1
|
Ji JX, Hoang LN, Cochrane DR, Lum A, Senz J, Farnell D, Tessier-Cloutier B, Huntsman DG, Klein Geltink RI. The unique metabolome of clear cell ovarian carcinoma. J Pathol 2024; 264:160-173. [PMID: 39096103 DOI: 10.1002/path.6329] [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: 02/25/2024] [Revised: 04/29/2024] [Accepted: 06/08/2024] [Indexed: 08/04/2024]
Abstract
Clear cell ovarian carcinoma (CCOC) is an aggressive malignancy affecting younger women. Despite ovarian cancer subtypes having diverse molecular and clinical characteristics, the mainstay of treatment for advanced stage disease remains cytotoxic chemotherapy. Late stage CCOC is resistant to conventional chemotherapy, which means a suboptimal outcome for patients affected. Despite detailed genomic, epigenomic, transcriptomic, and proteomic characterisation, subtype-specific treatment for CCOC has shown little progress. The unique glycogen accumulation defining CCOC suggests altered metabolic pathway activity and dependency. This study presents the first metabolomic landscape of ovarian cancer subtypes, including 42 CCOC, 20 high-grade serous and 21 endometrioid ovarian carcinomas, together comprising the three most common ovarian carcinoma subtypes. We describe a distinct metabolomic landscape of CCOC compared with other ovarian cancer subtypes, including alterations in energy utilisation and cysteine metabolism. In addition, we identify CCOC-specific alterations in metabolic pathways including serine biosynthesis and ROS-associated pathways that could serve as potential therapeutic targets. Our study provides the first in-depth study into the metabolome of ovarian cancers and a rich resource to support ongoing research efforts to identify subtype-specific therapeutic targets that could improve the dismal outcome for patients with this devastating malignancy. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jennifer X Ji
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lien N Hoang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Amy Lum
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Janine Senz
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - David Farnell
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Ramon I Klein Geltink
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
2
|
Cardona CJ, Kim Y, Chowanadisai W, Montgomery MR. Considerations for Using Neuroblastoma Cell Lines to Examine the Roles of Iron and Ferroptosis in Neurodegeneration. Cells 2024; 13:1541. [PMID: 39329725 PMCID: PMC11430288 DOI: 10.3390/cells13181541] [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: 08/16/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Ferroptosis is an iron-dependent form of programmed cell death that is influenced by biological processes such as iron metabolism and senescence. As brain iron levels increase with aging, ferroptosis is also implicated in the development of age-related pathologic conditions such as Alzheimer's disease (AD) and related dementias (ADRD). Indeed, inhibitors of ferroptosis have been shown to be protective in models of degenerative brain disorders like AD/ADRD. Given the inaccessibility of the living human brain for metabolic studies, the goal of this work was to characterize an in vitro model for understanding how aging and iron availability influence neuronal iron metabolism and ferroptosis. First, the human (SH-SY5Y) and mouse (Neuro-2a) neuroblastoma lines were terminally differentiated into mature neurons by culturing in all-trans-retinoic acid for at least 72 h. Despite demonstrating all signs of neuronal differentiation and maturation, including increased expression of the iron storage protein ferritin, we discovered that differentiation conferred ferroptosis resistance in both cell lines. Gene expression data indicates differentiated neurons increase their capacity to protect against iron-mediated oxidative damage by augmenting cystine import, and subsequently increasing intracellular cysteine levels, to promote glutathione production and glutathione peroxidase activity (GPX). In support of this hypothesis, we found that culturing differentiated neurons in cysteine-depleted media sensitized them to GPX4 inhibition, and that these effects are mitigated by cystine supplementation. Such findings are important as they provide guidance for the use of in vitro experimental models to investigate the role of ferroptosis in neurodegeneration in pathologies such as ADRD.
Collapse
Affiliation(s)
- Cameron J Cardona
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Yoo Kim
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Winyoo Chowanadisai
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - McKale R Montgomery
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
3
|
Guo K, Lu M, Bi J, Yao T, Gao J, Ren F, Zhu L. Ferroptosis: mechanism, immunotherapy and role in ovarian cancer. Front Immunol 2024; 15:1410018. [PMID: 39192972 PMCID: PMC11347334 DOI: 10.3389/fimmu.2024.1410018] [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: 03/31/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Ovarian cancer is currently the second most common malignant tumor among gynecological cancers worldwide, primarily due to challenges in early diagnosis, high recurrence rates, and resistance to existing treatments. Current therapeutic options are inadequate for addressing the needs of ovarian cancer patients. Ferroptosis, a novel form of regulated cell death with demonstrated tumor-suppressive properties, has gained increasing attention in ovarian malignancy research. A growing body of evidence suggests that ferroptosis plays a significant role in the onset, progression, and incidence of ovarian cancer. Additionally, it has been found that immunotherapy, an emerging frontier in tumor treatment, synergizes with ferroptosis in the context of ovarian cancer. Consequently, ferroptosis is likely to become a critical target in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ke Guo
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Miao Lu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianlei Bi
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Tianyu Yao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fang Ren
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
4
|
Chahla C, Kovacic H, Ferhat L, Leloup L. Pathological Impact of Redox Post-Translational Modifications. Antioxid Redox Signal 2024; 41:152-180. [PMID: 38504589 DOI: 10.1089/ars.2023.0252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Oxidative stress is involved in the development of several pathologies. The different reactive oxygen species (ROS) produced during oxidative stress are at the origin of redox post-translational modifications (PTMs) on proteins and impact nucleic acids and lipids. This review provides an overview of recent data on cysteine and methionine oxidation and protein carbonylation following oxidative stress in a pathological context. Oxidation, like nitration, is a selective process and not all proteins are impacted. It depends on multiple factors, including amino acid environment, accessibility, and physical and chemical properties, as well as protein structures. Thiols can undergo reversible oxidations and others that are irreversible. On the contrary, carbonylation represents irreversible PTM. To date, hundreds of proteins were shown to be modified by ROS and reactive nitrogen species (RNS). We reviewed recent advances in the impact of redox-induced PTMs on protein functions and activity, as well as its involvement in disease development or treatment. These data show a complex situation of the involvement of redox PTM on the function of targeted proteins. Many proteins can have their activity decreased by the oxidation of cysteine thiols or methionine S-methyl thioethers, while for other proteins, this oxidation will be activating. This complexity of redox PTM regulation suggests that a global antioxidant therapeutic approach, as often proposed, is unlikely to be effective. However, the specificity of the effect obtained by targeting a cysteine or methionine residue to be able to inactivate or activate a particular protein represents a major interest if it is possible to consider this targeting from a therapeutic point of view with our current pharmacological tools. Antioxid. Redox Signal. 41, 152-180.
Collapse
Affiliation(s)
- Charbel Chahla
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| | - Hervé Kovacic
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| | - Lotfi Ferhat
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| | - Ludovic Leloup
- Faculté de Médecine, INP, Institut de neurophysiopathologie, Aix Marseille Université, CNRS, Marseille, France
| |
Collapse
|
5
|
Ji M, Xu Q, Li X. Dietary methionine restriction in cancer development and antitumor immunity. Trends Endocrinol Metab 2024; 35:400-412. [PMID: 38383161 PMCID: PMC11096033 DOI: 10.1016/j.tem.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Methionine restriction (MR) has been shown to suppress tumor growth and improve the responses to various anticancer therapies. However, methionine itself is required for the proliferation, activation, and differentiation of T cells that are crucial for antitumor immunity. The dual impact of methionine, that influences both tumor and immune cells, has generated concerns regarding the potential consequences of MR on T cell immunity and its possible role in promoting cancer. In this review we systemically examine current literature on the interactions between dietary methionine, cancer cells, and immune cells. Based on recent findings on MR in immunocompetent animals, we further discuss how tumor stage-specific methionine dependence of immune cells and cancer cells in the tumor microenvironment could ultimately dictate the response of tumors to MR.
Collapse
Affiliation(s)
- Ming Ji
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Qing Xu
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
6
|
Vaziripour M, Faghihi M, Ranjbaran M, Asadi B, Abdi A, Kianian F, Hajiaqaei M, Seifi B. Exploring the Therapeutic Potential of Sodium Hydrosulfide in Alleviating Oxidative Stress and Ovarian Dysfunction in a Rat Model of Polycystic Ovary Syndrome. J Reprod Infertil 2024; 25:133-139. [PMID: 39157798 PMCID: PMC11327425 DOI: 10.18502/jri.v25i2.16007] [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: 11/07/2023] [Accepted: 04/07/2024] [Indexed: 08/20/2024] Open
Abstract
Background Oxidative stress is known to play a key role in the occurrence of polycystic ovary syndrome (PCOS) as the most common cause of anovulatory infertility. The purpose of the current study was to investigate whether diminished activity of ovarian enzymes responsible for hydrogen sulfide (H2S) production, cystathionine β-synthase (CBS), and cystathionine γ-lyase (CSE) contributes to oxidative stress in PCOS. The study also explored whether administration of sodium hydrosulfide (NaSH), an H2S donor, could ameliorate PCOS symptoms by reducing oxidative stress. Methods The total eighteen rats were randomly assigned into three groups (n=6): control, PCOS, and PCOS+NaSH. PCOS was induced by intramuscular injection of estradiol valerate to induce PCOS in the PCOS and PCOS+NaSH groups. The PCOS+NaSH group received 30 μmol/L of NaSH in drinking water for 27 days after PCOS induction. Ovarian tissue samples were analyzed for oxidative stress indices including malondialdehyde (MDA) levels and superoxide dismutase (SOD) activity. Additional analyses measured H2S levels, CBS, and CSE activity. Results PCOS induction led to a significant decrease in SOD activity, H2S levels, and CBS and CSE activity, accompanied by a significant increase in MDA levels (p<0.0001). Furthermore, PCOS caused severe histological alterations in the ovaries. However, administration of NaSH effectively restored all measured parameters to pre-PCOS induction levels (p<0.0001). Conclusion This study showed that the decrease in the activity of H2S-producing enzymes and H2S levels may contribute to oxidative stress in PCOS. Therefore, administration of NaSH as a H2S donor can be considered as a potential therapeutic strategy for PCOS patients.
Collapse
Affiliation(s)
- Maryam Vaziripour
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdieh Faghihi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Ranjbaran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Asadi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Abdi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Kianian
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Hajiaqaei
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behjat Seifi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Beddows I, Fan H, Heinze K, Johnson BK, Leonova A, Senz J, Djirackor S, Cho KR, Pearce CL, Huntsman DG, Anglesio MS, Shen H. Cell State of Origin Impacts Development of Distinct Endometriosis-Related Ovarian Carcinoma Histotypes. Cancer Res 2024; 84:26-38. [PMID: 37874327 PMCID: PMC10758692 DOI: 10.1158/0008-5472.can-23-1362] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/01/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Clear cell ovarian carcinoma (CCOC) and endometrioid ovarian carcinoma (ENOC) are ovarian carcinoma histotypes, which are both thought to arise from ectopic endometrial (or endometrial-like) cells through an endometriosis intermediate. How the same cell type of origin gives rise to two morphologically and biologically different histotypes has been perplexing, particularly given that recurrent genetic mutations are common to both and present in nonmalignant precursors. We used RNA transcription analysis to show that the expression profiles of CCOC and ENOC resemble those of normal endometrium at secretory and proliferative phases of the menstrual cycle, respectively. DNA methylation at the promoter of the estrogen receptor (ER) gene (ESR1) was enriched in CCOC, which could potentially lock the cells in the secretory state. Compared with normal secretory-type endometrium, CCOC was further defined by increased expression of cysteine and glutathione synthesis pathway genes and downregulation of the iron antiporter, suggesting iron addiction and highlighting ferroptosis as a potential therapeutic target. Overall, these findings suggest that while CCOC and ENOC arise from the same cell type, these histotypes likely originate from different cell states. This "cell state of origin" model may help to explain the presence of histologic and molecular cancer subtypes arising in other organs. SIGNIFICANCE Two cancer histotypes diverge from a common cell of origin epigenetically locked in different cell states, highlighting the importance of considering cell state to better understand the cell of origin of cancer.
Collapse
Affiliation(s)
- Ian Beddows
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
| | - Huihui Fan
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
| | - Karolin Heinze
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Anna Leonova
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Janine Senz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Kathleen R. Cho
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Celeste Leigh Pearce
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - David G. Huntsman
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S. Anglesio
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hui Shen
- Department of Epigenetics, Van Andel Institute, Grand Rapids, Michigan
| |
Collapse
|
8
|
Naha S, Velmathi S. A fluorescence turn "on-off" imaging probe for sequential detection of Al 3+ and L-Cysteine in HeLa cells. Methods 2024; 221:27-34. [PMID: 38008345 DOI: 10.1016/j.ymeth.2023.11.009] [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: 09/20/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023] Open
Abstract
At this "Aluminum Age", exposure to aluminum (metallic or ionic form) is inevitable and inestimable. The presence of aluminum in biological systems is evident but more often aluminum toxicity is less understood. Therefore, the presence of biologically reactive aluminum needs to be identified and quantified. Alongside metals, L-cysteine, an essential amino acid, plays a pivotal role in the homeostasis of cellular oxidative and reductive stress. However, excess (<7g) could be lethal and can lead to death. Thus, in-situ selective detection of aluminum and L-cysteine is of larger interest. Here we report a fluorogenic probe (R) for the sequential selective detection and quantification of Al3+ and L-cysteine in a semi-aqueous medium (3:7; water: DMSO). The probe (R) was synthesized by a one-step acid-mediated condensation reaction between pyridine-3,4-diamine and 2-hydroxy-1-napthaldehyde. The synthesized probe was characterized using 1H and 13C NMR, and HR-Mass spectroscopic techniques. The probe (R) is non-emissive in nature, but on recognition of Al3+, the probe R showed "turn-on" emission (bright yellow colour) showing two emission maxima (522 nm and 547 nm), and no naked eye observable color change. Other competing cations do not show any noticeable fluorescence outcome. The R + Al3+ ensemble can specifically detect L-cysteine among all the essential amino acids by showing a fluorescence "turn-off" response. The sensing mechanism of Al3+ is obeying the chelation-enhanced fluorescence (CHEF) effect. The binding constant of R + Al3+ is 0.3 × 104 M-1. The limit of detection (LoD) for Al3+ and L-cysteine are 2.02 × 10-7 M and 0.5 × 10-5 M respectively. The probe (R) can show maximum efficiency within the pH range (7.0-10.0). The probe is found non-toxic (>80 % cell viability with 15 µM concentration) and employed for the in-vitro fluorescence imaging in the HeLa cell.
Collapse
Affiliation(s)
- Sanay Naha
- Organic and Polymer Synthesis Laboratory, National Institute of Technology, Tiruchirappalli, Tamilnadu 620015, India.
| | - Sivan Velmathi
- Organic and Polymer Synthesis Laboratory, National Institute of Technology, Tiruchirappalli, Tamilnadu 620015, India.
| |
Collapse
|
9
|
Ruan D, Wen J, Fang F, Lei Y, Zhao Z, Miao Y. Ferroptosis in epithelial ovarian cancer: a burgeoning target with extraordinary therapeutic potential. Cell Death Discov 2023; 9:434. [PMID: 38040696 PMCID: PMC10692128 DOI: 10.1038/s41420-023-01721-6] [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: 04/25/2023] [Revised: 10/15/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is universally acknowledged as a terrifying women killer for its high mortality. Recent research advances support that ferroptosis, an emerging iron-dependent type of regulated cell death (RCD) triggered by the excessive accumulation of lipid peroxides probably possesses extraordinary therapeutic potential in EOC therapy. Herein, we firstly provide a very concise introduction of ferroptosis. Special emphasis will be put on the ferroptosis's vital role in EOC, primarily covering its role in tumorigenesis and progression of EOC, the capability of reversing chemotherapy resistance, and the research and development of related therapeutic strategies. Furthermore, the construction of ferroptosis-related prognostic prediction systems, and mechanisms of ferroptosis resistance in EOC are also discussed. Finally, we propose and highlight several important yet unanswered problems and some future research directions in this field.
Collapse
Affiliation(s)
- Danhua Ruan
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, West China Campus, Sichuan University, Chengdu, 610041, Sichuan Province, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Jirui Wen
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Fei Fang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Yuqin Lei
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, West China Campus, Sichuan University, Chengdu, 610041, Sichuan Province, China
| | - Zhiwei Zhao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| | - Yali Miao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, West China Campus, Sichuan University, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
10
|
Dong Y, Wei J, Yang F, Qu Y, Huang J, Shi D. Nutrient-Based Approaches for Melanoma: Prevention and Therapeutic Insights. Nutrients 2023; 15:4483. [PMID: 37892558 PMCID: PMC10609833 DOI: 10.3390/nu15204483] [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: 09/22/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Melanoma, a prevalent and lethal form of skin cancer, remains a formidable challenge in terms of prevention and treatment. While significant progress has been made in understanding its pathogenesis and treatment, the quest for effective prevention strategies and therapeutic approaches remains ongoing. Considering the increased advancements in understanding the dynamic interplay between nutrients and melanoma, we aim to offer a refreshed perspective on nutrient-based approaches for melanoma prevention and adjunctive therapy. In contrast to other studies, we have innovatively provided a detailed exposition of the nutrients' influences on melanoma prognosis and treatment. This review firstly examines various nutrients, including antioxidants (namely vitamins A, D, C, and E; selenium; and caffeine), polyunsaturated fatty acids, and flavonoids, for their effects and underlying mechanisms in reducing melanoma risk. Among these nutrients, caffeine shows the most promising potential, as it is supported by multiple cohort studies for its protective effect against melanoma. In contrast, there is a certain degree of inconsistency in the research of other nutrients, possibly due to inherent differences between animal studies and epidemiological research, as well as variations in the definition of nutrient intake. To comprehensively investigate the impact of nutrients on melanoma progression and therapeutic approaches, the following sections will explore how nutrients influence immune responses and other physiological processes. While there is robust support from cell and animal studies regarding the immunomodulatory attributes of vitamins D and zinc, the anti-angiogenic potential of polyphenols, and the cell growth-inhibitory effects of flavonoids, the limited availability of human-based research substantially constrains their practical relevance in clinical contexts. As for utilizing nutrients in adjuvant melanoma treatments, multiple approaches have garnered clinical research support, including the utilization of vitamin D to decrease the postoperative recurrence rates among melanoma patients and the adoption of a high-fiber diet to enhance the effectiveness of immunotherapy. In general, the effects of most nutrients on reducing the risk of melanoma are not entirely clear. However, several nutrients, including vitamin D and dietary fiber, have demonstrated their potential to improve the melanoma prognosis and enhance the treatment outcomes, making them particularly deserving of clinical attention. A personalized and interdisciplinary approach, involving dermatologists, oncologists, nutritionists, and researchers, holds the promise of optimizing melanoma treatment strategies.
Collapse
Affiliation(s)
- Yucheng Dong
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Jiaxin Wei
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Fan Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Yang Qu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Jiuzuo Huang
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Di Shi
- Department of Emergency Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| |
Collapse
|
11
|
Liu M, Wu K, Wu Y. The emerging role of ferroptosis in female reproductive disorders. Biomed Pharmacother 2023; 166:115415. [PMID: 37660655 DOI: 10.1016/j.biopha.2023.115415] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023] Open
Abstract
Iron, as an essential trace element for the organism, is vital for maintaining the organism's health. Excessive iron can promote reactive oxygen species (ROS) accumulation, thus damaging cells and tissues. Ferroptosis is a novel form of programmed cell death distinguished by iron overload and lipid peroxidation, which is unique from autophagy, apoptosis and necrosis, more and more studies are focusing on ferroptosis. Recent evidence suggests that ferroptosis is associated with the development of female reproductive disorders (FRDs), including polycystic ovary syndrome (PCOS), premature ovarian insufficiency (POI), endometriosis (EMs), ovarian cancer (OC), preeclampsia (PE) and spontaneous abortion (SA). Pathways and genes associated with ferroptosis may participate in processes that regulate granulosa cell proliferation and secretion, oocyte development, ovarian reserve function, early embryonic development and placental oxidative stress. However, its exact mechanism has not been fully revealed. Therefore, our review systematically elaborates the occurrence mechanism of ferroptosis and its research progress in the development of FRDs, with a view to providing literature references for clinical targeting of ferroptosis -related pathways and regulatory factors for the management of FRDs.
Collapse
Affiliation(s)
- Min Liu
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; Department of Gynecology, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Keming Wu
- Department of Gynecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China; Department of Gynecology, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China.
| | - Yeke Wu
- Department of Stomatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610032, China.
| |
Collapse
|
12
|
Nguyen M, Le Mignon M, Schnellbächer A, Wehsling M, Braun J, Baumgaertner J, Grabner M, Zimmer A. Mechanistic insights into the biological activity of S-Sulfocysteine in CHO cells using a multi-omics approach. Front Bioeng Biotechnol 2023; 11:1230422. [PMID: 37680342 PMCID: PMC10482334 DOI: 10.3389/fbioe.2023.1230422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/31/2023] [Indexed: 09/09/2023] Open
Abstract
S-Sulfocysteine (SSC), a bioavailable L-cysteine derivative (Cys), is known to be taken up and metabolized in Chinese hamster ovary (CHO) cells used to produce novel therapeutic biological entities. To gain a deeper mechanistic insight into the SSC biological activity and metabolization, a multi-omics study was performed on industrially relevant CHO-K1 GS cells throughout a fed-batch process, including metabolomic and proteomic profiling combined with multivariate data and pathway analyses. Multi-layered data and enzymatical assays revealed an intracellular SSC/glutathione mixed disulfide formation and glutaredoxin-mediated reduction, releasing Cys and sulfur species. Increased Cys availability was directed towards glutathione and taurine synthesis, while other Cys catabolic pathways were likewise affected, indicating that cells strive to maintain Cys homeostasis and cellular functions.
Collapse
Affiliation(s)
- Melanie Nguyen
- Upstream R&D, Merck Life Science KGaA, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Darmstadt, Germany
| | | | | | - Maria Wehsling
- Upstream R&D, Merck Life Science KGaA, Darmstadt, Germany
| | - Julian Braun
- Upstream R&D, Merck Life Science KGaA, Darmstadt, Germany
| | - Jens Baumgaertner
- Biomolecule Analytics and Proteomics, Merck KGaA, Darmstadt, Germany
| | | | - Aline Zimmer
- Upstream R&D, Merck Life Science KGaA, Darmstadt, Germany
| |
Collapse
|
13
|
Kobayashi H, Yoshimoto C, Matsubara S, Shigetomi H, Imanaka S. A comprehensive overview of recent developments on the mechanisms and pathways of ferroptosis in cancer: the potential implications for therapeutic strategies in ovarian cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:547-566. [PMID: 37842240 PMCID: PMC10571061 DOI: 10.20517/cdr.2023.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/03/2023] [Accepted: 08/07/2023] [Indexed: 10/17/2023]
Abstract
Cancer cells adapt to environmental changes and alter their metabolic pathways to promote survival and proliferation. Metabolic reprogramming not only allows tumor cells to maintain a reduction-oxidation balance by rewiring resources for survival, but also causes nutrient addiction or metabolic vulnerability. Ferroptosis is a form of regulated cell death characterized by the iron-dependent accumulation of lipid peroxides. Excess iron in ovarian cancer amplifies free oxidative radicals and drives the Fenton reaction, thereby inducing ferroptosis. However, ovarian cancer is characterized by ferroptosis resistance. Therefore, the induction of ferroptosis is an exciting new targeted therapy for ovarian cancer. In this review, potential metabolic pathways targeting ferroptosis were summarized to promote anticancer effects, and current knowledge and future perspectives on ferroptosis for ovarian cancer therapy were discussed. Two therapeutic strategies were highlighted in this review: directly inducing the ferroptosis pathway and targeting metabolic vulnerabilities that affect ferroptosis. The overexpression of SLC7A11, a cystine/glutamate antiporter SLC7A11 (also known as xCT), is involved in the suppression of ferroptosis. xCT inhibition by ferroptosis inducers (e.g., erastin) can promote cell death when carbon as an energy source of glucose, glutamine, or fatty acids is abundant. On the contrary, xCT regulation has been reported to be highly dependent on the metabolic vulnerability. Drugs that target intrinsic metabolic vulnerabilities (e.g., GLUT1 inhibitors, PDK4 inhibitors, or glutaminase inhibitors) predispose cancer cells to death, which is triggered by decreased nicotinamide adenine dinucleotide phosphate generation or increased reactive oxygen species accumulation. Therefore, therapeutic approaches that either directly inhibit the xCT pathway or target metabolic vulnerabilities may be effective in overcoming ferroptosis resistance. Real-time monitoring of changes in metabolic pathways may aid in selecting personalized treatment modalities. Despite the rapid development of ferroptosis-inducing agents, therapeutic strategies targeting metabolic vulnerability remain in their infancy. Thus, further studies must be conducted to comprehensively understand the precise mechanism linking metabolic rewiring with ferroptosis.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara 634-0813, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara 634-8522, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara 634-8522, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, Nara 630-8581, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara 634-8522, Japan
- Department of Medicine, Kei Oushin Clinic, Nishinomiya 663-8184, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara 634-8522, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, Nara 634-0001, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara 634-0813, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara 634-8522, Japan
| |
Collapse
|
14
|
Cardona CJ, Montgomery MR. Iron regulatory proteins: players or pawns in ferroptosis and cancer? Front Mol Biosci 2023; 10:1229710. [PMID: 37457833 PMCID: PMC10340119 DOI: 10.3389/fmolb.2023.1229710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
Cells require iron for essential functions like energy production and signaling. However, iron can also engage in free radical formation and promote cell proliferation thereby contributing to both tumor initiation and growth. Thus, the amount of iron within the body and in individual cells is tightly regulated. At the cellular level, iron homeostasis is maintained post-transcriptionally by iron regulatory proteins (IRPs). Ferroptosis is an iron-dependent form of programmed cell death with vast chemotherapeutic potential, yet while IRP-dependent targets have established roles in ferroptosis, our understanding of the contributions of IRPs themselves is still in its infancy. In this review, we present the growing circumstantial evidence suggesting that IRPs play critical roles in the adaptive response to ferroptosis and ferroptotic cell death and describe how this knowledge can be leveraged to target neoplastic iron dysregulation more effectively.
Collapse
|
15
|
Wang CK, Chen TJ, Tan GY, Chang FP, Sridharan S, Yu CHA, Chang YH, Chen YJ, Cheng LT, Hwang-Verslues WW. MEX3A Mediates p53 Degradation to Suppress Ferroptosis and Facilitate Ovarian Cancer Tumorigenesis. Cancer Res 2023; 83:251-263. [PMID: 36354374 PMCID: PMC9845988 DOI: 10.1158/0008-5472.can-22-1159] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 08/24/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022]
Abstract
Epithelial ovarian cancer is a highly heterogeneous and malignant female cancer with an overall low survival rate. Mutations in p53 are prevalent in the major ovarian cancer histotype, high-grade serous ovarian carcinoma (HGSOC), while p53 mutations are much less frequent in other ovarian cancer subtypes, particularly in ovarian clear cell carcinoma (OCCC). Advanced stage OCCC with wild-type (WT) p53 has a worse prognosis and increased drug resistance, metastasis, and recurrence than HGSOC. The mechanisms responsible for driving the aggressiveness of WT p53-expressing ovarian cancer remain poorly understood. Here, we found that upregulation of MEX3A, a dual-function protein containing a RING finger domain and an RNA-binding domain, was critical for tumorigenesis in WT p53-expressing ovarian cancer. MEX3A overexpression enhanced the growth and clonogenicity of OCCC cell lines. In contrast, depletion of MEX3A in OCCC cells, as well as ovarian teratocarcinoma cells, reduced cell survival and proliferative ability. MEX3A depletion also inhibited tumor growth and prolonged survival in orthotopic xenograft models. MEX3A depletion did not alter p53 mRNA level but did increase p53 protein stability. MEX3A-mediated p53 protein degradation was crucial to suppress ferroptosis and enhance tumorigenesis. Consistently, p53 knockdown reversed the effects of MEX3A depletion. Together, our observations identified MEX3A as an important oncogenic factor promoting tumorigenesis in ovarian cancer cells expressing WT p53. SIGNIFICANCE Degradation of p53 mediated by MEX3A drives ovarian cancer growth by circumventing p53 tumor suppressive functions, suggesting targeting MEX3A as a potential strategy for treating of ovarian cancer expressing WT p53.
Collapse
Affiliation(s)
- Cheng-Kai Wang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Tzu-Jou Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Grace Y.T. Tan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Fang-Pei Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | - Yen-Hou Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Jen Chen
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Li-Tzu Cheng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wendy W. Hwang-Verslues
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Corresponding Author: Wendy W. Hwang-Verslues, Genomics Research Center, Academia Sinica, No. 128, Sec. 2, Academia Road, Taipei 115, Taiwan. Phone: +886-2-2787-1246; Fax: +886-2-2789-9924; E-mail:
| |
Collapse
|
16
|
Tan Z, Huang H, Sun W, Li Y, Jia Y. Current progress of ferroptosis study in ovarian cancer. Front Mol Biosci 2022; 9:966007. [PMID: 36090052 PMCID: PMC9458863 DOI: 10.3389/fmolb.2022.966007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Tumors are the leading cause of death all over the world, among which ovarian cancer ranks the third in gynecological malignancies. The current treatment for ovarian cancer is liable to develop chemotherapy resistance and high recurrence rate, in which a new strategy is demanded. Ferroptosis, a newly discovered manner of regulatory cell death, is shown to be induced by massive iron-dependent accumulation of lipid reactive oxygen species. With the in-depth study of ferroptosis, its associated mechanism with various tumors is gradually elucidated, including ovarian tumor, which probably promotes the application of ferroptosis in treating ovarian cancer. To this end, this review will focus on the history and current research progress of ferroptosis, especially its regulation mechanism, and its potential application as a novel treatment strategy for ovarian cancer.
Collapse
|
17
|
Aragoneses-Cazorla G, Buendia-Nacarino MP, Mena ML, Luque-Garcia JL. A Multi-Omics Approach to Evaluate the Toxicity Mechanisms Associated with Silver Nanoparticles Exposure. NANOMATERIALS 2022; 12:nano12101762. [PMID: 35630985 PMCID: PMC9146515 DOI: 10.3390/nano12101762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022]
Abstract
Silver nanoparticles (AgNPs) are currently used in many different industrial, commercial and health fields, mainly due to their antibacterial properties. Due to this widespread use, humans and the environment are increasingly exposed to these types of nanoparticles, which is the reason why the evaluation of the potential toxicity associated with AgNPs is of great importance. Although some of the toxic effects induced by AgNPs have already been shown, the elucidation of more complete mechanisms is yet to be achieved. In this sense, and since the integration of metabolomics and transcriptomics approaches constitutes a very useful strategy, in the present study targeted and untargeted metabolomics and DNA microarrays assays have been combined to evaluate the molecular mechanisms involved in the toxicity induced by 10 nm AgNPs. The results have shown that AgNPs induce the synthesis of glutathione as a cellular defense mechanism to face the oxidative environment, while inducing the depletion of relevant molecules implicated in the synthesis of important antioxidants. In addition, it has been observed that AgNPs completely impair the intracellular energetic metabolism, especially affecting the production of adenosine triphosphate (ATP) and disrupting the tricarboxylic acids cycle. It has been demonstrated that AgNPs exposure also affects the glycolysis pathway. The effect on such pathway differs depending on the step of the cycle, which a significant increase in the levels of glucose as way to counterbalance the depleted levels of ATP.
Collapse
|
18
|
Moving beyond the Tip of the Iceberg: DJ-1 Implications in Cancer Metabolism. Cells 2022; 11:cells11091432. [PMID: 35563738 PMCID: PMC9103122 DOI: 10.3390/cells11091432] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
DJ-1, also called Parkinson’s protein 7 (PARK7), is ubiquitously expressed and plays multiple actions in different physiological and, especially, pathophysiological processes, as evidenced by its identification in neurodegenerative diseases and its high expression in different types of cancer. To date, the exact activity of DJ-1 in carcinogenesis has not been fully elucidated, however several recent studies disclosed its involvement in regulating fundamental pathways involved in cancer onset, development, and metastatization. At this purpose, we have dissected the role of DJ-1 in maintaining the transformed phenotype, survival, drug resistance, metastasis formation, and differentiation in cancer cells. Moreover, we have discussed the role of DJ-1 in controlling the redox status in cancer cells, along with the ability to attenuate reactive oxygen species (ROS)-dependent cell death, as well as to mediate ferropotosis. Finally, a mention to the development of therapeutic strategies targeting DJ-1 has been done. We have reported the most recent studies, aiming to shed light on the role played by DJ-1 in different cancer aspects and create the foundation for moving beyond the tip of the iceberg.
Collapse
|
19
|
Gu I, Gregory E, Atwood C, Lee SO, Song YH. Exploring the Role of Metabolites in Cancer and the Associated Nerve Crosstalk. Nutrients 2022; 14:nu14091722. [PMID: 35565690 PMCID: PMC9103817 DOI: 10.3390/nu14091722] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 02/05/2023] Open
Abstract
Since Otto Warburg's first report on the increased uptake of glucose and lactate release by cancer cells, dysregulated metabolism has been acknowledged as a hallmark of cancer that promotes proliferation and metastasis. Over the last century, studies have shown that cancer metabolism is complex, and by-products of glucose and glutamine catabolism induce a cascade of both pro- and antitumorigenic processes. Some vitamins, which have traditionally been praised for preventing and inhibiting the proliferation of cancer cells, have also been proven to cause cancer progression in a dose-dependent manner. Importantly, recent findings have shown that the nervous system is a key player in tumor growth and metastasis via perineural invasion and tumor innervation. However, the link between cancer-nerve crosstalk and tumor metabolism remains unclear. Here, we discuss the roles of relatively underappreciated metabolites in cancer-nerve crosstalk, including lactate, vitamins, and amino acids, and propose the investigation of nutrients in cancer-nerve crosstalk based on their tumorigenicity and neuroregulatory capabilities. Continued research into the metabolic regulation of cancer-nerve crosstalk will provide a more comprehensive understanding of tumor mechanisms and may lead to the identification of potential targets for future cancer therapies.
Collapse
Affiliation(s)
- Inah Gu
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72704, USA
| | - Emory Gregory
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Casey Atwood
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72704, USA
| | - Sun-Ok Lee
- Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72704, USA
| | - Young Hye Song
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
20
|
Kung WM, Lin CC, Chen WJ, Jiang LL, Sun YY, Hsieh KH, Lin MS. Anti-Inflammatory CDGSH Iron-Sulfur Domain 2: A Biomarker of Central Nervous System Insult in Cellular, Animal Models and Patients. Biomedicines 2022; 10:biomedicines10040777. [PMID: 35453528 PMCID: PMC9030396 DOI: 10.3390/biomedicines10040777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
Spinal cord injury (SCI) promotes brain inflammation; conversely, brain injury promotes spinal neuron loss. There is a need to identify molecular biomarkers and therapeutic targets for central nervous system (CNS) injury. CDGSH iron-sulfur structural domain 2 (CISD2), an NF-κB antagonist, is downregulated after injury in vivo and in vitro. We aimed to examine the diagnostic value of CISD2 in patients with CNS insult. Plasma and cerebrospinal fluid (CSF) CISD2 levels were decreased in 13 patients with CNS insult and were negatively correlated with plasma IL6 levels (associated with disease severity; r = −0.7062; p < 0.01). SCI-induced inflammatory mediators delivered through CSF promoted mouse brain inflammation at 1 h post-SCI. Anti-CISD2 antibody treatment exacerbated SCI-induced inflammation in mouse spine and brain. Lipopolysaccharide-stimulated siCISD2-transfected EOC microglial cells exhibited proinflammatory phenotypes (enhanced M1 polarization, decreased M2 polarization, and increased intranuclear NF-κB p65 translocation). Plasma and CSF CISD2 levels were increased in three patients with CNS insult post-therapeutic hypothermia. CISD2 levels were negatively correlated with plasma and CSF levels of inflammatory mediators. CISD2 inhibition and potentiation experiments in cells, animals, and humans revealed CISD2 as a biomarker for CNS insult and upregulation of CISD2 anti-inflammatory properties as a potential therapeutic strategy for CNS insult.
Collapse
Affiliation(s)
- Woon-Man Kung
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan;
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei 11114, Taiwan
| | - Chai-Ching Lin
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (C.-C.L.); (W.-J.C.); (L.-L.J.)
| | - Wei-Jung Chen
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (C.-C.L.); (W.-J.C.); (L.-L.J.)
| | - Li-Lin Jiang
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (C.-C.L.); (W.-J.C.); (L.-L.J.)
| | - Yu-Yo Sun
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan;
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA 22903, USA
| | - Kuang-Hui Hsieh
- Department of Laboratory Service, Kuang Tien General Hospital, Taichung 43303, Taiwan;
| | - Muh-Shi Lin
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan; (C.-C.L.); (W.-J.C.); (L.-L.J.)
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung 43303, Taiwan
- Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Correspondence:
| |
Collapse
|
21
|
Yue Y, Zhao T, Ma K, Huo F, Yin C. Endogenous cysteine fluorescence monitoring and its deployment in tumour demarcation. Chem Commun (Camb) 2022; 58:2311-2314. [PMID: 35076042 DOI: 10.1039/d1cc06765f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A cysteine-specific fluorescent probe with a wide concentration detection range was used to monitor changes in cysteine levels in HeLa cells under stress and to demarcate the boundary of a xenograft tumour.
Collapse
Affiliation(s)
- Yongkang Yue
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China.
| | - Tingting Zhao
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China.
| | - Kaiqing Ma
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China.
| | - Fangjun Huo
- Research Institute of Applied Chemistry, Shanxi University, Taiyuan 030006, China
| | - Caixia Yin
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
22
|
Guan Q, Zhou LL, Zhou W, Dong YB. A vinyl-decorated covalent organic framework for ferroptotic cancer therapy via visible-light-triggered cysteine depletion. J Mater Chem B 2022; 10:8894-8909. [DOI: 10.1039/d2tb01815b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Intracellular cysteine depletion induced by a COF-based click photoreaction achieves effective cancer therapy by ferroptosis.
Collapse
Affiliation(s)
- Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| | - Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| | - Wei Zhou
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
23
|
Li Z, Zhou S, Yang X, Li X, Yang GX, Chant J, Snyder M, Wang X. Broad Anti-Cancer Activity Produced by Targeted Nutrients Deprivation (TND) of Multiple Non-Essential Amino Acids. Nutr Cancer 2021; 74:2607-2621. [PMID: 34905997 DOI: 10.1080/01635581.2021.2013904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
It has been known for close to 100 years that the metabolism of cancer cells is altered and different than that of healthy cells in the body. On that basis, we have developed an entirely novel approach to managing cancer, termed Targeted Nutrients Deprivation (TND). TND employs a formulated diet depleted of multiple non-essential amino acids (NEAAs) that are required by tumor cells but not by normal cells. Cancer cells specifically require those NEAAs due to their heightened and rewired metabolism. We demonstrated that our first proprietary formulated TND diet-FTN203-significantly reduced the growth of multiple human tumor xenografts in mouse. In combination with chemotherapy and immunotherapy, FTN203 further enhanced therapeutic efficacy. Reliance on FTN203 as the sole nutrition source was shown to be safe without causing detrimental body-weight loss or internal organ damage. Our findings indicate that TND is a novel and safe approach to managing cancer.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.2013904 .
Collapse
Affiliation(s)
- Zehui Li
- Filtricine, Inc, Santa Clara, California, USA
| | - Shuang Zhou
- Filtricine, Inc, Santa Clara, California, USA
| | | | - Xiyan Li
- Filtricine, Inc, Santa Clara, California, USA
| | | | - John Chant
- Filtricine, Inc, Santa Clara, California, USA
| | | | - Xin Wang
- Filtricine, Inc, Santa Clara, California, USA
| |
Collapse
|
24
|
Wilder CS, Chen Z, DiGiovanni J. Pharmacologic approaches to amino acid depletion for cancer therapy. Mol Carcinog 2021; 61:127-152. [PMID: 34534385 DOI: 10.1002/mc.23349] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 11/09/2022]
Abstract
Cancer cells undergo metabolic reprogramming to support increased demands in bioenergetics and biosynthesis and to maintain reactive oxygen species at optimum levels. As metabolic alterations are broadly observed across many cancer types, metabolic reprogramming is considered a hallmark of cancer. A metabolic alteration commonly seen in cancer cells is an increased demand for certain amino acids. Amino acids are involved in a wide range of cellular functions, including proliferation, redox balance, bioenergetic and biosynthesis support, and homeostatic functions. Thus, targeting amino acid dependency in cancer is an attractive strategy for a number of cancers. In particular, pharmacologically mediated amino acid depletion has been evaluated as a cancer treatment option for several cancers. Amino acids that have been investigated for the feasibility of drug-induced depletion in preclinical and clinical studies for cancer treatment include arginine, asparagine, cysteine, glutamine, lysine, and methionine. In this review, we will summarize the status of current research on pharmacologically mediated amino acid depletion as a strategy for cancer treatment and potential chemotherapeutic combinations that synergize with amino acid depletion to further inhibit tumor growth and progression.
Collapse
Affiliation(s)
- Carly S Wilder
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Zhao Chen
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - John DiGiovanni
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA.,Center for Molecular Carcinogenesis and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
25
|
Li L, Qiu C, Hou M, Wang X, Huang C, Zou J, Liu T, Qu J. Ferroptosis in Ovarian Cancer: A Novel Therapeutic Strategy. Front Oncol 2021; 11:665945. [PMID: 33996593 PMCID: PMC8117419 DOI: 10.3389/fonc.2021.665945] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer (OVCA) is one of the most lethal malignancies with a five-year relative survival below 50% by virtue of its high recurrence rate and inadequate early detection methods. For OVCA patients, modern approaches include debulking surgery, chemotherapies, angiogenesis inhibitors, poly ADP-ribose polymerase (PARP) inhibitors, and immunotherapies depending on the histological type and staging of the tumor. However, in most cases, simple standard treatment is not satisfactory. Thus, a more effective way of treatment is needed. Ferroptosis is a newly recognized type of regulated cell death marked by lipid peroxidation, iron accumulation and glutathione deprivation, having a connection with a variety of disorders and showing great potential in anti-tumor therapy. Intriguingly, a possible connection between ferroptosis and OVCA is shown on the basis of previously published findings. Furthermore, a growing number of ferroptosis protection pathways have been identified during the past few years with increasing ferroptosis regulators being discovered. In this review, we summarized several major pathways involved in ferroptosis and the study foundation of ferroptosis and ovarian cancer, hoping to provide clues regarding OVCA treatment. And some important issues were also raised to point out future research directions.
Collapse
Affiliation(s)
- Lanyu Li
- Department of Obstetrics and Gynecology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Qiu
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Hou
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinyu Wang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changzhen Huang
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jialin Zou
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianyi Liu
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jinfeng Qu
- Department of Obstetrics and Gynecology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
26
|
The Multifaceted Regulation of Mitochondria in Ferroptosis. Life (Basel) 2021; 11:life11030222. [PMID: 33801920 PMCID: PMC8001967 DOI: 10.3390/life11030222] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/05/2021] [Indexed: 01/03/2023] Open
Abstract
Ferroptosis is characterized as a novel form of regulated cell death, which is initiated by the lethal accumulation of lipid peroxidation catalyzed by cellular labile free iron. This iron driven cell death sharply differs from other well characterized forms of regulated cell death at morphological, genetic and biochemical levels. Increasing research has elaborated a high relevance between dysregulated ferroptosis and the pathogenesis of degenerative diseases and organs injury in human patients. Additionally, targeted induction of ferroptosis is considered as a potentially therapeutic design for the clinical intervention of other therapy-resistant cancers. It is well understood that mitochondria, the cellular powerhouse, determine several types of regulated cell death. Recently, compromised mitochondrial morphology and functionalities have been primarily formulated in ferroptosis. Several mitochondria associated proteins and metabolic processes have been elaborated to fine-tune ferroptotic program. Herein, we critically review the recent advances in this booming field, with focus on summarizing the multifaceted mitochondrial regulation of ferroptosis and providing a perspective on the potential biochemical basis. Finally, we are attempting to shed light on an integrative view on the possibility of mitochondria- and ferroptosis-targeting therapeutics as novel treatment designs for the intervention of ferroptosis related diseases.
Collapse
|
27
|
Soula M, Weber RA, Zilka O, Alwaseem H, La K, Yen F, Molina H, Garcia-Bermudez J, Pratt DA, Birsoy K. Metabolic determinants of cancer cell sensitivity to canonical ferroptosis inducers. Nat Chem Biol 2020; 16:1351-1360. [PMID: 32778843 PMCID: PMC8299533 DOI: 10.1038/s41589-020-0613-y] [Citation(s) in RCA: 428] [Impact Index Per Article: 85.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
Cancer cells rewire their metabolism and rely on endogenous antioxidants to mitigate lethal oxidative damage to lipids. However, the metabolic processes that modulate the response to lipid peroxidation are poorly defined. Using genetic screens, we compared metabolic genes essential for proliferation upon inhibition of cystine uptake or glutathione peroxidase-4 (GPX4). Interestingly, very few genes were commonly required under both conditions, suggesting that cystine limitation and GPX4 inhibition may impair proliferation via distinct mechanisms. Our screens also identify tetrahydrobiopterin (BH4) biosynthesis as an essential metabolic pathway upon GPX4 inhibition. Mechanistically, BH4 is a potent radical-trapping antioxidant that protects lipid membranes from autoxidation, alone and in synergy with vitamin E. Dihydrofolate reductase catalyzes the regeneration of BH4, and its inhibition by methotrexate synergizes with GPX4 inhibition. Altogether, our work identifies the mechanism by which BH4 acts as an endogenous antioxidant and provides a compendium of metabolic modifiers of lipid peroxidation.
Collapse
Affiliation(s)
- Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Ross A Weber
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Omkar Zilka
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Hanan Alwaseem
- The Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Konnor La
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Frederick Yen
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Henrik Molina
- The Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Javier Garcia-Bermudez
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA.
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada.
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
28
|
Li Y, Xia J, Shao F, Zhou Y, Yu J, Wu H, Du J, Ren X. Sorafenib induces mitochondrial dysfunction and exhibits synergistic effect with cysteine depletion by promoting HCC cells ferroptosis. Biochem Biophys Res Commun 2020; 534:877-884. [PMID: 33162029 DOI: 10.1016/j.bbrc.2020.10.083] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant cancers worldwide. The prognosis of HCC remains poor. Currently, sorafenib is the first-line drug for advanced HCC. Although sorafenib's mechanism of action involving several established cancer-related protein kinase targets is well-characterized, the underlying molecular mechanism is still unclear. Here, we found that sorafenib inhibited viability, proliferation, and migration of HCC cells in a dose-dependent manner. Sorafenib treatment of HCC cells destroyed mitochondrial morphology, accompanied by decreased activity of oxidative phosphorylation, collapse of mitochondrial membrane potential, and reduced synthesis of ATP, with consequent cell death due to ferroptosis. Pharmacological utilization of glutathione (GSH) rescued the sorafenib-induced ferroptosis, eliminated the accumulation of cellular mitochondrial reactive oxygen species (ROS), and lipid peroxide. GSH depletion through cysteine deprivation or cysteinase inhibition exacerbated sorafenib-induced ferroptotic cell death and lipid peroxides generation, and enhanced oxidative stress and mitochondrial ROS accumulation. Collectively, these findings indicate that depletion of cysteine acts synergistically with sorafenib and renders HCC cells vulnerable to ferroptosis, presenting the potential value of new therapeutic combinations for advanced HCC.
Collapse
Affiliation(s)
- Yanchun Li
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Jun Xia
- Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Fangchun Shao
- Department of Respiratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yan Zhou
- Department of Laboratory Medicine, Guangxin District People's Hospital, Shangrao, Jiangxi, 334100, China
| | - Jiaqi Yu
- Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Hengyu Wu
- Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jing Du
- Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Xueying Ren
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|