1
|
Gentile R, Feudi D, Sallicandro L, Biagini A. Can the Tumor Microenvironment Alter Ion Channels? Unraveling Their Role in Cancer. Cancers (Basel) 2025; 17:1244. [PMID: 40227837 PMCID: PMC11988140 DOI: 10.3390/cancers17071244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/29/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Neoplastic cells are characterized by metabolic reprogramming, known as the Warburg effect, in which glucose metabolism is predominantly directed toward aerobic glycolysis, with reduced mitochondrial oxidative phosphorylation and increased lactate production even in the presence of oxygen. This phenomenon provides cancer cells with a proliferative advantage, allowing them to rapidly produce energy (in the form of ATP) and generate metabolic intermediates necessary for the biosynthesis of macromolecules essential for cell growth. It is important to understand the role of ion channels in the tumor context since they participate in various physiological processes and in the regulation of the tumor microenvironment. These changes may contribute to the development and transformation of cancer cells, as well as affect the communication between cells and the surrounding microenvironment, including impaired or altered expression and functionality of ion channels. Therefore, the aim of this review is to elucidate the impact of the tumor microenvironment on the electrical properties of the cellular membranes in several cancers as a possible therapeutic target.
Collapse
Affiliation(s)
- Rosaria Gentile
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
| | - Davide Feudi
- Department of Biostatistics, Epidemiology and Public Health, University of Padua, Via L. Loredan 18, 35131 Padova, Italy;
| | - Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06123 Perugia, Italy;
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| |
Collapse
|
2
|
Wang K, Shen M, Tang H, Zhou J, Liu Y, Niu D, Zeng Z, Pan L, Yao J, Sun C. Jingfang Granule promotes the tricarboxylic acid cycle to improve chronic fatigue syndrome by increasing the expression of Idh1 and Idh2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119241. [PMID: 39689747 DOI: 10.1016/j.jep.2024.119241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic fatigue syndrome (CFS), as a complex, multisystemic, and multisystemic disorder affecting multiple organs and systems, often accompanies by symptoms such as post-exercise discomfort, sleep disorders, cognitive difficulties, and orthostatic intolerance. Jingfang Granule (JFG) is a traditional Chinese medicine that have significant protective effects on CFS, but the mechanism is still vague. AIM OF STUDY This study was designed to evaluate the protective mechanism of JFG on mice with CFS. MATERIALS AND METHODS The combined stimuli method was used to establish the mice CFS model, and JFG was orally administered. The body weight, exhaustion swimming training and tail suspension test were assayed every 7 days to evaluate the improvement of JFG on CFS. Lactic acid, adenosine triphosphate (ATP), malondialdehyde (MDA), superoxide dismutase (SOD), reactive oxygen species (ROS), IL-1β, TNF-α, IL-6 in serum and liver glycogen, muscle glycogen in muscle were analyzed. Transmission electron microscopy was used to detect mitochondrial morphology. The regulatory networks were investigated by proteomics and central carbon metabolomics, which were verified by Western blot. RESULTS JFG reversed the loss of weight and reduce of exhaust swimming time (P < 0.05) induced by CFS in mice, and increased the tail suspension time (P < 0.05), indicating that JFG has an improving effect on CFS. Meanwhile, JFG increased the spleen index (P < 0.05), decreased the thymus index (P < 0.05) and cardiac index (P < 0.05), inhibited the secretion of Lactic acid (P < 0.05), and increased the content of liver glycogen (P < 0.05), muscle glycogen (P < 0.05), and ATP (P < 0.05), and improved mitochondrial morphology in mice with CFS. JFG also inhibited the release of TNF-α (P < 0.05), IL-1β (P < 0.05) and IL-6 (P < 0.05) in serum by inhibiting TLR4/NF-κB signaling pathway and NLRP3 inflammasome signaling pathway, and inhibited oxidative stress by activating Nrf2/HO-1/NQO1 axis. Integrated central carbon metabolomics, proteomics and Western blot showed that JFG intervened in CFS by increasing the expression of Idh1 (P < 0.05) and Idh2 (P < 0.01) to promote tricarboxylic acid (TCA) cycle. CONCLUSIONS This study confirmed that JFG promoted the TCA cycle by increasing the expression of Idh1 and Idh2, and then inhibited inflammation and oxidative stress to prevent CFS injury, which provided a potential drug candidate for CFS treatment.
Collapse
Affiliation(s)
- Kun Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China.
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Hongguang Tang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yan Liu
- School of Pharmacy, Qingdao University, Qingdao, 266071, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jingchun Yao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China; College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| |
Collapse
|
3
|
Sharma A, Virmani T, Kumar G, Sharma A, Virmani R, Gugulothu D, Singh K, Misra SK, Pathak K, Chitranshi N, Coutinho HDM, Jain D. Mitochondrial signaling pathways and their role in cancer drug resistance. Cell Signal 2024; 122:111329. [PMID: 39098704 DOI: 10.1016/j.cellsig.2024.111329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
Mitochondria, traditionally known as cellular powerhouses, now emerge as critical signaling centers influencing cancer progression and drug resistance. The review highlights the role that apoptotic signaling, DNA mutations, mitochondrial dynamics and metabolism play in the development of resistance mechanisms and the advancement of cancer. Targeted approaches are discussed, with an emphasis on managing mitophagy, fusion, and fission of the mitochondria to make resistant cancer cells more susceptible to traditional treatments. Additionally, metabolic reprogramming can be used to effectively target metabolic enzymes such GLUT1, HKII, PDK, and PKM2 in order to avoid resistance mechanisms. Although there are potential possibilities for therapy, the complex structure of mitochondria and their subtle role in tumor development hamper clinical translation. Novel targeted medicines are put forth, providing fresh insights on combating drug resistance in cancer. The study also emphasizes the significance of glutamine metabolism, mitochondrial respiratory complexes, and apoptotic pathways as potential targets to improve treatment effectiveness against drug-resistant cancers. Combining complementary and nanoparticle-based techniques to target mitochondria has demonstrated encouraging results in the treatment of cancer, opening doors to reduce resistance and enable individualized treatment plans catered to the unique characteristics of each patient. Suggesting innovative approaches such as drug repositioning and mitochondrial drug delivery to enhance the efficacy of mitochondria-targeting therapies, presenting a pathway for advancements in cancer treatment. This thorough investigation is a major step forward in the treatment of cancer and has the potential to influence clinical practice and enhance patient outcomes.
Collapse
Affiliation(s)
- Ashwani Sharma
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Tarun Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India.
| | - Girish Kumar
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India.
| | - Anjali Sharma
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India
| | - Reshu Virmani
- School of Pharmaceutical Sciences, MVN University, Palwal, Haryana 121105, India.
| | - Dalapathi Gugulothu
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Kuldeep Singh
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| | - Shashi Kiran Misra
- School of Pharmaceutical Sciences, CSJM University Kanpur, Kanpur 208024, India
| | - Kamla Pathak
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India
| | - Nitin Chitranshi
- Macquarie Medical School, Macquarie University, New South Wales, Australia; School of Science and Technology, the University of New England, Armidale, New South Wales, Australia.
| | | | - Divya Jain
- Department of Microbiology, School of Applied and Life Sciences, Uttaranchal University, Dehradun 248007, Uttarakhand, India
| |
Collapse
|
4
|
Tigu AB, Tomuleasa C. Exploring Novel Frontiers in Cancer Therapy. Biomedicines 2024; 12:1345. [PMID: 38927551 PMCID: PMC11202039 DOI: 10.3390/biomedicines12061345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer progression and initiation are sustained by a series of alterations in molecular pathways because of genetic errors, external stimuli and other factors, which lead to an abnormal cellular function that can be translated into uncontrolled cell growth and metastasis [...].
Collapse
Affiliation(s)
- Adrian Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400015 Cluj-Napoca, Romania
| |
Collapse
|
5
|
Zhang H, Song J, Ward R, Han Y, Hunt A, Shriwas P, Steed A, Edwards C, Cao Y, Co M, Chen X. Diverse temporal and spatial mechanisms work, partially through Stanniocalcin-1, V-ATPase and senescence, to activate the extracellular ATP-mediated drug resistance in human cancer cells. Front Oncol 2024; 14:1276092. [PMID: 38380370 PMCID: PMC10876858 DOI: 10.3389/fonc.2024.1276092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Introduction Resistance to drug therapies is associated with a large majority of cancer-related deaths. ATP-binding cassette (ABC) transporter-mediated drug efflux, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), glutathione (GSH), senescence, and vacuole-type ATPase (V-ATPase) all contribute to the resistance. We recently showed that extracellular ATP (eATP) induces and regulates EMT, CSC formation, and ABC transporters in human cancer cells and tumors. eATP also consistently upregulates Stanniocalcin-1 (STC1), a gene that significantly contributes to EMT, CSC formation, and tumor growth. We also found that eATP enhances drug resistance in cancer cells through eATP internalization mediated by macropinocytosis, leading to an elevation of intracellular ATP (iATP) levels, induction of EMT, and CSC formation. However, these factors have never been systematically investigated in the context of eATP-induced drug resistance. Methods In this study, we hypothesized that eATP increases drug resistance via inducing ABC efflux, EMT, CSCs, STC1, and their accompanied processes such as GSH reducing activity, senescence, and V-ATPase. RNA sequencing, metabolomics, gene knockdown and knockout, and functional assays were performed to investigate these pathways and processes. Results and discussion Our study results showed that, in multiple human cancer lines, eATP induced genes involved in drug resistance, elevated ABC transporters' efflux activity of anticancer drugs; generated transcriptomic and metabolic profiles representing a drug resistant state; upregulated activities of GSH, senescence, and V-ATPase to promote drug resistance. Collectively, these newly found players shed light on the mechanisms of eATP-induced as well as STC1- and V-ATPase-mediated drug resistance and offer potential novel targets for combating drug resistance in cancers.
Collapse
Affiliation(s)
- Haiyun Zhang
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Jingwen Song
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Ryan Ward
- The Honor Tutorial College, Ohio University, Athens, OH, United States
| | - Yong Han
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Arabella Hunt
- The Honor Tutorial College, Ohio University, Athens, OH, United States
| | - Pratik Shriwas
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Alexander Steed
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Cory Edwards
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Yanyang Cao
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Milo Co
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Xiaozhuo Chen
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- Department of Biomedical Science, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
6
|
Tirendi S, Marengo B, Domenicotti C, Bassi AM, Almonti V, Vernazza S. Colorectal cancer and therapy response: a focus on the main mechanisms involved. Front Oncol 2023; 13:1208140. [PMID: 37538108 PMCID: PMC10396348 DOI: 10.3389/fonc.2023.1208140] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/19/2023] [Indexed: 08/05/2023] Open
Abstract
Introduction The latest GLOBOCAN 2021 reports that colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. Most CRC cases are sporadic and associated with several risk factors, including lifestyle habits, gut dysbiosis, chronic inflammation, and oxidative stress. Aim To summarize the biology of CRC and discuss current therapeutic interventions designed to counteract CRC development and to overcome chemoresistance. Methods Literature searches were conducted using PubMed and focusing the attention on the keywords such as "Current treatment of CRC" or "chemoresistance and CRC" or "oxidative stress and CRC" or "novel drug delivery approaches in cancer" or "immunotherapy in CRC" or "gut microbiota in CRC" or "systematic review and meta-analysis of randomized controlled trials" or "CSCs and CRC". The citations included in the search ranged from September 1988 to December 2022. An additional search was carried out using the clinical trial database. Results Rounds of adjuvant therapies, including radiotherapy, chemotherapy, and immunotherapy are commonly planned to reduce cancer recurrence after surgery (stage II and stage III CRC patients) and to improve overall survival (stage IV). 5-fluorouracil-based chemotherapy in combination with other cytotoxic drugs, is the mainstay to treat CRC. However, the onset of the inherent or acquired resistance and the presence of chemoresistant cancer stem cells drastically reduce the efficacy. On the other hand, the genetic-molecular heterogeneity of CRC often precludes also the efficacy of new therapeutic approaches such as immunotherapies. Therefore, the CRC complexity made of natural or acquired multidrug resistance has made it necessary the search for new druggable targets and new delivery systems. Conclusion Further knowledge of the underlying CRC mechanisms and a comprehensive overview of current therapeutic opportunities can provide the basis for identifying pharmacological and biological barriers that render therapies ineffective and for identifying new potential biomarkers and therapeutic targets for advanced and aggressive CRC.
Collapse
Affiliation(s)
- Sara Tirendi
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Genoa, Italy
| | - Barbara Marengo
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Genoa, Italy
| | - Cinzia Domenicotti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Genoa, Italy
| | - Anna M. Bassi
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Genoa, Italy
| | - Vanessa Almonti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Stefania Vernazza
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Genoa, Italy
| |
Collapse
|
7
|
Cao Y, Chen E, Wang X, Song J, Zhang H, Chen X. An emerging master inducer and regulator for epithelial-mesenchymal transition and tumor metastasis: extracellular and intracellular ATP and its molecular functions and therapeutic potential. Cancer Cell Int 2023; 23:20. [PMID: 36750864 PMCID: PMC9903449 DOI: 10.1186/s12935-023-02859-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/24/2023] [Indexed: 02/09/2023] Open
Abstract
Despite the rapid development of therapeutic strategies in cancer treatment, metastasis remains the major cause of cancer-related death and scientific challenge. Epithelial-Mesenchymal Transition (EMT) plays a crucial role in cancer invasion and progression, a process by which tumor cells lose cell-cell adhesion and acquire increased invasiveness and metastatic activity. Recent work has uncovered some crucial roles of extracellular adenosine 5'- triphosphate (eATP), a major component of the tumor microenvironment (TME), in promoting tumor growth and metastasis. Intratumoral extracellular ATP (eATP), at levels of 100-700 µM, is 103-104 times higher than in normal tissues. In the current literature, eATP's function in promoting metastasis has been relatively poorly understood as compared with intracellular ATP (iATP). Recent evidence has shown that cancer cells internalize eATP via macropinocytosis in vitro and in vivo, promoting cell growth and survival, drug resistance, and metastasis. Furthermore, ATP acts as a messenger molecule that activates P2 purinergic receptors expressed on both tumor and host cells, stimulating downstream signaling pathways to enhance the invasive and metastatic properties of tumor cells. Here, we review recent progress in understanding eATP's role in each step of the metastatic cascade, including initiating invasion, inducing EMT, overcoming anoikis, facilitating intravasation, circulation, and extravasation, and eventually establishing metastatic colonization. Collectively, these studies reveal eATP's important functions in many steps of metastasis and identify new opportunities for developing more effective therapeutic strategies to target ATP-associated processes in cancer.
Collapse
Affiliation(s)
- Yanyang Cao
- grid.20627.310000 0001 0668 7841Department of Biological Sciences, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841The Edison Biotechnology Institute, Ohio University, Athens, OH USA
| | - Eileen Chen
- grid.20627.310000 0001 0668 7841Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA
| | - Xuan Wang
- grid.20627.310000 0001 0668 7841Department of Biological Sciences, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841The Edison Biotechnology Institute, Ohio University, Athens, OH USA
| | - Jingwen Song
- grid.20627.310000 0001 0668 7841Department of Biological Sciences, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841The Edison Biotechnology Institute, Ohio University, Athens, OH USA
| | - Haiyun Zhang
- grid.20627.310000 0001 0668 7841Department of Biological Sciences, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH USA ,grid.20627.310000 0001 0668 7841The Edison Biotechnology Institute, Ohio University, Athens, OH USA
| | - Xiaozhuo Chen
- Department of Biological Sciences, Ohio University, Athens, OH, USA. .,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH, USA. .,The Edison Biotechnology Institute, Ohio University, Athens, OH, USA. .,Department of Chemistry and Biochemistry, Ohio University, Athens, OH, USA. .,Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
8
|
Osum M, Kalkan R. Cancer Stem Cells and Their Therapeutic Usage. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1436:69-85. [PMID: 36689167 DOI: 10.1007/5584_2022_758] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cancer stem cells (CSC) have unique characteristics which include self-renewal, multi-directional differentiation capacity, quiescence/dormancy, and tumor-forming capability. These characteristics are referred to as the "stemness" properties. Tumor microenvironment contributes to CSC survival, function, and remaining them in an undifferentiated state. CSCs can form malignant tumors with heterogeneous phenotypes mediated by the tumor microenvironment. Therefore, the crosstalk between CSCs and tumor microenvironment can modulate tumor heterogeneity. CSCs play a crucial role in several biological processes, epithelial-mesenchymal transition (EMT), autophagy, and cellular stress response. In this chapter, we focused characteristics of cancer stem cells, reprogramming strategies cells into CSCs, and then we highlighted the contribution of CSCs to therapy resistance and cancer relapse and their potential of therapeutic targeting of CSCs.
Collapse
Affiliation(s)
- Meryem Osum
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Near East University, Nicosia, Cyprus
| | - Rasime Kalkan
- Department of Medical Genetics, Faculty of Medicine, Cyprus Health and Social Sciences University, Guzelyurt, Cyprus.
| |
Collapse
|
9
|
Seyfried TN, Arismendi-Morillo G, Zuccoli G, Lee DC, Duraj T, Elsakka AM, Maroon JC, Mukherjee P, Ta L, Shelton L, D'Agostino D, Kiebish M, Chinopoulos C. Metabolic management of microenvironment acidity in glioblastoma. Front Oncol 2022; 12:968351. [PMID: 36059707 PMCID: PMC9428719 DOI: 10.3389/fonc.2022.968351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma (GBM), similar to most cancers, is dependent on fermentation metabolism for the synthesis of biomass and energy (ATP) regardless of the cellular or genetic heterogeneity seen within the tumor. The transition from respiration to fermentation arises from the documented defects in the number, the structure, and the function of mitochondria and mitochondrial-associated membranes in GBM tissue. Glucose and glutamine are the major fermentable fuels that drive GBM growth. The major waste products of GBM cell fermentation (lactic acid, glutamic acid, and succinic acid) will acidify the microenvironment and are largely responsible for drug resistance, enhanced invasion, immunosuppression, and metastasis. Besides surgical debulking, therapies used for GBM management (radiation, chemotherapy, and steroids) enhance microenvironment acidification and, although often providing a time-limited disease control, will thus favor tumor recurrence and complications. The simultaneous restriction of glucose and glutamine, while elevating non-fermentable, anti-inflammatory ketone bodies, can help restore the pH balance of the microenvironment while, at the same time, providing a non-toxic therapeutic strategy for killing most of the neoplastic cells.
Collapse
Affiliation(s)
- Thomas N. Seyfried
- Biology Department, Boston College, Chestnut Hill, MA, United States
- *Correspondence: Thomas N. Seyfried,
| | - Gabriel Arismendi-Morillo
- Instituto de Investigaciones Biológicas, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Giulio Zuccoli
- The Program for the Study of Neurodevelopment in Rare Disorders (NDRD), University of Pittsburgh, Pittsburgh, PA, United States
| | - Derek C. Lee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Tomas Duraj
- Faculty of Medicine, Institute for Applied Molecular Medicine (IMMA), CEU San Pablo University, Madrid, Spain
| | - Ahmed M. Elsakka
- Neuro Metabolism, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Joseph C. Maroon
- Department of Neurosurgery, University of Pittsburgh, Medical Center, Pittsburgh, PA, United States
| | - Purna Mukherjee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Linh Ta
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | | | - Dominic D'Agostino
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, United States
| | | | | |
Collapse
|
10
|
Alam SR, Wallrabe H, Christopher KG, Siller KH, Periasamy A. Characterization of mitochondrial dysfunction due to laser damage by 2-photon FLIM microscopy. Sci Rep 2022; 12:11938. [PMID: 35831321 PMCID: PMC9279287 DOI: 10.1038/s41598-022-15639-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Mitochondria are the central organelles in cellular bio-energetics with key roles to play in energy metabolism and cell fate decisions. Fluorescence Lifetime Imaging microscopy (FLIM) is used to track metabolic changes by following the intrinsic co-enzymes NAD(P)H and FAD, present in metabolic pathways. FLIM records-lifetimes and the relative fractions of free (unbound) and bound states of NAD(P)H and FAD are achieved by multiphoton excitation of a pulsed femto-second infra-red laser. Optimization of multiphoton laser power levels is critical to achieve sufficient photon counts for correct lifetime fitting while avoiding phototoxic effects. We have characterized two photon (2p) laser induced changes at the intra-cellular level, specifically in the mitochondria, where damage was assessed at rising 2p laser average power excitation. Our results show that NAD(P)H-a2%—the lifetime-based enzyme bound fraction, an indicator of mitochondrial OXPHOS activity is increased by rising average power, while inducing changes in the mitochondria at higher power levels, quantified by different probes. Treatment response tracked by means of NAD(P)H-a2% can be confounded by laser-induced damage producing the same effect. Our study demonstrates that 2p-laser power optimization is critical by characterizing changes in the mitochondria at increasing laser average power.
Collapse
Affiliation(s)
- Shagufta Rehman Alam
- The W.M. Keck Center for Cellular Imaging, University of Virginia, Virginia, 22904, USA
| | - Horst Wallrabe
- The W.M. Keck Center for Cellular Imaging, University of Virginia, Virginia, 22904, USA
| | - Kathryn G Christopher
- The W.M. Keck Center for Cellular Imaging, University of Virginia, Virginia, 22904, USA
| | - Karsten H Siller
- Advanced Research Computing Services, University of Virginia, Virginia, 22904, USA
| | - Ammasi Periasamy
- The W.M. Keck Center for Cellular Imaging, University of Virginia, Virginia, 22904, USA. .,Departments of Biology and Biomedical Engineering, University of Virginia, Virginia, 22904, USA.
| |
Collapse
|
11
|
Pan Y, Zhou S, Liu C, Ma X, Xing J, Parshad B, Li W, Wu A, Haag R. Dendritic Polyglycerol-Conjugated Gold Nanostars for Metabolism Inhibition and Targeted Photothermal Therapy in Breast Cancer Stem Cells. Adv Healthc Mater 2022; 11:e2102272. [PMID: 34990518 PMCID: PMC11468648 DOI: 10.1002/adhm.202102272] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer stem cells (CSCs) are believed to be responsible for tumor initiation, invasion, metastasis, and recurrence, which lead to treatment failure. Thus, developing effective CSC-targeted therapeutic strategies is crucial for enhancing therapeutic efficacy. In this work, GNSs-dPG-3BP, TPP, and HA nanocomposite particles are developed by simultaneously conjugating hexokinase 2 (HK2) inhibitor 3-bromopyruvate (3BP), mitochondrial targeting molecule triphenyl phosphonium (TPP), and CSCs targeting agent hyaluronic acid (HA) onto gold nanostars-dendritic polyglycerol (GNSs-dPG) nanoplatforms for efficient eradication of CSCs. The nanocomposite particles possess good biocompatibility and exhibit superior mitochondrial-bound HK2 binding ability via 3BP to inhibit metabolism, and further induce cellular apoptosis by releasing the cytochrome c. Therefore, it enhanced the therapeutic efficacy of CSCs-specific targeted photothermal therapy (PTT), and achieved a synergistic effect for the eradication of breast CSCs. After administration of the synergistic treatment, the self-renewal of breast CSCs and the stemness gene expression are suppressed, CSC-driven mammosphere formation is diminished, the in vivo tumor growth is effectively inhibited, and CSCs are eradicated. Altogether, GNSs-dPG-3BP, TPP, and HA nanocomposite particles have been developed, which will provide a novel strategy for precise and highly efficient targeted eradication of CSCs.
Collapse
Affiliation(s)
- Yuanwei Pan
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Suqiong Zhou
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Chuang Liu
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Xuehua Ma
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Jie Xing
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Badri Parshad
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Wenzhong Li
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| | - Aiguo Wu
- Cixi Institute of Biomedical EngineeringCAS Key Laboratory of Magnetic Materials and Devices, & Key Laboratory of Additive Manufacturing Materials of Zhejiang ProvinceNingbo Institute of Materials Technology and EngineeringChinese Academy of SciencesNingbo315201P. R. China
| | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustrasse 3Berlin14195Germany
| |
Collapse
|
12
|
Network Biology and Artificial Intelligence Drive the Understanding of the Multidrug Resistance Phenotype in Cancer. Drug Resist Updat 2022; 60:100811. [DOI: 10.1016/j.drup.2022.100811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
|
13
|
Dadgar T, Ebrahimi N, Gholipour AR, Akbari M, Khani L, Ahmadi A, Hamblin MR. Targeting the metabolism of cancer stem cells by energy disruptor molecules. Crit Rev Oncol Hematol 2021; 169:103545. [PMID: 34838705 DOI: 10.1016/j.critrevonc.2021.103545] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 10/17/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) have been identified in various tumor types. CSCs are believed to contribute to tumor metastasis and resistance to conventional therapy. So targeting these cells could be an effective strategy to eliminate tumors and a promising new type of cancer treatment. Alterations in metabolism play an essential role in CSC biology and their resistance to treatment. The metabolic properties pathways in CSCs are different from normal cells, and to some extent, are different from regular tumor cells. Interestingly, CSCs can use other nutrients for their metabolism and growth. The different metabolism causes increased sensitivity of CSCs to agents that disrupt cellular homeostasis. Compounds that interfere with the central metabolic pathways are known as energy disruptors and can reduce CSC survival. This review highlights the differences between regular cancer cells and CSC metabolism and discusses the action mechanisms of energy disruptors at the cellular and molecular levels.
Collapse
Affiliation(s)
- Tahere Dadgar
- Department of Biology, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Nasim Ebrahimi
- Division of Genetics, Department of Cell and Molecular & Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Gholipour
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Akbari
- Department of Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khani
- Department of Immunology, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Amirhossein Ahmadi
- Department of Biological Science and Technology, Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr, 75169, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| |
Collapse
|
14
|
Ion Channels, Transporters, and Sensors Interact with the Acidic Tumor Microenvironment to Modify Cancer Progression. Rev Physiol Biochem Pharmacol 2021; 182:39-84. [PMID: 34291319 DOI: 10.1007/112_2021_63] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Solid tumors, including breast carcinomas, are heterogeneous but typically characterized by elevated cellular turnover and metabolism, diffusion limitations based on the complex tumor architecture, and abnormal intra- and extracellular ion compositions particularly as regards acid-base equivalents. Carcinogenesis-related alterations in expression and function of ion channels and transporters, cellular energy levels, and organellar H+ sequestration further modify the acid-base composition within tumors and influence cancer cell functions, including cell proliferation, migration, and survival. Cancer cells defend their cytosolic pH and HCO3- concentrations better than normal cells when challenged with the marked deviations in extracellular H+, HCO3-, and lactate concentrations typical of the tumor microenvironment. Ionic gradients determine the driving forces for ion transporters and channels and influence the membrane potential. Cancer and stromal cells also sense abnormal ion concentrations via intra- and extracellular receptors that modify cancer progression and prognosis. With emphasis on breast cancer, the current review first addresses the altered ion composition and the changes in expression and functional activity of ion channels and transporters in solid cancer tissue. It then discusses how ion channels, transporters, and cellular sensors under influence of the acidic tumor microenvironment shape cancer development and progression and affect the potential of cancer therapies.
Collapse
|
15
|
Zhang H, Steed A, Co M, Chen X. Cancer stem cells, epithelial-mesenchymal transition, ATP and their roles in drug resistance in cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:684-709. [PMID: 34322664 PMCID: PMC8315560 DOI: 10.20517/cdr.2021.32] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cancer stem cell (CSC) state and epithelial-mesenchymal transition (EMT) activation are tightly interconnected. Cancer cells that acquire the EMT/CSC phenotype are equipped with adaptive metabolic changes to maintain low reactive oxygen species levels and stemness, enhanced drug transporters, anti-apoptotic machinery and DNA repair system. Factors present in the tumor microenvironment such as hypoxia and the communication with non-cancer stromal cells also promote cancer cells to enter the EMT/CSC state and display related resistance. ATP, particularly the high levels of intratumoral extracellular ATP functioning through both signaling pathways and ATP internalization, induces and regulates EMT and CSC. The three of them work together to enhance drug resistance. New findings in each of these factors will help us explore deeper into mechanisms of drug resistance and suggest new resistance-associated markers and therapeutic targets.
Collapse
Affiliation(s)
- Haiyun Zhang
- Department of Biological Science, Ohio University, Athens, OH 45701, USA.,Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA
| | - Alexander Steed
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Milo Co
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Xiaozhuo Chen
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA.,Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA.,Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
16
|
Abstract
Genomic information is encoded on long strands of DNA, which are folded into chromatin and stored in a tiny nucleus. Nuclear chromatin is a negatively charged polymer composed of DNA, histones, and various nonhistone proteins. Because of its highly charged nature, chromatin structure varies greatly depending on the surrounding environment (e.g., cations, molecular crowding, etc.). New technologies to capture chromatin in living cells have been developed over the past 10 years. Our view on chromatin organization has drastically shifted from a regular and static one to a more variable and dynamic one. Chromatin forms numerous compact dynamic domains that act as functional units of the genome in higher eukaryotic cells and locally appear liquid-like. By changing DNA accessibility, these domains can govern various functions. Based on new evidences from versatile genomics and advanced imaging studies, we discuss the physical nature of chromatin in the crowded nuclear environment and how it is regulated.
Collapse
Affiliation(s)
- Kazuhiro Maeshima
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
- Department of Genetics, School of Life Science, Sokendai (Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Shiori Iida
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
- Department of Genetics, School of Life Science, Sokendai (Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Sachiko Tamura
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
17
|
Integrated Metabolomics and Transcriptomics Using an Optimised Dual Extraction Process to Study Human Brain Cancer Cells and Tissues. Metabolites 2021; 11:metabo11040240. [PMID: 33919944 PMCID: PMC8070957 DOI: 10.3390/metabo11040240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 11/18/2022] Open
Abstract
The integration of untargeted metabolomics and transcriptomics from the same population of cells or tissue enhances the confidence in the identified metabolic pathways and understanding of the enzyme–metabolite relationship. Here, we optimised a simultaneous extraction method of metabolites/lipids and RNA from ependymoma cells (BXD-1425). Relative to established RNA (mirVana kit) or metabolite (sequential solvent addition and shaking) single extraction methods, four dual-extraction techniques were evaluated and compared (methanol:water:chloroform ratios): cryomill/mirVana (1:1:2); cryomill-wash/Econospin (5:1:2); rotation/phenol-chloroform (9:10:1); Sequential/mirVana (1:1:3). All methods extracted the same metabolites, yet rotation/phenol-chloroform did not extract lipids. Cryomill/mirVana and sequential/mirVana recovered the highest amounts of RNA, at 70 and 68% of that recovered with mirVana kit alone. sequential/mirVana, involving RNA extraction from the interphase of our established sequential solvent addition and shaking metabolomics-lipidomics extraction method, was the most efficient approach overall. Sequential/mirVana was applied to study a) the biological effect caused by acute serum starvation in BXD-1425 cells and b) primary ependymoma tumour tissue. We found (a) 64 differentially abundant metabolites and 28 differentially expressed metabolic genes, discovering four gene-metabolite interactions, and (b) all metabolites and 62% lipids were above the limit of detection, and RNA yield was sufficient for transcriptomics, in just 10 mg of tissue.
Collapse
|
18
|
Sameiyan E, Bagheri E, Dehghani S, Ramezani M, Alibolandi M, Abnous K, Taghdisi SM. Aptamer-based ATP-responsive delivery systems for cancer diagnosis and treatment. Acta Biomater 2021; 123:110-122. [PMID: 33453405 DOI: 10.1016/j.actbio.2020.12.057] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/25/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022]
Abstract
In recent years, many stimuli-triggered drug delivery platforms have been designed to deliver drugs accurately to specific sites and reduce their side effects, improving "on-demand" therapeutic efficacy. Adenosine-5'-triphosphate (ATP)-responsive drug delivery methods are examples of these systems that use ATP molecules as a trigger for delivery of therapeutic agents. Since intra- and extra-cellular ATP concentrations are significantly different from each other (1-10 mM and <0.4 mM, respectively), the use of ATP can be a practical method for regulating drug release. Aptamers possess unique properties including, ligand-specific response, short sequence (~ 20-80 bases) and easy functionalization. Thus, their combination with ATP-responsive systems results in more accurate drug delivery systems and greater control of drug release. A wide range of nanoparticles, such as polymeric nanogels, liposomes, metallic nanoparticles, protein, or DNA nano-assemblies, have been employed in the fabrication of nanocarriers. In this review, we describe several ATP-responsive drug delivery systems based on the various carriers and discuss the challenges and strengths of each method.
Collapse
|
19
|
Oizel K, Yang C, Renoult O, Gautier F, Do QN, Joalland N, Gao X, Ko B, Vallette F, Ge WP, Paris F, DeBerardinis RJ, Pecqueur C. Glutamine uptake and utilization of human mesenchymal glioblastoma in orthotopic mouse model. Cancer Metab 2020; 8:9. [PMID: 32789014 PMCID: PMC7416393 DOI: 10.1186/s40170-020-00215-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 03/24/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) are highly heterogeneous on the cellular and molecular basis. It has been proposed that glutamine metabolism of primary cells established from human tumors discriminates aggressive mesenchymal GBM subtype to other subtypes. METHODS To study glutamine metabolism in vivo, we used a human orthotopic mouse model for GBM. Tumors evolving from the implanted primary GBM cells expressing different molecular signatures were analyzed using mass spectrometry for their metabolite pools and enrichment in carbon 13 (13C) after 13C-glutamine infusion. RESULTS Our results showed that mesenchymal GBM tumors displayed increased glutamine uptake and utilization compared to both control brain tissue and other GBM subtypes. Furthermore, both glutamine synthetase and transglutaminase-2 were expressed accordingly to GBM metabolic phenotypes. CONCLUSION Thus, our results outline the specific enhanced glutamine flux in vivo of the aggressive mesenchymal GBM subtype.
Collapse
Affiliation(s)
- Kristell Oizel
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Chendong Yang
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | | | - Fabien Gautier
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Quyen N. Do
- Department of Radiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9061 USA
| | - Noemie Joalland
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| | - Xiaofei Gao
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Bookyung Ko
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - François Vallette
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Woo-Ping Ge
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Pediatrics, Neuroscience, Neurology & Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - François Paris
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- Institut de Cancérologie de l’Ouest, Saint-Herblain, France
| | - Ralph J. DeBerardinis
- Children’s Research Institute, UT Southwestern Medical Center, Dallas, TX 75390 USA
- Howard Hughes Medical Institute, Chevy Chase, USA
| | - Claire Pecqueur
- Université de Nantes, CNRS, INSERM, CRCINA, Nantes, France
- LabEx IGO “Immunotherapy, Graft, Oncology”, Nantes, France
| |
Collapse
|
20
|
Heterogeneous Response in Rabbit Fetal Diaphragmatic Hernia Lungs After Tracheal Occlusion. J Surg Res 2020; 250:23-38. [PMID: 32014698 DOI: 10.1016/j.jss.2019.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/10/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Fetal tracheal occlusion (TO) is an experimental therapeutic approach to stimulate lung growth in the most severe congenital diaphragmatic hernia (CDH) cases. We have previously demonstrated a heterogeneous response of normal fetal rabbit lungs after TO with the appearance of at least two distinct zones. The aim of this study was to examine the fetal lung response after TO in a left CDH fetal rabbit model. METHODS Fetal rabbits at 25 d gestation underwent surgical creation of CDH followed by TO at 27 d and harvest on day 30. Morphometric analysis, global metabolomics, and fluorescence lifetime imaging microscopy (FLIM) were performed to evaluate structural and metabolic changes in control, CDH, and CDH + TO lungs. RESULTS Right and left lungs were different at the baseline and had a heterogeneous pulmonary growth response in CDH and after TO. The relative percent growth of the right lungs in CDH + TO was higher than the left lungs. Morphometric analyses revealed heterogeneous tissue-to-airspace ratios, in addition to size and number of airspaces within and between the lungs in the different groups. Global metabolomics demonstrated a slower rate of metabolism in the CDH group with the left lungs being less metabolically active. TO stimulated metabolic activity in both lungs to different degrees. FLIM analysis demonstrated local heterogeneity in glycolysis, oxidative phosphorylation (OXPHOS), and FLIM "lipid-surfactant" signal within and between the right and left lungs in all groups. CONCLUSIONS We demonstrate that TO leads to a heterogeneous morphologic and metabolic response within and between the right and left lungs in a left CDH rabbit model.
Collapse
|
21
|
Rabé M, Dumont S, Álvarez-Arenas A, Janati H, Belmonte-Beitia J, Calvo GF, Thibault-Carpentier C, Séry Q, Chauvin C, Joalland N, Briand F, Blandin S, Scotet E, Pecqueur C, Clairambault J, Oliver L, Perez-Garcia V, Nadaradjane A, Cartron PF, Gratas C, Vallette FM. Identification of a transient state during the acquisition of temozolomide resistance in glioblastoma. Cell Death Dis 2020; 11:19. [PMID: 31907355 PMCID: PMC6944699 DOI: 10.1038/s41419-019-2200-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 12/17/2022]
Abstract
Drug resistance limits the therapeutic efficacy in cancers and leads to tumor recurrence through ill-defined mechanisms. Glioblastoma (GBM) are the deadliest brain tumors in adults. GBM, at diagnosis or after treatment, are resistant to temozolomide (TMZ), the standard chemotherapy. To better understand the acquisition of this resistance, we performed a longitudinal study, using a combination of mathematical models, RNA sequencing, single cell analyses, functional and drug assays in a human glioma cell line (U251). After an initial response characterized by cell death induction, cells entered a transient state defined by slow growth, a distinct morphology and a shift of metabolism. Specific genes expression associated to this population revealed chromatin remodeling. Indeed, the histone deacetylase inhibitor trichostatin (TSA), specifically eliminated this population and thus prevented the appearance of fast growing TMZ-resistant cells. In conclusion, we have identified in glioblastoma a population with tolerant-like features, which could constitute a therapeutic target.
Collapse
Affiliation(s)
- Marion Rabé
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Solenne Dumont
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,GenoBiRD, SFR François Bonamy, Université de Nantes, Nantes, France
| | - Arturo Álvarez-Arenas
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | - Hicham Janati
- Laboratoire Jacques-Louis Lions, Inria, Mamba team and Sorbonne Université, Paris 6, UPMC, Paris, France
| | - Juan Belmonte-Beitia
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | - Gabriel F Calvo
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | | | - Quentin Séry
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France
| | - Cynthia Chauvin
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Noémie Joalland
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Floriane Briand
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Stéphanie Blandin
- Plate-Forme MicroPICell, SFR François Bonamy, Université de Nantes, Nantes, France
| | - Emmanuel Scotet
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Claire Pecqueur
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Jean Clairambault
- Laboratoire Jacques-Louis Lions, Inria, Mamba team and Sorbonne Université, Paris 6, UPMC, Paris, France
| | - Lisa Oliver
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,CHU Nantes, 44093, Nantes, France
| | - Victor Perez-Garcia
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | - Arulraj Nadaradjane
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France
| | - Pierre-François Cartron
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France
| | - Catherine Gratas
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,CHU Nantes, 44093, Nantes, France.
| | - François M Vallette
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France.
| |
Collapse
|
22
|
Perusina Lanfranca M, Thompson JK, Bednar F, Halbrook C, Lyssiotis C, Levi B, Frankel TL. Metabolism and epigenetics of pancreatic cancer stem cells. Semin Cancer Biol 2019; 57:19-26. [PMID: 30273655 PMCID: PMC6438777 DOI: 10.1016/j.semcancer.2018.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 09/26/2018] [Indexed: 02/06/2023]
Abstract
Pancreatic Cancer (PDA) is an aggressive malignancy characterized by early spread and a high mortality. Current studies suggest that a subpopulation of cells exist within tumors, cancer stem cell (CSC), which are capable of self-renewal and give rise to unique progeny which form the major neoplastic cellular component of tumors. While CSCs constitute a small cellular subpopulation within the tumor, their resistance to chemotherapy and radiation make them an important therapeutic target for eradication. Along with distinctive phenotypic properties, CSCs possess a unique metabolic plasticity allowing them to rapidly respond and adapt to environmental changes. These cells and their progeny also display a significantly altered epigenetic state with distinctive patterns of DNA methylation. Several mechanisms of cross-talk between epigenetic and metabolic pathways in PDA exist which ultimately contribute to the observed cellular plasticity and enhanced tumorigenesis. In this review we discuss various examples of this metabolic-epigenetic interplay and how it may constitute a new avenue for therapy specifically targeting CSCs in PDA.
Collapse
Affiliation(s)
| | - J K Thompson
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - F Bednar
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - C Halbrook
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States; Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - C Lyssiotis
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States; Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - B Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - T L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
23
|
Abstract
Cancer is the second leading cause of death in the US. Current major treatments for cancer management include surgery, cytotoxic chemotherapy, targeted therapy, radiation therapy, endocrine therapy and immunotherapy. Despite the endeavors and achievements made in treating cancers during the past decades, resistance to classical chemotherapeutic agents and/or novel targeted drugs continues to be a major problem in cancer therapies. Drug resistance, either existing before treatment (intrinsic) or generated after therapy (acquired), is responsible for most relapses of cancer, one of the major causes of death of the disease. Heterogeneity among patients and tumors, and the versatility of cancer to circumvent therapies make drug resistance more challenging to deal with. Better understanding the mechanisms of drug resistance is required to provide guidance to future cancer treatment and achieve better outcomes. In this review, intrinsic and acquired resistance will be discussed. In addition, new discoveries in mechanisms of drug resistance will be reviewed. Particularly, we will highlight roles of ATP in drug resistance by discussing recent findings of exceptionally high levels of intratumoral extracellular ATP as well as intracellular ATP internalized from extracellular environment. The complexity of drug resistance development suggests that combinational and personalized therapies, which should take ATP into consideration, might provide better strategies and improved efficacy for fighting drug resistance in cancer.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Haiyun Zhang
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA
| | - Xiaozhuo Chen
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, OH 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA.,Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
24
|
Kim P, Jia P, Zhao Z. Kinase impact assessment in the landscape of fusion genes that retain kinase domains: a pan-cancer study. Brief Bioinform 2019; 19:450-460. [PMID: 28013235 DOI: 10.1093/bib/bbw127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Indexed: 12/13/2022] Open
Abstract
Assessing the impact of kinase in gene fusion is essential for both identifying driver fusion genes (FGs) and developing molecular targeted therapies. Kinase domain retention is a crucial factor in kinase fusion genes (KFGs), but such a systematic investigation has not been done yet. To this end, we analyzed kinase domain retention (KDR) status in chimeric protein sequences of 914 KFGs covering 312 kinases across 13 major cancer types. Based on 171 kinase domain-retained KFGs including 101 kinases, we studied their recurrence, kinase groups, fusion partners, exon-based expression depth, short DNA motifs around the break points and networks. Our results, such as more KDR than 5'-kinase fusion genes, combinatorial effects between 3'-KDR kinases and their 5'-partners and a signal transduction-specific DNA sequence motif in the break point intronic sequences, supported positive selection on 3'-kinase fusion genes in cancer. We introduced a degree-of-frequency (DoF) score to measure the possible number of KFGs of a kinase. Interestingly, kinases with high DoF scores tended to undergo strong gene expression alteration at the break points. Furthermore, our KDR gene fusion network analysis revealed six of the seven kinases with the highest DoF scores (ALK, BRAF, MET, NTRK1, NTRK3 and RET) were all observed in thyroid carcinoma. Finally, we summarized common features of 'effective' (highly recurrent) kinases in gene fusions such as expression alteration at break point, redundant usage in multiple cancer types and 3'-location tendency. Collectively, our findings are useful for prioritizing driver kinases and FGs and provided insights into KFGs' clinical implications.
Collapse
Affiliation(s)
- Pora Kim
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
25
|
Jonnalagadda S, Jonnalagadda SK, Ronayne CT, Nelson GL, Solano LN, Rumbley J, Holy J, Mereddy VR, Drewes LR. Novel N,N-dialkyl cyanocinnamic acids as monocarboxylate transporter 1 and 4 inhibitors. Oncotarget 2019; 10:2355-2368. [PMID: 31040927 PMCID: PMC6481325 DOI: 10.18632/oncotarget.26760] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/22/2019] [Indexed: 12/25/2022] Open
Abstract
Potent and dual monocarboxylate transporter (MCT) 1 and 4 inhibitors have been developed for the first time as potential anticancer agents based on α-cyanocinnamic acid structural template. Candidate inhibitors 1-9 have been evaluated for in vitro cell proliferation against MCT1 and MCT4 expressing cancer cell lines. Potential MCT1 and MCT4 binding interactions of the lead compound 9 have been studied through homology modeling and molecular docking prediction. In vitro effects on extracellular flux via glycolysis and mitochondrial stress tests suggest that candidate compounds 3 and 9 disrupt glycolysis and OxPhos efficiently in MCT1 expressing colorectal adenocarcinoma WiDr and MCT4 expressing triple negative breast cancer MDA-MB-231 cells. Fluorescence microscopy analyses in these cells also indicate that compound 9 is internalized and concentrated near mitochondria. In vivo tumor growth inhibition studies in WiDr and MDA-MB-231 xenograft tumor models in mice indicate that the candidate compound 9 exhibits a significant single agent activity.
Collapse
Affiliation(s)
- Shirisha Jonnalagadda
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA
| | - Sravan K Jonnalagadda
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA
| | - Conor T Ronayne
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA
| | - Grady L Nelson
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA
| | - Lucas N Solano
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA
| | - Jon Rumbley
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA.,Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN 55812, USA
| | - Jon Holy
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA.,Department of Biomedical Sciences, Medical School Duluth, University of Minnesota, Duluth, MN 55812, USA
| | - Venkatram R Mereddy
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA.,Department of Pharmacy Practice & Pharmaceutical Sciences, University of Minnesota, Duluth, MN 55812, USA.,Department of Chemistry and Biochemistry, University of Minnesota, Duluth, MN 55812, USA
| | - Lester R Drewes
- Integrated Biosciences Graduate Program, University of Minnesota, Duluth, MN 55812, USA.,Department of Biomedical Sciences, Medical School Duluth, University of Minnesota, Duluth, MN 55812, USA
| |
Collapse
|
26
|
Garnier D, Renoult O, Alves-Guerra MC, Paris F, Pecqueur C. Glioblastoma Stem- Like Cells, Metabolic Strategy to Kill a Challenging Target. Front Oncol 2019; 9:118. [PMID: 30895167 PMCID: PMC6415584 DOI: 10.3389/fonc.2019.00118] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/11/2019] [Indexed: 01/25/2023] Open
Abstract
Over the years, substantial evidence has definitively confirmed the existence of cancer stem-like cells within tumors such as Glioblastoma (GBM). The importance of Glioblastoma stem-like cells (GSCs) in tumor progression and relapse clearly highlights that cancer eradication requires killing of GSCs that are intrinsically resistant to conventional therapies as well as eradication of the non-GSCs cells since GSCs emergence relies on a dynamic process. The past decade of research highlights that metabolism is a significant player in tumor progression and actually might orchestrate it. The growing interest in cancer metabolism reprogrammation can lead to innovative approaches exploiting metabolic vulnerabilities of cancer cells. These approaches are challenging since they require overcoming the compensatory and adaptive responses of GSCs. In this review, we will summarize the current knowledge on GSCs with a particular focus on their metabolic complexity. We will also discuss potential approaches targeting GSCs metabolism to potentially improve clinical care.
Collapse
Affiliation(s)
| | | | | | - François Paris
- CRCINA, INSERM CNRS, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest - René Gauducheau, St Herblain, France
| | - Claire Pecqueur
- CRCINA, INSERM CNRS, Université de Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
27
|
Co-Expression Analysis Reveals Mechanisms Underlying the Varied Roles of NOTCH1 in NSCLC. J Thorac Oncol 2018; 14:223-236. [PMID: 30408569 DOI: 10.1016/j.jtho.2018.10.162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 09/23/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Notch receptor family dysregulation can be tumor promoting or suppressing depending on cellular context. Our studies shed light on the mechanistic differences that are responsible for NOTCH1's opposing roles in lung adenocarcinoma and lung squamous cell carcinoma. METHODS We integrated transcriptional patient-derived datasets with gene co-expression analyses to elucidate mechanisms behind NOTCH1 function in subsets of NSCLC. Differential co-expression was examined using hierarchical clustering and principal component analysis. Enrichment analysis was used to examine pathways associated with the underlying transcriptional networks. These pathways were validated in vitro and in vivo. Endogenously epitope-tagged NOTCH1 was used to identify novel interacting proteins. RESULTS NOTCH1 co-expressed genes in lung adenocarcinoma and squamous carcinoma were distinct and associated with either angiogenesis and immune system pathways or cell cycle control and mitosis pathways, respectively. Tissue culture and xenograft studies of lung adenocarcinoma and lung squamous models with NOTCH1 knockdown showed growth differences and opposing effects on these pathways. Differential NOTCH1 interacting proteins were identified as potential mediators of these differences. CONCLUSIONS Recognition of the opposing role of NOTCH1 in lung cancer, downstream pathways, and interacting proteins in each context may help direct the development of rational NOTCH1 pathway-dependent targeted therapies for specific tumor subsets of NSCLC.
Collapse
|
28
|
Valentín-Guillama G, López S, Kucheryavykh YV, Chorna NE, Pérez J, Ortiz-Rivera J, Inyushin M, Makarov V, Valentín-Acevedo A, Quinones-Hinojosa A, Boukli N, Kucheryavykh LY. HIV-1 Envelope Protein gp120 Promotes Proliferation and the Activation of Glycolysis in Glioma Cell. Cancers (Basel) 2018; 10:cancers10090301. [PMID: 30200472 PMCID: PMC6162763 DOI: 10.3390/cancers10090301] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/14/2022] Open
Abstract
Patients infected with human immunodeficiency virus (HIV) are more prone to developing cancers, including glioblastomas (GBMs). The median survival for HIV positive GBM patients is significantly shorter than for those who are uninfected, despite the fact that they receive the same treatments. The nature of the GBM–HIV association remains poorly understood. In this study, we analyzed the effect of the HIV envelope glycoprotein gp120 on GBM cell proliferation. Specifically, we performed cell cycle, western blot, protein synthesis and metabolomics analysis as well as ATP production and oxygen consumption assays to evaluate proliferation and metabolic pathways in primary human glioma cell line, U87, A172 cells and in the HIVgp120tg/GL261 mouse model. Glioma cells treated with gp120 (100 ng/mL for 7–10 days) showed higher proliferation rates and upregulation in the expression of enolase 2, hexokinase and glyceraldehyde-3-phosphate dehydrogenase when compared to untreated cells. Furthermore, we detected an increase in the activity of pyruvate kinase and a higher glycolytic index in gp120 treated cells. Gp120 treated GBM cells also showed heightened lipid and protein synthesis. Overall, we demonstrate that in glioma cells, the HIV envelope glycoprotein promotes proliferation and activation of glycolysis resulting in increased protein and lipid synthesis.
Collapse
Affiliation(s)
- Gabriel Valentín-Guillama
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Sheila López
- Biomedical Proteomics Facility, Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Yuriy V Kucheryavykh
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Nataliya E Chorna
- Department of Biochemistry, University of Puerto Rico, School of Medicine, San Juan, PR 00936, USA.
| | - Jose Pérez
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Jescelica Ortiz-Rivera
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Michael Inyushin
- Department of Physiology, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA, .
| | - Vladimir Makarov
- Department of Physics, University of Puerto Rico, Río Piedras Campus, San Juan, PR 00931, USA.
| | - Aníbal Valentín-Acevedo
- Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Alfredo Quinones-Hinojosa
- Department of Neurologic Surgery, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA.
| | - Nawal Boukli
- Biomedical Proteomics Facility, Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| | - Lilia Y Kucheryavykh
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, Ave. Laurel, Santa Juanita, Bayamon, PR 00956, USA.
| |
Collapse
|
29
|
Zhang H, Guo X, Feng X, Wang T, Hu Z, Que X, Tian Q, Zhu T, Guo G, Huang W, Li X. MiRNA-543 promotes osteosarcoma cell proliferation and glycolysis by partially suppressing PRMT9 and stabilizing HIF-1α protein. Oncotarget 2018; 8:2342-2355. [PMID: 27911265 PMCID: PMC5356804 DOI: 10.18632/oncotarget.13672] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 11/06/2016] [Indexed: 01/17/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor, occurring frequently in adolescents and possessing a high malignant severity. MicroRNAs play critical roles during OS development. Thus, elucidation of the involvement of specific microRNAs in the development of OS may provide novel therapeutic targets for OS treatment. Here, we showed that in the OS specimens from patients, the levels of miR-543 were significantly increased whereas the levels of PRMT9 were significantly decreased, compared to the paired normal bone tissue. Moreover, miR-543 and PRMT9 inversely correlated in the OS cell lines. Bioinformatics analyses predicted that miR-543 may target the 3'-UTR of PRMT9 mRNA to inhibit its translation, which was confirmed by luciferase-reporter assay. MiR-543 promoted OS cell proliferation in vitro and in vivo. Mechanistically, miR-543 inhibited PRMT9-enhanced cell oxidative phosphorylation, while miR-543 depletion promoted PRMT9-increased HIF-1α instability and inhibited glycolysis in OS cells. Clinically, miR-543 expression was negatively correlated with PRMT9 expression in OS tissues. Together, our data provide important evidence for glycolysis in OS development, and suggest that targeting glycolytic pathway through miR-543/PRMT9/HIF-1α axis may represent a potential therapeutic strategy to eradicate OS cells.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Xiaofeng Guo
- Medical College of Xiamen University, Xiamen, Fujian, China
| | - Xing Feng
- Department of Molecular Pharmacology, Rutgers University, New Brunswick, New Jersey, USA
| | - Tingting Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Zhaohua Hu
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Xiangyong Que
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Qingsong Tian
- Department of Medicine, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Tianbo Zhu
- Department of Medicine, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Guixian Guo
- Department of Medicine, The Second Hospital Affiliated to Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Huang
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| | - Xinzhi Li
- Department of Orthopedics, Renhe Hospital, China Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
30
|
Segmented cell analyses to measure redox states of autofluorescent NAD(P)H, FAD & Trp in cancer cells by FLIM. Sci Rep 2018; 8:79. [PMID: 29311591 PMCID: PMC5758727 DOI: 10.1038/s41598-017-18634-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/13/2017] [Indexed: 01/07/2023] Open
Abstract
Multiphoton FLIM microscopy offers many opportunities to investigate processes in live cells, tissue and animal model systems. For redox measurements, FLIM data is mostly published by cell mean values and intensity-based redox ratios. Our method is based entirely on FLIM parameters generated by 3-detector time domain microscopy capturing autofluorescent signals of NAD(P)H, FAD and novel FLIM-FRET application of Tryptophan and NAD(P)H-a2%/FAD-a1% redox ratio. Furthermore, image data is analyzed in segmented cells thresholded by 2 × 2 pixel Regions of Interest (ROIs) to separate mitochondrial oxidative phosphorylation from cytosolic glycolysis in a prostate cancer cell line. Hundreds of data points allow demonstration of heterogeneity in response to intervention, identity of cell responders to treatment, creating thereby different sub-populations. Histograms and bar charts visualize differences between cells, analyzing whole cell versus mitochondrial morphology data, all based on discrete ROIs. This assay method allows to detect subtle differences in cellular and tissue responses, suggesting an advancement over means-based analyses.
Collapse
|
31
|
Norouzi-Barough L, Sarookhani MR, Sharifi M, Moghbelinejad S, Jangjoo S, Salehi R. Molecular mechanisms of drug resistance in ovarian cancer. J Cell Physiol 2018; 233:4546-4562. [PMID: 29152737 DOI: 10.1002/jcp.26289] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/14/2017] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the most lethal malignancy among the gynecological cancers, with a 5-year survival rate, mainly due to being diagnosed at advanced stages, recurrence and resistance to the current chemotherapeutic agents. Drug resistance is a complex phenomenon and the number of known involved genes and cross-talks between signaling pathways in this process is growing rapidly. Thus, discovering and understanding the underlying molecular mechanisms involved in chemo-resistance are crucial for management of treatment and identifying novel and effective drug targets as well as drug discovery to improve therapeutic outcomes. In this review, the major and recently identified molecular mechanisms of drug resistance in ovarian cancer from relevant literature have been investigated. In the final section of the paper, new approaches for studying detailed mechanisms of chemo-resistance have been briefly discussed.
Collapse
Affiliation(s)
- Leyla Norouzi-Barough
- Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Mohammadreza Sharifi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sahar Moghbelinejad
- Department of Biochemistry and Genetic, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Saranaz Jangjoo
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
32
|
Dichloroacetate induced intracellular acidification in glioblastoma: in vivo detection using AACID-CEST MRI at 9.4 Tesla. J Neurooncol 2017; 136:255-262. [DOI: 10.1007/s11060-017-2664-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022]
|
33
|
Wang X, Li Y, Qian Y, Cao Y, Shriwas P, Zhang H, Chen X. Extracellular ATP, as an energy and phosphorylating molecule, induces different types of drug resistances in cancer cells through ATP internalization and intracellular ATP level increase. Oncotarget 2017; 8:87860-87877. [PMID: 29152126 PMCID: PMC5675678 DOI: 10.18632/oncotarget.21231] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/16/2017] [Indexed: 11/25/2022] Open
Abstract
Cancer cells are able to uptake extracellular ATP (eATP) via macropinocytosis to elevate intracellular ATP (iATP) levels, enhancing their survival in drug treatment. However, the involved drug resistance mechanisms are unknown. Here we investigated the roles of eATP as either an energy or a phosphorylating molecule in general drug resistance mediated by ATP internalization and iATP elevation. We report that eATP increased iATP levels and promoted drug resistance to various tyrosine kinase inhibitors (TKIs) and chemo-drugs in human cancer cell lines of five cancer types. In A549 lung cancer cells, the resistance was downregulated by macropinocytosis inhibition or siRNA knockdown of PAK1, an essential macropinocytosis enzyme. The elevated iATP upregulated the efflux activity of ABC transporters in A549 and SK-Hep-1 cells as well as phosphorylation of PDGFRα and proteins in the PDGFR-mediated Akt-mTOR and Raf-MEK signaling pathways in A549 cells. Similar phosphorylation upregulations were found in A549 tumors. These results demonstrate that eATP induces different types of drug resistance by eATP internalization and iATP elevation, implicating the ATP-rich tumor microenvironment in cancer drug resistance, expanding our understanding of the roles of eATP in the Warburg effect and offering new anticancer drug resistance targets.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Biological Sciences, Ohio University, Athens, Ohio 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Yunsheng Li
- The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Yanrong Qian
- The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Yanyang Cao
- Department of Biological Sciences, Ohio University, Athens, Ohio 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Pratik Shriwas
- Department of Biological Sciences, Ohio University, Athens, Ohio 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Haiyun Zhang
- Department of Biological Sciences, Ohio University, Athens, Ohio 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA
| | - Xiaozhuo Chen
- Department of Biological Sciences, Ohio University, Athens, Ohio 45701, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Ohio University, Athens, Ohio 45701, USA.,The Edison Biotechnology Institute, Ohio University, Athens, Ohio 45701, USA.,Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio 45701, USA.,Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| |
Collapse
|
34
|
Angione C. Integrating splice-isoform expression into genome-scale models characterizes breast cancer metabolism. Bioinformatics 2017; 34:494-501. [DOI: 10.1093/bioinformatics/btx562] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 09/06/2017] [Indexed: 12/20/2022] Open
Affiliation(s)
- Claudio Angione
- Department of Computer Science and Information Systems, Teesside University, Middlesbrough, UK
| |
Collapse
|
35
|
Alam SR, Wallrabe H, Svindrych Z, Chaudhary AK, Christopher KG, Chandra D, Periasamy A. Investigation of Mitochondrial Metabolic Response to Doxorubicin in Prostate Cancer Cells: An NADH, FAD and Tryptophan FLIM Assay. Sci Rep 2017; 7:10451. [PMID: 28874842 PMCID: PMC5585313 DOI: 10.1038/s41598-017-10856-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/15/2017] [Indexed: 01/20/2023] Open
Abstract
Prostate cancer (PCa) is one of the leading cancers in men in the USA. Lack of experimental tools that predict therapy response is one of the limitations of current therapeutic regimens. Mitochondrial dysfunctions including defective oxidative phosphorylation (OXPHOS) in cancer inhibit apoptosis by modulating ROS production and cellular signaling. Thus, correction of mitochondrial dysfunction and induction of apoptosis are promising strategies in cancer treatment. We have used Fluorescence Lifetime Imaging Microscopy (FLIM) to quantify mitochondrial metabolic response in PCa cells by tracking auto-fluorescent NAD(P)H, FAD and tryptophan (Trp) lifetimes and their enzyme-bound fractions as markers, before and after treatment with anti-cancer drug doxorubicin. A 3-channel FLIM assay and quantitative analysis of these markers for cellular metabolism show in response to doxorubicin, NAD(P)H mean fluorescence lifetime (τm) and enzyme-bound (a2%) fraction increased, FAD enzyme-bound (a1%) fraction was decreased, NAD(P)H-a2%/FAD-a1% FLIM-based redox ratio and ROS increased, followed by induction of apoptosis. For the first time, a FRET assay in PCa cells shows Trp-quenching due to Trp-NAD(P)H interactions, correlating energy transfer efficiencies (E%) vs NAD(P)H-a2%/FAD-a1% as sensitive parameters in predicting drug response. Applying this FLIM assay as early predictor of drug response would meet one of the important goals in cancer treatment.
Collapse
Affiliation(s)
- Shagufta Rehman Alam
- The W.M. Keck Center for Cellular Imaging, Physical and Life Sciences Building, University of Virginia, 90 Geldard Dr., Charlottesville, Virginia, 22904, USA
| | - Horst Wallrabe
- The W.M. Keck Center for Cellular Imaging, Physical and Life Sciences Building, University of Virginia, 90 Geldard Dr., Charlottesville, Virginia, 22904, USA
| | - Zdenek Svindrych
- The W.M. Keck Center for Cellular Imaging, Physical and Life Sciences Building, University of Virginia, 90 Geldard Dr., Charlottesville, Virginia, 22904, USA
| | - Ajay K Chaudhary
- Roswell Park Cancer Institute, Centre for Genetics and Pharmacology, Department of Pharmacology and Therapeutics, Elm & Carlton Streets, Buffalo, New York, 14263, USA
| | - Kathryn G Christopher
- Departments of Biology and Biomedical Engineering, University of Virginia, 90 Geldard Dr., Charlottesville, Virginia, 22904, USA
| | - Dhyan Chandra
- Roswell Park Cancer Institute, Centre for Genetics and Pharmacology, Department of Pharmacology and Therapeutics, Elm & Carlton Streets, Buffalo, New York, 14263, USA
| | - Ammasi Periasamy
- The W.M. Keck Center for Cellular Imaging, Physical and Life Sciences Building, University of Virginia, 90 Geldard Dr., Charlottesville, Virginia, 22904, USA. .,Departments of Biology and Biomedical Engineering, University of Virginia, 90 Geldard Dr., Charlottesville, Virginia, 22904, USA.
| |
Collapse
|
36
|
Ahn SS, Hwang SH, Jung SM, Lee SW, Park YB, Yun M, Song JJ. The clinical utility of splenic fluorodeoxyglucose uptake for diagnosis and prognosis in patients with macrophage activation syndrome. Medicine (Baltimore) 2017; 96:e7901. [PMID: 28834911 PMCID: PMC5572033 DOI: 10.1097/md.0000000000007901] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The aim of the study was to evaluate splenic glucose metabolism in macrophage activation syndrome (MAS), characterized by overwhelming systemic inflammation. Splenic F-fluorodeoxyglucose (FDG) uptake was compared in patients with MAS and sepsis using positron emission tomography/computed tomography (PET/CT).Clinical and FDG-PET/CT findings from patients with MAS and those with culture-proven sepsis were evaluated. The standardized uptake value (SUV) for the spleen and liver were measured. The maximum of the spleen to liver SUV ratio (SLRmax) was calculated as spleen SUVmax/liver SUVmean. The radiological splenic volume was also measured, and splenic metabolic volume (MV) was defined as the total splenic volume with an SLRmean > 1.14. The association between clinical features, laboratory variables, and SLRmax was analyzed.The median SLRmax and splenic MV were significantly higher in patients with MAS (n = 38) than they were in those with sepsis (n = 15) (SLRmax: 1.51 vs 1.09, P = .001; MV: 346.0 vs 154.0, P = .015). Multivariate analyses revealed that SLRmax > 1.31 was useful for discriminating between MAS and sepsis. SLRmax positively correlated with ferritin and lactate dehydrogenase level in MAS. Furthermore, MAS patients with high splenic FDG uptake (SLRmax > 1.72) had higher in-hospital mortality compared to those with moderate to low splenic FDG uptake (P = .013).This study was the first to demonstrate that splenic FDG uptake is significantly elevated in patients with MAS compared to those with sepsis. This may be useful to differentiate between MAS and sepsis, and to predict poor prognosis in patients with MAS.
Collapse
Affiliation(s)
- Sung Soo Ahn
- Division of Rheumatology, Department of Internal Medicine
| | | | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
37
|
Yoon JD, Hwang SU, Kim E, Jin M, Kim S, Hyun SH. GDF8 activates p38 MAPK signaling during porcine oocyte maturation in vitro. Theriogenology 2017; 101:123-134. [PMID: 28708509 DOI: 10.1016/j.theriogenology.2017.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/30/2017] [Accepted: 06/04/2017] [Indexed: 02/07/2023]
Abstract
Growth Differentiation Factor 8 (GDF8) is a member of the transforming growth factor-β (TGF-β) family and has been identified as a strong physiological regulator. This factor is expressed as a paracrine factor in mural granulosa cells. To investigate the effects of GDF8 on the in vitro maturation (IVM) of porcine oocytes, we assessed the quality of matured oocytes as well as the specific gene transcription and protein activation levels in oocytes and cumulus cells (CCs) after IVM and subsequent embryonic development after in vitro fertilization (IVF) and parthenogenetic activation (PA). Supplemental concentrations (0, 1, 10, and 100 ng/ml) of GDF8 were provided in IVM medium. Supplementation with GDF8 during IVM induced transcription of specific TGF-β receptor genes, such as ActRIIb and Alk4/5, and the recognition of the GDF8 by these receptors induced phosphorylation of p38 MAPK. Activated p38 MAPK signaling changed oocyte maturation and cumulus expansion-related gene transcription: Nrf2 and Bcl-2 in oocytes and PCNA, Nrf2, Has2, Ptx3, and TNFAIP6 in CCs. The altered gene expression pattern during IVM resulted in a 10% lower level of intracellular ROS in mature oocytes. The improved cytoplasmic maturation led to an increase in the fertilization efficiency and subsequent embryonic developmental competence. The embryonic development showed increases in the blastocyst formation rate and higher transcription levels of POU5F1 and BCL-2 in the blastocysts. The present study suggests that supplementation of GDF8 during IVM synergistically improved the developmental potential of IVF- and PA-derived porcine embryos by reducing the intracellular ROS level in oocytes by altering the transcription of specific genes and increasing the phosphorylation of p38 MAPK during IVM. In conclusion, for the first time, our results demonstrate that GDF8 can act as a paracrine factor to modulate oocyte maturation by regulating p38 MAPK phosphorylation and intracellular ROS level during porcine IVM.
Collapse
Affiliation(s)
- Junchul David Yoon
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea; Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea
| | - Seon-Ung Hwang
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea; Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea
| | - Eunhye Kim
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea; Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea
| | - Minghui Jin
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea; Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea
| | - Soochong Kim
- Laboratory of Veterinary Pathology and Platelets Signaling, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea; Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju 28644, Republic of Korea.
| |
Collapse
|
38
|
Targeted apoptosis in ovarian cancer cells through mitochondrial dysfunction in response to Sambucus nigra agglutinin. Cell Death Dis 2017; 8:e2762. [PMID: 28471452 PMCID: PMC5520748 DOI: 10.1038/cddis.2017.77] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 12/14/2022]
Abstract
Ovarian carcinoma (OC) patients encounter the severe challenge of clinical management owing to lack of screening measures, chemoresistance and finally dearth of non-toxic therapeutics. Cancer cells deploy various defense strategies to sustain the tumor microenvironment, among which deregulated apoptosis remains a versatile promoter of cancer progression. Although recent research has focused on identifying agents capable of inducing apoptosis in cancer cells, yet molecules efficiently breaching their survival advantage are yet to be classified. Here we identify lectin, Sambucus nigra agglutinin (SNA) to exhibit selectivity towards identifying OC by virtue of its specific recognition of α-2, 6-linked sialic acids. Superficial binding of SNA to the OC cells confirm the hyper-sialylated status of the disease. Further, SNA activates the signaling pathways of AKT and ERK1/2, which eventually promotes de-phosphorylation of dynamin-related protein-1 (Drp-1). Upon its translocation to the mitochondrial fission loci Drp-1 mediates the central role of switch in the mitochondrial phenotype to attain fragmented morphology. We confirmed mitochondrial outer membrane permeabilization resulting in ROS generation and cytochrome-c release into the cytosol. SNA response resulted in an allied shift of the bioenergetics profile from Warburg phenotype to elevated mitochondrial oxidative phosphorylation, altogether highlighting the involvement of mitochondrial dysfunction in restraining cancer progression. Inability to replenish the SNA-induced energy crunch of the proliferating cancer cells on the event of perturbed respiratory outcome resulted in cell cycle arrest before G2/M phase. Our findings position SNA at a crucial juncture where it proves to be a promising candidate for impeding progression of OC. Altogether we unveil the novel aspect of identifying natural molecules harboring the inherent capability of targeting mitochondrial structural dynamics, to hold the future for developing non-toxic therapeutics for treating OC.
Collapse
|
39
|
Roy D, Sheng GY, Herve S, Carvalho E, Mahanty A, Yuan S, Sun L. Interplay between cancer cell cycle and metabolism: Challenges, targets and therapeutic opportunities. Biomed Pharmacother 2017; 89:288-296. [PMID: 28235690 DOI: 10.1016/j.biopha.2017.01.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/02/2017] [Accepted: 01/02/2017] [Indexed: 12/31/2022] Open
Abstract
A growing interest has emerged in the field of studying the cross-talk between cancer cell cycle and metabolism. In this review, we aimed to present how metabolism and cell cycle are correlated and how cancer cells get energy to drive cell cycle. Cell proliferation and cell death largely depend on the metabolic activity of the cell. Cell cycle proteins, e.g. cyclin D, cyclin dependent kinase (CDK), some pro-apoptotic and anti-apoptotic proteins, and P53 have been shown to be regulated by metabolic crosstalk. Dysregulation of this cross-talk between metabolism and cell cycle leads to degenerative disorder(s) and cancer. It is not fully understood the actual reason of aberration between metabolism and cell cycle, but it is a hallmark of cancer research. Herein, we discussed the role of some regulatory molecules relative of cell cycle and metabolism and highlight how they control the function of each other. We also pointed out, current therapeutic opportunities and some additional crucial therapeutic targets on these fields that could be a breakthrough in cancer research.
Collapse
Affiliation(s)
- Debmalya Roy
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| | - Gao Ying Sheng
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| | - Semukunzi Herve
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Evandro Carvalho
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Arpan Mahanty
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China.
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China.
| | - Li Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
40
|
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: 143] [Impact Index Per Article: 15.9] [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
|
41
|
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: 1.6] [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]
|
42
|
Chaudhary AK, Bhat TA, Kumar S, Kumar A, Kumar R, Underwood W, Koochekpour S, Shourideh M, Yadav N, Dhar S, Chandra D. Mitochondrial dysfunction-mediated apoptosis resistance associates with defective heat shock protein response in African-American men with prostate cancer. Br J Cancer 2016; 114:1090-100. [PMID: 27115471 PMCID: PMC4865976 DOI: 10.1038/bjc.2016.88] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/10/2016] [Accepted: 03/10/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND African-American (AA) patients with prostate cancer (PCa) respond poorly to current therapy compared with Caucasian American (CA) PCa patients. Although underlying mechanisms are not defined, mitochondrial dysfunction is a key reason for this disparity. METHODS Cell death, cell cycle, and mitochondrial function/stress were analysed by flow cytometry or by Seahorse XF24 analyzer. Expression of cellular proteins was determined using immunoblotting and real-time PCR analyses. Cell survival/motility was evaluated by clonogenic, cell migration, and gelatin zymography assays. RESULTS Glycolytic pathway inhibitor dichloroacetate (DCA) inhibited cell proliferation in both AA PCa cells (AA cells) and CA PCa cells (CA cells). AA cells possess reduced endogenous reactive oxygen species, mitochondrial membrane potential (mtMP), and mitochondrial mass compared with CA cells. DCA upregulated mtMP in both cell types, whereas mitochondrial mass was significantly increased in CA cells. DCA enhanced taxol-induced cell death in CA cells while sensitising AA cells to doxorubicin. Reduced expression of heat shock proteins (HSPs) was observed in AA cells, whereas DCA induced expression of CHOP, C/EBP, HSP60, and HSP90 in CA cells. AA cells are more aggressive and metastatic than CA cells. CONCLUSIONS Restoration of mitochondrial function may provide new option for reducing PCa health disparity among American men.
Collapse
Affiliation(s)
- Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Tariq A Bhat
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Anil Kumar
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Willie Underwood
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Shahriar Koochekpour
- Department of Urology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.,Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Mojgan Shourideh
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Shanta Dhar
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|
43
|
Lactate dehydrogenase inhibition: exploring possible applications beyond cancer treatment. Future Med Chem 2016; 8:713-25. [PMID: 27054686 DOI: 10.4155/fmc.16.10] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Lactate dehydrogenase (LDH) inhibition is considered a worthwhile attempt in the development of innovative anticancer strategies. Unfortunately, in spite of the involvement of several research institutions and pharma-companies, the discovery of LDH inhibitors with drug-like properties seems a hardly resolvable challenge. While awaiting new advancements, in the present review we will examine other pathologic conditions characterized by increased glycolysis and LDH activity, which could potentially benefit from LDH inhibition. The rationale for targeting LDH activity in these contexts is the same justifying the LDH-based approach in anticancer therapy: because of the enzyme position at the end of glycolytic pathway, LDH inhibitors are not expected to hinder glucose metabolism of normal cells. Moreover, we will summarize the latest contributions in the discovery of enzyme inhibitors and try to glance over the reasons underlying the complexity of this research.
Collapse
|
44
|
Lactate dehydrogenase-A inhibition induces human glioblastoma multiforme stem cell differentiation and death. Sci Rep 2015; 5:15556. [PMID: 26494310 PMCID: PMC4616042 DOI: 10.1038/srep15556] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/29/2015] [Indexed: 12/11/2022] Open
Abstract
Therapies that target the signal transduction and metabolic pathways of cancer stem cells (CSCs) are innovative strategies to effectively reduce the recurrence and significantly improve the outcome of glioblastoma multiforme (GBM). CSCs exhibit an increased rate of glycolysis, thus rendering them intrinsically more sensitive to prospective therapeutic strategies based on the inhibition of the glycolytic pathway. The enzyme lactate dehydrogenase-A (LDH-A), which catalyses the interconversion of pyruvate and lactate, is up-regulated in human cancers, including GBM. Although several papers have explored the benefits of targeting cancer metabolism in GBM, the effects of direct LDH-A inhibition in glial tumours have not yet been investigated, particularly in the stem cell subpopulation. Here, two representative LDH-A inhibitors (NHI-1 and NHI-2) were studied in GBM-derived CSCs and compared to differentiated tumour cells. LDH-A inhibition was particularly effective in CSCs isolated from different GBM cell lines, where the two compounds blocked CSC formation and elicited long-lasting effects by triggering both apoptosis and cellular differentiation. These data demonstrate that GBM, particularly the stem cell subpopulation, is sensitive to glycolytic inhibition and shed light on the therapeutic potential of LDH-A inhibitors in this tumour type.
Collapse
|
45
|
Valvona CJ, Fillmore HL, Nunn PB, Pilkington GJ. The Regulation and Function of Lactate Dehydrogenase A: Therapeutic Potential in Brain Tumor. Brain Pathol 2015; 26:3-17. [PMID: 26269128 PMCID: PMC8029296 DOI: 10.1111/bpa.12299] [Citation(s) in RCA: 379] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/05/2015] [Indexed: 12/14/2022] Open
Abstract
There are over 120 types of brain tumor and approximately 45% of primary brain tumors are gliomas, of which glioblastoma multiforme (GBM) is the most common and aggressive with a median survival rate of 14 months. Despite progress in our knowledge, current therapies are unable to effectively combat primary brain tumors and patient survival remains poor. Tumor metabolism is important to consider in therapeutic approaches and is the focus of numerous research investigations. Lactate dehydrogenase A (LDHA) is a cytosolic enzyme, predominantly involved in anaerobic and aerobic glycolysis (the Warburg effect); however, it has multiple additional functions in non‐neoplastic and neoplastic tissues, which are not commonly known or discussed. This review summarizes what is currently known about the function of LDHA and identifies areas that would benefit from further exploration. The current knowledge of the role of LDHA in the brain and its potential as a therapeutic target for brain tumors will also be highlighted. The Warburg effect appears to be universal in tumors, including primary brain tumors, and LDHA (because of its involvement with this process) has been identified as a potential therapeutic target. Currently, there are, however, no suitable LDHA inhibitors available for tumor therapies in the clinic.
Collapse
Affiliation(s)
- Cara J Valvona
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| | - Helen L Fillmore
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| | - Peter B Nunn
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| | - Geoffrey J Pilkington
- Cellular & Molecular Neuro-oncology Research Group, University of Portsmouth, School of Pharmacy & Biomedical Sciences, Portsmouth, UK
| |
Collapse
|
46
|
Abstract
Metabolic processes are altered in cancer cells, which obtain advantages from this metabolic reprogramming in terms of energy production and synthesis of biomolecules that sustain their uncontrolled proliferation. Due to the conceptual progresses in the last decade, metabolic reprogramming was recently included as one of the new hallmarks of cancer. The advent of high-throughput technologies to amass an abundance of omic data, together with the development of new computational methods that allow the integration and analysis of omic data by using genome-scale reconstructions of human metabolism, have increased and accelerated the discovery and development of anticancer drugs and tumor-specific metabolic biomarkers. Here we review and discuss the latest advances in the context of metabolic reprogramming and the future in cancer research.
Collapse
|
47
|
Othayoth R, Mathi P, Bheemanapally K, Kakarla L, Botlagunta M. Characterization of vitamin–cisplatin-loaded chitosan nano-particles for chemoprevention and cancer fatigue. J Microencapsul 2015. [DOI: 10.3109/02652048.2015.1065921] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Shen YA, Li WH, Chen PH, He CL, Chang YH, Chuang CM. Intraperitoneal delivery of a novel liposome-encapsulated paclitaxel redirects metabolic reprogramming and effectively inhibits cancer stem cells in Taxol(®)-resistant ovarian cancer. Am J Transl Res 2015; 7:841-55. [PMID: 26175846 PMCID: PMC4494136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/14/2015] [Indexed: 06/04/2023]
Abstract
Taxol(®) remained as the mainstay therapeutic agent in the treatment of ovarian cancer, however recurrence rate is still high. Cancer stem cells (CSCs) represent a subset of cells in the bulk of tumors and play a central role in inducing drug resistance and recurrence. Furthermore, cancer metabolism has been an area under intensive investigation, since accumulating evidence has shown that CSCs and cancer metabolism are closely linked, an effect named as metabolic reprogramming. In this work, we aimed to investigate the impacts of a novel liposome-encapsulated paclitaxel (Nano-Taxol) on the stemness phenotype and metabolic reprogramming. A paclitaxel-resistant cell line (TR) was established at first. Tumor growth was induced in the mice peritoneal cavity by inoculation of TR cells. A 2x2 factorial experiment was designed to test the therapeutic efficacy in which factor 1 represented the comparison of drugs (Taxol(®) versus Nano-Taxol), while factor 2 represented the delivery route (intravenous versus intraperitoneal delivery). In this work, we found that intraperitoneal delivery of Nano-Taxol redirects metabolic reprogramming, from glycolysis to oxidative phosphorylation, and effectively suppresses cancer stem cells. Also, intraperitoneal delivery of Nano-Taxol led to a significantly better control of tumor growth compared with intravenous delivery of Taxol(®) (current standard treatment). This translational research may serve as a novel pathway for the drug development of nanomedicine. In the future, this treatment modality may be extended to treat several relevant cancers that have been proved to be suitable for the loco-regional delivery of therapeutic agents, including colon cancer, gastric cancer, and pancreatic cancer.
Collapse
Affiliation(s)
- Yao-An Shen
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Taipei Veterans General HospitalTaipei, Taiwan
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming UniversityTaipei, Taiwan
| | - Wai-Hou Li
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Taipei Veterans General HospitalTaipei, Taiwan
| | - Po-Hung Chen
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Taipei Veterans General HospitalTaipei, Taiwan
| | - Chun-Lin He
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Taipei Veterans General HospitalTaipei, Taiwan
| | - Yen-Hou Chang
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Taipei Veterans General HospitalTaipei, Taiwan
- Institute of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming UniversityTaipei, Taiwan
| | - Chi-Mu Chuang
- Section of Gynecologic Oncology, Department of Obstetrics and Gynecology, Taipei Veterans General HospitalTaipei, Taiwan
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming UniversityTaipei, Taiwan
| |
Collapse
|
49
|
Cojoc M, Mäbert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin Cancer Biol 2015; 31:16-27. [DOI: 10.1016/j.semcancer.2014.06.004] [Citation(s) in RCA: 268] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/04/2014] [Accepted: 06/11/2014] [Indexed: 12/11/2022]
|
50
|
Forrester DM. Self-assembled multi-ring formations of glutamine and a possible link to erythema gyratum repens. Med Hypotheses 2015; 85:10-6. [PMID: 25804239 DOI: 10.1016/j.mehy.2015.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/07/2015] [Indexed: 11/29/2022]
Abstract
In the body L-glutamine is abundant and required for the proliferation of cells. Indeed human physiology is dependent upon having and maintaining the correct glutamine levels for a range of functions including neurological signalling and a healthy immune system. However, during tumourigenesis cell proliferation demands elevated levels of glutamine, which can ultimately lead to muscle atrophy. In some cases the skin provides the first indications of the underlying disease and erupts in a wave of complicated pattern formations. One such skin marker is erythema gyratum repens. We investigated the pattern formations associated with concentrations of glutamine in aqueous solutions at levels higher than that of a normal biological functionality. We find remarkable similarities between the patterns of erythema gyratum repens and the unusual self-assembled patterns of glutamine. The findings may lead to new therapeutics and understanding for those working in oncology and toxicology. Utilising the formations associated with glutamine could also assist in bio-functionalising micro and nanoparticles for high efficacy.
Collapse
Affiliation(s)
- Derek Michael Forrester
- Department of Chemical Engineering, Loughborough University, Loughborough LE11 3TU, United Kingdom.
| |
Collapse
|