101
|
Jin J, Byun JK, Choi YK, Park KG. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp Mol Med 2023; 55:706-715. [PMID: 37009798 PMCID: PMC10167356 DOI: 10.1038/s12276-023-00971-9] [Citation(s) in RCA: 200] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 04/04/2023] Open
Abstract
Proliferating cancer cells rely largely on glutamine for survival and proliferation. Glutamine serves as a carbon source for the synthesis of lipids and metabolites via the TCA cycle, as well as a source of nitrogen for amino acid and nucleotide synthesis. To date, many studies have explored the role of glutamine metabolism in cancer, thereby providing a scientific rationale for targeting glutamine metabolism for cancer treatment. In this review, we summarize the mechanism(s) involved at each step of glutamine metabolism, from glutamine transporters to redox homeostasis, and highlight areas that can be exploited for clinical cancer treatment. Furthermore, we discuss the mechanisms underlying cancer cell resistance to agents that target glutamine metabolism, as well as strategies for overcoming these mechanisms. Finally, we discuss the effects of glutamine blockade on the tumor microenvironment and explore strategies to maximize the utility of glutamine blockers as a cancer treatment.
Collapse
Affiliation(s)
- Jonghwa Jin
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea
| | - Jun-Kyu Byun
- BK21 FOUR Community-based Intelligent Novel Drug Discovery Education Unit, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, 41566, Korea
| | - Yeon-Kyung Choi
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, 41404, Korea.
| | - Keun-Gyu Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, South Korea.
| |
Collapse
|
102
|
Metabolomics analysis reveals cytotoxic effects of ouabain towards psoriatic keratinocytes via impairment of glutathione metabolism. Mol Genet Genomics 2023; 298:567-577. [PMID: 36856826 PMCID: PMC10133367 DOI: 10.1007/s00438-023-02001-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/17/2023] [Indexed: 03/02/2023]
Abstract
Ouabain is a cardiac glycoside long studied for treating heart diseases, but the attempts to evaluate its anti-psoriatic activity have not been reported. We aimed to explore the effects of ouabain on proliferation and metabolism towards psoriatic keratinocytes. In human HaCaT keratinocytes, ouabain potently decreased viability, promoted apoptosis and caused G2/M cycle arrest. Metabolomics analysis indicated that ouabain markedly impaired glutathione metabolism. The solute carrier family 7 member 11 (SLC7A11) is an amino acid transporter highly specific to cysteine, which is critical for glutathione synthesis. Ouabain downregulated SLC7A11, reduced cysteine uptake and subsequently inhibited glutathione synthesis, probably through inhibiting Akt/mTOR/beclin axis that regulate protein activity of SLC7A11. The impaired glutathione synthesis and oxidative stress caused by ouabain may contribute to its cytotoxicity towards psoriatic keratinocytes. Our results provide experimental evidence supporting further study of ouabain as a potential anti-psoriatic agent.
Collapse
|
103
|
Glutamine Metabolism in Cancer Stem Cells: A Complex Liaison in the Tumor Microenvironment. Int J Mol Sci 2023; 24:ijms24032337. [PMID: 36768660 PMCID: PMC9916789 DOI: 10.3390/ijms24032337] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
In this review we focus on the role of glutamine in control of cancer stem cell (CSC) fate. We first provide an overview of glutamine metabolism, and then summarize relevant studies investigating how glutamine metabolism modulates the CSC compartment, concentrating on solid tumors. We schematically describe how glutamine in CSC contributes to several metabolic pathways, such as redox metabolic pathways, ATP production, non-essential aminoacids and nucleotides biosynthesis, and ammonia production. Furthermore, we show that glutamine metabolism is a key regulator of epigenetic modifications in CSC. Finally, we briefly discuss how cancer-associated fibroblasts, adipocytes, and senescent cells in the tumor microenvironment may indirectly influence CSC fate by modulating glutamine availability. We aim to highlight the complexity of glutamine's role in CSC, which supports our knowledge about metabolic heterogeneity within the CSC population.
Collapse
|
104
|
Overcoming cancer chemotherapy resistance by the induction of ferroptosis. Drug Resist Updat 2023; 66:100916. [PMID: 36610291 DOI: 10.1016/j.drup.2022.100916] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022]
Abstract
Development of resistance to chemotherapy in cancer continues to be a major challenge in cancer management. Ferroptosis, a unique type of cell death, is mechanistically and morphologically different from other forms of cell death. Ferroptosis plays a pivotal role in inhibiting tumour growth and has presented new opportunities for treatment of chemotherapy-insensitive tumours in recent years. Emerging studies have suggested that ferroptosis can regulate the therapeutic responses of tumours. Accumulating evidence supports ferroptosis as a potential target for chemotherapy resistance. Pharmacological induction of ferroptosis could reverse drug resistance in tumours. In this review article, we first discuss the key principles of chemotherapeutic resistance in cancer. We then provide a brief overview of the core mechanisms of ferroptosis in cancer chemotherapeutic drug resistance. Finally, we summarise the emerging data that supports the fact that chemotherapy resistance in different types of cancers could be subdued by pharmacologically inducing ferroptosis. This review article suggests that pharmacological induction of ferroptosis by bioactive compounds (ferroptosis inducers) could overcome chemotherapeutic drug resistance. This article also highlights some promising therapeutic avenues that could be used to overcome chemotherapeutic drug resistance in cancer.
Collapse
|
105
|
Wang J, Hao S, Song G, Wang Y, Hao Q. The prognostic and clinicopathological significance of SLC7A11 in human cancers: a systematic review and meta-analysis. PeerJ 2023; 11:e14931. [PMID: 36874967 PMCID: PMC9979827 DOI: 10.7717/peerj.14931] [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: 11/28/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023] Open
Abstract
Objective It is of great importance to recognize bio-markers for cancer prognosis. However, the association between solute carrier family 7 member 11 (SLC7A11) and prognosis is still controversial. Therefore, we conducted this systematic review and meta-analysis to identify the prognostic and clinicopathological significance of SLC7A11 in human cancers. Methods PubMed, Web of Science, Scopus, the Cochrane Library and Embase database were searched from database inceptions to March 19th 2022. Hand searches were also conducted in references. Prognosis and clinicopathological data were extracted and analyzed. Results A total of 12 eligible studies with 1,955 patients were included. The results indicated that SLC7A11 expression is associated with unfavorable overall survival (OS), unfavorable recurrence-free survival (RFS) and unfavorable progression free survival (PFS). And SLC7A11 expression is also associated with more advanced tumor stage. Conclusions SLC7A11 expression is associated with more unfavorable prognosis and more advanced tumor stage. Therefore, SLC7A11 could be a potential biomarker for human cancer prognosis.
Collapse
Affiliation(s)
- Jiantao Wang
- Sichuan University, State Key Laboratory of Biotherapy & Department of Lung Cancer Center and Department of Radiation Oncology, West China Hospital, Chengdu, China
| | - Siyuan Hao
- Sichuan University, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Chengdu, China
| | - Guojiao Song
- Sichuan University, State Key Laboratory of Biotherapy & Department of Lung Cancer Center and Department of Radiation Oncology, West China Hospital, Chengdu, China
| | - Yan Wang
- Sichuan University, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Chengdu, China
| | - Qiukui Hao
- McMaster University, Faculty of Health Sciences, School of Rehabilitation Science, Ontario, Canada
| |
Collapse
|
106
|
Zhao S, Wang L, Ding W, Ye B, Cheng C, Shao J, Liu J, Zhou H. Crosstalk of disulfidptosis-related subtypes, establishment of a prognostic signature and immune infiltration characteristics in bladder cancer based on a machine learning survival framework. Front Endocrinol (Lausanne) 2023; 14:1180404. [PMID: 37152941 PMCID: PMC10154596 DOI: 10.3389/fendo.2023.1180404] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Background Bladder cancer (BLCA) is the most common malignancy of the urinary tract. On the other hand, disulfidptosis, a mechanism of disulfide stress-induced cell death, is closely associated with tumorigenesis and progression. Here, we investigated the impact of disulfidptosis-related genes (DRGs) on the prognosis of BLCA, identified various DRG clusters, and developed a risk model to assess patient prognosis, immunological profile, and treatment response. Methods The expression and mutational characteristics of four DRGs were first analyzed in bulk RNA-Seq and single-cell RNA sequencing data, IHC staining identified the role of DRGs in BLCA progression, and two DRG clusters were identified by consensus clustering. Using the differentially expressed genes (DEGs) from these two clusters, we transformed ten machine learning algorithms into more than 80 combinations and finally selected the best algorithm to construct a disulfidptosis-related prognostic signature (DRPS). We based this selection on the mean C-index of three BLCA cohorts. Furthermore, we explored the differences in clinical characteristics, mutational landscape, immune cell infiltration, and predicted efficacy of immunotherapy between high and low-risk groups. To visually depict the clinical value of DRPS, we employed nomograms. Additionally, we verified whether DRPS predicts response to immunotherapy in BLCA patients by utilizing the Tumour Immune Dysfunction and Rejection (TIDE) and IMvigor 210 cohorts. Results In the integrated cohort, we identified several DRG clusters and DRG gene clusters that differed significantly in overall survival (OS) and tumor microenvironment. After the integration of clinicopathological features, DRPS showed robust predictive power. Based on the median risk score associated with disulfidptosis, BLCA patients were divided into low-risk (LR) and high-risk (HR) groups, with patients in the LR group having a better prognosis, a higher tumor mutational load and being more sensitive to immunotherapy and chemotherapy. Conclusion Our study, therefore, provides a valuable tool to further guide clinical management and tailor the treatment of BLCA patients, offering new insights into individualized treatment.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Urology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Lanyu Wang
- Department of Urology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Wei Ding
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bicheng Ye
- School of Clinical Medicine, Yangzhou Polytechnic College, Yangzhou, China
| | - Chao Cheng
- Department of Neurosurgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jianfeng Shao
- Department of Urology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
- *Correspondence: Jianfeng Shao, ; Jinhui Liu, ; Hongyi Zhou,
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Jianfeng Shao, ; Jinhui Liu, ; Hongyi Zhou,
| | - Hongyi Zhou
- Department of Urology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
- *Correspondence: Jianfeng Shao, ; Jinhui Liu, ; Hongyi Zhou,
| |
Collapse
|
107
|
Drüeke TB, Massy ZA. Vascular calcification in chronic kidney disease: contribution of ferroptosis? Kidney Int 2022; 102:1209-1211. [PMID: 36411014 DOI: 10.1016/j.kint.2022.08.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022]
Abstract
Vascular calcification associated with chronic kidney disease (CKD) is an active, regulated process. Apoptosis of vascular smooth muscle cells has long been known to play a major role in its pathogenesis, with apoptotic bodies derived from these cells acting as nucleating structures for calcium crystal formation and deposition. Ye et al. now show in experimental models in vitro and in vivo that ferroptosis can also contribute to the development of vascular calcification in CKD.
Collapse
Affiliation(s)
- Tilman B Drüeke
- Inserm U-1018, Centre de recherche en épidémiologie et santé des populations, Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, Université de Versailles Saint-Quentin-en-Yvelines), and Paris Saclay University, Villejuif, France.
| | - Ziad A Massy
- Inserm U-1018, Centre de recherche en épidémiologie et santé des populations, Versailles Saint-Quentin-en-Yvelines University (Paris-Ile-de-France-Ouest University, Université de Versailles Saint-Quentin-en-Yvelines), and Paris Saclay University, Villejuif, France; Department of Nephrology, Ambroise Paré University Hospital, Assistance Publique - Hôpitaux de Paris, Boulogne-Billancourt, Paris, France
| |
Collapse
|
108
|
Lin KJ, Chen SD, Lin KL, Liou CW, Lan MY, Chuang YC, Wang PW, Lee JJ, Wang FS, Lin HY, Lin TK. Iron Brain Menace: The Involvement of Ferroptosis in Parkinson Disease. Cells 2022; 11:3829. [PMID: 36497089 PMCID: PMC9735800 DOI: 10.3390/cells11233829] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson disease (PD) is the second-most common neurodegenerative disease. The characteristic pathology of progressive dopaminergic neuronal loss in people with PD is associated with iron accumulation and is suggested to be driven in part by the novel cell death pathway, ferroptosis. A unique modality of cell death, ferroptosis is mediated by iron-dependent phospholipid peroxidation. The mechanisms of ferroptosis inhibitors enhance antioxidative capacity to counter the oxidative stress from lipid peroxidation, such as through the system xc-/glutathione (GSH)/glutathione peroxidase 4 (GPX4) axis and the coenzyme Q10 (CoQ10)/FSP1 pathway. Another means to reduce ferroptosis is with iron chelators. To date, there is no disease-modifying therapy to cure or slow PD progression, and a recent topic of research seeks to intervene with the development of PD via regulation of ferroptosis. In this review, we provide a discussion of different cell death pathways, the molecular mechanisms of ferroptosis, the role of ferroptosis in blood-brain barrier damage, updates on PD studies in ferroptosis, and the latest progress of pharmacological agents targeting ferroptosis for the intervention of PD in clinical trials.
Collapse
Affiliation(s)
- Kai-Jung Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Family Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Shang-Der Chen
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center of Parkinson’s Disease, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Kai-Lieh Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chia-Wei Liou
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center of Parkinson’s Disease, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Min-Yu Lan
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center of Parkinson’s Disease, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Yao-Chung Chuang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center of Parkinson’s Disease, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Neurology, Pao Chien Hospital, Pingtung 90064, Taiwan
- Department of Biological Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| | - Pei-Wen Wang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Metabolism, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jong-Jer Lee
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Feng-Sheng Wang
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Hung-Yu Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Tsu-Kung Lin
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Center of Parkinson’s Disease, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| |
Collapse
|
109
|
Zhang X, Ammous F, Lin L, Ratliff SM, Ware EB, Faul JD, Zhao W, Kardia SLR, Smith JA. The Interplay of Epigenetic, Genetic, and Traditional Risk Factors on Blood Pressure: Findings from the Health and Retirement Study. Genes (Basel) 2022; 13:1959. [PMID: 36360196 PMCID: PMC9689874 DOI: 10.3390/genes13111959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/11/2022] [Accepted: 10/19/2022] [Indexed: 01/21/2023] Open
Abstract
The epigenome likely interacts with traditional and genetic risk factors to influence blood pressure. We evaluated whether 13 previously reported DNA methylation sites (CpGs) are associated with systolic (SBP) or diastolic (DBP) blood pressure, both individually and aggregated into methylation risk scores (MRS), in 3070 participants (including 437 African ancestry (AA) and 2021 European ancestry (EA), mean age = 70.5 years) from the Health and Retirement Study. Nine CpGs were at least nominally associated with SBP and/or DBP after adjusting for traditional hypertension risk factors (p < 0.05). MRSSBP was positively associated with SBP in the full sample (β = 1.7 mmHg per 1 standard deviation in MRSSBP; p = 2.7 × 10-5) and in EA (β = 1.6; p = 0.001), and MRSDBP with DBP in the full sample (β = 1.1; p = 1.8 × 10-6), EA (β = 1.1; p = 7.2 × 10-5), and AA (β = 1.4; p = 0.03). The MRS and BP-genetic risk scores were independently associated with blood pressure in EA. The effects of both MRSs were weaker with increased age (pinteraction < 0.01), and the effect of MRSDBP was higher among individuals with at least some college education (pinteraction = 0.02). In AA, increasing MRSSBP was associated with higher SBP in females only (pinteraction = 0.01). Our work shows that MRS is a potential biomarker of blood pressure that may be modified by traditional hypertension risk factors.
Collapse
Affiliation(s)
- Xinman Zhang
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lisha Lin
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott M. Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erin B. Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
110
|
Guo L, Zhang Q, Liu Y. The role of microRNAs in ferroptosis. Front Mol Biosci 2022; 9:1003045. [PMID: 36310600 PMCID: PMC9596748 DOI: 10.3389/fmolb.2022.1003045] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Ferroptosis is a newly discovered type of programmed cell death, which is closely related to the imbalance of iron metabolism and oxidative stress. Ferroptosis has become an important research topic in the fields of cardiomyopathy, tumors, neuronal injury disorders, and ischemia perfusion disorders. As an important part of non-coding RNA, microRNAs regulate various metabolic pathways in the human body at the post-transcriptional level and play a crucial role in the occurrence and development of many diseases. The present review introduces the mechanisms of ferroptosis and describes the relevant pathways by which microRNAs affect cardiomyopathy, tumors, neuronal injury disorders and ischemia perfusion disorders through regulating ferroptosis. In addition, it provides important insights into ferroptosis-related microRNAs, aiming to uncover new methods for treatment of the above diseases, and discusses new ideas for the implementation of possible microRNA-based ferroptosis-targeted therapies in the future.
Collapse
Affiliation(s)
- Liqing Guo
- Department of Otolaryngology, The Second Affiliated Hospital of Nanchang University, NanChang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
| | - Qingkun Zhang
- Department of Otolaryngology, The Second Affiliated Hospital of Nanchang University, NanChang, China
| | - Yuehui Liu
- Department of Otolaryngology, The Second Affiliated Hospital of Nanchang University, NanChang, China
- *Correspondence: Yuehui Liu,
| |
Collapse
|
111
|
Zhang G, Fang Y, Li X, Zhang Z. Ferroptosis: A novel therapeutic strategy and mechanism of action in glioma. Front Oncol 2022; 12:947530. [PMID: 36185243 PMCID: PMC9520297 DOI: 10.3389/fonc.2022.947530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Glioma is the most common malignant tumor of the central nervous system and resistance is easily developed to chemotherapy drugs during the treatment process, resulting in high mortality and short survival in glioma patients. Novel therapeutic approaches are urgently needed to improve the therapeutic efficacy of chemotherapeutic drugs and to improve the prognosis of patients with glioma. Ferroptosis is a novel regulatory cell death mechanism that plays a key role in cancer, neurodegenerative diseases, and other diseases. Studies have found that ferroptosis-related regulators are closely related to the survival of patients with glioma, and induction of ferroptosis can improve glioma resistance to chemotherapy drugs. Therefore, induction of tumor cell ferroptosis may be an effective therapeutic strategy for glioma. This review summarizes the relevant mechanisms of ferroptosis, systematically summarizes the key role of ferroptosis in the treatment of glioma and outlines the relationship between ferroptosis-related ncRNAs and the progression of glioma.
Collapse
|
112
|
Galluccio M, Scalise M, Pappacoda G, Scarpelli M, Bonanomi M, Gaglio D, Indiveri C. Production of recombinant human xCT (SLC7A11) and reconstitution in proteoliposomes for functional studies. Front Physiol 2022; 13:993626. [PMID: 36148304 PMCID: PMC9485625 DOI: 10.3389/fphys.2022.993626] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
The plasma membrane transporter xCT belongs to the SLC7 family and has the physiological role of mediating the exchange of glutamate and cystine across the cell plasma membrane, being crucial for redox control. The xCT protein forms a heterodimer with the ancillary protein CD98. Over the years, xCT became a hot pharmacological target due to the documented over-expression in virtually all human cancers, which rely on cystine availability for their progression. Notwithstanding, several unknown aspects of xCT biology still exist that require a suitable single protein experimental model, to be addressed. To this aim, the recombinant host Escherichia coli has been exploited to over-express the human isoform of xCT. In this widely used and low-cost system, the optimization for growth and protein production has been achieved by acting on the metabolic needs of the bacterial strains. Then, the His-tagged protein has been purified by Ni2+-chelating chromatography and reconstituted in proteoliposomes for transport activity assays. The expressed protein was in a folded/active state allowing functional and kinetic characterization. Interestingly, the features of the recombinant protein meet those of the native one extracted from intact cells, further confirming the suitability of E. coli as a host for the expression of human proteins. This study opens perspectives for elucidating other molecular aspects of xCT, as well as for studying the interaction with endogenous and exogenous compounds, relevant to human health.
Collapse
Affiliation(s)
- Michele Galluccio
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia Ecologia Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia Ecologia Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Gilda Pappacoda
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia Ecologia Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Martina Scarpelli
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia Ecologia Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Marcella Bonanomi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
- ISBE.IT/Centre of Systems Biology, Milan, Italy
| | - Daniela Gaglio
- ISBE.IT/Centre of Systems Biology, Milan, Italy
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), Milan, Italy
| | - Cesare Indiveri
- Unit of Biochemistry and Molecular Biotechnology, Department DiBEST (Biologia Ecologia Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
- CNR Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies (IBIOM), Bari, Italy
- *Correspondence: Cesare Indiveri,
| |
Collapse
|
113
|
Hormesis and Oxidative Distress: Pathophysiology of Reactive Oxygen Species and the Open Question of Antioxidant Modulation and Supplementation. Antioxidants (Basel) 2022; 11:antiox11081613. [PMID: 36009331 PMCID: PMC9405171 DOI: 10.3390/antiox11081613] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Alterations of redox homeostasis leads to a condition of resilience known as hormesis that is due to the activation of redox-sensitive pathways stimulating cell proliferation, growth, differentiation, and angiogenesis. Instead, supraphysiological production of reactive oxygen species (ROS) exceeds antioxidant defence and leads to oxidative distress. This condition induces damage to biomolecules and is responsible or co-responsible for the onset of several chronic pathologies. Thus, a dietary antioxidant supplementation has been proposed in order to prevent aging, cardiovascular and degenerative diseases as well as carcinogenesis. However, this approach has failed to demonstrate efficacy, often leading to harmful side effects, in particular in patients affected by cancer. In this latter case, an approach based on endogenous antioxidant depletion, leading to ROS overproduction, has shown an interesting potential for enhancing susceptibility of patients to anticancer therapies. Therefore, a deep investigation of molecular pathways involved in redox balance is crucial in order to identify new molecular targets useful for the development of more effective therapeutic approaches. The review herein provides an overview of the pathophysiological role of ROS and focuses the attention on positive and negative aspects of antioxidant modulation with the intent to find new insights for a successful clinical application.
Collapse
|
114
|
Mejía-Hernández JO, Raghu D, Caramia F, Clemons N, Fujihara K, Riseborough T, Teunisse A, Jochemsen AG, Abrahmsén L, Blandino G, Russo A, Gamell C, Fox SB, Mitchell C, Takano EA, Byrne D, Miranda PJ, Saleh R, Thorne H, Sandhu S, Williams SG, Keam SP, Haupt Y, Haupt S. Targeting MDM4 as a Novel Therapeutic Approach in Prostate Cancer Independent of p53 Status. Cancers (Basel) 2022; 14:3947. [PMID: 36010941 PMCID: PMC9405814 DOI: 10.3390/cancers14163947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Metastatic prostate cancer is a lethal disease in patients incapable of responding to therapeutic interventions. Invasive prostate cancer spread is caused by failure of the normal anti-cancer defense systems that are controlled by the tumour suppressor protein, p53. Upon mutation, p53 malfunctions. Therapeutic strategies to directly re-empower the growth-restrictive capacities of p53 in cancers have largely been unsuccessful, frequently because of a failure to discriminate responses in diseased and healthy tissues. Our studies sought alternative prostate cancer drivers, intending to uncover new treatment targets. We discovered the oncogenic potency of MDM4 in prostate cancer cells, both in the presence and absence of p53 and also its mutation. We uncovered that sustained depletion of MDM4 is growth inhibitory in prostate cancer cells, involving either apoptosis or senescence, depending on the cell and genetic context. We identified that the potency of MDM4 targeting could be potentiated in prostate cancers with mutant p53 through the addition of a first-in-class small molecule drug that was selected as a p53 reactivator and has the capacity to elevate oxidative stress in cancer cells to drive their death.
Collapse
Affiliation(s)
- Javier Octavio Mejía-Hernández
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Dinesh Raghu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - Franco Caramia
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicholas Clemons
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kenji Fujihara
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Thomas Riseborough
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Amina Teunisse
- Department of Cell and Chemical Biology, Leiden University Medical Centre, 2333 Leiden, The Netherlands
| | - Aart G. Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Centre, 2333 Leiden, The Netherlands
| | | | - Giovanni Blandino
- Translational Oncology Research Unit, IRCSS Regina Elena National Cancer Institute, 0144 Rome, Italy
| | - Andrea Russo
- Surgical Pathology Unit, Department of Research, Advanced Diagnostics and Technological Innovation, IRCSS Regina Elena National Cancer Institute, 0144 Rome, Italy
| | - Cristina Gamell
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stephen B. Fox
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Catherine Mitchell
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Elena A. Takano
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - David Byrne
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Pathology Department, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Panimaya Jeffreena Miranda
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Reem Saleh
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Heather Thorne
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Scott G. Williams
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Parkville, VIC 3000, Australia
| | - Simon P. Keam
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ygal Haupt
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sue Haupt
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Tumour Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
115
|
Piccini I, Sousa M, Altendorf S, Jimenez F, Rossi A, Funk W, Bíró T, Paus R, Seibel J, Jakobs M, Yesilkaya T, Edelkamp J, Bertolini M. Intermediate Hair Follicles from Patients with Female Pattern Hair Loss Are Associated with Nutrient Insufficiency and a Quiescent Metabolic Phenotype. Nutrients 2022; 14:3357. [PMID: 36014862 PMCID: PMC9416027 DOI: 10.3390/nu14163357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/28/2022] Open
Abstract
Female pattern hair loss (FPHL) is a non-scarring alopecia resulting from the progressive conversion of the terminal (t) scalp hair follicles (HFs) into intermediate/miniaturized (i/m) HFs. Although data supporting nutrient deficiency in FPHL HFs are lacking, therapeutic strategies are often associated with nutritional supplementation. Here, we show by metabolic analysis that selected nutrients important for hair growth such as essential amino acids and vitamins are indeed decreased in affected iHFs compared to tHFs in FPHL scalp skin, confirming nutrient insufficiency. iHFs also displayed a more quiescent metabolic phenotype, as indicated by altered metabolite abundance in freshly collected HFs and release/consumption during organ culture of products/substrates of TCA cycle, aerobic glycolysis, and glutaminolysis. Yet, as assessed by exogenous nutrient supplementation ex vivo, nutrient uptake mechanisms are not impaired in affected FPHL iHFs. Moreover, blood vessel density is not diminished in iHFs versus tHFs, despite differences in tHFs from different FPHL scalp locations or versus healthy scalp or changes in the expression of angiogenesis-associated growth factors. Thus, our data reveal that affected iHFs in FPHL display a relative nutrient insufficiency and dormant metabolism, but are still capable of absorbing nutrients, supporting the potential of nutritional supplementation as an adjunct therapy for FPHL.
Collapse
Affiliation(s)
- Ilaria Piccini
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
| | - Marta Sousa
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
| | - Sabrina Altendorf
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
| | - Francisco Jimenez
- Mediteknia Hair Transplant Clinic and Hair Lab, Universidad Fernando Pessoa Canarias, Gran Canaria, Canary Islands, 35450 Guía, Spain
| | - Alfredo Rossi
- Department of Clinical Internal Anesthesiological and Cardiovascular Sciences, “Sapienza” University of Rome, 00161 Rome, Italy
| | | | - Tamás Bíró
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
| | - Ralf Paus
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
- Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | - Janin Edelkamp
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
| | - Marta Bertolini
- Monasterium Laboratory Skin & Hair Research Solutions GmbH, 48149 Münster, Germany
| |
Collapse
|
116
|
Ascenção K, Szabo C. Emerging roles of cystathionine β-synthase in various forms of cancer. Redox Biol 2022; 53:102331. [PMID: 35618601 PMCID: PMC9168780 DOI: 10.1016/j.redox.2022.102331] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of the reverse transsulfuration enzyme cystathionine-β-synthase (CBS) is markedly increased in many forms of cancer, including colorectal, ovarian, lung, breast and kidney, while in other cancers (liver cancer and glioma) it becomes downregulated. According to the clinical database data in high-CBS-expressor cancers (e.g. colon or ovarian cancer), high CBS expression typically predicts lower survival, while in the low-CBS-expressor cancers (e.g. liver cancer), low CBS expression is associated with lower survival. In the high-CBS expressing tumor cells, CBS, and its product hydrogen sulfide (H2S) serves as a bioenergetic, proliferative, cytoprotective and stemness factor; it also supports angiogenesis and epithelial-to-mesenchymal transition in the cancer microenvironment. The current article reviews the various tumor-cell-supporting roles of the CBS/H2S axis in high-CBS expressor cancers and overviews the anticancer effects of CBS silencing and pharmacological CBS inhibition in various cancer models in vitro and in vivo; it also outlines potential approaches for biomarker identification, to support future targeted cancer therapies based on pharmacological CBS inhibition.
Collapse
|
117
|
Criscuolo D, Morra F, Celetti A. A xCT role in tumour-associated ferroptosis shed light on novel therapeutic options. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:570-581. [PMID: 36338517 PMCID: PMC9630094 DOI: 10.37349/etat.2022.00101] [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: 05/13/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022] Open
Abstract
Solute carrier family 7 member 11 (SLC7A11; also known as xCT), a key component of the cystine/glutamate antiporter, is essential for the maintenance of cellular redox status and the regulation of tumor-associated ferroptosis. Accumulating evidence has demonstrated that xCT overexpression, resulting from different oncogenic and tumor suppressor signaling, promotes tumor progression and multidrug resistance partially via suppressing ferroptosis. In addition, recent studies have highlighted the role of xCT in regulating the metabolic flexibility in cancer cells. In this review, the xCT activities in intracellular redox balance and in ferroptotic cell death have been summarized. Moreover, the role of xCT in promoting tumor development, drug resistance, and nutrient dependency in cancer cells has been explored. Finally, different therapeutic strategies, xCT-based, for anti-cancer treatments have been discussed.
Collapse
Affiliation(s)
- Daniela Criscuolo
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, 80131 Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Francesco Morra
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, 80131 Naples, Italy,Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Angela Celetti
- Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, 80131 Naples, Italy,Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy,Correspondence: Angela Celetti, Institute for the Experimental Endocrinology and Oncology, Research National Council, CNR, 80131 Naples, Italy.
| |
Collapse
|