicate that, IS inhibits aberrant energy homeostasis. This inhibition is found to be due to suppression of mammalian target of rapamycin complex 1 (mTORC1)-eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) axis through blocking the assembly of mTORC1. Furthermore, IS inhibits the cap-dependent translation of c-myc through mTORC1-4E-BP1 axis which links the relationship between mRNA translation and glycolysis. Inhibition of aberrant energy homeostasis by IS, contributes to its in vitro and in vivo anti-proliferation activity. These data indicate that IS disrupts aberrant energy homeostasis of sarcoma cells through suppression of mTORC1-4E-BP1 axis, providing a novel mechanism of IS to inhibit cell proliferation in sarcoma cells.
Collapse
Affiliation(s)
- Chao Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Lei Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Ya-di Geng
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Fa-Liang An
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Yuan-Zheng Xia
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Chao Guo
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Jian-Guang Luo
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Lu-Yong Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Qing-Long Guo
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Ling-Yi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
65
|
Roles of PFKFB3 in cancer. Signal Transduct Target Ther 2017; 2:17044. [PMID: 29263928 PMCID: PMC5701083 DOI: 10.1038/sigtrans.2017.44] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/22/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022] Open
Abstract
The understanding of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFK-2/FBPase 3, PFKFB3) has advanced considerably since its initial identification in human macrophages in the mid-1990s. As a vital regulator of glycolysis, accumulating studies have suggested that PFKFB3 is associated with many aspects of cancer, including carcinogenesis, cancer cell proliferation, vessel aggressiveness, drug resistance and tumor microenvironment. In this review, we summarize current knowledge of PFKFB3 regulation by several signal pathways and its function in cancer development in different cell types in cancer tissues. Ubiquitous PFKFB3 has emerged as a potential target for anti-neoplastic therapy.
Collapse
|
66
|
Thomas TM, Yu JS. Metabolic regulation of glioma stem-like cells in the tumor micro-environment. Cancer Lett 2017; 408:174-181. [PMID: 28743531 PMCID: PMC5790120 DOI: 10.1016/j.canlet.2017.07.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/11/2017] [Accepted: 07/15/2017] [Indexed: 12/18/2022]
Abstract
Cancer metabolism has emerged as one of the most interesting old ideas being revisited from a new perspective. In the early 20th century Otto Warburg declared metabolism the prime cause in a disease of many secondary causes, and this statement seems more prescient in view of modern expositions into the true nature of tumor evolution. As the complexity of tumor heterogeneity becomes more clear from a genetic perspective, it is important to consider the inevitably heterogeneous metabolic components of the tumor and the tumor microenvironment. High grade gliomas remain one of the most difficult to treat solid tumors, due in part to the highly vascularized nature of the tumor and the maintenance of more resistant stem-like subpopulations within the tumor. Maintenance of glioma stem cells (GSCs) requires specific alterations within the cells and the greater tumor microenvironment with regards to signaling and metabolism. Specific niches within gliomas help foster the survival of stem-like sub-populations of cells with high tumorigenicity and high metabolic plasticity. Understanding these maintenance pathways and the metabolic dependencies within the niche may highlight potential avenues of addressing tumor resistance and recurrence in glioma patients.
Collapse
Affiliation(s)
- Tom M Thomas
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - John S Yu
- Maxine-Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
67
|
Paiva-Oliveira DI, Martins-Neves SR, Abrunhosa AJ, Fontes-Ribeiro C, Gomes CMF. Therapeutic potential of the metabolic modulator Metformin on osteosarcoma cancer stem-like cells. Cancer Chemother Pharmacol 2017; 81:49-63. [PMID: 29086064 DOI: 10.1007/s00280-017-3467-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 10/21/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE Osteosarcoma is the most common primary bone tumour appearing in children and adolescents. Recent studies demonstrate that osteosarcoma possesses a stem-like cell subset, so-called cancer stem-like cells, refractory to conventional chemotherapeutics and pointed out as responsible for relapses frequently observed in osteosarcoma patients. Here, we explored the therapeutic potential of Metformin on osteosarcoma stem-like cells, alone and as a chemosensitizer of doxorubicin. METHODS Stem-like cells were isolated from human osteosarcoma cell lines, MNNG/HOS and MG-63, using the sphere-forming assay. Metformin cytotoxicity alone and combined with doxorubicin were evaluated using MTT/BrdU assays. Protein levels of AMPK and AKT were evaluated by Western Blot. Cellular metabolic status was assessed based on [18F]-FDG uptake and lactate production measurements. Sphere-forming efficiency and expression of pluripotency transcription factors analysed by qRT-PCR were tested as readout of Metformin effects on stemness features. RESULTS Metformin induced a concentration-dependent decrease in the metabolic activity and proliferation of sphere-forming cells and improved doxorubicin-induced cytotoxicity. This drug also down-regulated the expression of master regulators of pluripotency (OCT4, SOX2, NANOG), and decreased spheres' self-renewal ability. Metformin effects on mitochondria led to the activation and phosphorylation of the energetic sensor AMPK along with an upregulation of the pro-survival AKT pathway in both cell populations. Furthermore, Metformin-induced mitochondrial stress increased [18F]-FDG uptake and lactate production in parental cells but not in the quiescent stem-like cells, suggesting the inability of the latter to cope with the energy crisis induced by metformin. CONCLUSIONS This preclinical study suggests that Metformin may be a potentially useful therapeutic agent and chemosensitizer of osteosarcoma stem-like cells to doxorubicin.
Collapse
Affiliation(s)
- Daniela I Paiva-Oliveira
- Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta. Comba, Celas, 3000-354, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Sara R Martins-Neves
- Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta. Comba, Celas, 3000-354, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Antero J Abrunhosa
- Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Carlos Fontes-Ribeiro
- Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta. Comba, Celas, 3000-354, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Célia M F Gomes
- Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta. Comba, Celas, 3000-354, Coimbra, Portugal. .,CNC.IBILI, University of Coimbra, Coimbra, Portugal. .,Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
68
|
Tirinato L, Pagliari F, Limongi T, Marini M, Falqui A, Seco J, Candeloro P, Liberale C, Di Fabrizio E. An Overview of Lipid Droplets in Cancer and Cancer Stem Cells. Stem Cells Int 2017; 2017:1656053. [PMID: 28883835 PMCID: PMC5572636 DOI: 10.1155/2017/1656053] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 06/08/2017] [Accepted: 07/05/2017] [Indexed: 02/06/2023] Open
Abstract
For decades, lipid droplets have been considered as the main cellular organelles involved in the fat storage, because of their lipid composition. However, in recent years, some new and totally unexpected roles have been discovered for them: (i) they are active sites for synthesis and storage of inflammatory mediators, and (ii) they are key players in cancer cells and tissues, especially in cancer stem cells. In this review, we summarize the main concepts related to the lipid droplet structure and function and their involvement in inflammatory and cancer processes.
Collapse
Affiliation(s)
- L. Tirinato
- German Cancer Research Center (DKFZ), Heidelberg, Baden-Württemberg, Germany
- Physical Science and Engineering (PSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - F. Pagliari
- Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - T. Limongi
- Physical Science and Engineering (PSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Department of Applied Science and Technology (DISAT), Politecnico di Torino, Torino, Italy
| | - M. Marini
- Physical Science and Engineering (PSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - A. Falqui
- Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - J. Seco
- German Cancer Research Center (DKFZ), Heidelberg, Baden-Württemberg, Germany
| | - P. Candeloro
- BioNEM Lab, Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - C. Liberale
- Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - E. Di Fabrizio
- Physical Science and Engineering (PSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
69
|
Ma R, Karthik GM, Lövrot J, Haglund F, Rosin G, Katchy A, Zhang X, Viberg L, Frisell J, Williams C, Linder S, Fredriksson I, Hartman J. Estrogen Receptor β as a Therapeutic Target in Breast Cancer Stem Cells. J Natl Cancer Inst 2017; 109:1-14. [PMID: 28376210 PMCID: PMC5441302 DOI: 10.1093/jnci/djw236] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 09/20/2016] [Indexed: 12/14/2022] Open
Abstract
Background Breast cancer cells with tumor-initiating capabilities (BSCs) are considered to maintain tumor growth and govern metastasis. Hence, targeting BSCs will be crucial to achieve successful treatment of breast cancer. Methods We characterized mammospheres derived from more than 40 cancer patients and two breast cancer cell lines for the expression of estrogen receptors (ERs) and stem cell markers. Mammosphere formation and proliferation assays were performed on cells from 19 cancer patients and five healthy individuals after incubation with ER-subtype selective ligands. Transcriptional analysis was performed to identify pathways activated in ERβ-stimulated mammospheres and verified using in vitro experiments. Xenograft models (n = 4 or 5 per group) were used to study the role of ERs during tumorigenesis. Results We identified an absence of ERα but upregulation of ERβ in BSCs associated with phenotypic stem cell markers and responsible for the proliferative role of estrogens. Knockdown of ERβ caused a reduction of mammosphere formation in cell lines and in patient-derived cancer cells (40.7%, 26.8%, and 39.1%, respectively). Gene set enrichment analysis identified glycolysis-related pathways (false discovery rate < 0.001) upregulated in ERβ-activated mammospheres. We observed that tamoxifen or fulvestrant alone was insufficient to block proliferation of patient-derived BSCs while this could be accomplished by a selective inhibitor of ERβ (PHTPP; 53.7% in luminal and 45.5% in triple-negative breast cancers). Furthermore, PHTPP reduced tumor initiation in two patient-derived xenografts (75.9% and 59.1% reduction in tumor volume, respectively) and potentiated tamoxifen-mediated inhibition of tumor growth in MCF7 xenografts. Conclusion We identify ERβ as a mediator of estrogen action in BSCs and a novel target for endocrine therapy.
Collapse
Affiliation(s)
- Ran Ma
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Govindasamy-Muralidharan Karthik
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - John Lövrot
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Felix Haglund
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| | - Gustaf Rosin
- Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Anne Katchy
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Xiaonan Zhang
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Lisa Viberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Jan Frisell
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Williams
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.,Science for Life Laboratory, Department of Proteomics, KTH, Royal Institute of Technology, Stockholm, Sweden
| | - Stig Linder
- Department of Medical and Health Sciences, Department of Medicine and Health, Linköping University, Linköping, Sweden
| | - Irma Fredriksson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.,Department of Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| |
Collapse
|
70
|
Dai J, Ji Y, Wang W, Kim D, Fai LY, Wang L, Luo J, Zhang Z. Loss of fructose-1,6-bisphosphatase induces glycolysis and promotes apoptosis resistance of cancer stem-like cells: an important role in hexavalent chromium-induced carcinogenesis. Toxicol Appl Pharmacol 2017. [PMID: 28624442 DOI: 10.1016/j.taap.2017.06.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hexavalent chromium (Cr(VI)) compounds are confirmed human carcinogens for lung cancer. Our previous studies has demonstrated that chronic exposure of human bronchial epithelial BEAS-2B cells to low dose of Cr(VI) causes malignant cell transformation. The acquisition of cancer stem cell-like properties is involved in the initiation of cancers. The present study has observed that a small population of cancer stem-like cells (BEAS-2B-Cr-CSC) exists in the Cr(VI)-transformed cells (BEAS-2B-Cr). Those BEAS-2B-Cr-CSC exhibit extremely reduced capability of generating reactive oxygen species (ROS) and apoptosis resistance. BEAS-2B-Cr-CSC are metabolic inactive as evidenced by reductions in oxygen consumption, glucose uptake, ATP production, and lactate production. Most importantly, BEAS-2B-Cr-CSC are more tumorigenic with high levels of cell self-renewal genes, Notch1 and p21. Further study has found that fructose-1,6-bisphosphatase (FBP1), an rate-limiting enzyme driving glyconeogenesis, was lost in BEAS-2B-Cr-CSC. Forced expression of FBP1 in BEAS-2B-Cr-CSC restored ROS generation, resulting in increased apoptosis, leading to inhibition of tumorigenesis. In summary, the present study suggests that loss of FBP1 is a critical event in tumorigenesis of Cr(VI)-transformed cells.
Collapse
Affiliation(s)
- Jin Dai
- Department of Toxicology and Cancer Biology, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Yanli Ji
- Department of Toxicology and Cancer Biology, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Wei Wang
- Department of Toxicology and Cancer Biology, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Donghern Kim
- Department of Toxicology and Cancer Biology, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Leonard Yenwong Fai
- Department of Toxicology and Cancer Biology, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Diseases, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Department of Toxicology and Cancer Biology, 1095 Veterans Drive, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
71
|
Wong TL, Che N, Ma S. Reprogramming of central carbon metabolism in cancer stem cells. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1728-1738. [PMID: 28502706 DOI: 10.1016/j.bbadis.2017.05.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/23/2017] [Accepted: 05/10/2017] [Indexed: 12/19/2022]
Abstract
Cancer metabolism has been studied for years and adopted in the clinic for monitoring disease progression and therapy response. Despite our growing knowledge of a distinctly altered metabolic behavior in cancer, drugs targeting cancer metabolism have remained less than promising. Recent efforts in cancer stem cell (CSC) biology suggest that a subpopulation of tumor-initiating cells within the tumor bulk represents the root of tumor recurrence and therapy resistance. In recent years, metabolic phenotype of CSCs of various tumor types has been identified. This breakthrough has shed light on the underlying mechanism by which CSCs maintain stemness, confer resistance to therapies and initiate tumor relapse. The distinct metabolic characteristics of CSCs compared to non-CSCs provide an opportunity to target CSCs more specifically and have become a major focus in cancer research in recent years with substantial efforts conducted towards discovering clinical targets. This perspective article summarizes the current knowledge of CSC metabolism in carcinogenesis and highlights the potential of targeting CSC metabolism for therapy.
Collapse
Affiliation(s)
- Tin Lok Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Noélia Che
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; State Key Laboratory for Liver Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
72
|
Sun L, Moritake T, Ito K, Matsumoto Y, Yasui H, Nakagawa H, Hirayama A, Inanami O, Tsuboi K. Metabolic analysis of radioresistant medulloblastoma stem-like clones and potential therapeutic targets. PLoS One 2017; 12:e0176162. [PMID: 28426747 PMCID: PMC5398704 DOI: 10.1371/journal.pone.0176162] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/06/2017] [Indexed: 12/11/2022] Open
Abstract
Medulloblastoma is a fatal brain tumor in children, primarily due to the presence of treatment-resistant medulloblastoma stem cells. The energy metabolic pathway is a potential target of cancer therapy because it is often different between cancer cells and normal cells. However, the metabolic properties of medulloblastoma stem cells, and whether specific metabolic pathways are essential for sustaining their stem cell-like phenotype and radioresistance, remain unclear. We have established radioresistant medulloblastoma stem-like clones (rMSLCs) by irradiation of the human medulloblastoma cell line ONS-76. Here, we assessed reactive oxygen species (ROS) production, mitochondria function, oxygen consumption rate (OCR), energy state, and metabolites of glycolysis and tricarboxylic acid cycle in rMSLCs and parental cells. rMSLCs showed higher lactate production and lower oxygen consumption rate than parental cells. Additionally, rMSLCs had low mitochondria mass, low endogenous ROS production, and existed in a low-energy state. Treatment with the metabolic modifier dichloroacetate (DCA) resulted in mitochondria dysfunction, glycolysis inhibition, elongated mitochondria morphology, and increased ROS production. DCA also increased radiosensitivity by suppression of the DNA repair capacity through nuclear oxidization and accelerated the generation of acetyl CoA to compensate for the lack of ATP. Moreover, treatment with DCA decreased cancer stem cell-like characters (e.g., CD133 positivity and sphere-forming ability) in rMSLCs. Together, our findings provide insights into the specific metabolism of rMSLCs and illuminate potential metabolic targets that might be exploited for therapeutic benefit in medulloblastoma.
Collapse
Affiliation(s)
- Lue Sun
- Department of Radiological Health Science, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Takashi Moritake
- Department of Radiological Health Science, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- * E-mail:
| | - Kazuya Ito
- Department of Radiobiology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshitaka Matsumoto
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hironobu Yasui
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hidehiko Nakagawa
- Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Aki Hirayama
- Center for Integrative Medicine, Tsukuba University of Technology, Tsukuba, Ibaraki, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Koji Tsuboi
- Proton Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
73
|
Knockdown of stem cell regulator Oct4A in ovarian cancer reveals cellular reprogramming associated with key regulators of cytoskeleton-extracellular matrix remodelling. Sci Rep 2017; 7:46312. [PMID: 28406185 PMCID: PMC5390261 DOI: 10.1038/srep46312] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/14/2017] [Indexed: 12/17/2022] Open
Abstract
Oct4A is a master regulator of self-renewal and pluripotency in embryonic stem cells. It is a well-established marker for cancer stem cell (CSC) in malignancies. Recently, using a loss of function studies, we have demonstrated key roles for Oct4A in tumor cell survival, metastasis and chemoresistance in in vitro and in vivo models of ovarian cancer. In an effort to understand the regulatory role of Oct4A in tumor biology, we employed the use of an ovarian cancer shRNA Oct4A knockdown cell line (HEY Oct4A KD) and a global mass spectrometry (MS)-based proteomic analysis to investigate novel biological targets of Oct4A in HEY samples (cell lysates, secretomes and mouse tumor xenografts). Based on significant differential expression, pathway and protein network analyses, and comprehensive literature search we identified key proteins involved with biologically relevant functions of Oct4A in tumor biology. Across all preparations of HEY Oct4A KD samples significant alterations in protein networks associated with cytoskeleton, extracellular matrix (ECM), proliferation, adhesion, metabolism, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) and drug resistance was observed. This comprehensive proteomics study for the first time presents the Oct4A associated proteome and expands our understanding on the biological role of this stem cell regulator in carcinomas.
Collapse
|
74
|
Morandi A, Taddei ML, Chiarugi P, Giannoni E. Targeting the Metabolic Reprogramming That Controls Epithelial-to-Mesenchymal Transition in Aggressive Tumors. Front Oncol 2017; 7:40. [PMID: 28352611 PMCID: PMC5348536 DOI: 10.3389/fonc.2017.00040] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 02/28/2017] [Indexed: 01/06/2023] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) process allows the trans-differentiation of a cell with epithelial features into a cell with mesenchymal characteristics. This process has been reported to be a key priming event for tumor development and therefore EMT activation is now considered an established trait of malignancy. The transcriptional and epigenetic reprogramming that governs EMT has been extensively characterized and reviewed in the last decade. However, increasing evidence demonstrates a correlation between metabolic reprogramming and EMT execution. The aim of the current review is to gather the recent findings that illustrate this correlation to help deciphering whether metabolic changes are causative or just a bystander effect of EMT activation. The review is divided accordingly to the catabolic and anabolic pathways that characterize carbohydrate, aminoacid, and lipid metabolism. Moreover, at the end of each part, we have discussed a series of potential metabolic targets involved in EMT promotion and execution for which drugs are either available or that could be further investigated for therapeutic intervention.
Collapse
Affiliation(s)
- Andrea Morandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence , Florence , Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence , Florence , Italy
| | - Paola Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Excellence Centre for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| | - Elisa Giannoni
- Department of Experimental and Clinical Biomedical Sciences, University of Florence , Florence , Italy
| |
Collapse
|
75
|
Li Y, Atkinson K, Zhang T. Combination of chemotherapy and cancer stem cell targeting agents: Preclinical and clinical studies. Cancer Lett 2017; 396:103-109. [PMID: 28300634 DOI: 10.1016/j.canlet.2017.03.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/12/2022]
Abstract
The cancer stem cell model claims that the initiation, maintenance, and growth of a tumor are driven by a small population of cancer cells termed cancer stem cells. Cancer stem cells possess a variety of phenotypes associated with therapeutic resistance and often cause recurrence of the diseases. Several strategies have been investigated to target cancer stem cells in a variety of cancers, such as blocking one or more self-renewal signaling pathways, reducing the expression of drug efflux and ATP-binding cassette efflux transporters, modulating epigenetic aberrations, and promoting cancer stem cell differentiation. A number of cell and animal studies strongly support the potential benefits of combining chemotherapeutic drugs with cancer stem cell targeting agents. Clinical trials are still underway to address the pharmacokinetics, safety, and efficacy of combination treatment. This mini-review provides an updated discussion of these preclinical and clinical studies.
Collapse
Affiliation(s)
- Yanyan Li
- College of Science and Humanities, Husson University, 1 College Circle, Bangor, ME, 04401, USA.
| | - Katharine Atkinson
- College of Science and Humanities, Husson University, 1 College Circle, Bangor, ME, 04401, USA
| | - Tao Zhang
- School of Pharmacy, Husson University, 1 College Circle, Bangor, ME, 04401, USA
| |
Collapse
|
76
|
Qureshi-Baig K, Ullmann P, Haan S, Letellier E. Tumor-Initiating Cells: a criTICal review of isolation approaches and new challenges in targeting strategies. Mol Cancer 2017; 16:40. [PMID: 28209178 PMCID: PMC5314476 DOI: 10.1186/s12943-017-0602-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/20/2017] [Indexed: 02/07/2023] Open
Abstract
Most cancers contain a subpopulation of highly tumorigenic cells, known as cancer stem cells (CSCs) or tumor-initiating cells (TICs). Targeting TICs may be essential to achieve cure, because of their self-renewal and tumorigenic properties as well as their resistance to conventional therapies. Despite significant advances in TIC biology, their isolation and identification remain largely disputed and incompletely established. In this review, we discuss the latest developments in isolation and culturing approaches of TICs, with focus on colorectal cancer (CRC). We feature recent findings on TIC-relevant signaling pathways and the metabolic identity of TICs, as well as their current clinical implications. Lastly, we highlight the influence of inter- and intra-tumoral heterogeneity on TIC function and targeting approaches.
Collapse
Affiliation(s)
- Komal Qureshi-Baig
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg
| | - Pit Ullmann
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg
| | - Serge Haan
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg.
| |
Collapse
|
77
|
Zhang HL, Wang MD, Zhou X, Qin CJ, Fu GB, Tang L, Wu H, Huang S, Zhao LH, Zeng M, Liu J, Cao D, Guo LN, Wang HY, Yan HX, Liu J. Blocking preferential glucose uptake sensitizes liver tumor-initiating cells to glucose restriction and sorafenib treatment. Cancer Lett 2016; 388:1-11. [PMID: 27894955 DOI: 10.1016/j.canlet.2016.11.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/03/2023]
Abstract
Cancer cells display altered metabolic phenotypes characterized by a high level of glycolysis, even under normoxic conditions. Because of a high rate of glycolytic flux and inadequate vascularization, tumor cells often suffer from nutrient deficiency and require metabolic adaptations to address such stresses. Although tumor-initiating cells (T-ICs) have been identified in various malignancies, the cells' metabolic phenotypes remain elusive. In this study, we observed that liver T-ICs preferentially survived under restricted glucose treatment. These cell populations compete successfully for glucose uptake by preferentially expressing glucose transporters (GLUT1 and GLUT3), whereas inhibition of GLUT1 or GLUT3 abolished the survival advantage and suppressed the tumorigenic potential of liver T-ICs. Among signaling pathways related to T-ICs, IL-6/STAT3 was identified to be responsible for the elevation of glucose uptake in liver T-ICs under glucose limitation. Further investigation revealed that IL-6 stimulation upregulated GLUT1 and GLUT3 expressions in CD133+ cells, particularly during glucose deprivation. More importantly, inhibition of glucose uptake sensitized liver T-ICs to sorafenib treatment and enhanced the therapeutic efficacy in vivo. Our findings suggest that blocking IL-6/STAT3-mediated preferential glucose uptake might be exploited for novel therapeutic targets during hepatocellular carcinoma (HCC) progression.
Collapse
Affiliation(s)
- Hui-Lu Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ming-Da Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Xu Zhou
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Chen-Jie Qin
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Gong-Bo Fu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Liang Tang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Han Wu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Shuai Huang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Ling-Hao Zhao
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Min Zeng
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Jiao Liu
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Dan Cao
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Lin-Na Guo
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Hong-Yang Wang
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China.
| | - He-Xin Yan
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China.
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
78
|
Deshmukh A, Deshpande K, Arfuso F, Newsholme P, Dharmarajan A. Cancer stem cell metabolism: a potential target for cancer therapy. Mol Cancer 2016; 15:69. [PMID: 27825361 PMCID: PMC5101698 DOI: 10.1186/s12943-016-0555-x] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/01/2016] [Indexed: 12/19/2022] Open
Abstract
Cancer Stem cells (CSCs) are a unipotent cell population present within the tumour cell mass. CSCs are known to be highly chemo-resistant, and in recent years, they have gained intense interest as key tumour initiating cells that may also play an integral role in tumour recurrence following chemotherapy. Cancer cells have the ability to alter their metabolism in order to fulfil bio-energetic and biosynthetic requirements. They are largely dependent on aerobic glycolysis for their energy production and also are associated with increased fatty acid synthesis and increased rates of glutamine utilisation. Emerging evidence has shown that therapeutic resistance to cancer treatment may arise due to dysregulation in glucose metabolism, fatty acid synthesis, and glutaminolysis. To propagate their lethal effects and maintain survival, tumour cells alter their metabolic requirements to ensure optimal nutrient use for their survival, evasion from host immune attack, and proliferation. It is now evident that cancer cells metabolise glutamine to grow rapidly because it provides the metabolic stimulus for required energy and precursors for synthesis of proteins, lipids, and nucleic acids. It can also regulate the activities of some of the signalling pathways that control the proliferation of cancer cells. This review describes the key metabolic pathways required by CSCs to maintain a survival advantage and highlights how a combined approach of targeting cellular metabolism in conjunction with the use of chemotherapeutic drugs may provide a promising strategy to overcome therapeutic resistance and therefore aid in cancer therapy.
Collapse
Affiliation(s)
- Abhijeet Deshmukh
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia
| | - Kedar Deshpande
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia.
| |
Collapse
|
79
|
Bulfoni M, Turetta M, Del Ben F, Di Loreto C, Beltrami AP, Cesselli D. Dissecting the Heterogeneity of Circulating Tumor Cells in Metastatic Breast Cancer: Going Far Beyond the Needle in the Haystack. Int J Mol Sci 2016; 17:ijms17101775. [PMID: 27783057 PMCID: PMC5085799 DOI: 10.3390/ijms17101775] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Revised: 10/08/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Although the enumeration of circulating tumor cells (CTC) defined as expressing both epithelial cell adhesion molecule and cytokeratins (EpCAM+/CK+) can predict prognosis and response to therapy in metastatic breast, colon and prostate cancer, its clinical utility (i.e., the ability to improve patient outcome by guiding therapy) has not yet been proven in clinical trials. Therefore, scientists are now focusing on the molecular characterization of CTC as a way to explore its possible use as a “surrogate” of tumor tissues to non-invasively assess the genomic landscape of the cancer and its evolution during treatment. Additionally, evidences confirm the existence of CTC in epithelial-to-mesenchymal transition (EMT) characterized by a variable loss of epithelial markers. Since the EMT process can originate cells with enhanced invasiveness, stemness and drug-resistance, the enumeration and characterization of this population, perhaps the one truly responsible of tumor recurrence and progression, could be more clinically useful. For these reasons, several devices able to capture CTC independently from the expression of epithelial markers have been developed. In this review, we will describe the types of heterogeneity so far identified and the key role played by the epithelial-to-mesenchymal transition in driving CTC heterogeneity. The clinical relevance of detecting CTC-heterogeneity will be discussed as well.
Collapse
Affiliation(s)
- Michela Bulfoni
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| | - Matteo Turetta
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| | - Fabio Del Ben
- Department of Clinical Pathology, CRO Aviano National Cancer Institute, via F. Gallini 2, 33081 Aviano, Italy.
| | - Carla Di Loreto
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
- Institute of Pathology, University Hospital of Udine-ASUIUD, Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy.
| | - Antonio Paolo Beltrami
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| | - Daniela Cesselli
- Department of Medical and Biological Sciences, University of Udine, Piazzale M. Kolbe 4, 33100 Udine, Italy.
| |
Collapse
|
80
|
Song K, Kwon H, Han C, Zhang J, Dash S, Lim K, Wu T. Active glycolytic metabolism in CD133(+) hepatocellular cancer stem cells: regulation by MIR-122. Oncotarget 2016; 6:40822-35. [PMID: 26506419 PMCID: PMC4747371 DOI: 10.18632/oncotarget.5812] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/23/2015] [Indexed: 02/07/2023] Open
Abstract
Although altered metabolic pathway is an important diagnostic maker and therapeutic target in cancer, it is poorly understood in cancer stem cells (CSCs). Here we show that the CD133 (+) hepatocellular CSCs have distinct metabolic properties, characterized by more active glycolysis over oxidative phosphorylation, compared to the CD133 (−) cells. Inhibition of PDK4 and LDHA markedly suppresses CD133 (+) stemness characteristics and overcome resistance to sorafenib (current chemotherapeutic agent for hepatocellular cancer). Addition of glucose or lactate to CD133 (−) cells promotes CSC phenotypes, as evidenced by increased CD133 (+) cell population, elevated stemness gene expression and enhanced spheroid formation. Furthermore, the liver-specific miRNA, miR-122, inhibits CSC phenotypes by regulating glycolysis through targeting PDK4. Our findings suggest that enhanced glycolysis is associated with CD133 (+) stem-like characteristics and that metabolic reprogramming through miR-122 or PDK4 may represent a novel therapeutic approach for the treatment of hepatocellular cancer.
Collapse
Affiliation(s)
- Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hyunjoo Kwon
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Chang Han
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kyu Lim
- Department of Biochemistry, College of Medicine, Cancer Research Institute and Infection Signaling Network Research Center, Chungnam National University, Daejeon, Korea
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
81
|
Intraperitoneal 188Re-Liposome delivery switches ovarian cancer metabolism from glycolysis to oxidative phosphorylation and effectively controls ovarian tumour growth in mice. Radiother Oncol 2016; 119:282-90. [DOI: 10.1016/j.radonc.2016.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 12/24/2015] [Accepted: 02/01/2016] [Indexed: 01/02/2023]
|
82
|
Pharmacological or genetic inhibition of LDHA reverses tumor progression of pediatric osteosarcoma. Biomed Pharmacother 2016; 81:388-393. [PMID: 27261617 DOI: 10.1016/j.biopha.2016.04.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 04/11/2016] [Accepted: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
Reprogrammed energy metabolism is an emerging hallmark of cancer. Lactate dehydrogenase A (LDHA), a key enzyme involved in anaerobic glycolysis, is frequently deregulated in human malignancies. However, limited knowledge is known about its roles in the progression of osteosarcoma (OS). In this study, we found that LDHA is commonly upregulated in four OS cell lines compared with the normal osteoblast cells (hFOB1.19). Treatment with FX11, a specific inhibitor of LDHA, significantly reduced LDHA activity, and inhibited cell proliferation and invasive potential in a dose dependent manner. Genetic silencing of LDHA resulted in a decreased lactate level in the culture medium, reduced cell viability and decreased cell invasion ability. Meanwhile, silencing of LDHA also compromised tumorigenesis in vivo. Furthermore, knockdown of LDHA remarkably reduced extracellular acidification rate (ECAR) as well as glucose consumption. In the presence of 2-DG, a glycolysis inhibitor, LDHA-mediated cell proliferation and invasion were completely blocked, indicating the oncogenic activities of LDHA may dependent on Warburg effect. Finally, pharmacological inhibition of c-Myc or HIF1α significantly attenuated LDHA expression. Taken together, upregulated LDHA facilitates tumor progression of OS and might be a potential target for OS treatment.
Collapse
|
83
|
Polyphenols as Modulator of Oxidative Stress in Cancer Disease: New Therapeutic Strategies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6475624. [PMID: 26649142 PMCID: PMC4663347 DOI: 10.1155/2016/6475624] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/21/2015] [Indexed: 12/19/2022]
Abstract
Cancer onset and progression have been linked to oxidative stress by increasing DNA mutations or inducing DNA damage, genome instability, and cell proliferation and therefore antioxidant agents could interfere with carcinogenesis. It is well known that conventional radio-/chemotherapies influence tumour outcome through ROS modulation. Since these antitumour treatments have important side effects, the challenge is to develop new anticancer therapeutic strategies more effective and less toxic for patients. To this purpose, many natural polyphenols have emerged as very promising anticancer bioactive compounds. Beside their well-known antioxidant activities, several polyphenols target epigenetic processes involved in cancer development through the modulation of oxidative stress. An alternative strategy to the cytotoxic treatment is an approach leading to cytostasis through the induction of therapy-induced senescence. Many anticancer polyphenols cause cellular growth arrest through the induction of a ROS-dependent premature senescence and are considered promising antitumour therapeutic tools. Furthermore, one of the most innovative and interesting topics is the evaluation of efficacy of prooxidant therapies on cancer stem cells (CSCs). Several ROS inducers-polyphenols can impact CSCs metabolisms and self-renewal related pathways. Natural polyphenol roles, mainly in chemoprevention and cancer therapies, are described and discussed in the light of the current literature data.
Collapse
|
84
|
Shen YA, Wang CY, Hsieh YT, Chen YJ, Wei YH. Metabolic reprogramming orchestrates cancer stem cell properties in nasopharyngeal carcinoma. Cell Cycle 2015; 14:86-98. [PMID: 25483072 DOI: 10.4161/15384101.2014.974419] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation of tumor cells endowed with self-renewal capacity and are considered as an underlying cause of tumor recurrence and metastasis. The metabolic signatures of CSCs and the mechanisms involved in the regulation of their stem cell-like properties still remain elusive. We utilized nasopharyngeal carcinoma (NPC) CSCs as a model to dissect their metabolic signatures and found that CSCs underwent metabolic shift and mitochondrial resetting distinguished from their differentiated counterparts. In metabolic shift, CSCs showed a greater reliance on glycolysis for energy supply compared with the parental cells. In mitochondrial resetting, the quantity and function of mitochondria of CSCs were modulated by the biogenesis of the organelles, and the round-shaped mitochondria were distributed in a peri-nuclear manner similar to those seen in the stem cells. In addition, we blocked the glycolytic pathway, increased the ROS levels, and depolarized mitochondrial membranes of CSCs, respectively, and examined the effects of these metabolic factors on CSC properties. Intriguingly, the properties of CSCs were curbed when we redirected the quintessential metabolic reprogramming, which indicates that the plasticity of energy metabolism regulated the balance between acquisition and loss of the stemness status. Taken together, we suggest that metabolic reprogramming is critical for CSCs to sustain self-renewal, deter from differentiation and enhance the antioxidant defense mechanism. Characterization of metabolic reprogramming governing CSC properties is paramount to the design of novel therapeutic strategies through metabolic intervention of CSCs.
Collapse
Key Words
- ATP6, ATP synthase 6
- COX, cytochrome c oxidase
- Cu/ZnSOD, copper/zinc superoxide dismutase
- GLUT1, glucose transporter 1
- GPI, glucose-6-phosphate isomerase
- GR, glutathione reductase
- HK, hexokinase
- MnSOD, manganese superoxide dismutase
- ND1, NADH dehydrogenase subunit 1
- PDH, pyruvate dehydrogenase
- PDK, pyruvate dehydrogenase kinase
- PGC-1α, peroxisome proliferator-activated receptor gamma coactivator 1α
- POLG, mitochondrial DNA polymerase gamma
- TFAM, mitochondrial transcription factor A
- cancer stem cells
- metabolic reprogramming
- metabolic shift
- mitochondrial membrane potential
- mitochondrial resetting
- nasopharyngeal carcinoma
- reactive oxygen species
Collapse
Affiliation(s)
- Yao-An Shen
- a Institute of Biochemistry and Molecular Biology ; Taipei , Taiwan
| | | | | | | | | |
Collapse
|
85
|
Martins-Neves SR, Corver WE, Paiva-Oliveira DI, van den Akker BEWM, Briaire-de-Bruijn IH, Bovée JVMG, Gomes CMF, Cleton-Jansen AM. Osteosarcoma Stem Cells Have Active Wnt/β-catenin and Overexpress SOX2 and KLF4. J Cell Physiol 2015; 231:876-86. [DOI: 10.1002/jcp.25179] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Sara R. Martins-Neves
- Department of Pathology; Leiden University Medical Center; Leiden The Netherlands
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
- Center of Investigation in Environment; Genetics and Oncobiology; CIMAGO - Faculty of Medicine; University of Coimbra, Coimbra; Celas Portugal
| | - Willem E. Corver
- Department of Pathology; Leiden University Medical Center; Leiden The Netherlands
| | - Daniela I. Paiva-Oliveira
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
| | | | | | | | - Célia M. F. Gomes
- Pharmacology and Experimental Therapeutics; Institute for Biomedical Imaging and Life Sciences (IBILI); Faculty of Medicine; University of Coimbra; Coimbra Portugal
- CNC.IBILI; University of Coimbra; Coimbra Portugal
- Center of Investigation in Environment; Genetics and Oncobiology; CIMAGO - Faculty of Medicine; University of Coimbra, Coimbra; Celas Portugal
| | | |
Collapse
|
86
|
Dando I, Dalla Pozza E, Biondani G, Cordani M, Palmieri M, Donadelli M. The metabolic landscape of cancer stem cells. IUBMB Life 2015; 67:687-93. [DOI: 10.1002/iub.1426] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 08/14/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Ilaria Dando
- Department of Life and Reproduction Sciences; Section of Biochemistry, University of Verona; Verona Italy
| | - Elisa Dalla Pozza
- Department of Life and Reproduction Sciences; Section of Biochemistry, University of Verona; Verona Italy
| | - Giulia Biondani
- Department of Life and Reproduction Sciences; Section of Biochemistry, University of Verona; Verona Italy
| | - Marco Cordani
- Department of Life and Reproduction Sciences; Section of Biochemistry, University of Verona; Verona Italy
| | - Marta Palmieri
- Department of Life and Reproduction Sciences; Section of Biochemistry, University of Verona; Verona Italy
| | - Massimo Donadelli
- Department of Life and Reproduction Sciences; Section of Biochemistry, University of Verona; Verona Italy
| |
Collapse
|
87
|
Glycolytic enzyme upregulation and numbness of mitochondrial activity characterize the early phase of apoptosis in cerebellar granule cells. Apoptosis 2015; 20:10-28. [PMID: 25351440 DOI: 10.1007/s10495-014-1049-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) and cancer proceed via one or more common molecular mechanisms: a metabolic shift from oxidative phosphorylation to glycolysis-corresponding to the activation of the Warburg effect-occurs in both diseases. The findings reported in this paper demonstrate that, in the early phase of apoptosis, glucose metabolism is enhanced, i.e. key proteins which internalize and metabolize glucose-glucose transporter, hexokinase and phosphofructokinase-are up-regulated, in concomitance with a parallel decrease in oxygen consumption by mitochondria and increase of L-lactate accumulation. Reversal of the glycolytic phenotype occurs in the presence of dichloroacetate, inhibitor of the pyruvate dehydrogenase kinase enzyme, which speeds up apoptosis of cerebellar granule cells, reawakening mitochondria and then modulating glycolytic enzymes. Loss of the adaptive advantage afforded by aerobic glycolysis, which occurs in the late phase of apoptosis, exacerbates the pathological processes underlying neurodegeneration, leading inevitably the cell to death. In conclusion, the data propose that both aerobic, i.e. Warburg effect, essentially due to the protective numbness of mitochondria, and anaerobic glycolysis, rather due to the mitochondrial impairment, characterize the entire time frame of apoptosis, from the early to the late phase, which mimics the development of AD.
Collapse
|
88
|
Wang YH, Scadden DT. Harnessing the apoptotic programs in cancer stem-like cells. EMBO Rep 2015; 16:1084-98. [PMID: 26253117 DOI: 10.15252/embr.201439675] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 06/19/2015] [Indexed: 12/12/2022] Open
Abstract
Elimination of malignant cells is an unmet challenge for most human cancer types even with therapies targeting specific driver mutations. Therefore, a multi-pronged strategy to alter cancer cell biology on multiple levels is increasingly recognized as essential for cancer cure. One such aspect of cancer cell biology is the relative apoptosis resistance of tumor-initiating cells. Here, we provide an overview of the mechanisms affecting the apoptotic process in tumor cells emphasizing the differences in the tumor-initiating or stem-like cells of cancer. Further, we summarize efforts to exploit these differences to design therapies targeting that important cancer cell population.
Collapse
Affiliation(s)
- Ying-Hua Wang
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA Harvard Stem Cell Institute, Cambridge, MA, USA Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
89
|
Corominas-Faja B, Cuyàs E, Gumuzio J, Bosch-Barrera J, Leis O, Martin ÁG, Menendez JA. Chemical inhibition of acetyl-CoA carboxylase suppresses self-renewal growth of cancer stem cells. Oncotarget 2015; 5:8306-16. [PMID: 25246709 PMCID: PMC4226684 DOI: 10.18632/oncotarget.2059] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cancer stem cells (CSC) may take advantage of the Warburg effect-induced siphoning of metabolic intermediates into de novo fatty acid biosynthesis to increase self-renewal growth. We examined the anti-CSC effects of the antifungal polyketide soraphen A, a specific inhibitor of the first committed step of lipid biosynthesis catalyzed by acetyl-CoA carboxylase (ACACA). The mammosphere formation capability of MCF-7 cells was reduced following treatment with soraphen A in a dose-dependent manner. MCF-7 cells engineered to overexpress the oncogene HER2 (MCF-7/HER2 cells) were 5-fold more sensitive than MCF-7 parental cells to soraphen A-induced reductions in mammosphere-forming efficiency. Soraphen A treatment notably decreased aldehyde dehydrogenase (ALDH)-positive CSC-like cells and impeded the HER2's ability to increase the ALDH+-stem cell population. The following results confirmed that soraphen A-induced suppression of CSC populations occurred through ACACA-driven lipogenesis: a.) exogenous supplementation with supraphysiological concentrations of oleic acid fully rescued mammosphere formation in the presence of soraphen A and b.) mammosphere cultures of MCF-7 cells with stably silenced expression of the cytosolic isoform ACACA1, which specifically participates in de novo lipogenesis, were mostly refractory to soraphen A treatment. Our findings reveal for the first time that ACACA may constitute a previously unrecognized target for novel anti-breast CSC therapies.
Collapse
Affiliation(s)
- Bruna Corominas-Faja
- Metabolism and Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia Spain. Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia Spain
| | - Elisabet Cuyàs
- Metabolism and Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia Spain. Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia Spain
| | - Juan Gumuzio
- Fundación Inbiomed, San Sebastián, Gipuzkoa Spain
| | | | - Olatz Leis
- StemTek Therapeutics, Bilbao, Biscay Spain
| | | | - Javier A Menendez
- Metabolism and Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia Spain. Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia Spain
| |
Collapse
|
90
|
Cuyàs E, Corominas-Faja B, Menendez JA. The nutritional phenome of EMT-induced cancer stem-like cells. Oncotarget 2015; 5:3970-82. [PMID: 24994116 PMCID: PMC4147299 DOI: 10.18632/oncotarget.2147] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The metabolic features of cancer stem (CS) cells and the effects of specific nutrients or metabolites on CS cells remain mostly unexplored. A preliminary study to delineate the nutritional phenome of CS cells exploited the landmark observation that upon experimental induction into an epithelial-to-mesenchymal (EMT) transition, the proportion of CS-like cells drastically increases within a breast cancer cell population. EMT-induced CS-like cells (HMLERshEcad) and isogenic parental cells (HMLERshCntrol) were simultaneously screened for their ability to generate energy-rich NADH when cultured in a standardized high-throughput metabolic phenotyping platform comprising >350 wells that were pre-loaded with different carbohydrates/starches, alcohols, fatty acids, ketones, carboxylic acids, amino acids, and bi-amino acids. The generation of “phenetic maps” of the carbon and nitrogen utilization patterns revealed that the acquisition of a CS-like cellular state provided an enhanced ability to utilize additional catabolic fuels, especially under starvation conditions. Crucially, the acquisition of cancer stemness activated a metabolic infrastructure that enabled the vectorial transfer of high-energy nutrients such as glycolysis end products (pyruvate, lactate) and bona fide ketone bodies (β-hydroxybutyrate) from the extracellular microenvironment to support mitochondrial energy production in CS-like cells. Metabolic reprogramming may thus constitute an efficient adaptive strategy through which CS-like cells would rapidly obtain an advantage in hostile conditions such as nutrient starvation following the inhibition of tumor angiogenesis. By understanding how specific nutrients could bioenergetically boost EMT-CS-like phenotypes, “smart foods” or systemic “metabolic nichotherapies” may be tailored to specific nutritional CSC phenomes, whereas high-resolution heavy isotope-labeled nutrient tracking may be developed to monitor the spatiotemporal distribution and functionality of CS-like cells in real time.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- Metabolism & Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia, SPAIN; Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia, SPAIN
| | - Bruna Corominas-Faja
- Metabolism & Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia, SPAIN; Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia, SPAIN
| | - Javier A Menendez
- Metabolism & Cancer Group, Translational Research Laboratory, Catalan Institute of Oncology, Girona, Catalonia, SPAIN; Girona Biomedical Research Institute (IDIBGI), Girona, Catalonia, SPAIN
| |
Collapse
|
91
|
Li Y, Luo S, Ma R, Liu J, Xu P, Zhang H, Tang K, Ma J, Zhang Y, Liang X, Sun Y, Ji T, Wang N, Huang B. Upregulation of cytosolic phosphoenolpyruvate carboxykinase is a critical metabolic event in melanoma cells that repopulate tumors. Cancer Res 2015; 75:1191-6. [PMID: 25712344 DOI: 10.1158/0008-5472.can-14-2615] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/22/2015] [Indexed: 12/23/2022]
Abstract
Although metabolic defects have been investigated extensively in differentiated tumor cells, much less attention has been directed to the metabolic properties of stem-like cells that repopulate tumors [tumor-repopulating cells (TRC)]. Here, we show that melanoma TRCs cultured in three-dimensional soft fibrin gels reprogram glucose metabolism by hijacking the cytosolic enzyme phosphoenolpyruvate carboxykinase (PCK1), a key player in gluconeogenesis. Surprisingly, upregulated PCK1 in TRCs did not mediate gluconeogenesis but promoted glucose side-branch metabolism, including in the serine and glycerol-3-phosphate pathways. Moreover, this retrograde glucose carbon flow strengthened rather than antagonized glycolysis and glucose consumption. Silencing PCK1 or inhibiting its enzymatic activity slowed the growth of TRCs in vitro and impeded tumorigenesis in vivo. Overall, our work unveiled metabolic features of TRCs in melanoma that have implications for targeting a unique aspect of this disease.
Collapse
Affiliation(s)
- Yong Li
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shunqun Luo
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruihua Ma
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Liu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingwei Xu
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huafeng Zhang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ke Tang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingwei Ma
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Zhang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyu Liang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanling Sun
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Ji
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Wang
- Laboratory for Cell Biomechanics and Regenerative Medicine, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China. Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Bo Huang
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
92
|
OVCAR-3 spheroid-derived cells display distinct metabolic profiles. PLoS One 2015; 10:e0118262. [PMID: 25688563 PMCID: PMC4331360 DOI: 10.1371/journal.pone.0118262] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 01/07/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction Recently, multicellular spheroids were isolated from a well-established epithelial ovarian cancer cell line, OVCAR-3, and were propagated in vitro. These spheroid-derived cells displayed numerous hallmarks of cancer stem cells, which are chemo- and radioresistant cells thought to be a significant cause of cancer recurrence and resultant mortality. Gene set enrichment analysis of expression data from the OVCAR-3 cells and the spheroid-derived putative cancer stem cells identified several metabolic pathways enriched in differentially expressed genes. Before this, there had been little previous knowledge or investigation of systems-scale metabolic differences between cancer cells and cancer stem cells, and no knowledge of such differences in ovarian cancer stem cells. Methods To determine if there were substantial metabolic changes corresponding with these transcriptional differences, we used two-dimensional gas chromatography coupled to mass spectrometry to measure the metabolite profiles of the two cell lines. Results These two cell lines exhibited significant metabolic differences in both intracellular and extracellular metabolite measurements. Principal components analysis, an unsupervised dimensional reduction technique, showed complete separation between the two cell types based on their metabolite profiles. Pathway analysis of intracellular metabolomics data revealed close overlap with metabolic pathways identified from gene expression data, with four out of six pathways found enriched in gene-level analysis also enriched in metabolite-level analysis. Some of those pathways contained multiple metabolites that were individually statistically significantly different between the two cell lines, with one of the most broadly and consistently different pathways, arginine and proline metabolism, suggesting an interesting hypothesis about cancerous and stem-like metabolic phenotypes in this pair of cell lines. Conclusions Overall, we demonstrate for the first time that metabolism in an ovarian cancer stem cell line is distinct from that of more differentiated isogenic cancer cells, supporting the potential importance of metabolism in the differences between cancer cells and cancer stem cells.
Collapse
|
93
|
Du JY, Wang LF, Wang Q, Yu LD. miR-26b inhibits proliferation, migration, invasion and apoptosis induction via the downregulation of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 driven glycolysis in osteosarcoma cells. Oncol Rep 2015; 33:1890-8. [PMID: 25672572 DOI: 10.3892/or.2015.3797] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/09/2015] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are differentially expressed and play crucial roles in cancer development and progression. Elevated glycolysis provides survival advantage and metastatic phenotype. Emerging evidence indicates that glycolysis in cancers can be regulated by miRNAs. In the present study, the role of miR-26b in the proliferation, invasion and glycolytic phenotype of osteosarcoma (OS) cells was investigated. miR-26b was reported to be downregulated in OS tissues, however, the effect of miR-26b on OS has not been distinctly evaluated. The present study therefore investigated the miR-26b sensitivity mechanism in OS. To determine the role of miR-26, we reinstated its expression in the U2OS OS cell line through transfection with miR-26b mimics and examined the effects on cell proliferation, migration, invasion, cell cycle progression and glycolytic parameters. The computational prediction tool was employed to identify the molecular target of miR-26b and was confirmed experimentally. Restoration of miR-26b expression inhibited cell proliferation, migration and invasion, arrested cell cycle progression, and induced cell apoptosis accompanied by the downregulation of glycolytic phenotype. Moreover, the binding site for miR-26b was predicted in the 3'UTR of gene 6-phosphofructo-2-kinase/fructose‑2,6-bisphosphatase-3 (PFKFB3), suggesting a role for miR-26b in metabolic alteration in OS cells. Further studies showed that overexpression of miR-26b repressed PFKFB3 mRNA and protein levels followed by modulation of the expression of glycolytic components (LDHA, GLUT-1) and markers of invasion and cell cycle such as MMP-9, MMP-2, cyclin D1 and p27. Collectively, the data suggested the tumor suppressive role of miR-26b which functions by targeting the glycolytic metabolism in OS cells, and providing a possible therapeutic strategy for OS patients by targeting miRNA expression.
Collapse
Affiliation(s)
- Jing-Yu Du
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Li-Feng Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Quan Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Lie-Dao Yu
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
94
|
Vlashi E, Pajonk F. The metabolic state of cancer stem cells-a valid target for cancer therapy? Free Radic Biol Med 2015; 79:264-8. [PMID: 25450330 PMCID: PMC4339632 DOI: 10.1016/j.freeradbiomed.2014.10.732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 12/26/2022]
Abstract
In the 1920s Otto Warburg first described high glucose uptake, aerobic glycolysis, and high lactate production in tumors. Since then high glucose uptake has been utilized in the development of PET imaging for cancer. However, despite a deepened understanding of the molecular underpinnings of glucose metabolism in cancer, this fundamental difference between normal and malignant tissue has yet to be employed in targeted cancer therapy in the clinic. In this review, we highlight attempts in the recent literature to target cancer cell metabolism and elaborate on the challenges and controversies of these strategies in general and in the context of tumor cell heterogeneity in cancer.
Collapse
Affiliation(s)
- Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, and Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, and Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095-1714, USA.
| |
Collapse
|
95
|
Pistollato F, Giampieri F, Battino M. The use of plant-derived bioactive compounds to target cancer stem cells and modulate tumor microenvironment. Food Chem Toxicol 2015; 75:58-70. [DOI: 10.1016/j.fct.2014.11.004] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 12/18/2022]
|
96
|
Anderson AS, Roberts PC, Frisard MI, Hulver MW, Schmelz EM. Ovarian tumor-initiating cells display a flexible metabolism. Exp Cell Res 2014; 328:44-57. [PMID: 25172556 DOI: 10.1016/j.yexcr.2014.08.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 08/14/2014] [Accepted: 08/17/2014] [Indexed: 01/06/2023]
Abstract
An altered metabolism during ovarian cancer progression allows for increased macromolecular synthesis and unrestrained growth. However, the metabolic phenotype of cancer stem or tumor-initiating cells, small tumor cell populations that are able to recapitulate the original tumor, has not been well characterized. In the present study, we compared the metabolic phenotype of the stem cell enriched cell variant, MOSE-LFFLv (TIC), derived from mouse ovarian surface epithelial (MOSE) cells, to their parental (MOSE-L) and benign precursor (MOSE-E) cells. TICs exhibit a decrease in glucose and fatty acid oxidation with a concomitant increase in lactate secretion. In contrast to MOSE-L cells, TICs can increase their rate of glycolysis to overcome the inhibition of ATP synthase by oligomycin and can increase their oxygen consumption rate to maintain proton motive force when uncoupled, similar to the benign MOSE-E cells. TICs have an increased survival rate under limiting conditions as well as an increased survival rate when treated with AICAR, but exhibit a higher sensitivity to metformin than MOSE-E and MOSE-L cells. Together, our data show that TICs have a distinct metabolic profile that may render them flexible to adapt to the specific conditions of their microenvironment. By better understanding their metabolic phenotype and external environmental conditions that support their survival, treatment interventions can be designed to extend current therapy regimens to eradicate TICs.
Collapse
Affiliation(s)
- Angela S Anderson
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Paul C Roberts
- Biomedical Science and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Madlyn I Frisard
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA
| | - Matthew W Hulver
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA.
| | - Eva M Schmelz
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
97
|
Pacini N, Borziani F. Cancer stem cell theory and the warburg effect, two sides of the same coin? Int J Mol Sci 2014; 15:8893-930. [PMID: 24857919 PMCID: PMC4057766 DOI: 10.3390/ijms15058893] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 04/28/2014] [Accepted: 05/12/2014] [Indexed: 12/12/2022] Open
Abstract
Over the last 100 years, many studies have been performed to determine the biochemical and histopathological phenomena that mark the origin of neoplasms. At the end of the last century, the leading paradigm, which is currently well rooted, considered the origin of neoplasms to be a set of genetic and/or epigenetic mutations, stochastic and independent in a single cell, or rather, a stochastic monoclonal pattern. However, in the last 20 years, two important areas of research have underlined numerous limitations and incongruities of this pattern, the hypothesis of the so-called cancer stem cell theory and a revaluation of several alterations in metabolic networks that are typical of the neoplastic cell, the so-called Warburg effect. Even if this specific “metabolic sign” has been known for more than 85 years, only in the last few years has it been given more attention; therefore, the so-called Warburg hypothesis has been used in multiple and independent surveys. Based on an accurate analysis of a series of considerations and of biophysical thermodynamic events in the literature, we will demonstrate a homogeneous pattern of the cancer stem cell theory, of the Warburg hypothesis and of the stochastic monoclonal pattern; this pattern could contribute considerably as the first basis of the development of a new uniform theory on the origin of neoplasms. Thus, a new possible epistemological paradigm is represented; this paradigm considers the Warburg effect as a specific “metabolic sign” reflecting the stem origin of the neoplastic cell, where, in this specific metabolic order, an essential reason for the genetic instability that is intrinsic to the neoplastic cell is defined.
Collapse
Affiliation(s)
- Nicola Pacini
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| | - Fabio Borziani
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| |
Collapse
|
98
|
Anti-spasmogenic effect of cyproheptadine on guinea-pig ileum. Cancers (Basel) 1984; 11:cancers11070965. [PMID: 31324052 PMCID: PMC6678244 DOI: 10.3390/cancers11070965] [Citation(s) in RCA: 111] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/04/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer that lacks targeted therapy options, and patients diagnosed with TNBC have poorer outcomes than patients with other breast cancer subtypes. Emerging evidence suggests that breast cancer stem cells (BCSCs), which have tumor-initiating potential and possess self-renewal capacity, may be responsible for this poor outcome by promoting therapy resistance, metastasis, and recurrence. TNBC cells have been consistently reported to display cancer stem cell (CSC) signatures at functional, molecular, and transcriptional levels. In recent decades, CSC-targeting strategies have shown therapeutic effects on TNBC in multiple preclinical studies, and some of these strategies are currently being evaluated in clinical trials. Therefore, understanding CSC biology in TNBC has the potential to guide the discovery of novel therapeutic agents in the future. In this review, we focus on the self-renewal signaling pathways (SRSPs) that are aberrantly activated in TNBC cells and discuss the specific signaling components that are involved in the tumor-initiating potential of TNBC cells. Additionally, we describe the molecular mechanisms shared by both TNBC cells and CSCs, including metabolic plasticity, which enables TNBC cells to switch between metabolic pathways according to substrate availability to meet the energetic and biosynthetic demands for rapid growth and survival under harsh conditions. We highlight CSCs as potential key regulators driving the aggressiveness of TNBC. Thus, the manipulation of CSCs in TNBC can be a targeted therapeutic strategy for TNBC in the future.
Collapse
|
99
|
Yadav UP, Singh T, Kumar P, Sharma P, Kaur H, Sharma S, Singh S, Kumar S, Mehta K. [Morbidity in primary medical services in the jurisdiction of Huamantla, Tlaxcala]. SALUD PUBLICA DE MEXICO 1982; 10:1010. [PMID: 32670883 PMCID: PMC7330710 DOI: 10.3389/fonc.2020.01010] [Citation(s) in RCA: 81] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Umesh Prasad Yadav
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Tashvinder Singh
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Pramit Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Praveen Sharma
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Harsimrat Kaur
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
- Desh Bhagat Dental College, Mandi Gobindgarh, India
| | - Sadhana Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Sandeep Singh
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Santosh Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, Patna, India
| | - Kapil Mehta
- Department of Experimental Therapeutics, MD Anderson Cancer Centre, The University of Texas, Houston, TX, United States
| |
Collapse
|
100
|
Degeneration and regeneration of the nerves of the heart after transplantation. Transplantation 1973; 5:e1336. [PMID: 25032859 PMCID: PMC4123079 DOI: 10.1038/cddis.2014.285] [Citation(s) in RCA: 192] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/21/2014] [Accepted: 05/27/2014] [Indexed: 12/11/2022]
Abstract
A number of studies suggest that cancer stem cells are essential for tumour growth, and failure to target these cells can result in tumour relapse. As this population of cells has been shown to be resistant to radiation and chemotherapy, it is essential to understand their biology and identify new therapeutic approaches. Targeting cancer metabolism is a potential alternative strategy to counteract tumour growth and recurrence. Here we applied a proteomic and targeted metabolomic analysis in order to point out the main metabolic differences between breast cancer cells grown as spheres and thus enriched in cancer stem cells were compared with the same cells grown in adherent differentiating conditions. This integrated approach allowed us to identify a metabolic phenotype associated with the stem-like condition and shows that breast cancer stem cells (BCSCs) shift from mitochondrial oxidative phosphorylation towards fermentative glycolysis. Functional validation of proteomic and metabolic data provide evidences for increased activities of key enzymes of anaerobic glucose fate such as pyruvate kinase M2 isoform, lactate dehydrogenase and glucose 6-phopshate dehydrogenase in cancer stem cells as well as different redox status. Moreover, we show that treatment with 2-deoxyglucose, a well known inhibitor of glycolysis, inhibits BCSC proliferation when used alone and shows a synergic effect when used in combination with doxorubicin. In conclusion, we suggest that inhibition of glycolysis may be a potentially effective strategy to target BCSCs.
Collapse
|