1
|
Li Y, Xu H, Ma Z, Li Q, Xiong Y, Xiong X, Li J, Lan D, Fu W. Comprehensive cognition of yak ( Bos grunniens) AIFM2 gene and its anti-ferroptosis role in bisphenol A-induced fetal fibroblast model. Anim Biotechnol 2024; 35:2377209. [PMID: 39037081 DOI: 10.1080/10495398.2024.2377209] [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] [Indexed: 07/23/2024]
Abstract
Apoptosis-inducing factor mitochondrion-associated 2 (AIFM2) has been identified as a gene with anti-ferroptosis properties. To explore whether AIFM2 exerts anti-ferroptosis role in yaks (Bos grunniens), we cloned yak AIFM2 gene and analyzed its biological characteristics. The coding region of AIFM2 had 1122 bp and encoded 373 amino acids, which was conserved in mammals. Next, RT-qPCR results showed an extensive expression of AIMF2 in yak tissues. Furthermore, we isolated yak skin fibroblasts (YSFs) and established a bisphenol A (BPA)-induced ferroptosis model to further investigate the role of AIFM2. BPA elevated oxidative stress (reactive oxygen species, ROS) and lipid peroxidation (malondialdehyde, MDA and BODIPY), and reduced cell viability and antioxidant capacity (glutathione, GSH), with the severity depending on the dosage. Of note, a supplement of Ferrostatin-1 (Fer), an inhibitor of ferroptosis, restored the previously mentioned indicators. Subsequently, we constructed an AIFM2 overexpression vector and designed AIFM2 specific interfering siRNAs, which were transfected into YSFs. The results showed that overexpressing AIFM2 alleviated ferroptosis, characterizing by significant changes of cell viability, ROS, BODIPY, MDA and GSH. Meanwhile, interfering AIFM2 aggravated ferroptosis, demonstrating the critical anti-ferroptosis role of the yak AIFM2 gene. This study shed light on further exploring the molecular mechanism of AIFM2 in plateau adaptability.
Collapse
Affiliation(s)
- Yueyue Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Hongmei Xu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Zifeng Ma
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Qiao Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Xianrong Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Jian Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Daoliang Lan
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Wei Fu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Ministry of Education, Southwest Minzu University, Chengdu, China
- Key Laboratory of Animal Science of National Ethnic Affairs Commission of China, Southwest Minzu University, Chengdu, China
- Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization Key Laboratory of Sichuan Province, Southwest Minzu University, Chengdu, China
| |
Collapse
|
2
|
Wu C, Bao S, Sun H, Chen X, Yang L, Li R, Peng Y. Noncoding RNAs regulating ferroptosis in cardiovascular diseases: novel roles and therapeutic strategies. Mol Cell Biochem 2024; 479:2827-2841. [PMID: 38064139 PMCID: PMC11473578 DOI: 10.1007/s11010-023-04895-w] [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: 03/30/2023] [Accepted: 11/06/2023] [Indexed: 10/15/2024]
Abstract
The morbidity and mortality rates of cardiovascular diseases (CVDs) are increasing; thus, they impose substantial health and economic burdens worldwide, and effective interventions are needed for immediate resolution of this issue. Recent studies have suggested that noncoding RNAs (ncRNAs) play critical roles in the occurrence and development of CVDs and are potential therapeutic targets and novel biomarkers for these diseases. Newly discovered modes of cell death, including necroptosis, pyroptosis, apoptosis, autophagy-dependent cell death and ferroptosis, also play key roles in CVD progression. However, ferroptosis, which differs from the other aforementioned forms of regulated cell death in terms of cell morphology, biochemistry and inhereditability, is a unique iron-dependent mode of nonapoptotic cell death induced by abnormal iron metabolism and excessive accumulation of iron-dependent lipid peroxides and reactive oxygen species (ROS). Increasing evidence has confirmed that ncRNA-mediated ferroptosis is involved in regulating tissue homeostasis and CVD-related pathophysiological conditions, such as cardiac ischemia/reperfusion (I/R) injury, myocardial infarction (MI), atrial fibrillation (AF), cardiomyopathy and heart failure (HF). In this review, we summarize the underlying mechanism of ferroptosis, discuss the pathophysiological effects of ncRNA-mediated ferroptosis in CVDs and provide ideas for effective therapeutic strategies.
Collapse
Affiliation(s)
- Changyong Wu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Suli Bao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Huang Sun
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaocui Chen
- Department of Gastroenterology, Affiliated Hospital of Panzhihua University, Panzhihua, China
| | - Lu Yang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruijie Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Yunzhu Peng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Oh SJ, Shin YY, Ahn JS, Park HJ, Kang MJ, Shin TH, Lee BC, Kim WK, Oh JM, Lee D, Kim YH, Kim JM, Sung ES, Lee EW, Jeong JH, Lee BJ, Seo Y, Kim HS. TGFβ2-Driven Ferritin Degradation and Subsequent Ferroptosis Underlie Salivary Gland Dysfunction in Postmenopausal Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2400660. [PMID: 39481440 DOI: 10.1002/advs.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/05/2024] [Indexed: 11/02/2024]
Abstract
Despite the high incidence of dry mouth in postmenopausal women, its underlying mechanisms and therapeutic interventions remain underexplored. Using ovariectomized (OVX) mouse models, here this study identifies ferroptosis, an iron-dependent regulated cell death, as a central mechanism driving postmenopausal salivary gland (SG) dysfunction. In the OVX-SGs, TGFβ signaling pathway is enhanced with the aberrant TGFβ2 expression in SG mesenchymal cells. Intriguingly, TGFβ2 treatment reduces iron-storing ferritin levels, leading to lipid peroxidation and ferroptotic death in SG epithelial organoids (SGOs). Mechanistically, TGFβ2 promotes the autophagy-mediated ferritin degradation, so-called ferritinophagy. A notable overexpression of the type III TGFβ receptor (TβRIII) is found in the OVX-SGs and TGFβ2-treated SGOs, while the silencing of TβRIII mitigates the ferroptosis-mediated deleterious effects of TGFβ2 on SGOs. Finally, administration of ferroptosis inhibitor, Liproxstatin-1 (Lip-1), improves saliva secretion in OVX mice. Present findings collectively suggest a link between TGFβ signaling, ferroptosis, and SG injury, offering new therapeutic avenues for postmenopausal xerostomia.
Collapse
Affiliation(s)
- Su-Jeong Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ye Young Shin
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Seoul, 08590, Republic of Korea
| | - Ji-Su Ahn
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hee-Jeong Park
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Min-Jung Kang
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Tae-Hoon Shin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju-si, 63243, Republic of Korea
| | - Byung-Chul Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Won Kyu Kim
- Natural Product Research Center, Korea Institute of Science andTechnology (KIST), Gangneung, 25451, Republic of Korea
- Department of Convergence Medicine, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
- Division of Natural Products Applied Science, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jung-Min Oh
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Ji Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Republic of Korea
| | - Eui-Suk Sung
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Pusan National University School of Medicine, Yangsan Pusan National University Hospital, Yangsan, 50612, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Byung-Joo Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Republic of Korea
| | - Yoojin Seo
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hyung-Sik Kim
- Department of Oral Biochemistry, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
- Education and Research Team for Life Science on Dentistry, Pusan National University, Yangsan, 50612, Republic of Korea
| |
Collapse
|
4
|
Zhang Y, Zhang C, Dai Q, Ma R. Continuous Theta Burst Stimulation Inhibits Oxidative Stress-Induced Inflammation and Autophagy in Hippocampal Neurons by Activating Glutathione Synthesis Pathway, Improving Cognitive Impairment in Sleep-Deprived Mice. Neuromolecular Med 2024; 26:40. [PMID: 39388015 DOI: 10.1007/s12017-024-08807-z] [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: 05/08/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024]
Abstract
Sleep deprivation (SD) has been reported to have a negative impact on cognitive function. Continuous theta burst stimulation (cTBS) shows certain effects in improving sleep and neurological diseases, and its molecular or cellular role in SD-induced cognition impairment still need further exploration. In this study, C57BL/6 mice were subjected to 48 h of SD and cTBS treatment, and cTBS treatment significantly improved SD-triggered impairment of spatial learning and memory abilities in mice. Additionally, cTBS reduced malondialdehyde levels, increased superoxide dismutase activities, and inhibited the production of inflammatory cytokines, alleviating oxidative stress and inflammation levels in hippocampal tissues of SD model mice. cTBS decreased LC3II/LC3I ratio, Beclin1 protein levels, and LC3B puncta intensity, and elevated p62 protein levels to suppress excessive autophagy in hippocampal tissues of SD-stimulated mice. Then, we proved that inhibiting oxidative stress alleviated inflammation, autophagy, and death of hippocampal neuron cells through an in vitro cellular model for oxidative stress, and cTBS treatment promoted the production of glutathione (GSH), the nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and the mRNA expression of GSH synthesis-related genes to enhance antioxidant capacity in hippocampal tissues of SD mice. An Nrf2 inhibitor ML385 or a GSH synthesis inhibitor BSO reversed the alleviating effects of cTBS treatment on oxidative stress-associated damage of hippocampal tissues and cognitive impairment in SD model mice. Altogether, our study demonstrated that cTBS mitigates oxidative stress-associated inflammation and autophagy through activating the Nrf2-mediated GSH synthesis pathway, improving cognitive impairment in SD mice.
Collapse
Affiliation(s)
- Yi Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Cheng Zhang
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China
| | - Qing Dai
- Anesthesiology Department, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Rui Ma
- Clinical Psychology Department, the People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, 830001, China.
| |
Collapse
|
5
|
Xiao H, Chen C, Yuan X, Yang L, Zheng Y, Yuan J, Huang S, Liang J, Yuan S, Li M, Wang J. Gingerenone A induces ferroptosis in colorectal cancer via targeting suppression of SLC7A11 signaling pathway. Biomed Pharmacother 2024; 180:117529. [PMID: 39393329 DOI: 10.1016/j.biopha.2024.117529] [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: 08/06/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common and fatal diseases, yet effective therapeutic drugs are lacking in clinical settings. Gingerenone A (GA) is an active compound derived from ginger, has demonstrated anti-tumor properties. However, the efficacy of GA against CRC and its primary mechanism of action remain unclear. MATERIALS AND METHODS MTT assay and colony formation assay were employed to evaluate cell viability. Transwell assays were utilized to assess the migratory and invasive capabilities of the cells. The effects of GA on ferroptosis related proteins were analyzed using Western blot. Levels of glutathione (GSH), malondialdehyde (MDA), Fe2+, and 4-hydroxynonenal (4-HNE) levels were measured with a biochemical index determination kit. Cellular reactive oxygen species (ROS) were quantified using flow cytometry. CETSA, pull-down, and co-immunoprecipitation (Co-IP) assays confirmed the interactions between GA and SLC7A11, as well as the ubiquitination promoted by SLC7A11. A xenograft mouse model was employed to validate the anticancer effect of GA in vivo. RESULTS We observed that GA significantly suppressed proliferation in human CRC cells. Additionally, GA treatment inhibited the migration, invasion, and colony formation of CRC cells. Subsequently, through the use of specific inhibitors, we discovered that the suppression of CRC cells by GA was dependent on ferroptosis rather than autophagy or apoptosis. Previous research has demonstrated that GA treatment significantly triggers ferroptosis. Mechanistically, GA treatment promotes the degradation of the SLC7A11 protein, which plays a crucial role in ferroptosis. Notably, the knockdown of SLC7A11 abolished the detrimental effects of GA on the proliferation of CRC cells and reversed GA-induced ferroptosis in CRC cells both in vivo and in vitro. Further research has shown that GA can directly bind to the SLC7A11 protein and promote its ubiquitination. CONCLUSION Our research provides compelling evidence that GA may serve as a potential agent for suppressing the progression of CRC by inducing ferroptosis and promoting the ubiquitination and degradation of SLC7A11.
Collapse
Affiliation(s)
- Hongyu Xiao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Department of General Surgery, Chongqing General Hospital, Chongqing 400013, China
| | - Chen Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan, Guangdong 510006, China
| | - Xin Yuan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Limei Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Yun Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jin Yuan
- Pu'er Traditional Chinese Medicine Hospital in Yunnan Province, Puer, Yunnan 665099, China
| | - Song Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jian Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Shengliang Yuan
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Affiliated Gaozhou People's Hospital, Guangdong Medical University, Gaozhou, Guangdong 525200, China.
| | - Meifen Li
- Guangdong Women and Children Hospital, Guangzhou 511400, PR China.
| | - Junyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
6
|
Long Y, Xu Z, Yu J, Hu X, Xie Y, Duan X, Li N, Yan Y, Wang Y, Qin J. Targeting xCT with sulfasalazine suppresses triple-negative breast cancer growth via inducing autophagy and coordinating cell cycle and proliferation. Anticancer Drugs 2024; 35:830-843. [PMID: 39016262 DOI: 10.1097/cad.0000000000001630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
There is a substantial unmet need for effective treatment strategies in triple-negative breast cancer (TNBC). Recently, renewed attention has been directed towards targeting glutamine (Gln) metabolism to enhance the efficacy of cancer treatment. Nonetheless, a comprehensive exploration into the mechanistic implications of targeting Gln metabolism in TNBC is lacking. In this study, our objective was to probe the sensitivity of TNBC to alterations in Gln metabolism, using representative TNBC cell lines: MDA-MB-231, MDA-MB-468, and 4T1. Through an integration of bioinformatics, in-vitro, and in-vivo investigations, we demonstrated that sulfasalazine (SAS), like erastin (a known xCT inhibitor), effectively suppressed the expression and transport function of xCT, resulting in a depletion of glutathione levels in MDA-MB-231 and MDA-MB-468 cells. Furthermore, both xCT knockdown and SAS treatment demonstrated the promotion of cellular autophagy. We unveiled a positive correlation between xCT and the autophagy-related molecule p62, their co-expression indicating poor survival outcomes in breast cancer patients. In addition, our research revealed the influence of SAS and xCT on the expression of proteins regulating cell cycle and proliferation. Treatment with SAS or xCT knockdown led to the inhibition of MYC, CDK1, and CD44 expression. Significantly, the combined administration of SAS and rapamycin exhibited a synergistic inhibitory effect on the growth of transplanted breast tumor in mouse models constructed from murine-derived 4T1 cells. Taken together, our findings suggested the potential and clinical relevance of the SAS and rapamycin combination in the treatment of TNBC.
Collapse
Affiliation(s)
- Yaping Long
- Department of Immunology, School of Medicine, Nankai University
| | - Zizheng Xu
- Department of Immunology, School of Medicine, Nankai University
| | - Jing Yu
- Department of Immunology, School of Medicine, Nankai University
| | - Xiao Hu
- Department of Immunology, School of Medicine, Nankai University
| | - Yu Xie
- Department of Immunology, School of Medicine, Nankai University
| | - Xianxian Duan
- Department of Immunology, School of Medicine, Nankai University
| | - Ning Li
- School of Disaster and Emergency Medicine, Faculty of Medicine, Tianjin University
| | - Yan Yan
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Yue Wang
- Department of Immunology, School of Medicine, Nankai University
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Hospital of Stomatology, Nankai University, Tianjin, China
| | - Junfang Qin
- Department of Immunology, School of Medicine, Nankai University
| |
Collapse
|
7
|
Song M, Huang S, Wu X, Zhao Z, Liu X, Wu C, Wang M, Gao J, Ke Z, Ma X, He W. UBR5 mediates colorectal cancer chemoresistance by attenuating ferroptosis via Lys 11 ubiquitin-dependent stabilization of Smad3-SLC7A11 signaling. Redox Biol 2024; 76:103349. [PMID: 39260061 PMCID: PMC11415886 DOI: 10.1016/j.redox.2024.103349] [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: 07/07/2024] [Revised: 08/24/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024] Open
Abstract
Chemoresistance remains a principal culprit for the treatment failure in colorectal cancer (CRC), especially for patients with recurrent or metastatic disease. Deciphering the molecular basis of chemoresistance may lead to novel therapeutic strategies for this fatal disease. Here, UBR5, an E3 ubiquitin ligase frequently overexpressed in human CRC, is demonstrated to mediate chemoresistance principally by inhibiting ferroptosis. Paradoxically, UBR5 shields oxaliplatin-activated Smad3 from proteasome-dependent degradation via Lys 11-linked polyubiquitination. This novel chemical modification of Smad3 facilitates the transcriptional repression of ATF3, induction of SLC7A11 and inhibition of ferroptosis, contributing to chemoresistance. Consequently, targeting UBR5 in combination with a ferroptosis inducer synergistically sensitizes CRC to oxaliplatin-induced cell death and control of tumor growth. This study reveals, for the first time, a major clinically relevant chemoresistance mechanism in CRC mediated by UBR5 in sustaining TGFβ-Smad3 signaling and tuning ferroptosis, unveiling its potential as a viable therapeutic target for chemosensitization.
Collapse
Affiliation(s)
- Mei Song
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China.
| | - Shuting Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Xiaoxue Wu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Ziyi Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Xiaoting Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Chong Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Mengru Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Jialing Gao
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Zunfu Ke
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China
| | - Xiaojing Ma
- Department of Microbiology and Immunology, Weill Cornell Medicine, NY, 10065, USA
| | - Weiling He
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510275, China; School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, 361000, China.
| |
Collapse
|
8
|
Shi JX, Zhang ZC, Yin HZ, Piao XJ, Liu CH, Liu QJ, Zhang JC, Zhou WX, Liu FC, Yang F, Wang YF, Liu H. RNA m6A modification in ferroptosis: implications for advancing tumor immunotherapy. Mol Cancer 2024; 23:213. [PMID: 39342168 PMCID: PMC11437708 DOI: 10.1186/s12943-024-02132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
The pursuit of innovative therapeutic strategies in oncology remains imperative, given the persistent global impact of cancer as a leading cause of mortality. Immunotherapy is regarded as one of the most promising techniques for systemic cancer therapies among the several therapeutic options available. Nevertheless, limited immune response rates and immune resistance urge us on an augmentation for therapeutic efficacy rather than sticking to conventional approaches. Ferroptosis, a novel reprogrammed cell death, is tightly correlated with the tumor immune environment and interferes with cancer progression. Highly mutant or metastasis-prone tumor cells are more susceptible to iron-dependent nonapoptotic cell death. Consequently, ferroptosis-induction therapies hold the promise of overcoming resistance to conventional treatments. The most prevalent post-transcriptional modification, RNA m6A modification, regulates the metabolic processes of targeted RNAs and is involved in numerous physiological and pathological processes. Aberrant m6A modification influences cell susceptibility to ferroptosis, as well as the expression of immune checkpoints. Clarifying the regulation of m6A modification on ferroptosis and its significance in tumor cell response will provide a distinct method for finding potential targets to enhance the effectiveness of immunotherapy. In this review, we comprehensively summarized regulatory characteristics of RNA m6A modification on ferroptosis and discussed the role of RNA m6A-mediated ferroptosis on immunotherapy, aiming to enhance the effectiveness of ferroptosis-sensitive immunotherapy as a treatment for immune-resistant malignancies.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Cheng-Hu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qian-Jia Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jia-Cheng Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Wen-Xuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu-Chen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
9
|
Chen YS, Lee CH, Hsieh YH, Chiou HL, Hung MC, Lee HL. Sorafenib, a Tyrosine Kinase Inhibitor, Synergistically Enhances the Ferroptosis Effects of Asiatic Acid in Hepatocellular Carcinoma Cells. ENVIRONMENTAL TOXICOLOGY 2024. [PMID: 39264136 DOI: 10.1002/tox.24415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 09/13/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Asiatic acid (AA) is a natural triterpene, which is recognized as effect of antioxidant and antitumor. Sorafenib (Sor), an orally target drug, has been applicate for the HCC therapy. However, the synergistic effect of AA and Sor on human HCC is still unclear. Here, we explore the effect of combined treatment with AA and Sor in the HCC cell line SK-HEP-1 and HepG2. Compared with treating alone, our results demonstrated that AA combined with Sor synergistically inhibited proliferative rates in MTT assay and colony formation assay. We also found that AA combined with Sor in HCC cells strongly caused cell cycle arrest in G0/G1 phase and affected the protein level of cyclin D1 and SKP2. Furthermore, combination treatment strongly enhanced ferroptosis through cellular accumulation of iron ions, lipid peroxidation, and ferroptosis-related proteins (GPX4 and FTH1) in HCC cells. In addition, the combined treatment resulted in higher phosphorylation of JNK1/2 in the promotion of ferroptosis than drug treatment alone. These results indicate that AA combined with Sor synergistically improved ferroptosis in HCC cells through the regulation of JNK1/2 signaling. Taken together, the combinatorial strategy may serve as the potential treatment in HCC.
Collapse
Affiliation(s)
- Yong-Syuan Chen
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Hsing Lee
- Division of Pediatric Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- Department of Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Chun Hung
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
10
|
Zhang S, Guo L, Tao R, Liu S. Ferroptosis-targeting drugs in breast cancer. J Drug Target 2024:1-18. [PMID: 39225187 DOI: 10.1080/1061186x.2024.2399181] [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: 05/19/2024] [Revised: 07/07/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
In 2020, breast cancer surpassed lung cancer as the most common cancer in the world for the first time. Due to the resistance of some breast cancer cell lines to apoptosis, the therapeutic effect of anti-breast cancer drugs is limited. According to recent report, the susceptibility of breast cancer cells to ferroptosis affects the progress, prognosis and drug resistance of breast cancer. For instance, roblitinib induces ferroptosis of trastuzumab-resistant human epidermal growth factor receptor 2 (HER2)-positive breast cancer cells by diminishing fibroblast growth factor receptor 4 (FGFR4) expression, thereby augmenting the susceptibility of these cells to HER2-targeted therapies. In tamoxifen-resistant breast cancer cells, Fascin exacerbates their resistance by repressing solute carrier family 7 member 11 (SLC7A11) expression, which in turn heightens their responsiveness to tamoxifen. In recent years, Chinese herbs extracts and therapeutic drugs have been demonstrated to elicit ferroptosis in breast cancer cells by modulating a spectrum of regulatory factors pertinent to ferroptosis, including SLC7A11, glutathione peroxidase 4 (GPX4), acyl-CoA synthetase long chain family member 4 (ACSL4), and haem oxygenase 1 (HO-1). Here, we review the roles and mechanisms of Chinese herbal extracts and therapeutic drugs in regulating ferroptosis in breast cancer, providing potential therapeutic options for anti-breast cancer.
Collapse
Affiliation(s)
- Shuxian Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Lijuan Guo
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, China
| | - Shuangping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, China
| |
Collapse
|
11
|
Sun LL, He HY, Li W, Jin WL, Wei YJ. The solute carrier transporters (SLCs) family in nutrient metabolism and ferroptosis. Biomark Res 2024; 12:94. [PMID: 39218897 PMCID: PMC11367818 DOI: 10.1186/s40364-024-00645-2] [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: 07/12/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Ferroptosis is a novel form of programmed cell death caused by damage to lipid membranes due to the accumulation of lipid peroxides in response to various stimuli, such as high levels of iron, oxidative stress, metabolic disturbance, etc. Sugar, lipid, amino acid, and iron metabolism are crucial in regulating ferroptosis. The solute carrier transporters (SLCs) family, known as the "metabolic gating" of cells, is responsible for transporting intracellular nutrients and metabolites. Recent studies have highlighted the significant role of SLCs family members in ferroptosis by controlling the transport of various nutrients. Here, we summarized the function and mechanism of SLCs in ferroptosis regulated by ion, metabolic control of nutrients, and multiple signaling pathways, with a focus on SLC-related transporters that primarily transport five significant components: glucose, amino acid, lipid, trace metal ion, and other ion. Furthermore, the potential clinical applications of targeting SLCs with ferroptosis inducers for various diseases, including tumors, are discussed. Overall, this paper delves into the novel roles of the SLCs family in ferroptosis, aiming to enhance our understanding of the regulatory mechanisms of ferroptosis and identify new therapeutic targets for clinical applications.
Collapse
Affiliation(s)
- Li-Li Sun
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Hai-Yan He
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P. R. China
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Yi-Ju Wei
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
12
|
Zhou Z, Mai Y, Zhang G, Wang Y, Sun P, Jing Z, Li Z, Xu Y, Han B, Liu J. Emerging role of immunogenic cell death in cancer immunotherapy: Advancing next-generation CAR-T cell immunotherapy by combination. Cancer Lett 2024; 598:217079. [PMID: 38936505 DOI: 10.1016/j.canlet.2024.217079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Immunogenic cell death (ICD) is a stress-driven form of regulated cell death (RCD) in which dying tumor cells' specific signaling pathways are activated to release damage-associated molecular patterns (DAMPs), leading to the robust anti-tumor immune response as well as a reversal of the tumor immune microenvironment from "cold" to "hot". Chimeric antigen receptor (CAR)-T cell therapy, as a landmark in anti-tumor immunotherapy, plays a formidable role in hematologic malignancies but falls short in solid tumors. The Gordian knot of CAR-T cells for solid tumors includes but is not limited to, tumor antigen heterogeneity or absence, physical and immune barriers of tumors. The combination of ICD induction therapy and CAR-T cell immunotherapy is expected to promote the intensive use of CAR-T cell in solid tumors. In this review, we summarize the characteristics of ICD, stress-responsive mechanism, and the synergistic effect of various ICD-based therapies with CAR-T cells to effectively improve anti-tumor capacity.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yumiao Mai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Henan Province Key Laboratory of Cardiac Injury and Repair, Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, 450052, China
| | - Yingjie Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Pan Sun
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhaohe Jing
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jian Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
13
|
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
|
14
|
Lu KC, Tsai KW, Wang YK, Hu WC. Types of cell death and their relations to host immunological pathways. Aging (Albany NY) 2024; 16:11755-11768. [PMID: 39120579 PMCID: PMC11346778 DOI: 10.18632/aging.206035] [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: 02/15/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024]
Abstract
Various immune pathways have been identified in the host, including TH1, TH2, TH3, TH9, TH17, TH22, TH1-like, and THαβ immune reactions. While TH2 and TH9 responses primarily target multicellular parasites, host immune pathways directed against viruses, intracellular microorganisms (such as bacteria, protozoa, and fungi), and extracellular microorganisms can employ programmed cell death mechanisms to initiate immune responses or execute effective strategies for pathogen elimination. The types of programmed cell death involved include apoptosis, autophagy, pyroptosis, ferroptosis, necroptosis, and NETosis. Specifically, apoptosis is associated with host anti-virus eradicable THαβ immunity, autophagy with host anti-virus tolerable TH3 immunity, pyroptosis with host anti-intracellular microorganism eradicable TH1 immunity, ferroptosis with host anti-intracellular microorganism tolerable TH1-like immunity, necroptosis with host anti-extracellular microorganism eradicable TH22 immunity, and NETosis with host anti-extracellular microorganism tolerable TH17 immunity.
Collapse
Affiliation(s)
- Kuo-Cheng Lu
- Department of Medicine, Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
- Department of Medicine, Division of Nephrology, Fu Jen Catholic University Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, ROC
| | - Kuo-Wang Tsai
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan, ROC
| | - Yu-Kuen Wang
- Department of Obstetrics and Gynecology, Taoyuan Armed Forced General Hospital, Taiwan, ROC
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Wan-Chung Hu
- Department of Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan, ROC
- Department of Clinical pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan, ROC
- Department of Biotechnology, Ming Chuan University, Taoyuan City 333, Taiwan, ROC
| |
Collapse
|
15
|
Lu Y, Xie X, Luo L. Ferroptosis crosstalk in anti-tumor immunotherapy: molecular mechanisms, tumor microenvironment, application prospects. Apoptosis 2024:10.1007/s10495-024-01997-8. [PMID: 39008197 DOI: 10.1007/s10495-024-01997-8] [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] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Immunotherapies for cancer, specifically immune checkpoint inhibition (ICI), have shown potential in reactivating the body's immune response against tumors. However, there are challenges to overcome in addressing drug resistance and improving the effectiveness of these treatments. Recent research has highlighted the relationship between ferroptosis and the immune system within immune cells and the tumor microenvironment (TME), suggesting that combining targeted ferroptosis with immunotherapy could enhance anti-tumor effects. This review explores the potential of using immunotherapy to target ferroptosis either alone or in conjunction with other therapies like immune checkpoint blockade (ICB) therapy, radiotherapy, and nanomedicine synergistic treatments. It also delves into the roles of different immune cell types in promoting anti-tumor immune responses through ferroptosis. Together, these findings provide a comprehensive understanding of synergistic immunotherapy focused on ferroptosis and offer innovative strategies for cancer treatment.
Collapse
Affiliation(s)
- Yining Lu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Xiaoting Xie
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
16
|
Zeng L, Yang K, Yu G, Hao W, Zhu X, Ge A, Chen J, Sun L. Advances in research on immunocyte iron metabolism, ferroptosis, and their regulatory roles in autoimmune and autoinflammatory diseases. Cell Death Dis 2024; 15:481. [PMID: 38965216 PMCID: PMC11224426 DOI: 10.1038/s41419-024-06807-2] [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: 02/10/2024] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
Autoimmune diseases commonly affect various systems, but their etiology and pathogenesis remain unclear. Currently, increasing research has highlighted the role of ferroptosis in immune regulation, with immune cells being a crucial component of the body's immune system. This review provides an overview and discusses the relationship between ferroptosis, programmed cell death in immune cells, and autoimmune diseases. Additionally, it summarizes the role of various key targets of ferroptosis, such as GPX4 and TFR, in immune cell immune responses. Furthermore, the release of multiple molecules, including damage-associated molecular patterns (DAMPs), following cell death by ferroptosis, is examined, as these molecules further influence the differentiation and function of immune cells, thereby affecting the occurrence and progression of autoimmune diseases. Moreover, immune cells secrete immune factors or their metabolites, which also impact the occurrence of ferroptosis in target organs and tissues involved in autoimmune diseases. Iron chelators, chloroquine and its derivatives, antioxidants, chloroquine derivatives, and calreticulin have been demonstrated to be effective in animal studies for certain autoimmune diseases, exerting anti-inflammatory and immunomodulatory effects. Finally, a brief summary and future perspectives on the research of autoimmune diseases are provided, aiming to guide disease treatment strategies.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China.
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Wensa Hao
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Junpeng Chen
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
- Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, USA.
- College of Mechanical Engineering, Hunan University of Science and Technology, Xiangtan, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
17
|
Zhu X, Sha X, Zang Y, Ren Q, Zhang S, Ma D, Wang L, Yao J, Zhou X, Yu L, Li T. Current Progress of Ferroptosis Study in Hepatocellular Carcinoma. Int J Biol Sci 2024; 20:3621-3637. [PMID: 38993573 PMCID: PMC11234204 DOI: 10.7150/ijbs.96014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/08/2024] [Indexed: 07/13/2024] Open
Abstract
Ferroptosis, an emerging type of programmed cell death, is initiated by iron-dependent and excessive ROS-mediated lipid peroxidation, which eventually leads to plasma membrane rupture and cell death. Many canonical signalling pathways and biological processes are involved in ferroptosis. Furthermore, cancer cells are more susceptible to ferroptosis due to the high load of ROS and unique metabolic characteristics, including iron requirements. Recent investigations have revealed that ferroptosis plays a crucial role in the progression of tumours, especially HCC. Specifically, the induction of ferroptosis can not only inhibit the growth of hepatoma cells, thereby reversing tumorigenesis, but also improves the efficacy of immunotherapy and enhances the antitumour immune response. Therefore, triggering ferroptosis has become a new therapeutic strategy for cancer therapy. In this review, we summarize the characteristics of ferroptosis based on its underlying mechanism and role in HCC and provide possible therapeutic applications.
Collapse
Affiliation(s)
- Xinyue Zhu
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Xudong Sha
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Zang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Qiaohui Ren
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Shubing Zhang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Dongyue Ma
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Lianzi Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Junxiao Yao
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Xinyi Zhou
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| | - Li Yu
- Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Tao Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Shushan District, No. 218 Jixi Road, Hefei, 230032, Anhui, China
| |
Collapse
|
18
|
Liu D, Hu Z, Lu J, Yi C. Redox-Regulated Iron Metabolism and Ferroptosis in Ovarian Cancer: Molecular Insights and Therapeutic Opportunities. Antioxidants (Basel) 2024; 13:791. [PMID: 39061859 PMCID: PMC11274267 DOI: 10.3390/antiox13070791] [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: 05/01/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Ovarian cancer (OC), known for its lethality and resistance to chemotherapy, is closely associated with iron metabolism and ferroptosis-an iron-dependent cell death process, distinct from both autophagy and apoptosis. Emerging evidence suggests that dysregulation of iron metabolism could play a crucial role in OC by inducing an imbalance in the redox system, which leads to ferroptosis, offering a novel therapeutic approach. This review examines how disruptions in iron metabolism, which affect redox balance, impact OC progression, focusing on its essential cellular functions and potential as a therapeutic target. It highlights the molecular interplay, including the role of non-coding RNAs (ncRNAs), between iron metabolism and ferroptosis, and explores their interactions with key immune cells such as macrophages and T cells, as well as inflammation within the tumor microenvironment. The review also discusses how glycolysis-related iron metabolism influences ferroptosis via reactive oxygen species. Targeting these pathways, especially through agents that modulate iron metabolism and ferroptosis, presents promising therapeutic prospects. The review emphasizes the need for deeper insights into iron metabolism and ferroptosis within the redox-regulated system to enhance OC therapy and advocates for continued research into these mechanisms as potential strategies to combat OC.
Collapse
Affiliation(s)
- Dan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| | - Zewen Hu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| | - Jinzhi Lu
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
- Department of Laboratory Medicine, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China
| | - Cunjian Yi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Yangtze University, Jingzhou 434000, China; (D.L.); (Z.H.)
- Hubei Provincial Clinical Research Center for Personalized Diagnosis and Treatment of Cancer, Jingzhou 434000, China
| |
Collapse
|
19
|
Lv H, Yu J, Qian X, Shu J, Qian Q, Shen L, Shi D, Tao Z, Fan G, Zhuang B, Lu B. USP7 upregulated by TGF-β1 promotes ferroptosis via inhibiting LATS1-YAP axis in sepsis-induced acute lung injury. iScience 2024; 27:109667. [PMID: 38966570 PMCID: PMC11223090 DOI: 10.1016/j.isci.2024.109667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/27/2023] [Accepted: 04/02/2024] [Indexed: 07/06/2024] Open
Abstract
Our work aimed to investigate the interactive roles of transforming growth factor β1 (TGF-β1), ubiquitin-specific-processing protease 7 (USP7), and Yes-associated protein (YAP) in ferroptosis during sepsis-secondary acute lung injury (ALI). Our study demonstrated that ferroptosis was aggravated by TGF-β1 in both cellular and animal models of acute lung injury. Additionally, YAP upregulated glutathione peroxidase 4 (GPX4) and SLC7A11 by regulating the binding of TEAD4 to GPX4/SLC7A11 promoters. Furthermore, large tumor suppressor kinase 1 (LATS1) knockdown resulted in YAP expression stimulation, while USP7 downregulated YAP via deubiquitinating and stabilizing LATS1/2. YAP overexpression or USP7/LATS1 silencing reduced ferroptosis process, which regulated YAP through a feedback loop. However, TGF-β1 annulled the repression of ferroptosis by YAP overexpression or LATS1/USP7 knockdown. By elucidating the molecular interactions between TGF-β1, USP7, LATS1/2, and YAP, we identified a new regulatory axis of ferroptosis in sepsis-secondary ALI. Our study sheds light on the pathophysiology of ferroptosis and proposes a potential therapeutic approach for sepsis-induced ALI.
Collapse
Affiliation(s)
- Hong Lv
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Jing Yu
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Xingjia Qian
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Jun Shu
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Qiuhong Qian
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Luhong Shen
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Dongfang Shi
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Zhengzheng Tao
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| | - Guiqin Fan
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
- Soochow University School of Medicine, Suzhou, Jiangsu Province 215031, P.R. China
| | - Bufeng Zhuang
- Department of Thoracic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, P.R. China
| | - Bing Lu
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu Province 215499, P.R. China
| |
Collapse
|
20
|
Yang Q, Xia Y, Chen K, Wang Y, Song D, Zhu J, Tong J, Shen Y. Blue light induced ferroptosis via STAT3/GPX4/SLC7A11/FTH1 in conjunctiva epithelium in vivo and in vitro. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 255:112908. [PMID: 38663336 DOI: 10.1016/j.jphotobiol.2024.112908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 05/13/2024]
Abstract
The prevalence of Light-emitting diodes (LEDs) has exposed us to an excessive amount of blue light (BL) which causes various ophthalmic diseases. Previous studies have shown that conjunctiva is vulnerable to BL. In this study, we aimed to investigate the underlying mechanism of BL-induced injury in conjunctiva. We placed C57BL/6 mice and human conjunctival epithelial cell lines (HCECs) under BL (440 nm ± 15 nm, 0.2 mW/cm2) to establish a BL injury model in vivo and in vitro. Immunohistochemistry and MDA assay were used to identify lipid peroxidation (LPO) in vivo. HE staining was applied to detect morphological damage of conjunctival epithelium. DCFH-DA, C11-BODIPY 581/591, Calcein-AM, and FeRhoNox™-1 probes were performed to identify ferroptosis levels in vitro. Real-time qPCR and Western blotting techniques were employed to uncover signaling pathways of blue light-induced ferroptosis. Our findings demonstrated that BL affected tear film instability and induced conjunctival epithelium injury in vivo. Ferrostatin-1 significantly alleviated blue light-induced ferroptosis in vivo and in vitro. BL downregulates the levels of solute carrier family 7 member 11 (SLC7A11), Ferritin heavy chain (FTH1), and glutathione peroxidase (GPX4) by inhibiting the activation and translocation of the Signal transducer and activator of transcription 3 (STAT3) from inducing Fe2+ burst, ROS and LPO accumulation, ultimately resulting in ferroptosis. This study will offer new insight into BL-induced conjunctival injury and LED-induced dry eye.
Collapse
Affiliation(s)
- Qianjie Yang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yutong Xia
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Kuangqi Chen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yinhao Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Dongjie Song
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jiru Zhu
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jianping Tong
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ye Shen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
21
|
Zhou H, Cheng Y, Huang Q, Xiao J. Regulation of ferroptosis by nanotechnology for enhanced cancer immunotherapy. Expert Opin Drug Deliv 2024; 21:921-943. [PMID: 39014916 DOI: 10.1080/17425247.2024.2379937] [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: 02/05/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
INTRODUCTION This review explores the innovative intersection of ferroptosis, a form of iron-dependent cell death, with cancer immunotherapy. Traditional cancer treatments face limitations in efficacy and specificity. Ferroptosis as a new paradigm in cancer biology, targets metabolic peculiarities of cancer cells and may potentially overcome such limitations, enhancing immunotherapy. AREA COVERED This review centers on the regulation of ferroptosis by nanotechnology to augment immunotherapy. It explores how nanoparticle-modulated ferroptotic cancer cells impact the TME and immune responses. The dual role of nanoparticles in modulating immune response through ferroptosis are also discussed. Additionally, it investigates how nanoparticles can be integrated with various immunotherapeutic strategies, to optimize ferroptosis induction and cancer treatment efficacy. The literature search was conducted using PubMed and Google Scholar, covering articles published up to March 2024. EXPERT OPINION The manuscript underscores the promising yet intricate landscape of ferroptosis in immunotherapy. It emphasizes the need for a nuanced understanding of ferroptosis' impact on immune cells and the TME to develop more effective cancer treatments, highlighting the potential of nanoparticles in enhancing the efficacy of ferroptosis and immunotherapy. It calls for deeper exploration into the molecular mechanisms and clinical potential of ferroptosis to fully harness its therapeutic benefits in immunotherapy.
Collapse
Affiliation(s)
- Haohan Zhou
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, PR China
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yiyun Cheng
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Quan Huang
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, PR China
| | - Jianru Xiao
- Department of Orthopedic Oncology, Changzheng Hospital, Naval Medical University, Shanghai, PR China
| |
Collapse
|
22
|
Li Y, Tuerxun H, Zhao Y, Liu X, Li X, Wen S, Zhao Y. The new era of lung cancer therapy: Combining immunotherapy with ferroptosis. Crit Rev Oncol Hematol 2024; 198:104359. [PMID: 38615871 DOI: 10.1016/j.critrevonc.2024.104359] [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: 10/29/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
Ferroptosis is an unconventional programmed cell death mode caused by phospholipid peroxidation dependent on iron. Emerging immunotherapies (especially immune checkpoint inhibitors) have the potential to enhance lung cancer patients' long-term survival. Although immunotherapy has yielded significant positive applications in some patients, there are still many mechanisms that can cause lung cancer cells to evade immunity, thus leading to the failure of targeted therapies. Immune-tolerant cancer cells are insensitive to conventional death pathways such as apoptosis and necrosis, whereas mesenchymal and metastasis-prone cancer cells are particularly vulnerable to ferroptosis, which plays a vital role in mediating immune tolerance resistance by tumors and immune cells. As a result, triggering lung cancer cell ferroptosis holds significant therapeutic potential for drug-resistant malignancies. Here, we summarize the mechanisms underlying the suppression of ferroptosis in lung cancer, highlight its function in the lung cancer immune microenvironment, and propose possible therapeutic strategies.
Collapse
Affiliation(s)
- Yawen Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Halahati Tuerxun
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xingyu Liu
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xi Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Shuhui Wen
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
23
|
Liao S, Wei C, Wei G, Liang H, Peng F, Zhao L, Li Z, Liu C, Zhou Q. Cyclophosphamide activates ferroptosis-induced dysfunction of Leydig cells via SMAD2 pathway†. Biol Reprod 2024; 110:1012-1024. [PMID: 38320204 DOI: 10.1093/biolre/ioae020] [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: 02/20/2023] [Revised: 07/17/2023] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
Cyclophosphamide (CP) is a widely used chemotherapeutic drug and immunosuppressant in the clinic, and the hypoandrogenism caused by CP is receiving more attention. Some studies found that ferroptosis is a new mechanism of cell death closely related to chemotherapeutic drugs and plays a key role in regulating reproductive injuries. The purpose of this study is to explore ferroptosis' role in testicular Leydig cell dysfunction and molecular mechanisms relating to it. In this study, the level of ferroptosis in the mouse model of testicular Leydig cell dysfunction induced by CP was significantly increased and further affected testosterone synthesis. The ferroptosis inhibitors ferrostatin-1 (Fer-1) and iron chelator deferoxamine (DFO) can improve injury induced by CP. The results of immunohistochemistry showed that Fer-1 and DFO could improve the structural disorder of seminiferous tubules and the decrease of the number of Leydig cells in testicular tissue induced by CP. Immunofluorescence and western blot confirmed that Fer-1 and DFO could improve the expression of key enzymes in testosterone synthesis. The activation of SMAD family member 2 (Smad2)/cyclin-dependent kinase inhibitor 1A (Cdkn1a) pathway can improve the ferroptosis of Leydig cells induced by CP and protect the function of Leydig cells. By inhibiting the Smad2/Cdkn1a signal pathway, CP can regulate ferroptosis, resulting in testicular Leydig cell dysfunction. In this study, CP-induced hypoandrogenism is explained theoretically and a potential therapeutic strategy is provided.
Collapse
Affiliation(s)
- Senlin Liao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Cun Wei
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Guanyang Wei
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Haoyu Liang
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Fan Peng
- Department of Urology, Shenzhen Baoan District Central Hospital, Shenzhen, PR China
| | - Lei Zhao
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Ziguang Li
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Cundong Liu
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Qizhao Zhou
- Department of Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, PR China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
24
|
Li D, Zhang M, Liu J, Li Z, Ni B. Potential therapies for HCC involving targeting the ferroptosis pathway. Am J Cancer Res 2024; 14:1446-1465. [PMID: 38726269 PMCID: PMC11076240 DOI: 10.62347/sigp9279] [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: 11/19/2023] [Accepted: 03/15/2024] [Indexed: 05/12/2024] Open
Abstract
Liver cancer ranks as the third leading cause of cancer-related mortality worldwide, predominantly in the form of hepatocellular carcinoma (HCC). Conventional detection and treatment approaches have proven inadequate for addressing the elevated incidence and mortality rates associated with HCC. However, a significant body of research suggests that combating HCC through the induction of ferroptosis is possible. Ferroptosis is a regulated cell death process characterized by elevated levels of reactive oxygen species (ROS) and lipid peroxide accumulation, both of which are dependent on iron levels. In recent years, there has been an increasing focus on investigating ferroptosis, revealing its potential as an inhibitory mechanism against various diseases, including tumors. Therefore, ferroptosis induction holds great promise for treating multiple types of cancers, including HCC. This article provides a review of the key mechanisms involved in ferroptosis and explores the potential application of multiple targets and pathways associated with ferroptosis in HCC treatment to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Denghui Li
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical UniversityChongqing 400038, China
| | - Mengjie Zhang
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical UniversityChongqing 400038, China
| | - Ju Liu
- Department of Foreign Languages, College of Basic Medical Sciences, Third Military Medical UniversityChongqing 400038, China
| | - Zhifang Li
- Department of Foreign Languages, College of Basic Medical Sciences, Third Military Medical UniversityChongqing 400038, China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical UniversityChongqing 400038, China
| |
Collapse
|
25
|
Lei G, Zhuang L, Gan B. The roles of ferroptosis in cancer: Tumor suppression, tumor microenvironment, and therapeutic interventions. Cancer Cell 2024; 42:513-534. [PMID: 38593779 DOI: 10.1016/j.ccell.2024.03.011] [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] [Received: 11/30/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
In cancer treatment, the recurrent challenge of inducing apoptosis through conventional therapeutic modalities, often thwarted by therapy resistance, emphasizes the critical need to explore alternative cell death pathways. Ferroptosis, an iron-dependent form of regulated cell death triggered by the lethal accumulation of lipid peroxides on cellular membranes, has emerged as one such promising frontier in oncology. Induction of ferroptosis not only suppresses tumor growth but also holds potential for augmenting immunotherapy responses and surmounting resistance to existing cancer therapies. This review navigates the role of ferroptosis in tumor suppression. Furthermore, we delve into the complex role of ferroptosis within the tumor microenvironment and its interplay with antitumor immunity, offering insights into the prospect of targeting ferroptosis as a strategic approach in cancer therapy.
Collapse
Affiliation(s)
- Guang Lei
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhuang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
26
|
Sun C, Zhan J, Li Y, Zhou C, Huang S, Zhu X, Huang K. Non-apoptotic regulated cell death mediates reprogramming of the tumour immune microenvironment by macrophages. J Cell Mol Med 2024; 28:e18348. [PMID: 38652105 PMCID: PMC11037416 DOI: 10.1111/jcmm.18348] [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: 11/24/2023] [Revised: 02/23/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
Tumour immune microenvironment (TIME) plays an indispensable role in tumour progression, and tumour-associated macrophages (TAMs) are the most abundant immune cells in TIME. Non-apoptotic regulated cell death (RCD) can avoid the influence of tumour apoptosis resistance on anti-tumour immune response. Specifically, autophagy, ferroptosis, pyroptosis and necroptosis mediate the crosstalk between TAMs and tumour cells in TIME, thus reprogram TIME and affect the progress of tumour. In addition, although some achievements have been made in immune checkpoint inhibitors (ICIs), there is still defect that ICIs are only effective for some people because non-apoptotic RCD can bypass the apoptosis resistance of tumour. As a result, ICIs combined with targeting non-apoptotic RCD may be a promising solution. In this paper, the basic molecular mechanism of non-apoptotic RCD, the way in which non-apoptotic RCD mediates crosstalk between TAMs and tumour cells to reprogram TIME, and the latest research progress in targeting non-apoptotic RCD and ICIs are reviewed.
Collapse
Affiliation(s)
- Chengpeng Sun
- Department of NeurosurgeryThe Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiP. R. China
- HuanKui Academy, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Jianhao Zhan
- HuanKui Academy, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Yao Li
- The First Clinical Medical College, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Chulin Zhou
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Shuo Huang
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiChina
| | - Xingen Zhu
- Department of NeurosurgeryThe Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiP. R. China
- Institute of Neuroscience, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- JXHC Key Laboratory of Neurological MedicineNanchangJiangxiP. R. China
| | - Kai Huang
- Department of NeurosurgeryThe Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiP. R. China
- Institute of Neuroscience, Jiangxi Medical College, Nanchang UniversityNanchangJiangxiP. R. China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular DiseasesNanchangChina
- JXHC Key Laboratory of Neurological MedicineNanchangJiangxiP. R. China
| |
Collapse
|
27
|
Chen HD, Ye Z, Hu HF, Fan GX, Hu YH, Li Z, Li BR, Ji SR, Zhou CJ, Xu XW, Yu XJ, Qin Y. SMAD4 endows TGF-β1-induced highly invasive tumor cells with ferroptosis vulnerability in pancreatic cancer. Acta Pharmacol Sin 2024; 45:844-856. [PMID: 38057506 PMCID: PMC10943101 DOI: 10.1038/s41401-023-01199-z] [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: 07/25/2023] [Accepted: 11/12/2023] [Indexed: 12/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy prone to recurrence and metastasis. Studies show that tumor cells with increased invasive and metastatic potential are more likely to undergo ferroptosis. SMAD4 is a critical molecule in the transforming growth factor β (TGF-β) pathway, which affects the TGF-β-induced epithelial-mesenchymal transition (EMT) status. SMAD4 loss is observed in more than half of patients with PDAC. In this study, we investigated whether SMAD4-positive PDAC cells were prone to ferroptosis because of their high invasiveness. We showed that SMAD4 status almost determined the orientation of transforming growth factor β1 (TGF-β1)-induced EMT via the SMAD4-dependent canonical pathway in PDAC, which altered ferroptosis vulnerability. We identified glutathione peroxidase 4 (GPX4), which inhibited ferroptosis, as a SMAD4 down-regulated gene by RNA sequencing. We found that SMAD4 bound to the promoter of GPX4 and decreased GPX4 transcription in PDAC. Furthermore, TGF-β1-induced high invasiveness enhanced sensitivity of SMAD4-positive organoids and pancreas xenograft models to the ferroptosis inducer RAS-selective lethal 3 (RSL3). Moreover, SMAD4 enhanced the cytotoxic effect of gemcitabine combined with RSL3 in highly invasive PDAC cells. This study provides new ideas for the treatment of PDAC, especially SMAD4-positive PDAC.
Collapse
Affiliation(s)
- Hai-di Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Hai-Feng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Gui-Xiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Yu-Heng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Zheng Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Bo-Rui Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Shun-Rong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Chen-Jie Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China
| | - Xiao-Wu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
28
|
Shirahama H, Tani Y, Tsukahara S, Okamoto Y, Hasebe A, Noda T, Ando S, Ushijima M, Matsuura M, Tomida A. Induction of stearoyl-CoA desaturase confers cell density-dependent ferroptosis resistance in melanoma. J Cell Biochem 2024; 125:e30542. [PMID: 38362828 DOI: 10.1002/jcb.30542] [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: 10/20/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Ferroptosis is a form of regulated cell death that is induced by inhibiting glutathione peroxidase 4 (GPX4), which eliminates lipid peroxidation. Ferroptosis induction is influenced by the cell environment. However, the cellular states altering ferroptosis susceptibility remain largely unknown. We found that melanoma cell lines became resistant to ferroptosis as cell density increased. Comparative transcriptome and metabolome analyses revealed that cell density-dependent ferroptosis resistance was coupled with a shift toward a lipogenic phenotype accompanied by strong induction of stearoyl-CoA desaturase (SCD). Database analysis of gene dependency across hundreds of cancer cell lines uncovered a negative correlation between GPX4 and SCD dependency. Importantly, SCD inhibition, either pharmacologically or through genetic knockout, sensitized melanoma cells to GPX4 inhibition, thereby attenuating ferroptosis resistance in cells at high density. Our findings indicate that transition to an SCD-inducing, lipogenic cell state produces density-dependent resistance to ferroptosis, which may provide a therapeutic strategy against melanoma.
Collapse
Affiliation(s)
- Hitomi Shirahama
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuri Tani
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satomi Tsukahara
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuka Okamoto
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akiko Hasebe
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomomiki Noda
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Shuji Ando
- Department of Information Sciences, Faculty of Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Masaru Ushijima
- Clinical Research and Development Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masaaki Matsuura
- Graduate School of Public Health, Teikyo University, Tokyo, Japan
- Division of Cancer Genomics, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akihiro Tomida
- Division of Genome Research, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
Xie J, Wang H, Xie W, Liu Y, Chen Y. Gallic acid promotes ferroptosis in hepatocellular carcinoma via inactivating Wnt/β-catenin signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2437-2445. [PMID: 37847411 DOI: 10.1007/s00210-023-02770-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023]
Abstract
Hepatocellular carcinoma (HCC) has high morbidity and mortality, and effective therapies are lacking. Gallic acid (GA), a natural phenolic compound derived from plants, has been reported to prevent the onset and progression of various cancers. However, there is limited elaboration on the potential mechanisms and anticancer effects of GA on hepatocellular carcinoma. Inducing ferroptosis of tumor cells has become one of the most promising ways to eradicate tumor cells. However, the effect of GA on HCC ferroptosis remains unknown. We evaluated the impact of GA on cell viability, migration, and mitochondrial morphology in HepG2 cells. Our study identified a critical role of GA in inducing ferroptosis in HepG2 cells. Mechanistically, we found that GA could inhibit the expression of a ferroptosis-related protein SLC7A11 and GPX4 in HepG2, by blocking β-catenin transport from nuclear to the cytoplasm, thus inducing the inactivation of the Wnt/β-catenin pathway. Our study has confirmed that GA is a novel ferroptosis inducer of HC, suggesting GA could be a promising candidate for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Jingyi Xie
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Haijiao Wang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Wuxing Xie
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yongping Liu
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| | - Yi Chen
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
30
|
Zhang X, Hu Y, Wang B, Yang S. Ferroptosis: Iron-mediated cell death linked to disease pathogenesis. J Biomed Res 2024; 38:1-23. [PMID: 38808552 PMCID: PMC11461536 DOI: 10.7555/jbr.37.20230224] [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: 09/19/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 05/30/2024] Open
Abstract
Ferroptosis is an iron-mediated regulatory cell death pattern characterized by oxidative damage. The molecular regulating mechanisms are related to iron metabolism, lipid peroxidation, and glutathione metabolism. Additionally, some immunological signaling pathways, such as the cyclic GMP-AMP synthase-stimulator ofinterferon genes axis, Janus kinase-signal transducer and activator of transcription 1 axis, and transforming growth factor beta 1-Smad3 axis may also participate in the regulation of ferroptosis. Studies have shown that ferroptosis is closely related to many diseases such as cancer, neurodegenerative diseases, inflammatory diseases, and autoimmune diseases. Considering the pivotal role of ferroptosis-regulating signaling in the pathogenesis of diverse diseases, the development of ferroptosis inducers or inhibitors may have significant clinical potential for the treatment of the aforementioned conditions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingchao Hu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
31
|
Guo M, Liu M, Li W, Wang C, Zhang L, Zhang H. Osteopontin promotes tumor growth and metastasis and GPX4-mediated anti-lipid peroxidation in triple-negative breast cancer by activating the PI3k/Akt/mTOR pathway. J Cancer Res Clin Oncol 2024; 150:155. [PMID: 38526702 PMCID: PMC10963528 DOI: 10.1007/s00432-024-05658-w] [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: 11/13/2023] [Accepted: 02/20/2024] [Indexed: 03/27/2024]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) features high aggressiveness, metastasis rate, drug resistance as well as poor prognosis. Osteopontin (OPN) is a key protein in the process of osteogenesis and has emerged as a new tumor marker in recent years. METHODS Cell viability was tested with the CCK-8 kit. Transwell and wound healing were adopted to test cell invasive and migratory abilities. Tumor sphere formation was detected by tumor sphere formation assay. Human umbilical vein endothelial cell (HUVEC) tube formation assay was used to measure the angiogenesis of tumor cells. Western blot was applied for the estimation of the expression of cancer stem cell markers, angiogenesis-, signaling pathway-related proteins as well as OPN. Bioinformatics tools predicted OPN expression in breast cancer tissues. The levels of oxidative stress-related markers were assessed with ELISA. Following the overexpression of OPN in MD-MB-436 cells and the addition of the PI3K/AKT/mTOR pathway inhibitor LY294002, the aforementioned functional experiments were implemented again to investigate the mechanism. Finally, in vivo experiments of tumor-bearing mice were performed for further verification. RESULTS The proliferative, invasive, migratory and tumor sphere formation capabilities as well as angiogenesis of TNBC cells were conspicuously increased in contrast to non-TNBC cell lines. OPN expression in TNBC tissues and cells was dramatically enhanced. OPN upregulation significantly elevated cell proliferative, invasive and migratory capabilities as well as tumor sphere formation and angiogenesis. The mechanism might be achieved by activating PI3K/AKT/mTOR signaling to regulate glutathione peroxidase 4 (GPX4)-mediated anti-lipid peroxidation. CONCLUSION OPN promoted tumor sphere formation and angiogenesis in TNBC by activating the PI3K/AKT/mTOR pathway to regulate GPX4-mediated anti-lipid peroxidation levels.
Collapse
Affiliation(s)
- Man Guo
- Department of Thyroid and Breast Surgery, Nanyang Central Hospital, No. 312 Gongnong Road, Wancheng District, Nanyang City, 473005, Henan Province, China
| | - Mengyue Liu
- Xinxiang Medical University, Xinxiang City, 453003, Henan Province, China
| | - Weihan Li
- Department of Thyroid and Breast Surgery, Nanyang Central Hospital, No. 312 Gongnong Road, Wancheng District, Nanyang City, 473005, Henan Province, China
| | - Cao Wang
- Department of Thyroid and Breast Surgery, Nanyang Central Hospital, No. 312 Gongnong Road, Wancheng District, Nanyang City, 473005, Henan Province, China
| | - Lu Zhang
- Department of Thyroid and Breast Surgery, Nanyang Central Hospital, No. 312 Gongnong Road, Wancheng District, Nanyang City, 473005, Henan Province, China
| | - Hao Zhang
- Department of Thyroid and Breast Surgery, Nanyang Central Hospital, No. 312 Gongnong Road, Wancheng District, Nanyang City, 473005, Henan Province, China.
| |
Collapse
|
32
|
Chen Q, Zhang T, Zeng R, Zhang K, Li B, Zhu Z, Ma X, Zhang Y, Li L, Zhu J, Zhang G. The E3 ligase TRIM7 suppresses the tumorigenesis of gastric cancer by targeting SLC7A11. Sci Rep 2024; 14:6655. [PMID: 38509147 PMCID: PMC10954695 DOI: 10.1038/s41598-024-56746-3] [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: 11/30/2023] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
Tripartite motif-containing protein 7 (TRIM7), as an E3 ligase, plays an important regulatory role in various physiological and pathological processes. However, the role of TRIM7 in gastric cancer (GC) is still undefined. Our study detected the expression of TRIM7 in clinical specimens and investigated the regulatory effect and molecular mechanism of TRIM7 on GC progression through in vitro and in vivo experiments. Our finding showed that TRIM7 was significantly downregulated in GC, and patients with high expression of TRIM7 showed long overall survival. Both in vitro and in vivo experiments showed that TRIM7 dramatically suppressed the malignant progression of GC. Further investigation showed that ferroptosis was the major death type mediated by TRIM7. Mechanistically, TRIM7 interacted with SLC7A11 through its B30.2/SPRY domain and promoted Lys48-linked polyubiquitination of SLC7A11, which effectively suppressing SLC7A11/GPX4 axis and inducing ferroptosis in GC cells. In vivo experiments and correlation analysis based on clinical specimens further confirmed that TRIM7 inhibited tumor growth through suppressing SLC7A11/GPX4 axis. In conclusion, our investigation demonstrated for the first time that TRIM7, as a tumor suppressor, induced ferroptosis via targeting SLC7A11 in GC, which provided a new strategy for the molecular therapy of GC by upregulating TRIM7.
Collapse
Affiliation(s)
- Qishuai Chen
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Tongtong Zhang
- Department of Laboratory Medical, Zibo Central Hospital, Zibo, 255000, Shandong Province, People's Republic of China
| | - Runzhi Zeng
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Kunmiao Zhang
- Department of General Surgery, Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, People's Republic of China
| | - Bingjun Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Zhenguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Xiaomin Ma
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Yun Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Linchuan Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Jiankang Zhu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China
| | - Guangyong Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University, No.16766 Jingshi Road, Jinan, 250014, Shandong Province, People's Republic of China.
| |
Collapse
|
33
|
Mo Y, Zou Z, Chen E. Targeting ferroptosis in hepatocellular carcinoma. Hepatol Int 2024; 18:32-49. [PMID: 37880567 DOI: 10.1007/s12072-023-10593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 09/04/2023] [Indexed: 10/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor with complex survival mechanism and drug resistance, resulting in cancer-related high mortality in the world. Ferroptosis represents a form of regulated cell death, typically distinguished by iron-dependent lipid peroxidation. Cancer cells often employ antioxidant defenses to evade the harmful effects of excess iron. Recent research has proposed that directing interventions towards ferroptosis could serve as an effective strategy in curbing the proliferation and invasion of HCC. Immunotherapy has made some preliminary progress in the remodeling of immune microenvironment, but it has not completely inhibited HCC growth, invasion and drug resistance. Furthermore, ferroptosis is widely observed in the formation of immune microenvironment of HCC and mediates the response of many targeted drugs and immunotherapy. Clarifying the role of ferroptosis in these complex processes is expected to provide a new prospect for HCC treatment. In this review, we outline the mechanisms by which HCC develops invasiveness and drug resistance by evading iron-dependent death, and paint a comprehensive landscape of ferroptosis in different cell types in the HCC immune microenvironment.
Collapse
Affiliation(s)
- Yuqian Mo
- School of Public Health, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhilin Zou
- Department of Ophthalmology, Affiliated Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Erbao Chen
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong, China.
| |
Collapse
|
34
|
Ko MJ, Min S, Hong H, Yoo W, Joo J, Zhang YS, Kang H, Kim DH. Magnetic nanoparticles for ferroptosis cancer therapy with diagnostic imaging. Bioact Mater 2024; 32:66-97. [PMID: 37822917 PMCID: PMC10562133 DOI: 10.1016/j.bioactmat.2023.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/06/2023] [Accepted: 09/23/2023] [Indexed: 10/13/2023] Open
Abstract
Ferroptosis offers a novel method for overcoming therapeutic resistance of cancers to conventional cancer treatment regimens. Its effective use as a cancer therapy requires a precisely targeted approach, which can be facilitated by using nanoparticles and nanomedicine, and their use to enhance ferroptosis is indeed a growing area of research. While a few review papers have been published on iron-dependent mechanism and inducers of ferroptosis cancer therapy that partly covers ferroptosis nanoparticles, there is a need for a comprehensive review focusing on the design of magnetic nanoparticles that can typically supply iron ions to promote ferroptosis and simultaneously enable targeted ferroptosis cancer nanomedicine. Furthermore, magnetic nanoparticles can locally induce ferroptosis and combinational ferroptosis with diagnostic magnetic resonance imaging (MRI). The use of remotely controllable magnetic nanocarriers can offer highly effective localized image-guided ferroptosis cancer nanomedicine. Here, recent developments in magnetically manipulable nanocarriers for ferroptosis cancer nanomedicine with medical imaging are summarized. This review also highlights the advantages of current state-of-the-art image-guided ferroptosis cancer nanomedicine. Finally, image guided combinational ferroptosis cancer therapy with conventional apoptosis-based therapy that enables synergistic tumor therapy is discussed for clinical translations.
Collapse
Affiliation(s)
- Min Jun Ko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Woojung Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital Harvard Medical School, Cambridge, MA, 02139, USA
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, 60607, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
35
|
Wang J, Li J, Liu J, Chan KY, Lee HS, Lin KN, Wang CC, Lau TS. Interplay of Ferroptosis and Cuproptosis in Cancer: Dissecting Metal-Driven Mechanisms for Therapeutic Potentials. Cancers (Basel) 2024; 16:512. [PMID: 38339263 PMCID: PMC10854932 DOI: 10.3390/cancers16030512] [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: 12/06/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Iron (Fe) and copper (Cu), essential transition metals, play pivotal roles in various cellular processes critical to cancer biology, including cell proliferation, mitochondrial respiration, distant metastases, and oxidative stress. The emergence of ferroptosis and cuproptosis as distinct forms of non-apoptotic cell death has heightened their significance, particularly in connection with these metal ions. While initially studied separately, recent evidence underscores the interdependence of ferroptosis and cuproptosis. Studies reveal a link between mitochondrial copper accumulation and ferroptosis induction. This interconnected relationship presents a promising strategy, especially for addressing refractory cancers marked by drug tolerance. Harnessing the toxicity of iron and copper in clinical settings becomes crucial. Simultaneous targeting of ferroptosis and cuproptosis, exemplified by the combination of sorafenib and elesclomol-Cu, represents an intriguing approach. Strategies targeting mitochondria further enhance the precision of these approaches, providing hope for improving treatment outcomes of drug-resistant cancers. Moreover, the combination of iron chelators and copper-lowering agents with established therapeutic modalities exhibits a synergy that holds promise for the augmentation of anti-tumor efficacy in various malignancies. This review elaborates on the complex interplay between ferroptosis and cuproptosis, including their underlying mechanisms, and explores their potential as druggable targets in both cancer research and clinical settings.
Collapse
Affiliation(s)
- Jinjiang Wang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Jiaxi Li
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jiao Liu
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Kit-Ying Chan
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Ho-Sze Lee
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Kenneth Nansheng Lin
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| | - Tat-San Lau
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong; (J.W.); (K.N.L.); (C.-C.W.)
| |
Collapse
|
36
|
Mehta A, Stanger BZ. Lineage Plasticity: The New Cancer Hallmark on the Block. Cancer Res 2024; 84:184-191. [PMID: 37963209 PMCID: PMC10841583 DOI: 10.1158/0008-5472.can-23-1067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/12/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023]
Abstract
Plasticity refers to the ability of cells to adopt a spectrum of states or phenotypes. In cancer, it is a critical contributor to tumor initiation, progression, invasiveness, and therapy resistance, and it has recently been recognized as an emerging cancer hallmark. Plasticity can occur as a result of cell-intrinsic factors (e.g., genetic, transcriptional, or epigenetic fluctuations), or through cell-extrinsic cues (e.g., signaling from components of the tumor microenvironment or selective pressure from therapy). Over the past decade, technological advances, analysis of patient samples, and studies in mouse model systems have led to a deeper understanding of how such plastic states come about. In this review, we discuss: (i) the definition of plasticity; (ii) methods to measure and quantify plasticity; (iii) the clinical relevance of plasticity; and (iv) therapeutic hypotheses to modulate plasticity in the clinic.
Collapse
Affiliation(s)
- Arnav Mehta
- Department of Medicine, Massachusetts General Hospital, Boston, MA
- Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ben Z. Stanger
- Abramson Family Cancer Research Institute, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
- Department of Cell and Developmental Biology, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
37
|
Xiao L, Xian M, Zhang C, Guo Q, Yi Q. Lipid peroxidation of immune cells in cancer. Front Immunol 2024; 14:1322746. [PMID: 38259464 PMCID: PMC10800824 DOI: 10.3389/fimmu.2023.1322746] [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: 10/16/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Growing evidence indicates that cellular metabolism is a critical determinant of immune cell viability and function in antitumor immunity and lipid metabolism is important for immune cell activation and adaptation to the tumor microenvironment (TME). Lipid peroxidation is a process in which oxidants attack lipid-containing carbon-carbon double bonds and is an important part of lipid metabolism. In the past decades, studies have shown that lipid peroxidation participates in signal transduction to control cell proliferation, differentiation, and cell death, which is essential for cell function execution and human health. More importantly, recent studies have shown that lipid peroxidation affects immune cell function to modulate tumor immunity and antitumor ability. In this review, we briefly overview the effect of lipid peroxidation on the adaptive and innate immune cell activation and function in TME and discuss the effectiveness and sensitivity of the antitumor ability of immune cells by regulating lipid peroxidation.
Collapse
Affiliation(s)
| | | | | | | | - Qing Yi
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston Methodist, Houston, TX, United States
| |
Collapse
|
38
|
Huo K, Yang Y, Yang T, Zhang W, Shao J. Identification of Drug Targets and Agents Associated with Ferroptosis-related Osteoporosis through Integrated Network Pharmacology and Molecular Docking Technology. Curr Pharm Des 2024; 30:1103-1114. [PMID: 38509680 PMCID: PMC11348511 DOI: 10.2174/0113816128288225240318045050] [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: 10/19/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Osteoporosis is a systemic bone disease characterized by progressive reduction of bone mineral density and degradation of trabecular bone microstructure. Iron metabolism plays an important role in bone; its imbalance leads to abnormal lipid oxidation in cells, hence ferroptosis. In osteoporosis, however, the exact mechanism of ferroptosis has not been fully elucidated. OBJECTIVE The main objective of this project was to identify potential drug target proteins and agents for the treatment of ferroptosis-related osteoporosis. METHODS In the current study, we investigated the differences in gene expression of bone marrow mesenchymal stem cells between osteoporosis patients and normal individuals using bioinformatics methods to obtain ferroptosis-related genes. We could predict their protein structure based on the artificial intelligence database of AlphaFold, and their target drugs and binding sites with the network pharmacology and molecular docking technology. RESULTS We identified five genes that were highly associated with osteoporosis, such as TP53, EGFR, TGFB1, SOX2 and MAPK14, which, we believe, can be taken as the potential markers and targets for the diagnosis and treatment of osteoporosis. Furthermore, we observed that these five genes were highly targeted by resveratrol to exert a therapeutic effect on ferroptosis-related osteoporosis. CONCLUSION We examined the relationship between ferroptosis and osteoporosis based on bioinformatics and network pharmacology, presenting a promising direction to the pursuit of the exact molecular mechanism of osteoporosis so that a new target can be discovered for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Kailun Huo
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Yinchuan, Ningxia Hui-Autonomous Region 750004, China
| | - Yiqian Yang
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Yinchuan, Ningxia Hui-Autonomous Region 750004, China
| | - Tieyi Yang
- Department of Orthopedics, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Weiwei Zhang
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Jin Shao
- Department of Orthopedics, Shanghai Pudong New Area Gongli Hospital, Shanghai 200135, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai 200093, China
| |
Collapse
|
39
|
Cai J, Xu X, Saw PE. Nanomedicine targeting ferroptosis to overcome anticancer therapeutic resistance. SCIENCE CHINA. LIFE SCIENCES 2024; 67:19-40. [PMID: 37728804 DOI: 10.1007/s11427-022-2340-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 09/21/2023]
Abstract
A potential reason for the failure of tumor therapies is treatment resistance. Resistance to chemotherapy, radiotherapy, and immunotherapy continues to be a major obstacle in clinic, resulting in tumor recurrence and metastasis. The major mechanisms of therapy resistance are inhibitions of cell deaths, like apoptosis and necrosis, through drug inactivation and excretion, repair of DNA damage, tumor heterogeneity, or changes in tumor microenvironment, etc. Recent studies have shown that ferroptosis play a major role in therapies resistance by inducing phospholipid peroxidation and iron-dependent cell death. Some ferroptosis inducers in combination with clinical treatment techniques have been used to enhance the effect in tumor therapy. Notably, versatile ferroptosis nanoinducers exhibit an extensive range of functions in reversing therapy resistance, including directly triggering ferroptosis and feedback regulation. Herein, we provide a detailed description of the design, mechanism, and therapeutic application of ferroptosis-mediated synergistic tumor therapeutics. We also discuss the prospect and challenge of nanomedicine in tumor therapy resistance by regulating ferroptosis and combination therapy.
Collapse
Affiliation(s)
- Jing Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Foshan, 528200, China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Foshan, 528200, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Foshan, 528200, China.
| |
Collapse
|
40
|
Zhou N, Wei S, Sun T, Xie S, Liu J, Li W, Zhang B. Recent progress in the role of endogenous metal ions in doxorubicin-induced cardiotoxicity. Front Pharmacol 2023; 14:1292088. [PMID: 38143497 PMCID: PMC10748411 DOI: 10.3389/fphar.2023.1292088] [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: 09/11/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Doxorubicin is a widely used anticancer drug in clinical practice for the treatment of various human tumors. However, its administration is associated with cardiotoxicity. Administration of doxorubicin with low side effects for cancer treatment and prevention are, accordingly, urgently required. The human body harbors various endogenous metal ions that exert substantial influences. Consequently, extensive research has been conducted over several decades to investigate the potential of targeting endogenous metal ions to mitigate doxorubicin's side effects and impede tumor progression. In recent years, there has been a growing body of research indicating the potential efficacy of metal ion-associated therapeutic strategies in inhibiting doxorubicin-induced cardiotoxicity (DIC). These strategies offer a combination of favorable safety profiles and potential clinical utility. Alterations in intracellular levels of metal ions have been found to either facilitate or mitigate the development of DIC. For instance, ferroptosis, a cellular death mechanism, and metal ions such as copper, zinc, and calcium have been identified as significant contributors to DIC. This understanding can contribute to advancements in cancer treatment and provide valuable insights for mitigating the cardiotoxic effects of other therapeutic drugs. Furthermore, potential therapeutic strategies have been investigated to alleviate DIC in clinical settings. The ultimate goal is to improve the efficacy and safety of Dox and offer valuable insights for future research in this field.
Collapse
Affiliation(s)
- Ni Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| | - Shanshan Wei
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Taoli Sun
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Suifen Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| | - Jian Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Institute of Clinical Pharmacy, Central South University, Changsha, Hunan, China
- School of Pharmacy, Central South University, Changsha, Hunan, China
| |
Collapse
|
41
|
Wang Y, Lv MN, Zhao WJ. Research on ferroptosis as a therapeutic target for the treatment of neurodegenerative diseases. Ageing Res Rev 2023; 91:102035. [PMID: 37619619 DOI: 10.1016/j.arr.2023.102035] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
Ferroptosis is an iron- and lipid peroxidation (LPO)-mediated programmed cell death type. Recently, mounting evidence has indicated the involvement of ferroptosis in neurodegenerative diseases, especially in Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and so on. Treating ferroptosis presents opportunities as well as challenges for neurodegenerative diseases. This review provides a comprehensive overview of typical features of ferroptosis and the underlying mechanisms that contribute to its occurrence, as well as their implications in the pathogenesis and advancement of major neurodegenerative disorders. Meanwhile, we summarize the utilization of ferroptosis inhibition in both experimental and clinical approaches for the treatment of major neurodegenerative disorders. In addition, we specifically summarize recent advances in developing therapeutic means targeting ferroptosis in these diseases, which may guide future approaches for the effective management of these devastating medical conditions.
Collapse
Affiliation(s)
- Yi Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Meng-Nan Lv
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China
| | - Wei-Jiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China; Department of Cell Biology, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, PR China.
| |
Collapse
|
42
|
Guo Y, Wang J, Hua Y, Jiang M, Xu W, Shi Y, Yang J, Wan H, Yang R. Network pharmacology and in vitro experimental verification to reveal the mechanism of Astragaloside IV against kidney ischemia-reperfusion injury. Heliyon 2023; 9:e21711. [PMID: 38027853 PMCID: PMC10660051 DOI: 10.1016/j.heliyon.2023.e21711] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Ischemic acute kidney injury (AKI) is a prevalent disorder among hospitalized patients worldwide. Astragaloside IV (AS-IV) has been shown to protect against ischemic AKI. However, the specific effects and mechanisms of AS-IV on alleviating kidney ischemia-reperfusion (I/R) injury remain unclear. The objective of this research was to elucidate the regulatory targets and mechanisms through which AS-IV protects kidney I/R injury. A combination of network pharmacology, molecular docking, molecular dynamics (MD) simulation, pharmacodynamic study and Western blot were employed to explore the underlying mechanisms. Network pharmacology revealed that ferroptosis was a potential mechanism of AS-IV against kidney I/R injury. Molecular docking and MD simulations demonstrated strong binding affinity between the GPX4/SLC7A11 and AS-IV. The experimental verification demonstrated that AS-IV improved cell proliferation, decreased the level of ROS and Fe2+, and increased the expressions of GPX4 and SLC7A11 as same as Ferrostatin-1 in OGD/R-injured HUVECs. In conclusion, AS-IV had a significant inhibition on ferroptosis in kidney I/R injury, providing a new perspective for drug development on kidney I/R injury. Definitely, further exploration in vivo is necessary to fully understand whether AS-IV alleviates kidney I/R injury through inhibiting endothelial ferroptosis.
Collapse
Affiliation(s)
- Yan Guo
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, 310053, China
| | - Jinfu Wang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yanjie Hua
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Mengya Jiang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, 310053, China
| | - Wanyue Xu
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, 310053, China
| | - Yanpeng Shi
- Linping Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, Zhejiang, 310053, China
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Ruchun Yang
- Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, 310053, China
| |
Collapse
|
43
|
Shan Q, Zhang C, Li Y, Li Q, Zhang Y, Li X, Shi J, Hu F. SLC7A11, a potential immunotherapeutic target in lung adenocarcinoma. Sci Rep 2023; 13:18302. [PMID: 37880315 PMCID: PMC10600206 DOI: 10.1038/s41598-023-45284-z] [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: 07/05/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023] Open
Abstract
SLC7A11 has significant translational value in cancer treatment. However, there are few studies on whether SLC7A11 affects the immune status of lung adenocarcinoma (LUAD). Information on SLC7A11 expression and its impact on prognosis was obtained from the cancer genome atlas and gene expression omnibus databases. The differentially expressed genes (DEGs) were analysed by GO and KEGG. GSEA enrichment analysis was performed in the SLC7A11-high and SLC7A11-low groups. The relationship between SLC7A11 and tumour immunity, immune checkpoints, and immune cell infiltration was studied using R language. We analysed the correlation between SLC7A11 and chemotactic factors (CFs) and chemokine receptors using the TISIDB database. SLC7A11 is overexpressed in many tumours, including LUAD. The 5-year overall survival of patients in the SLC7A11-high group was lower than in the SLC7A11-low group. KEGG analysis found that the DEGs were enriched in ferroptosis signaling pathways. GSEA analysis found that the survival-related signaling pathways were enriched in the SLC7A11-low group. The SLC7A11-low group had higher immune scores and immune checkpoint expression. SLC7A11 was negatively correlated with many immune cells (CD8+ T cells, immature dendritic cells), CFs, chemokine receptors (such as CCL17/19/22/23, CXCL9/10/11/14, CCR4/6, CX3CR1, CXCR3) and MHCs (major histocompatibility complex). SLC7A11 may regulate tumour immunity and could be a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
- Qingqing Shan
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Chi Zhang
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Yangke Li
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China.
| | - Qunying Li
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China.
| | - Yifan Zhang
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Xue Li
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Junqing Shi
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China
| | - Fengying Hu
- Department of Respiration, Chengdu First People's Hospital, No. 18, Wangxiang North Road, High-Tech Zone, Chengdu, 610041, Sichuan Province, People's Republic of China
| |
Collapse
|
44
|
Wang L, Li Z, Li Z, Ren Y, Qian L, Yu Y, Shi W, Xiong Y. Identification of A Novel Gene Signature Combining Ferroptosis- and Immunity-Related Genes for Prognostic Prediction, Immunotherapy and Potential Therapeutic Targets in Gastric Cancer. J Cancer 2023; 14:3457-3476. [PMID: 38021154 PMCID: PMC10647194 DOI: 10.7150/jca.87223] [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: 06/16/2023] [Accepted: 09/17/2023] [Indexed: 12/01/2023] Open
Abstract
Gastric cancer (GC) is one of the most prevalent cancers worldwide. Ferroptosis and the immune status of tumor tissue play vital roles in the initiation and progression of GC. However, the role and functional mechanisms of ferroptosis- and immunity-related genes (FIRGs) in GC pathogenesis and their correlations with GC prognosis have not been elucidated. We aim to establish a prognostic prediction model based on the FIRGs signature for GC patients. Differentially expressed genes were screened from the Cancer Genome Atlas (TCGA) GC cohorts. The least absolute shrinkage and selection operator (LASSO) regression was performed to establish a FIRGs-based risk model. This gene signature with 7 FIRGs was identified as an independent prognostic factor. A nomogram incorporating clinical parameters and the FIRG signature was constructed to individualize outcome predictions. Finally, we provided in vivo and in vitro evidence to verify the reliability of FIRG signature for GC prognosis, and validate the expression and function of FIRGs contributing to the development and progression of GC. Herein, our work represents great therapeutic and prognostic potentials for GC.
Collapse
Affiliation(s)
- Liwei Wang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Zi Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| | - Wenzhen Shi
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Medical Research Center, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi, P.R. China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, P.R. China
| |
Collapse
|
45
|
Bao W, Wang J, Fan K, Gao Y, Chen J. PIAS3 promotes ferroptosis by regulating TXNIP via TGF-β signaling pathway in hepatocellular carcinoma. Pharmacol Res 2023; 196:106915. [PMID: 37689128 DOI: 10.1016/j.phrs.2023.106915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/11/2023]
Abstract
Ferroptosis has been suggested to play a potential role in cancer therapy as an iron-dependent programmed cell death mechanism distinct from other forms. Hepatocellular carcinoma (HCC) remains a great threat, with high mortality and limited therapeutic options. The induction of ferroptosis has emerged as a novel and promising therapeutic strategy for HCC. In the present study, we identified protein inhibitor of activated STAT3 (PIAS3) as a driver of ferroptosis in HCC using TMT-based quantitative proteomics and ferroptosis-related functional assays. Mechanistically, thioredoxin-interacting protein (TXNIP) was confirmed to be PIAS3 in promoting ferroptotic cell death, based on RNA-seq analysis. Knockdown of TXNIP degrades ferroptotic susceptibility caused by PIAS3-overexpression, whereas transfection-forced reexpression of TXNIP restores sensitivity to ferroptosis in PIAS3-downregulated cells. PIAS3 interacts with SMAD2/3 to activate transforming growth factor (TGF)-β signaling, leading to increased TXNIP expression. Our study revealed the critical role of PIAS3 in ferroptosis and a novel actionable axis-PIAS3/TGF-β/TXNIP that could govern ferroptotic sensitivity, paving the path for using ferroptosis as an efficient approach in HCC therapies.
Collapse
Affiliation(s)
- Wenfang Bao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Jialin Wang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Kailing Fan
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| | - Jingde Chen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Department of Oncology, Ji'an Hospital, Shanghai East Hospital, Ji'an 343000, China.
| |
Collapse
|
46
|
Wang X, Zhu W, Xing M, Zhu H, Chen E, Zhou J. Matrine disrupts Nrf2/GPX4 antioxidant system and promotes hepatocyte ferroptosis. Chem Biol Interact 2023; 384:110713. [PMID: 37716422 DOI: 10.1016/j.cbi.2023.110713] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/14/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Matrine (MT) is an alkaloid isolated from Sophora flavescens with various bioactivities and is widely used clinically. However, the broader its clinical use, the greater its toxicity concerns. We investigate the role of ferroptosis in MT-induced liver injury caused by an imbalance in the antioxidant pathway. Our results showed that MT could cause pathological changes in liver tissues and lead to a significant reduction in L02 cell viability. MT also reduced superoxide dismutase (SOD) and glutathione (GSH), increased malondialdehyde (MDA), reactive oxygen species (ROS), and lipid peroxidation levels, and disrupted iron homeostasis, leading to ferroptosis. In addition, MT decreased the protein levels of FTH, Nrf2, xCT, GPX4, HO-1 and ferroptosis suppressor protein 1 (FSP1) and increased the protein levels of TRF1 and DMT1, characteristic indicators of ferroptosis. Interestingly, the cytotoxic effects of MT were alleviated by ferroptosis inhibitor, Nrf2 agonist, or selenium supplementation. These results revealed that MT triggers hepatocyte ferroptosis by inhibiting the Nrf2/GPX4 antioxidant system.
Collapse
Affiliation(s)
- Xi Wang
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Wenjing Zhu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Miao Xing
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Haiyan Zhu
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Enqing Chen
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China
| | - Jie Zhou
- School of Medicine, Yichun University, 576 XueFu Road, Yuanzhou District, Yichun, 336000, PR China.
| |
Collapse
|
47
|
Zhang J, Luo Z, Zheng Y, Cai Q, Jiang J, Zhang H, Duan M, Chen Y, Xia J, Qiu Z, Zeng J, Huang C. A bibliometric study and visualization analysis of ferroptosis-inducing cancer therapy. Heliyon 2023; 9:e19801. [PMID: 37809417 PMCID: PMC10559163 DOI: 10.1016/j.heliyon.2023.e19801] [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: 05/06/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Ferroptosis is a form of regulated cell death that was first formally proposed a decade ago. While its role in cancer cell death was initially understudied, it has recently gained considerable interest from researchers. In recent years, a growing number of studies have focused on the role of ferroptosis in cancer progression, with the goal of developing novel ferroptosis-inducing cancer therapies. This study aims to present the developmental trend and hotspots of research on ferroptosis-inducing cancer therapy using bibliometric analysis. A literature search was conducted using the Web of Science Core Collection on October 1st, 2022, to retrieve articles and reviews pertaining to ferroptosis and cancer published from 2012 to 2022. Microsoft Excel 2016, VOSviewer 1.6.18 and CiteSpace (version 6.1. R6) were utilized to conduct the bibliometric analysis of publication trends, authorship, and citation networks, with a focus on identifying countries, institutions, journals, and authors contributing to the field. These analyses were used to predict future trends in this area. A total of 2839 articles were identified and extracted for analysis. The number of publications has increased almost every year, with a sharp increase after 2018. China produced the most publications in this area, followed by the United States. Central South University was the institution that published the most papers. Frontiers in Oncology was the journal with the highest number of publications, while Cell had the greatest impact factor. Daolin Tang was the most productive author and Dixon SJ was the most influential author. Co-occurrence and burst analyses of keywords and references were conducted to identify the developmental trends and hotspots in ferroptosis-inducing cancer therapy research. Main research directions have shifted from investigating the mechanism of ferroptosis to developing novel ferroptosis-targeting cancer therapies. Emerging topicsfocus on the role of ferroptosis in solid tumor therapy. Based on our bibliometric analysis, we predict that research on ferroptosis in cancer therapy will continue to be a hot topic in the future, with a growing number of treatment modalities related to ferroptosis being developed. Our study provides valuable insights into the current state and future trends of research in this field, serving as a useful guide for researchers seeking to make important contributions in this area.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zai Luo
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yang Zheng
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qianqian Cai
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jie Jiang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Haoliang Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Mingyu Duan
- Department of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yanmin Chen
- Department of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jiayang Xia
- Department of Education, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhengjun Qiu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jvdan Zeng
- Department of Obstetrics and Gynecology, The International Peace Maternity & Child Health Hospital of China Welfare Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Chen Huang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- The Affiliated Chuzhou Hospital of Anhui Medical University, Anhui, 239000, China
| |
Collapse
|
48
|
Tang J, Long G, Li X, Zhou L, Zhou Y, Wu Z. The deubiquitinase EIF3H promotes hepatocellular carcinoma progression by stabilizing OGT and inhibiting ferroptosis. Cell Commun Signal 2023; 21:198. [PMID: 37559097 PMCID: PMC10413709 DOI: 10.1186/s12964-023-01220-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/09/2023] [Indexed: 08/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal human malignancies, and with quite limited treatment alternatives. The proteasome is responsible for most of the protein degradation in eukaryotic cells and required for the maintenance of intracellular homeostasis. However, its potential role in HCC is largely unknown. In the current study, we identified eukaryotic translation initiation factor 3 subunit H (EIF3H), belonging to the JAB1/MPN/MOV34 (JAMM) superfamily, as a bona fide deubiquitylase of O-GlcNAc transferase (OGT) in HCC. We explored that EIF3H was positively associated with OGT in HCC and was related to the unfavorable prognosis. EIF3H could interact with, deubiquitylate, and stabilize OGT in a deubiquitylase-dependent manner. Specifically, EIF3H was associated with the GT domain of ERα via its JAB/MP domain, thus inhibiting the K48-linked ubiquitin chain on OGT. Besides, we demonstrated that the knockdown of EIF3H significantly reduced OGT protein expression, cell proliferation and invasion, and caused G1/S arrest of HCC. We also found that the deletion of EIF3H prompted ferroptosis in HCC cells. Finally, the effects of EIF3H depletion could be reversed by further OGT overexpression, implying that the OGT status is indispensable for EIF3H function in HCC carcinogenesis. In summary, our study described the oncogenic function of EIF3H and revealed an interesting post-translational mechanism between EIF3H, OGT, and ferroptosis in HCC. Targeting the EIF3H may be a promising approach in HCC. Video Abstract.
Collapse
Affiliation(s)
- Jianing Tang
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guo Long
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xuanxuan Li
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ledu Zhou
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yangying Zhou
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Zheyu Wu
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Orthopedics, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200000, China.
| |
Collapse
|
49
|
Choi J, Choi H, Chung J. Icariin Supplementation Suppresses the Markers of Ferroptosis and Attenuates the Progression of Nonalcoholic Steatohepatitis in Mice Fed a Methionine Choline-Deficient Diet. Int J Mol Sci 2023; 24:12510. [PMID: 37569885 PMCID: PMC10419585 DOI: 10.3390/ijms241512510] [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: 06/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Icariin, a flavonoid abundant in the herb Epimedium, exhibits anti-ferroptotic activity. However, its impact on nonalcoholic steatohepatitis (NASH) development remains unclear. This study aimed to investigate the potential role of icariin in mitigating methionine choline-deficient (MCD) diet-induced NASH in C57BL/6J mice. The results showed that icariin treatment significantly reduced serum alanine aminotrasferase and aspartate aminotransferase activities while improving steatosis, inflammation, ballooning, and fibrosis in the liver tissues of mice fed the MCD diet. These improvements were accompanied by a substantial reduction in the hepatic iron contents and levels of malondialdehyde and 4-hydroxynonenal, as well as an increase in the activities of catalase and superoxide dismutase. Notably, icariin treatment suppressed the hepatic protein levels of ferroptosis markers such as acyl-CoA synthetase long-chain family member 4 and arachidonate 12-lipoxygenase, which were induced by the MCD diet. Furthermore, transmission electron microscopy confirmed the restoration of morphological changes in the mitochondria, a hallmark characteristic of ferroptosis, by icariin. Additionally, icariin treatment significantly increased the protein levels of Nrf2, a cystine/glutamate transporter (xCT), and glutathione peroxidase 4 (GPX4). In conclusion, our study suggests that icariin has the potential to attenuate NASH, possibly by suppressing ferroptosis via the Nrf2-xCT/GPX4 pathway.
Collapse
Affiliation(s)
| | | | - Jayong Chung
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea; (J.C.); (H.C.)
| |
Collapse
|
50
|
Wang X, Kong X, Feng X, Jiang DS. Effects of DNA, RNA, and Protein Methylation on the Regulation of Ferroptosis. Int J Biol Sci 2023; 19:3558-3575. [PMID: 37497000 PMCID: PMC10367552 DOI: 10.7150/ijbs.85454] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
Ferroptosis is a form of programmed cell death characterized by elevated intracellular ferrous ion levels and increased lipid peroxidation. Since its discovery and characterization in 2012, considerable progress has been made in understanding the regulatory mechanisms and pathophysiological functions of ferroptosis. Recent findings suggest that numerous organ injuries (e.g., ischemia/reperfusion injury) and degenerative pathologies (e.g., aortic dissection and neurodegenerative disease) are driven by ferroptosis. Conversely, insufficient ferroptosis has been linked to tumorigenesis. Furthermore, a recent study revealed the effect of ferroptosis on hematopoietic stem cells under physiological conditions. The regulatory mechanisms of ferroptosis identified to date include mainly iron metabolism, such as iron transport and ferritinophagy, and redox systems, such as glutathione peroxidase 4 (GPX4)-glutathione (GSH), ferroptosis-suppressor-protein 1 (FSP1)-CoQ10, FSP1-vitamin K (VK), dihydroorotate dehydrogenase (DHODH)-CoQ, and GTP cyclohydrolase 1 (GCH1)-tetrahydrobiopterin (BH4). Recently, an increasing number of studies have demonstrated the important regulatory role played by epigenetic mechanisms, especially DNA, RNA, and protein methylation, in ferroptosis. In this review, we provide a critical analysis of the molecular mechanisms and regulatory networks of ferroptosis identified to date, with a focus on the regulatory role of DNA, RNA, and protein methylation. Furthermore, we discuss some debated findings and unanswered questions that should be the foci of future research in this field.
Collapse
Affiliation(s)
- Xiancan Wang
- Department of Cardiovascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Xianghai Kong
- Department of Intervention & Vascular Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and echnology, Wuhan, 430014, Hubei, China
| | - Xin Feng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|