1
|
Yan D, Hou Y, Lei X, Xiao H, Zeng Z, Xiong W, Fan C. The Impact of Polyunsaturated Fatty Acids in Cancer and Therapeutic Strategies. Curr Nutr Rep 2025; 14:46. [PMID: 40085324 DOI: 10.1007/s13668-025-00639-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
PURPOSE OF REVIEW Cancer is a disease influenced by both genetic and environmental factors, with dietary lipids being a significant contributing factor. This review summarizes the role of polyunsaturated fatty acids (PUFAs) in the mechanism of tumor occurrence and development, and elucidate the role of PUFAs in tumor treatment. RECENT FINDINGS PUFAs exert their impact on cancer through altering lipid composition in cell membranes, interacting with cell membrane lipid receptors, directly modulating gene expression in the cell nucleus, and participating in the metabolism of lipid mediators. Most omega-3 PUFAs are believed to inhibit cell proliferation, promote cancer cell death, suppress cancer metastasis, alter energy metabolism, inhibit tumor microenvironment inflammation, and regulate immune responses involving macrophages, T cells, NK cells, and others. However, certain omega-6 PUFAs exhibit weaker anti-tumor effects and may even promote tumor development, such as by fostering inflammatory tumor microenvironment and enhancing tumor cell proliferation. PUFAs play important roles in hallmarks of cancer including tumor cell proliferation, cell death, migration and invasion, energy metabolism remodeling, epigenetics, and immunity. These findings provide insights into the mechanisms of cancer development and offers options for dietary management of cancer.
Collapse
Affiliation(s)
- Dong Yan
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Yingshan Hou
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Xinyi Lei
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Hao Xiao
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China
| | - Zhaoyang Zeng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
- Department of Histology and Embryology, School of Basic Medicine Sciences, Central South University, Changsha, 410013, Hunan Province, China.
| |
Collapse
|
2
|
Bahraini F, Sayadi M, Safarpour H, Zarban A, Mesbahzadeh B, Sajjadi SM. n-3 polyunsaturated fatty acids enhanced efficacy of cytarabine in iron-overloaded NALM-6 cells via apoptotic and oxidative pathways. Toxicol In Vitro 2025; 103:105976. [PMID: 39613235 DOI: 10.1016/j.tiv.2024.105976] [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: 03/02/2024] [Revised: 10/05/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Despite progress in treating acute lymphoblastic leukemia (ALL), the adverse effects of chemotherapy toxicity and iron overload from transfusions continue to affect patients' quality of life. Polyunsaturated fatty acids (PUFAs) exhibit both antitumor and anti-inflammatory properties in leukemia. This study investigated the influence of n-3 PUFA on the efficacy of cytarabine in cells with iron overload. Iron overload was induced in NALM-6 cells using ferric ammonium citrate (FAC) and quantified through atomic absorption spectroscopy (AAS). The impact of n-3 PUFA on NALM-6 cells' response to cytarabine was evaluated using MTT, lactate dehydrogenase (LDH), apoptosis, and cell cycle assays. Additionally, gene expression analyses were performed on apoptotic, anti-apoptotic, and inflammatory genes, along with oxidative stress markers such as reactive oxygen species (ROS) and malondialdehyde (MDA) levels. The administration of n-3 PUFA significantly enhanced the effectiveness of cytarabine in iron-overloaded NALM-6 cells, leading to increased LDH secretion, elevated apoptosis rates, and G1 phase cell cycle arrest. These effects were associated with the upregulation of apoptotic genes such as P53 and caspase-8, the downregulation of the anti-apoptotic gene Bcl2, and a decrease in the inflammatory gene TNF-α. Furthermore, there was a notable increase in ROS and MDA levels. Overall, n-3 PUFA treatment improved cytarabine's efficacy in iron-overloaded NALM-6 cells by activating apoptotic processes and oxidative stress pathways.
Collapse
Affiliation(s)
| | - Mahtab Sayadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Asghar Zarban
- Clinical Biochemistry Department, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran.
| | - Behzad Mesbahzadeh
- Department of Physiology, School of Allied Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran.
| | - Seyed Mehdi Sajjadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
3
|
Wang S, Bai H, Liu T, Yang J, Wang Z. Optimization of concentrations of different n-3PUFAs on antioxidant capacity in mouse hepatocytes. Lipids Health Dis 2024; 23:214. [PMID: 38982376 PMCID: PMC11232338 DOI: 10.1186/s12944-024-02202-0] [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: 03/20/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), mainly including α-linolenic acid (ALA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), possess antioxidant properties and play a crucial role in growth and development. However, the combined effects of ALA, EPA, and DHA at different concentrations have rarely been reported. This work explored the effects of EPA, ALA, and DHA on the viability and antioxidant capacity of mouse hepatocytes, with the objective of enhancing the antioxidant capacity. Within the appropriate concentration range, cell viability and the activity of glutathione S-transferase, superoxide dismutase, and catalase were increased, while the oxidation products of malondialdehyde and the level of intracellular reactive oxygen species were obviously reduced. Thus, oxidative stress was relieved, and cellular antioxidant levels were improved. Finally, response surface optimization was carried out for EPA, ALA, and DHA, and the model was established. The antioxidant capacity of the cells was highest at EPA, ALA, and DHA concentrations of 145.46, 405.05, and 551.52 µM, respectively. These findings lay the foundation for further exploration of the interactive mechanisms of n-3 PUFAs in the body, as well as their applications in nutraceutical food.
Collapse
Affiliation(s)
- Shuting Wang
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China
| | - Huasong Bai
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China
| | - Tong Liu
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China
| | - Jiayi Yang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Zhanzhong Wang
- Nourse Science Centre for Pet Nutrition, Wuhu, 241200, China.
| |
Collapse
|
4
|
Nair B, Adithya JK, Chandrababu G, Lakshmi PK, Koshy JJ, Manoj SV, Ambiliraj DB, Vinod BS, Sethi G, Nath LR. Modulation of carcinogenesis with selected GRAS nutraceuticals via Keap1-Nrf2 signaling pathway. Phytother Res 2023; 37:4398-4413. [PMID: 37468211 DOI: 10.1002/ptr.7940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/16/2023] [Accepted: 06/25/2023] [Indexed: 07/21/2023]
Abstract
Keap1-Nrf2 is a fundamental signaling cascade known to promote or prevent carcinogenesis. Extensive studies identify the key target of modulatory aspects of Keap1-Nrf2 signaling against cancer. Nutraceuticals are those dietary agents with many health benefits that have immense potential for cancer chemoprevention. The nutritional supplements known as nutraceuticals are found to be one of the most promising chemoprevention agents. Upon investigating the dual nature of Nrf2, it became clear that, in addition to shielding normal cells from numerous stresses, Nrf2 may also promote the growth of tumors. In the present review, we performed a systematic analysis of the role of 12 different nutraceuticals like curcumin, sulforaphane, resveratrol, polyunsaturated fatty acids (PUFA) from fish oil, lycopene, soybean, kaempferol, allicin, thymoquinone, quercetin, gingerol, and piperine in modulating the Nrf2/Keap1 signaling mechanism. Among these, 12 Generally Recognized As Safe (GRAS) certified nutraceuticals, sulforaphane is the most extensively studied compound in modulating Keap1-Nrf signaling. Even though there is much evidence at preclinical levels, further high-quality research is still required to validate the potential role of these nutraceuticals in Keap1-Nrf2 modulation.
Collapse
Affiliation(s)
- Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Jayaprakash K Adithya
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Gopika Chandrababu
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - P K Lakshmi
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Joel Joy Koshy
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | | | - D B Ambiliraj
- Department of Chemistry, Sree Narayana College, Chempazhanthy, India
| | | | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
5
|
Montecillo-Aguado M, Tirado-Rodriguez B, Huerta-Yepez S. The Involvement of Polyunsaturated Fatty Acids in Apoptosis Mechanisms and Their Implications in Cancer. Int J Mol Sci 2023; 24:11691. [PMID: 37511450 PMCID: PMC10380946 DOI: 10.3390/ijms241411691] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is a significant global public health issue and, despite advancements in detection and treatment, the prognosis remains poor. Cancer is a complex disease characterized by various hallmarks, including dysregulation in apoptotic cell death pathways. Apoptosis is a programmed cell death process that efficiently eliminates damaged cells. Several studies have indicated the involvement of polyunsaturated fatty acids (PUFAs) in apoptosis, including omega-3 PUFAs such as alpha-linolenic acid, docosahexaenoic acid, and eicosapentaenoic acid. However, the role of omega-6 PUFAs, such as linoleic acid, gamma-linolenic acid, and arachidonic acid, in apoptosis is controversial, with some studies supporting their activation of apoptosis and others suggesting inhibition. These PUFAs are essential fatty acids, and Western populations today have a high consumption rate of omega-6 to omega-3 PUFAs. This review focuses on presenting the diverse molecular mechanisms evidence in both in vitro and in vivo models, to help clarify the controversial involvement of omega-3 and omega-6 PUFAs in apoptosis mechanisms in cancer.
Collapse
Affiliation(s)
- Mayra Montecillo-Aguado
- Unidad de Investigacion en Enfermedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, Mexico City 06720, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de Mexico (UNAM), Mexico City 04510, Mexico
| | - Belen Tirado-Rodriguez
- Unidad de Investigacion en Enfermedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, Mexico City 06720, Mexico
| | - Sara Huerta-Yepez
- Unidad de Investigacion en Enfermedades Oncologicas, Hospital Infantil de Mexico, Federico Gomez, Mexico City 06720, Mexico
| |
Collapse
|
6
|
Lo Presti C, Yamaryo-Botté Y, Mondet J, Berthier S, Nutiu D, Botté C, Mossuz P. Variation in Lipid Species Profiles among Leukemic Cells Significantly Impacts Their Sensitivity to the Drug Targeting of Lipid Metabolism and the Prognosis of AML Patients. Int J Mol Sci 2023; 24:ijms24065988. [PMID: 36983080 PMCID: PMC10054724 DOI: 10.3390/ijms24065988] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Several studies have linked bad prognoses of acute myeloid leukemia (AML) to the ability of leukemic cells to reprogram their metabolism and, in particular, their lipid metabolism. In this context, we performed "in-depth" characterization of fatty acids (FAs) and lipid species in leukemic cell lines and in plasma from AML patients. We firstly showed that leukemic cell lines harbored significant differences in their lipid profiles at steady state, and that under nutrient stress, they developed common mechanisms of protection that led to variation in the same lipid species; this highlights that the remodeling of lipid species is a major and shared mechanism of adaptation to stress in leukemic cells. We also showed that sensitivity to etomoxir, which blocks fatty acid oxidation (FAO), was dependent on the initial lipid profile of cell lines, suggesting that only a particular "lipidic phenotype" is sensitive to the drug targeting of FAO. We then showed that the lipid profiles of plasma samples from AML patients were significantly correlated with the prognosis of patients. In particular, we highlighted the impact of phosphocholine and phosphatidyl-choline metabolism on patients' survival. In conclusion, our data show that balance between lipid species is a phenotypic marker of the diversity of leukemic cells that significantly influences their proliferation and resistance to stress, and thereby, the prognosis of AML patients.
Collapse
Affiliation(s)
- Caroline Lo Presti
- Team "Epigenetic and Cellular Signaling", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
- Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX 9, France
| | - Yoshiki Yamaryo-Botté
- Team "Apicolipid", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
| | - Julie Mondet
- Team "Epigenetic and Cellular Signaling", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
- Department of Molecular Pathology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX 9, France
| | - Sylvie Berthier
- Platform of Cytometry, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX 9, France
| | - Denisa Nutiu
- Team "Epigenetic and Cellular Signaling", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
| | - Cyrille Botté
- Team "Apicolipid", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
| | - Pascal Mossuz
- Team "Epigenetic and Cellular Signaling", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700 Grenoble, France
- Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043 Grenoble, CEDEX 9, France
| |
Collapse
|
7
|
Ramzy A, ElSafy S, Elshoky HA, Soliman A, Youness R, Mansour S, Sebak A. Drugless nanoparticles tune-up an array of intertwined pathways contributing to immune checkpoint signaling and metabolic reprogramming in triple-negative breast cancer. Biomed Mater 2022; 18. [PMID: 36541457 DOI: 10.1088/1748-605x/aca85d] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 12/02/2022] [Indexed: 12/03/2022]
Abstract
Metabolic reprogramming 'Warburg effect' and immune checkpoint signaling are immunosuppressive hallmarks of triple-negative breast cancer (TNBC) contributing to the limited clinical applicability of immunotherapy. Biomaterials arise as novel tools for immunomodulation of the tumor microenvironment that can be used alongside conventional immunotherapeutics. Chitosan and lecithin are examples of versatile biomaterials with interesting immunomodulatory properties. In this study, we aimed at investigation of the role of carefully designed hybrid nanoparticles (NPs) on common mediators of both programmed death ligand 1 (PD-L1) expression and glycolytic metabolism. Hybrid lecithin-chitosan NPs were prepared and characterized. Their intracellular concentration, localization and effect on the viability of MDA-MB-231 cells were assessed. Glycolytic metabolism was quantified by measuring glucose consumption, adenosine triphosphate (ATP) generation, lactate production and extracellular acidification. Nitric oxide production was quantified using Greiss reagent. Gene expression of inducible nitric oxide synthase (iNOS), phosphatidylinositol-3-kinase (PI3K), protein kinase B (PKB or Akt), mammalian target of rapamycin (mTOR), hypoxia-inducible factor 1α(HIF-1α) and PD-L1 was quantified by quantitative reverse transcription polymerase chain reaction (q-RT-PCR). Chitosan, lecithin and the NPs-formulated forms have been shown to influence the 'Warburg effect' and immune checkpoint signaling of TNBC cells differently. The composition of the hybrid systems dictated their subcellular localization and hence the positive or negative impact on the immunosuppressive characteristics of TNBC cells. Carefully engineered hybrid lecithin-chitosan NPs could convert the immune-suppressive microenvironment of TNBC to an immune-active microenvironment via reduction of PD-L1 expression and reversal of the Warburg effect.
Collapse
Affiliation(s)
- Asmaa Ramzy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt.,Department of Chemistry, School of Sciences & Engineering, The American University in Cairo, AUC Avenue, New Cairo 11835, Egypt
| | - Sara ElSafy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt
| | - Hisham A Elshoky
- Nanotechnology and Advanced Materials Central Lab. (NAMCL), Agricultural Research Center, Giza, Egypt.,Regional Center for Food and Feed, Agricultural Research Center, Giza, Egypt.,Department of Research, Tumor Biology Research Program, Basic Research Unit, Children's Cancer Hospital Egypt 57357, Cairo 11441, Egypt
| | - Aya Soliman
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt
| | - Rana Youness
- Department of Pharmaceutical Biology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt.,Department of Biology and Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo 11578, Egypt
| | - Samar Mansour
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt
| | - Aya Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt.,Immunopharmacology of Cancer, School of Pharmaceutical Sciences, University of Geneva, Geneva 1211, Switzerland
| |
Collapse
|
8
|
Yan CH, Liu HW, Tian XX, Li J, Ding Y, Li Y, Mei Z, Zou MH, Han YL. AMPKα2 controls the anti-atherosclerotic effects of fish oils by modulating the SUMOylation of GPR120. Nat Commun 2022; 13:7721. [PMID: 36513627 PMCID: PMC9747961 DOI: 10.1038/s41467-022-34996-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/15/2022] [Indexed: 12/15/2022] Open
Abstract
Consuming fish oils (FO) is linked to reduced risk of cardiovascular disease in certain populations. However, FO failed to exhibit therapeutic effects in some patients with cardiovascular disease. This study aimed to determine the possible reasons for the inconsistent effects of FO. AMP-activated protein kinase (AMPK) α2 is an important energy metabolic sensor, which was reported to involve in FO mediated regulation of lipid and glucose metabolism. In an in vivo study, FO administration significantly reduced the aortic lesions and inflammation in the Ldlr-/- mouse model of atherosclerosis, but not in Ldlr-/-/Prkaa2-/-and Ldlr-/-/Prkaa2-/-Sm22Cre mice. Mechanistically, inactivation of AMPKα2 increased the SUMOylation of the fatty acid receptor GPR120 to block FO-induced internalization and binding to β-arrestin. In contrast, activation of AMPKα2 can phosphorylate the C-MYC at Serine 67 to inhibit its trans-localization into the nuclei and transcription of SUMO-conjugating E2 enzyme UBC9 and SUMO2/3 in vascular smooth muscle cells (VSMCs), which result in GPR120 SUMOylation. In human arteries, AMPKα2 levels were inversely correlated with UBC9 expression. In a cohort of patients with atherosclerosis, FO concentrations did not correlate with atherosclerotic severity, however, in a subgroup analysis a negative correlation between FO concentrations and atherosclerotic severity was found in patients with higher AMPKα2 levels. These data indicate that AMPKα2 is required for the anti-inflammatory and anti-atherosclerotic effects of FO.
Collapse
Affiliation(s)
- Cheng-hui Yan
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| | - Hai-Wei Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| | - Xiao-xiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| | - Jiayin Li
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| | - Ye Ding
- grid.256304.60000 0004 1936 7400Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303 USA
| | - Yi Li
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| | - Zhu Mei
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| | - Ming-Hui Zou
- grid.256304.60000 0004 1936 7400Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303 USA
| | - Ya-ling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, 110016 China
| |
Collapse
|
9
|
Khodakarami A, Adibfar S, Karpisheh V, Abolhasani S, Jalali P, Mohammadi H, Gholizadeh Navashenaq J, Hojjat-Farsangi M, Jadidi-Niaragh F. The molecular biology and therapeutic potential of Nrf2 in leukemia. Cancer Cell Int 2022; 22:241. [PMID: 35906617 PMCID: PMC9336077 DOI: 10.1186/s12935-022-02660-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 07/19/2022] [Indexed: 02/07/2023] Open
Abstract
NF-E2-related factor 2 (Nrf2) transcription factor has contradictory roles in cancer, which can act as a tumor suppressor or a proto-oncogene in different cell conditions (depending on the cell type and the conditions of the cell environment). Nrf2 pathway regulates several cellular processes, including signaling, energy metabolism, autophagy, inflammation, redox homeostasis, and antioxidant regulation. As a result, it plays a crucial role in cell survival. Conversely, Nrf2 protects cancerous cells from apoptosis and increases proliferation, angiogenesis, and metastasis. It promotes resistance to chemotherapy and radiotherapy in various solid tumors and hematological malignancies, so we want to elucidate the role of Nrf2 in cancer and the positive point of its targeting. Also, in the past few years, many studies have shown that Nrf2 protects cancer cells, especially leukemic cells, from the effects of chemotherapeutic drugs. The present paper summarizes these studies to scrutinize whether targeting Nrf2 combined with chemotherapy would be a therapeutic approach for leukemia treatment. Also, we discussed how Nrf2 and NF-κB work together to control the cellular redox pathway. The role of these two factors in inflammation (antagonistic) and leukemia (synergistic) is also summarized.
Collapse
Affiliation(s)
- Atefeh Khodakarami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Adibfar
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Abolhasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.,Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Gyan E, Pigneux A, Hunault M, Peterlin P, Carré M, Bay JO, Bonmati C, Gallego-Hernanz MP, Lioure B, Bertrand P, Vallet N, Ternant D, Darrouzain F, Picou F, Béné MC, Récher C, Hérault O. Adjunction of a fish oil emulsion to cytarabine and daunorubicin induction chemotherapy in high-risk AML. Sci Rep 2022; 12:9748. [PMID: 35697729 PMCID: PMC9192636 DOI: 10.1038/s41598-022-13626-y] [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: 09/28/2021] [Accepted: 05/26/2022] [Indexed: 12/04/2022] Open
Abstract
The treatment of acute myeloid leukemia (AML) with unfavorable cytogenetics treatment remains a challenge. We previously established that ex vivo exposure of AML blasts to eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), or fish oil emulsion (FO) induces Nrf2 pathway activation, metabolic switch, and cell death. The FILO group launched a pilot clinical study to evaluate the feasibility, safety, and efficacy of the adjunction of a commercial FO emulsion to 3 + 7 in untreated AML with unfavorable cytogenetics. The primary objective was complete response (CR). Thirty patients were included. FO administration raised the plasma levels of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acids (p < 0.001). The pharmacokinetics of cytarabine and daunorubicin were unaffected. A historical comparison to the LAM2001 trial (Lioure et al. Blood 2012) found a higher frequency of grade 3 serious adverse events, with no drug-related unexpected toxicity. The CR rate was 77%, and the partial response (PR) 10%, not significantly superior to that of the previous study (CR 72%, PR 1%). RT-qPCR analysis of Nrf2 target genes and antioxidant enzymes did not show a significant in vivo response. Overall, FO emulsion adjunction to 3 + 7 is feasible. An improvement in CR was not shown in this cohort of high-risk patients. The present data does not support the use of FO in adjunction with 3 + 7 in high-risk AML patients. ClinicalTrials.gov identifier: NCT01999413.
Collapse
Affiliation(s)
- Emmanuel Gyan
- Service d'Hématologie et Thérapie Cellulaire, Centre Hospitalier Universitaire de Tours, Hôpital Bretonneau, Bâtiment Kaplan, 2, boulevard Tonnellé, 37044, Tours Cedex 09, France. .,ERL CNRS 7001, Leukemic Niche and Redox Metabolism (LNOx), Faculté de Médecine, Université de Tours, Tours, France. .,Centre d'Investigation Clinique, INSERM U1415, Centre Hospitalier Universitaire, Tours, France.
| | - Arnaud Pigneux
- Clinique d'Hématologie, Université de Bordeaux, Hôpital Haut-Levêque, Pessac, France
| | - Mathilde Hunault
- Service des Maladies du Sang, FHU GOAL, CRCINA, INSERM Angers, Centre Hospitalier Universitaire, Tours, France
| | - Pierre Peterlin
- Service d'Hématologie, Centre Hospitalier Universitaire, Nantes, France
| | - Martin Carré
- Service d'Hématologie, Centre Hospitalier Universitaire, Grenoble, France
| | - Jacques-Olivier Bay
- Service d'Hématologie, Centre Hospitalier Universitaire, Clermont-Ferrand, France
| | - Caroline Bonmati
- Service d'Hématologie, Centre Hospitalier Universitaire, Nancy, France
| | | | - Bruno Lioure
- Service d'Hématologie, Centre Hospitalier Universitaire, Strasbourg, France
| | - Philippe Bertrand
- Laboratoire de Biostatistiques, Faculté de Médecine, Université de Tours, Tours, France
| | - Nicolas Vallet
- Service d'Hématologie et Thérapie Cellulaire, Centre Hospitalier Universitaire de Tours, Hôpital Bretonneau, Bâtiment Kaplan, 2, boulevard Tonnellé, 37044, Tours Cedex 09, France.,ERL CNRS 7001, Leukemic Niche and Redox Metabolism (LNOx), Faculté de Médecine, Université de Tours, Tours, France
| | - David Ternant
- Laboratoire de Pharmacologie-Toxicologie, Centre Hospitalier Universitaire, Tours, France
| | | | - Frédéric Picou
- ERL CNRS 7001, Leukemic Niche and Redox Metabolism (LNOx), Faculté de Médecine, Université de Tours, Tours, France
| | | | - Christian Récher
- Service d'Hématologie, Institut Universitaire de Cancérologie de Toulouse, Toulouse, France
| | - Olivier Hérault
- ERL CNRS 7001, Leukemic Niche and Redox Metabolism (LNOx), Faculté de Médecine, Université de Tours, Tours, France. .,Service d'Hématologie Biologique, FHU GOAL, Centre Hospitalier Universitaire de Tours, Hôpital Bretonneau, Bâtiment B2A, 2, boulevard Tonnellé, 37044, Tours Cedex 09, France.
| |
Collapse
|
11
|
Guéguinou M, Ibrahim S, Bourgeais J, Robert A, Pathak T, Zhang X, Crottès D, Dupuy J, Ternant D, Monbet V, Guibon R, Flores-Romero H, Lefèvre A, Lerondel S, Le Pape A, Dumas JF, Frank PG, Girault A, Chautard R, Guéraud F, García-Sáez AJ, Ouaissi M, Emond P, Sire O, Hérault O, Fromont-Hankard G, Vandier C, Tougeron D, Trebak M, Raoul W, Lecomte T. Curcumin and NCLX inhibitors share anti-tumoral mechanisms in microsatellite-instability-driven colorectal cancer. Cell Mol Life Sci 2022; 79:284. [PMID: 35526196 PMCID: PMC11072810 DOI: 10.1007/s00018-022-04311-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/05/2022] [Accepted: 04/15/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS Recent evidences highlight a role of the mitochondria calcium homeostasis in the development of colorectal cancer (CRC). To overcome treatment resistance, we aimed to evaluate the role of the mitochondrial sodium-calcium-lithium exchanger (NCLX) and its targeting in CRC. We also identified curcumin as a new inhibitor of NCLX. METHODS We examined whether curcumin and pharmacological compounds induced the inhibition of NCLX-mediated mitochondrial calcium (mtCa2+) extrusion, the role of redox metabolism in this process. We evaluated their anti-tumorigenic activity in vitro and in a xenograft mouse model. We analyzed NCLX expression and associations with survival in The Cancer Genome Atlas (TCGA) dataset and in tissue microarrays from 381 patients with microsatellite instability (MSI)-driven CRC. RESULTS In vitro, curcumin exerted strong anti-tumoral activity through its action on NCLX with mtCa2+ and reactive oxygen species overload associated with a mitochondrial membrane depolarization, leading to reduced ATP production and apoptosis. NCLX inhibition with pharmacological and molecular approaches reproduced the effects of curcumin. NCLX inhibitors decreased CRC tumor growth in vivo. Both transcriptomic analysis of TCGA dataset and immunohistochemical analysis of tissue microarrays demonstrated that higher NCLX expression was associated with MSI status, and for the first time, NCLX expression was significantly associated with recurrence-free survival. CONCLUSIONS Our findings highlight a novel anti-tumoral mechanism of curcumin through its action on NCLX and mitochondria calcium overload that could benefit for therapeutic schedule of patients with MSI CRC.
Collapse
Affiliation(s)
- Maxime Guéguinou
- EA 7501 GICC, Université de Tours, Tours, France.
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France.
| | | | | | - Alison Robert
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Trayambak Pathak
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, 500 University Dr, Hershey, PA, 17033, USA
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, 500 University Dr, Hershey, PA, 17033, USA
| | - David Crottès
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Jacques Dupuy
- TOXALIM (Research Centre in Food Toxicology)-Team E9-PPCA, Université de Toulouse, UMR 1331 INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - David Ternant
- EA 7501 GICC, Université de Tours, Tours, France
- EA4245 Transplant Immunology and Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Valérie Monbet
- IRMAR Mathematics Research Institute of Rennes, UMR-CNRS 6625, Rennes, France
| | - Roseline Guibon
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Hector Flores-Romero
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Antoine Lefèvre
- UMR 1253, iBrain, Université de Tours, Inserm, 37000, Tours, France
| | | | | | - Jean-François Dumas
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Philippe G Frank
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Alban Girault
- Laboratory of Cellular and Molecular Physiology, UR UPJV 4667, University of Picardie Jules Verne, Amiens, France
| | | | - Françoise Guéraud
- TOXALIM (Research Centre in Food Toxicology)-Team E9-PPCA, Université de Toulouse, UMR 1331 INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Ana J García-Sáez
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Interfaculty Institute of Biochemistry, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
| | - Mehdi Ouaissi
- EA4245 Transplant Immunology and Inflammation, Université de Tours, 10 Boulevard Tonnellé, 37032, Tours, France
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, Inserm, 37000, Tours, France
| | - Olivier Sire
- IRDL Institut de Recherche Dupuy de Lôme, UMR-CNRS, 06027, Vannes, France
| | | | - Gaëlle Fromont-Hankard
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Christophe Vandier
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - David Tougeron
- Hepato-Gastroenterology Department, Poitiers University Hospital and Faculty of Medicine of Poitiers, 86000, Poitiers, France
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, College of Medicine, The Pennsylvania State University, 500 University Dr, Hershey, PA, 17033, USA
| | - William Raoul
- EA 7501 GICC, Université de Tours, Tours, France
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France
| | - Thierry Lecomte
- EA 7501 GICC, Université de Tours, Tours, France.
- N2C, Nutrition Growth and Cancer, Faculté de Médecine, Université de Tours, Inserm, UMR 1069, Tours, France.
- Department of Hepato-Gastroenterology and Digestive Oncology, CHRU de Tours, Tours, France.
| |
Collapse
|
12
|
Wei L, Wu Z, Chen YQ. Multi-targeted therapy of cancer by omega-3 fatty acids-an update. Cancer Lett 2022; 526:193-204. [PMID: 34843864 DOI: 10.1016/j.canlet.2021.11.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022]
Abstract
Low in dietary ω3 polyunsaturated fatty acid (PUFA) consumption has been associated with increased incidence of cancers. Many basic and clinical studies have been conducted over the last several decades. We previously reviewed multi-targeted therapy of cancer by omega-3 fatty acids in 2008, and since hundreds of new clinical trials are being conducted to validate the effectiveness of ω3 PUFA in cancer therapy. Because of the availability of such large amount of clinical trial data, in this update we summarize clinical data, sort out trials that show promising results, and discuss potential mechanism(s) responsible for the clinical outcomes. It appears that ω3 PUFA mainly affects cancer-associated symptoms, namely cachexia, inflammation, neuropathy, post operative complications and quality of life. Mechanisms responsible for these effects are possible regulation of skeletal muscle protein turnover, inflammatory response and neuron cell survive by ω3 PUFA.
Collapse
Affiliation(s)
- Lengyun Wei
- Wuxi School of Medicine, Jiangnan University, Jiangsu Province, 214122, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China; School of Food Science and Technology, Jiangnan University, Jiangsu Province, 214122, China
| | - Zhipeng Wu
- Wuxi School of Medicine, Jiangnan University, Jiangsu Province, 214122, China
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, Jiangsu Province, 214122, China; Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China; School of Food Science and Technology, Jiangnan University, Jiangsu Province, 214122, China.
| |
Collapse
|
13
|
Baroud M, Lepeltier E, El-Makhour Y, Lautram N, Bejaud J, Thepot S, Duval O. Azacitidine Omega-3 Self-Assemblies: Synthesis, Characterization, and Potent Applications for Myelodysplastic Syndromes. Pharmaceuticals (Basel) 2021; 14:1317. [PMID: 34959720 PMCID: PMC8706301 DOI: 10.3390/ph14121317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 11/30/2022] Open
Abstract
5-Azacitidine, a cytidine analogue used as a hypomethylating agent, is one of the main drugs for the treatment of myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML) in the elderly. However, after administration, it exhibits several limitations, including restricted diffusion and cellular internalization due to its hydrophilicity, and a rapid enzymatic degradation by adenosine deaminase. The aim of this study was to improve the drug cell diffusion and protect it from metabolic degradation via the synthesis of amphiphilic prodrugs and their potential self-assembly. Azacitidine was conjugated to two different omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). The carboxylic acid group of the omega-3 fatty acids was effectively conjugated to the amine group of the azacitidine base, yielding two amphiphilic prodrugs. Nanoprecipitation of the obtained prodrugs was performed and self-assemblies were successfully obtained for both prodrugs, with a mean diameter of 190 nm, a polydispersity index below 0.2 and a positive zeta potential. The formation of self-assemblies was confirmed using pyrene as a fluorescent dye, and the critical aggregation concentrations were determined: 400 µM for AzaEPA and 688 µM for AzaDHA. Additionally, the stability of the obtained self-assemblies was studied and after 5 days their final stable arrangement was reached. Additionally, cryo-TEM revealed that the self-assemblies attain a multilamellar vesicle supramolecular structure. Moreover, the obtained self-assemblies presented promising cytotoxicity on a leukemia human cell line, having a low IC50 value, comparable to that of free azacitidine.
Collapse
Affiliation(s)
- Milad Baroud
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Elise Lepeltier
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Yolla El-Makhour
- Environmental Health Research Lab, Faculty of Science, Lebanese University, Nabatieh 1700, Lebanon;
| | - Nolwenn Lautram
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Jerome Bejaud
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Sylvain Thepot
- Department of Hematology, University Hospital of Angers, 49933 Angers, France;
- Federation Hospital of Universitaire Grand Ouest Acute Leukemia (FHU GOAL), 49933 Angers, France
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM, University of Angers, 49933 Angers, France
| | - Olivier Duval
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
- Department of Hematology, University Hospital of Angers, 49933 Angers, France;
| |
Collapse
|
14
|
Xiao B, Li Y, Lin Y, Lin J, Zhang L, Wu D, Zeng J, Li J, Liu JW, Li G. Eicosapentaenoic acid (EPA) exhibits antioxidant activity via mitochondrial modulation. Food Chem 2021; 373:131389. [PMID: 34710690 DOI: 10.1016/j.foodchem.2021.131389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/25/2021] [Accepted: 10/09/2021] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) are mitochondrial respiration byproducts, the accumulation of which may cause oxidative damage and is associated with several chronic health problems. As an essential unsaturated fatty acid, eicosapentaenoic acid (EPA) provides various physiological functions; however, its exact regulatory role remains elusive. The current study aimed to address how EPA regulates cellular antioxidant capacity and the possible mechanisms of action. Upon 48 h of EPA treatment, the ROS levels of HepG2 cells were reduced by at least 40%; the total cellular antioxidant capacity was increased by approximately 50-70%, accompanied by enhanced activities and expression of major antioxidant enzymes. Furthermore, the mitochondrial membrane potential and the mitochondrial biogenesis were dramatically improved in EPA-treated cells. These data suggest that EPA improves cellular antioxidant capacity by enhancing mitochondrial function and biogenesis, which sheds light on EPA as a dietary complement to relieve the oxidative damage caused by chronic diseases.
Collapse
Affiliation(s)
- Baoping Xiao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Yuanyuan Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Yanqi Lin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Jingyu Lin
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Daren Wu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Jun Zeng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China
| | - Jing Wen Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China.
| | - Guiling Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China; Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen, Fujian 361021, PR China.
| |
Collapse
|
15
|
Wu S, Chen Y, Wang X, Weng S, Zhou W, Liu Z. Effect of EPA on Hsp90 and GRα protein expression in multiple myeloma drug-resistant cells. BMC Cancer 2021; 21:1076. [PMID: 34600510 PMCID: PMC8487534 DOI: 10.1186/s12885-021-08804-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 09/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Approximately 20% of MM patients harbor glucocorticoid (GC) resistance and are not responsive to therapeutic effect. Chaperoneheat-shock proteins Hsp90 is needed for ligand docking, The imbalance of Hsp90/GRα (glucocorticoid receptor α) may be an important cause of GC resistance. Recent studies have indicated that EPA could repress cancer cell growth by regulating critical influential factors in progression of cancer, consisting of resistance to drugs, chemosensitivity. The aim of the present study was to test the cytotoxic effects of EPA alone or EPA + Dexamethasone in dexamethasone-resistant MM cell (MM.1R) and investigate whether DHA can induce apoptosis and reverse acquired glucocorticoid resistance in dexamethasone-resistant MM cell (MM.1R). METHODS Cell Counting Kit-8 (CCK-8) was used to detect the proliferation of MM.1R cells after treating with EPA alone and EPA combined with DEX. Mitochondrial membrane potential was measured by flow cytometry and GRα and Hsp90 protein expression were assessed by western blot analysis. RESULTS EPA alone was able to inhibit cell proliferation as evidenced by CCK-8 assay and the tumor growth was remarkably suppressed by EPA + Dexamethasone, Cell apoptosis after EPA treatment was obviously observed by Flow cytometry analysis of the mitochondrial membrane potential. Analysis of Hsp90 and GRα proteins in MM.1R cells incubated with EPA revealed down-regulation of Hsp90 and up-regulation of GRα. Accordingly, the Hsp90/GRα ratio was significantly decreased with the increase of EPA concentration. CONCLUSIONS EPA might be used as a new effective treatment for reversal of glucocorticoid-resistance in multiple myeloma.
Collapse
Affiliation(s)
- Shenghao Wu
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China.
| | - Yuemiao Chen
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Xueshuang Wang
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Shanshan Weng
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Wenjin Zhou
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| | - Zhen Liu
- Department of Hematology, The Second Affiliated Hospital of Shanghai University (The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou Central Hospital), No.252 East Baili Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, China
| |
Collapse
|
16
|
Bourgeais J, Hérault O. In Vitro Analysis of Energy Metabolism in Bone-Marrow Mesenchymal Stromal Cells. Methods Mol Biol 2021; 2308:59-70. [PMID: 34057714 DOI: 10.1007/978-1-0716-1425-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Bone marrow mesenchymal stromal cells (MSCs) play an essential role in the regulation of normal and leukemic hematopoiesis. Their multipotent potential of differentiation also makes them an interesting therapeutic tool. Among factors involved in the regulation of MSCs, energy metabolism plays a key role in their proliferation and differentiation. Seahorse Bioscience introduced extracellular flux technology to the life sciences market in 2006. This methodology allows, in living cells and in real time, the concomitant determination of basal oxygen consumption, glycolysis rates, ATP production, and respiratory capacity in a single experiment. Here we describe the protocol used to study concomitantly the respiratory and glycolytic metabolism of primary MSCs from the determination of oxygen consumption (OCR) and extracellular acidification (ECAR) rates.
Collapse
Affiliation(s)
- Jérôme Bourgeais
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, Faculty of Medicine, Tours University, Tours, France.,Department of Biolocical Hematology, Tours University Hospital, Tours, France
| | - Olivier Hérault
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France. .,EA7501 GICC, Faculty of Medicine, Tours University, Tours, France. .,Department of Biolocical Hematology, Tours University Hospital, Tours, France.
| |
Collapse
|
17
|
Varela-López A, Vera-Ramírez L, Giampieri F, Navarro-Hortal MD, Forbes-Hernández TY, Battino M, Quiles JL. The central role of mitochondria in the relationship between dietary lipids and cancer progression. Semin Cancer Biol 2021; 73:86-100. [DOI: 10.1016/j.semcancer.2021.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/31/2020] [Accepted: 01/01/2021] [Indexed: 12/20/2022]
|
18
|
Li G, Li Y, Xiao B, Cui D, Lin Y, Zeng J, Li J, Cao MJ, Liu J. Antioxidant Activity of Docosahexaenoic Acid (DHA) and Its Regulatory Roles in Mitochondria. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:1647-1655. [PMID: 33497204 DOI: 10.1021/acs.jafc.0c07751] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Reactive oxygen species (ROS) are single-electron-bearing oxidation-reduction products that are mainly produced in mitochondria. Excessive ROS accumulation may lead to oxidative damage. Docosahexaenoic acid (DHA) is an essential component of brain phospholipids and is mainly derived from the diet. Its antioxidant activities have been extensively studied. However, its regulatory roles in mitochondria and the underlying mechanism remain to be elucidated. In this study, the DHA's effect on cellular antioxidant capacity and mitochondrial functions was examined in HepG2 cells. The results showed that 100 μM DHA decreased cellular and mitochondrial ROS levels to 75.2 ± 9.4% (P < 0.05) and 55.1 ± 1.4% (P < 0.01), respectively. It also increased the total antioxidant capacity by 55.6 ± 0.1 and 49.2 ± 1.1% (P < 0.05), based on ABTS and FRAP assay results, respectively. Consistently, it increased the activities and gene expression of major antioxidant enzymes by at least 35 and 40% (P < 0.05), respectively. Furthermore, DHA promoted mitochondrial functions and biogenesis. These data suggested that DHA's antioxidant activity can be attributed to its enhancement of mitochondrial functions and biogenesis. This study may shed light on the molecular mechanisms underlying DHA's function in improving resistance to and relieving the symptoms of chronic disease.
Collapse
Affiliation(s)
- Guiling Li
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Yuanyuan Li
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Baoping Xiao
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Dongyue Cui
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Yanqi Lin
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Jun Zeng
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Jian Li
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Min-Jie Cao
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
- Fujian Marine Functional Food Engineering Technology Research Center, Xiamen, Fujian 361021, P. R. China
| | - Jingwen Liu
- College of Food and Biological Engineering, Jimei University, No. 43 Yindou Road, Jimei District, Xiamen, Fujian 361021, P. R. China
| |
Collapse
|
19
|
Kang GJ, Kim EJ, Lee CH. Therapeutic Effects of Specialized Pro-Resolving Lipids Mediators on Cardiac Fibrosis via NRF2 Activation. Antioxidants (Basel) 2020; 9:antiox9121259. [PMID: 33321955 PMCID: PMC7764646 DOI: 10.3390/antiox9121259] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Heart disease is the number one mortality disease in the world. In particular, cardiac fibrosis is considered as a major factor causing myocardial infarction and heart failure. In particular, oxidative stress is a major cause of heart fibrosis. In order to control such oxidative stress, the importance of nuclear factor erythropoietin 2 related factor 2 (NRF2) has recently been highlighted. In this review, we will discuss the activation of NRF2 by docosahexanoic acid (DHA), eicosapentaenoic acid (EPA), and the specialized pro-resolving lipid mediators (SPMs) derived from polyunsaturated lipids, including DHA and EPA. Additionally, we will discuss their effects on cardiac fibrosis via NRF2 activation.
Collapse
Affiliation(s)
- Gyeoung Jin Kang
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (G.J.K.); (E.J.K.)
| | - Eun Ji Kim
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (G.J.K.); (E.J.K.)
- College of Pharmacy, Dongguk University, Seoul 04620, Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Korea
- Correspondence: ; Tel.: +82-31-961-5213
| |
Collapse
|
20
|
Cortés Fuentes IA, Burotto M, Retamal MA, Frelinghuysen M, Caglevic C, Gormaz JG. Potential use of n-3 PUFAs to prevent oxidative stress-derived ototoxicity caused by platinum-based chemotherapy. Free Radic Biol Med 2020; 160:263-276. [PMID: 32827639 DOI: 10.1016/j.freeradbiomed.2020.07.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Platinum-based compounds are widely used for the treatment of different malignancies due to their high effectiveness. Unfortunately, platinum-based treatment may lead to ototoxicity, an often-irreversible side effect without a known effective treatment and prevention plan. Platinum-based compound-related ototoxicity results mainly from the production of toxic levels of reactive oxygen species (ROS) rather than DNA-adduct formation, which has led to test strategies based on direct ROS scavengers to ameliorate hearing loss. However, favorable clinical results have been associated with several complications, including potential interactions with chemotherapy efficacy. To understand the contribution of the different cytotoxic mechanisms of platinum analogues on malignant cells and auditory cells, the particular susceptibility and response of both kinds of cells to molecules that potentially interfere with these mechanisms, is fundamental to develop innovative strategies to prevent ototoxicity without affecting antineoplastic effects. The n-3 long-chain polyunsaturated fatty acids (n-3 PUFAs) have been tried in different clinical settings, including with cancer patients. Nevertheless, their use to decrease cisplatin-induced ototoxicity has not been explored to date. In this hypothesis paper, we address the mechanisms of platinum compounds-derived ototoxicity, focusing on the differences between the effects of these compounds in neoplastic versus auditory cells. We discuss the basis for a strategic use of n-3 PUFAs to potentially protect auditory cells from platinum-derived injury without affecting neoplastic cells and chemotherapy efficacy.
Collapse
Affiliation(s)
- Ignacio A Cortés Fuentes
- Otorhinolaryngology Service, Hospital Barros Luco-Trudeau, San Miguel, Santiago, Chile; Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Mauricio Burotto
- Oncology Department, Clínica Universidad de Los Andes, Santiago, Chile; Bradford Hill, Clinical Research Center, Santiago, Chile
| | - Mauricio A Retamal
- Universidad Del Desarrollo, Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana, Santiago, Chile.
| | | | - Christian Caglevic
- Cancer Research Department, Fundación Arturo López Pérez, Santiago, Chile
| | - Juan G Gormaz
- Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
21
|
Chen Y, Qiu X, Yang J. Comparing the In Vitro Antitumor, Antioxidant and Anti-Inflammatory Activities between Two New Very Long Chain Polyunsaturated Fatty Acids, Docosadienoic Acid (DDA) and Docosatrienoic Acid (DTA), and Docosahexaenoic Acid (DHA). Nutr Cancer 2020; 73:1697-1707. [PMID: 32781843 DOI: 10.1080/01635581.2020.1804949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Very long chain polyunsaturated fatty acids (VLCPUFAs) are widely used as nutraceutical supplements for human health. Recently, a high level of two new VLCPUFAs, docosadienoic acid (DDA, 22:2n-6) and docosatrienoic acid (DTA, 22:3n-3), was produced in oilseed crop Brassica carinata using a biotechnology approach. This study investigated the functional properties of these two VLCPUFAs in human cells. Compared to docosahexaenoic acid (DHA), the golden standard in evaluating the health-promoting activities of VLCPUFAs, both DDA and DTA exhibited comparable or even better antitumor and antioxidant effects against human breast cancer SK-BR-3 and MDA-MB-231 cells. Especially, DTA elicited much stronger antioxidant and pro-apoptotic effects than DHA. Furthermore, DDA and DTA showed strong anti-inflammatory effects in human macrophages differentiated from monocyte THP-1 cells through lowering the protein expression levels of pro-inflammatory cytokines interleukin-1β (IL-1β), interleukin-6 (IL-6), interferon γ (IFN-γ), monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor α (TNF-α). Future In Vivo and In Vivo studies are warranted to identify the mechanism of action (MOA) for the antitumor, antioxidant and anti-inflammatory functions of DDA and DTA and explore potential applications of these two VLCPUFAs as novel nutraceutical supplements in preventing inflammatory conditions, aging and even cancer.
Collapse
Affiliation(s)
- Yi Chen
- Department of Food and Bioproduct Sciences, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Xiao Qiu
- Department of Food and Bioproduct Sciences, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Jian Yang
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
22
|
Bojková B, Winklewski PJ, Wszedybyl-Winklewska M. Dietary Fat and Cancer-Which Is Good, Which Is Bad, and the Body of Evidence. Int J Mol Sci 2020; 21:ijms21114114. [PMID: 32526973 PMCID: PMC7312362 DOI: 10.3390/ijms21114114] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
A high-fat diet (HFD) induces changes in gut microbiota leading to activation of pro-inflammatory pathways, and obesity, as a consequence of overnutrition, exacerbates inflammation, a known risk factor not only for cancer. However, experimental data showed that the composition of dietary fat has a greater impact on the pathogenesis of cancer than the total fat content in isocaloric diets. Similarly, human studies did not prove that a decrease in total fat intake is an effective strategy to combat cancer. Saturated fat has long been considered as harmful, but the current consensus is that moderate intake of saturated fatty acids (SFAs), including palmitic acid (PA), does not pose a health risk within a balanced diet. In regard to monounsaturated fat, plant sources are recommended. The consumption of plant monounsaturated fatty acids (MUFAs), particularly from olive oil, has been associated with lower cancer risk. Similarly, the replacement of animal MUFAs with plant MUFAs decreased cancer mortality. The impact of polyunsaturated fatty acids (PUFAs) on cancer risk depends on the ratio between ω-6 and ω-3 PUFAs. In vivo data showed stimulatory effects of ω-6 PUFAs on tumour growth while ω-3 PUFAs were protective, but the results of human studies were not as promising as indicated in preclinical reports. As for trans FAs (TFAs), experimental data mostly showed opposite effects of industrially produced and natural TFAs, with the latter being protective against cancer progression, but human data are mixed, and no clear conclusion can be made. Further studies are warranted to establish the role of FAs in the control of cell growth in order to find an effective strategy for cancer prevention/treatment.
Collapse
Affiliation(s)
- Bianka Bojková
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, P.J. Šafárik University in Košice, 041 54 Košice, Slovakia;
| | - Pawel J. Winklewski
- Department of Human Physiology, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Department of Anatomy and Physiology, Pomeranian University of Slupsk, 76-200 Slupsk, Poland
- Correspondence: ; Tel./Fax: +48-58-3491515
| | | |
Collapse
|
23
|
Henry E, Souissi-Sahraoui I, Deynoux M, Lefèvre A, Barroca V, Campalans A, Ménard V, Calvo J, Pflumio F, Arcangeli ML. Human hematopoietic stem/progenitor cells display reactive oxygen species-dependent long-term hematopoietic defects after exposure to low doses of ionizing radiations. Haematologica 2019; 105:2044-2055. [PMID: 31780635 PMCID: PMC7395291 DOI: 10.3324/haematol.2019.226936] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/27/2019] [Indexed: 01/16/2023] Open
Abstract
Hematopoietic stem cells are responsible for life-long blood cell production and are highly sensitive to exogenous stresses. The effects of low doses of ionizing radiations on radiosensitive tissues such as the hematopoietic tissue are still unknown despite their increasing use in medical imaging. Here, we study the consequences of low doses of ionizing radiations on differentiation and self-renewal capacities of human primary hematopoietic stem/progenitor cells (HSPC). We found that a single 20 mGy dose impairs the hematopoietic reconstitution potential of human HSPC but not their differentiation properties. In contrast to high irradiation doses, low doses of irradiation do not induce DNA double strand breaks in HSPC but, similar to high doses, induce a rapid and transient increase of reactive oxygen species (ROS) that promotes activation of the p38MAPK pathway. HSPC treatment with ROS scavengers or p38MAPK inhibitor prior exposure to 20 mGy irradiation abolishes the 20 mGy-induced defects indicating that ROS and p38MAPK pathways are transducers of low doses of radiation effects. Taken together, these results show that a 20 mGy dose of ionizing radiation reduces the reconstitution potential of HSPC suggesting an effect on the self-renewal potential of human hematopoietic stem cells and pinpointing ROS or the p38MAPK as therapeutic targets. Inhibition of ROS or the p38MAPK pathway protects human primary HSPC from low-dose irradiation toxicity.
Collapse
Affiliation(s)
- Elia Henry
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Inès Souissi-Sahraoui
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Margaux Deynoux
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Andréas Lefèvre
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Vilma Barroca
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Anna Campalans
- UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay.,CEA, DRF-JACOB-IRCM-SIGRR-LRIG, UMR "Genetic stability, Stem Cells and Radiation"
| | - Véronique Ménard
- UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay.,UMR "Genetic stability, Stem Cells and Radiation", F-92265 Fontenay-aux-Roses, France
| | - Julien Calvo
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Françoise Pflumio
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis".,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| | - Marie-Laure Arcangeli
- INSERM, U1274, Laboratory "Niche, Cancer and Hematopoiesis" .,CEA, DRF-JACOB-IRCM-SCSR-LSHL, UMR "Genetic stability, Stem Cells and Radiation".,UMR "Genetic stability, Stem Cells and Radiation" Université de Paris.,UMR "Genetic stability, Stem Cells and Radiation", Université Paris-Saclay
| |
Collapse
|
24
|
Kouzi F, Zibara K, Bourgeais J, Picou F, Gallay N, Brossaud J, Dakik H, Roux B, Hamard S, Le Nail LR, Hleihel R, Foucault A, Ravalet N, Rouleux-Bonnin F, Gouilleux F, Mazurier F, Bene MC, Akl H, Gyan E, Domenech J, El-Sabban M, Herault O. Disruption of gap junctions attenuates acute myeloid leukemia chemoresistance induced by bone marrow mesenchymal stromal cells. Oncogene 2019; 39:1198-1212. [PMID: 31649334 PMCID: PMC7002301 DOI: 10.1038/s41388-019-1069-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 01/09/2023]
Abstract
The bone marrow (BM) niche impacts the progression of acute myeloid leukemia (AML) by favoring the chemoresistance of AML cells. Intimate interactions between leukemic cells and BM mesenchymal stromal cells (BM-MSCs) play key roles in this process. Direct intercellular communications between hematopoietic cells and BM-MSCs involve connexins, components of gap junctions. We postulated that blocking gap junction assembly could modify cell–cell interactions in the leukemic niche and consequently the chemoresistance. The comparison of BM-MSCs from AML patients and healthy donors revealed a specific profile of connexins in BM-MSCs of the leukemic niche and the effects of carbenoxolone (CBX), a gap junction disruptor, were evaluated on AML cells. CBX presents an antileukemic effect without affecting normal BM-CD34+ progenitor cells. The proapoptotic effect of CBX on AML cells is in line with the extinction of energy metabolism. CBX acts synergistically with cytarabine (Ara-C) in vitro and in vivo. Coculture experiments of AML cells with BM-MSCs revealed that CBX neutralizes the protective effect of the niche against the Ara-C-induced apoptosis of leukemic cells. Altogether, these results suggest that CBX could be of therapeutic interest to reduce the chemoresistance favored by the leukemic niche, by targeting gap junctions, without affecting normal hematopoiesis.
Collapse
Affiliation(s)
- Farah Kouzi
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,PRASE, DSST, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE, DSST, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Jerome Bourgeais
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Frederic Picou
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Nathalie Gallay
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Julie Brossaud
- Department of Nuclear Medicine, Bordeaux University Hospital, Pessac, France
| | - Hassan Dakik
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Benjamin Roux
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Sophie Hamard
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | | | - Rita Hleihel
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Amelie Foucault
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Noemie Ravalet
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Florence Rouleux-Bonnin
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Fabrice Gouilleux
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Frederic Mazurier
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France
| | - Marie C Bene
- Department of Biological Hematology, Nantes University Hospital, CRCINA, Nantes, France
| | - Haidar Akl
- PRASE, DSST, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Emmanuel Gyan
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Hematology and Cell Therapy, Tours University Hospital, Tours, France
| | - Jorge Domenech
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France.,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Olivier Herault
- CNRS ERL7001 LNOx "Leukemic Niche & Redox Metabolism", Tours, France. .,EA7501 GICC, University of Tours, Faculty of Medicine, Tours, France. .,Department of Biological Hematology, Tours University Hospital, Tours, France.
| |
Collapse
|
25
|
Mortaz E, Moloudizargari M, Khosravi A, Asghari MH, Movassaghi M, Varahram M, Vaezi M, Redegeld FA, Garssen J. EPA and DHA have selective toxicity for PBMCs from multiple myeloma patients in a partly caspase-dependent manner. Clin Nutr 2019; 39:2137-2143. [PMID: 31558292 DOI: 10.1016/j.clnu.2019.08.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
Poly-unsaturated fatty acids (PUFAs) have been shown to have cytotoxic effects in both solid and non-solid tumors. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are among the most studied PUFAs. The aim of the present study was to evaluate the cytotoxic effects of these two fatty acids (FAs) in the peripheral blood mononuclear cells (PBMCs) obtained from untreated patients (new cases) with confirmed symptomatic multiple myeloma (MM). Our results showed that EPA at the concentration of 100 μM and DHA at 50 and 100 μM induce potent apoptotic effects in the PBMCs of MM patients (P < 0.05) as evidenced by Annexin V and propidium iodide (PI) staining, while they have little or no effects on the PBMCs isolated from healthy donors (P > 0.05). The observed effects were concentration- and time-dependent and 72 h treatment with DHA at a concentration of 100 μM had the strongest effect (P < 0.01). CD138 + cells isolated from MM patients showed great sensitivity to EPA/DHA. EPA- and DHA-induced apoptosis was significantly inhibited by the pan-caspase inhibitor (Z-VAD-FMK), indicating that cell death was at least partly dependent on caspase activation. The results of the present study showed that EPA and DHA have selective toxicities for malignant human plasma cells from MM patients, but not for mononuclear cells of healthy donors. These results warrant further studies with larger study populations to investigate the usefulness of PUFAs as a promising adjunctive therapy in the treatment of MM.
Collapse
Affiliation(s)
- Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Moloudizargari
- Department of Immunology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Adnan Khosravi
- Chronic Respiratory Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Asghari
- Department of Pharmacology, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mehrnaz Movassaghi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Varahram
- Mycobacteriology Research Center (MRC) National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Frank A Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands; Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| |
Collapse
|