101
|
Hushmandi K, Klionsky DJ, Aref AR, Bonyadi M, Reiter RJ, Nabavi N, Salimimoghadam S, Saadat SH. Ferroptosis contributes to the progression of female-specific neoplasms, from breast cancer to gynecological malignancies in a manner regulated by non-coding RNAs: Mechanistic implications. Noncoding RNA Res 2024; 9:1159-1177. [PMID: 39022677 PMCID: PMC11250880 DOI: 10.1016/j.ncrna.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/27/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis, a recently identified type of non-apoptotic cell death, triggers the elimination of cells in the presence of lipid peroxidation and in an iron-dependent manner. Indeed, ferroptosis-stimulating factors have the ability of suppressing antioxidant capacity, leading to the accumulation of reactive oxygen species (ROS) and the subsequent oxidative death of the cells. Ferroptosis is involved in the pathophysiological basis of different maladies, such as multiple cancers, among which female-oriented malignancies have attracted much attention in recent years. In this context, it has also been unveiled that non-coding RNA transcripts, including microRNAs, long non-coding RNAs, and circular RNAs have regulatory interconnections with the ferroptotic flux, which controls the pathogenic development of diseases. Furthermore, the potential of employing these RNA transcripts as therapeutic targets during the onset of female-specific neoplasms to modulate ferroptosis has become a research hotspot; however, the molecular mechanisms and functional alterations of ferroptosis still require further investigation. The current review comprehensively highlights ferroptosis and its association with non-coding RNAs with a focus on how this crosstalk affects the pathogenesis of female-oriented malignancies, from breast cancer to ovarian, cervical, and endometrial neoplasms, suggesting novel therapeutic targets to decelerate and even block the expansion and development of these tumors.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Mojtaba Bonyadi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
102
|
Alrouji M, Anwar S, Venkatesan K, Shahwan M, Hassan MI, Islam A, Shamsi A. Iron homeostasis and neurodegeneration in the ageing brain: Insight into ferroptosis pathways. Ageing Res Rev 2024; 102:102575. [PMID: 39515619 DOI: 10.1016/j.arr.2024.102575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/25/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Ageing is a major risk factor for various chronic diseases and offers a potential target for developing novel and broadly effective preventatives or therapeutics for age-related conditions, including those affecting the brain. Mechanisms contributing to ageing have been summarized as the hallmarks of ageing, with iron imbalance being one of the major factors. Ferroptosis, an iron-mediated lipid peroxidation-induced programmed cell death, has recently been implicated in neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Addressing ferroptosis offers both opportunities and challenges for treating neurodegenerative diseases, though the specific mechanisms remain unclear. This research explores the key processes behind how ferroptosis contributes to brain ageing, with a focus on the complex signaling networks that are involved. The current article aims to uncover that how ferroptosis, a specific type of cell death, may drive age-related changes in the brain. Additionally, the article also unveils its role in neurodegenerative diseases, discussing how understanding these mechanisms could open up new therapeutic avenues.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Saleha Anwar
- Center for Global Health Research, Saveetha medical college, Saveetha institute of Medical and Technical Sciences, Chennai, India.
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| | - Md Imtaiyaz Hassan
- Center for Interdsicplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Asimul Islam
- Center for Interdsicplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Anas Shamsi
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| |
Collapse
|
103
|
Peng Q, Deng Y, Xu Z, Duan R, Wang W, Wang S, Hong Y, Wang Q, Zhang Y. Fat mass and obesity-associated protein alleviates cerebral ischemia/reperfusion injury by inhibiting ferroptosis via miR-320-3p/SLC7A11 axis. Biochem Pharmacol 2024; 230:116603. [PMID: 39486461 DOI: 10.1016/j.bcp.2024.116603] [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: 07/11/2024] [Revised: 09/27/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Fat mass and obesity-associated protein (FTO) is a demethylase and has recently been found to have a protective effect in acute ischemic stroke (AIS), but the underlying mechanism is unclear to a large extent. New studies have found that the expression of certain miRNAs may be affected by N6-methyladenosine (m6A) levels. Here, using high-throughput sequencing and quantitative polymerase chain reaction, we found miR-320-3p was significantly up-regulated in AIS patients. miR-320-3p aggravated the neurobehavioral manifestation, infarct volume and histopathology of middle cerebral artery occlusion/reperfusion model mice. Mechanically, miR-320-3p binds to the 3' untranslated region of solute carrier family 7 member 11 (SLC7A11) mRNA, promoting oxidative stress and ferroptosis induced by oxygen-glucose deprivation/reoxygenation in neurons. FTO inhibited the m6A methylation of the primary transcript pri-miR-320 and the maturation of miR-320-3p, thus having a protective effect on cerebral ischemia/reperfusion injury after AIS. Clinically, we also confirmed the down-regulation of FTO and SLC7A11 mRNA in the peripheral blood of AIS patients and their correlation with the expression of miR-320-3p. Our study found that FTO inhibits ferroptosis through miR-320-3p/SLC7A11 axis in an m6A-dependent manner, and thus has a protective effect on cerebral ischemic reperfusion injury. Our results provided a promising therapeutic target of cerebral ischemia/reperfusion injury after AIS.
Collapse
Affiliation(s)
- Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Yang Deng
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210006, Jiangsu, China
| | - Zhaohan Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Wei Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Shiyao Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| | - Qingguang Wang
- Department of Neurology, Jiangyin Hospital Affiliated to Nantong University, Wuxi 214400, Jiangsu, China.
| | - Yingdong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| |
Collapse
|
104
|
Furuya-Ikude C, Kitta A, Tomonobu N, Kawasaki Y, Sakaguchi M, Kondo E. NCF-1 plays a pivotal role in the survival of adenocarcinoma cells of pancreatic and gastric origins. In Vitro Cell Dev Biol Anim 2024; 60:1151-1159. [PMID: 39666242 DOI: 10.1007/s11626-024-00994-0] [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: 10/06/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024]
Abstract
Reactive oxygen species (ROS) play a pivotal biological role in cells, with ROS function differing depending on cellular conditions and the extracellular environment. Notably, ROS act as cytotoxic factors to eliminate infectious pathogens or promote cell death under cellular stress, while also facilitating cell growth (via ROS-sensing pathways) by modifying gene expression. Among ROS-related genes, neutrophil cytosolic factor-1 (NCF-1; p47phox) was identified as a ROS generator in neutrophils. This product is a subunit of a cytosolic NADPH oxidase complex activated in response to pathogens such as bacteria and viruses. NCF-1 has been examined primarily in terms of ROS-production pathways in macrophages and neutrophils; however, the expression of this protein and its biological role in cancer cells remain unclear. Here, we report expression of NCF-1 in pancreatic and gastric cancers, and demonstrate its biological significance in these tumor cells. Abundant expression of NCF-1 was observed in pancreatic adenocarcinoma (PDAC) lines and in patient tissues, as well as in gastric adenocarcinomas. Accumulation of the protein was also detected in the invasive/metastatic foci of these tumors. Unexpectedly, BxPC-3 underwent apoptotic cell death when transfected with a small interfering RNA (siRNA) specific to NCF-1, whereas the cells treated with a control siRNA proliferated in a time-dependent manner. A similar phenomenon was observed in HSC-58, a poorly differentiated gastric adenocarcinoma line. Consequently, the tumor cells highly expressing NCF-1 obtained coincident accumulation of ROS and reduced glutathione (GSH) with expression of glutathione peroxidase 4 (GPX4), a quencher involved in ferroptosis. Unlike the conventional role of ROS as a representative cytotoxic factor, these findings suggest that NCF-1-mediated ROS generation may be required for expansive growth of PDAC and gastric cancers.
Collapse
Affiliation(s)
- Chiemi Furuya-Ikude
- Division of Tumor Pathology, NIR-PIT Research Institute, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Akane Kitta
- Division of Tumor Pathology, NIR-PIT Research Institute, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Naoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Yoshihiro Kawasaki
- Division of Tumor Pathology, NIR-PIT Research Institute, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan.
| | - Eisaku Kondo
- Division of Tumor Pathology, NIR-PIT Research Institute, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| |
Collapse
|
105
|
Zhang M, Xu L, Zhu C, Zhang Y, Luo R, Ren J, Yu J, Zhang Y, Liang G, Zhang Y. Magnoflorine ameliorates hepatic fibrosis and hepatic stellate cell activation by regulating ferroptosis signaling pathway. Heliyon 2024; 10:e39892. [PMID: 39634391 PMCID: PMC11615489 DOI: 10.1016/j.heliyon.2024.e39892] [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: 07/30/2024] [Revised: 10/25/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
Liver fibrosis is a chronic liver disease that brings a heavy economic burden to the world and has attracted global attention. Although the pathological mechanisms and treatment strategies of liver fibrosis have been extensively studied, there are currently no effective targeted drugs for the prevention and treatment of liver fibrosis in clinical practice. Therefore, it is imperative to seek and develop effective treatment strategies and drugs for liver fibrosis. Magnoflorine (MAG) is a natural product with multiple pharmacological activities. Thus, in this study, we will explore the effect of MAG on alleviating liver fibrosis in mice and its mechanism of action. Our study indicates that MAG can alleviate liver damage, improve liver collagen deposition, and significantly reduced the expression levels of hepatic stellate cells (HSCs) activation markers in vivo. Additionally, the findings of this study indicate that MAG can inhibit the transforming growth factor-beta (TGF-β)/Smad signaling pathway. Bioinformatics analysis suggests that the alleviating effect of MAG on liver fibrosis may be associated with ferroptosis. Interestingly, in vitro experiments have demonstrated that MAG slows down the progression of liver fibrosis by inhibiting the activation of HSCs, and further confirms that MAG promotes ferroptosis in ROS-mediated activated HSCs. In short, MAG has a good alleviating effect on liver fibrosis and will be a potential candidate drug for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Lenan Xu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Chengkai Zhu
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Yawen Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Ruixiang Luo
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Juan Ren
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Jie Yu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Yanmei Zhang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, 310012, China
| |
Collapse
|
106
|
Liu D, Zhu Y. Unveiling Smyd-2's Role in Cytoplasmic Nrf-2 Sequestration and Ferroptosis Induction in Hippocampal Neurons After Cerebral Ischemia/Reperfusion. Cells 2024; 13:1969. [PMID: 39682718 PMCID: PMC11639856 DOI: 10.3390/cells13231969] [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: 10/25/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
SET and MYND Domain-Containing 2 (Smyd-2), a specific protein lysine methyltransferase (PKMT), influences both histones and non-histones. Its role in cerebral ischemia/reperfusion (CIR), particularly in ferroptosis-a regulated form of cell death driven by lipid peroxidation-remains poorly understood. This study identifies the expression of Smyd-2 in the brain and investigates its relationship with neuronal programmed cell death (PCD). We specifically investigated how Smyd-2 regulates ferroptosis in CIR through its interaction with the Nuclear Factor Erythroid-2-related Factor-2 (Nrf-2)/Kelch-like ECH-associated protein (Keap-1) pathway. Smyd-2 knockout protects HT-22 cells from Erastin-induced ferroptosis but not TNF-α + Smac-mimetic-induced apoptosis/necroptosis. This neuroprotective effect of Smyd-2 knockout in HT-22 cells after Oxygen-Glucose Deprivation/Reperfusion (OGD/R) was reversed by Erastin. Smyd-2 knockout in HT-22 cells shows neuroprotection primarily via the Nuclear Factor Erythroid-2-related Factor-2 (Nrf-2)/Kelch-like ECH-associated protein (Keap-1) pathway, despite the concurrent upregulation of Smyd-2 and Nrf-2 observed in both the middle cerebral artery occlusion (MCAO) and OGD/R models. Interestingly, vivo experiments demonstrated that Smyd-2 knockout significantly reduced ferroptosis and lipid peroxidation in hippocampal neurons following CIR. Moreover, the Nrf-2 inhibitor ML-385 abolished the neuroprotective effects of Smyd-2 knockout, confirming the pivotal role of Nrf-2 in ferroptosis regulation. Cycloheximide (CHX) fails to reduce Nrf-2 expression in Smyd-2 knockout HT-22 cells. Smyd-2 knockout suppresses Nrf-2 lysine methylation, thereby promoting the Nrf-2/Keap-1 pathway without affecting the PKC-δ/Nrf-2 pathway. Conversely, Smyd-2 overexpression disrupts Nrf-2 nuclear translocation, exacerbating ferroptosis and oxidative stress, highlighting its dual regulatory role. This study underscores Smyd-2's potential for ischemic stroke treatment by disrupting the Smyd-2/Nrf-2-driven antioxidant capacity, leading to hippocampal neuronal ferroptosis. By clarifying the intricate interplay between ferroptosis and oxidative stress via the Nrf-2/Keap-1 pathway, our findings provide new insights into the molecular mechanisms of CIR and identify Smyd-2 as a promising therapeutic target.
Collapse
Affiliation(s)
- Daohang Liu
- School of Pharmacy, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 201203, China;
| | - Yizhun Zhu
- School of Pharmacy, Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 201203, China;
- School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
107
|
Ge A, Xiang W, Li Y, Zhao D, Chen J, Daga P, Dai CC, Yang K, Yan Y, Hao M, Zhang B, Xiao W. Broadening horizons: the multifaceted role of ferroptosis in breast cancer. Front Immunol 2024; 15:1455741. [PMID: 39664391 PMCID: PMC11631881 DOI: 10.3389/fimmu.2024.1455741] [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: 06/27/2024] [Accepted: 10/21/2024] [Indexed: 12/13/2024] Open
Abstract
Breast cancer poses a serious threat to women's health globally. Current radiotherapy and chemotherapy regimens can induce drug-resistance effects in cancer tissues, such as anti-apoptosis, anti-pyroptosis, and anti-necroptosis, leading to poor clinical outcomes in the treatment of breast cancer. Ferroptosis is a novel programmed cell death modality characterized by iron overload, excessive generation of reactive oxygen species, and membrane lipid peroxidation. The occurrence of ferroptosis results from the imbalance between intracellular peroxidation mechanisms (executive system) and antioxidant mechanisms (defensive system), specifically involving iron metabolism pathways, amino acid metabolism pathways, and lipid metabolism pathways. In recent years, it has been found that ferroptosis is associated with the progression of various diseases, including tumors, hypertension, diabetes, and Alzheimer's disease. Studies have confirmed that triggering ferroptosis in breast cancer cells can significantly inhibit cancer cell proliferation and invasion, and improve cancer cell sensitivity to radiotherapy and chemotherapy, making induction of ferroptosis a potential strategy for the treatment of breast cancer. This paper reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including signaling pathways such as GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, and GCH1-BH4) in breast cancer disease, the latest research progress, and summarizes the research on ferroptosis in breast cancer disease within the framework of metabolism, reactive oxygen biology, and iron biology. The key regulatory factors and mechanisms of ferroptosis in breast cancer disease, as well as important concepts and significant open questions in the field of ferroptosis and related natural compounds, are introduced. It is hoped that future research will make further breakthroughs in the regulatory mechanisms of ferroptosis and the use of ferroptosis in treating breast cancer cells. Meanwhile, natural compounds may also become a new direction for potential drug development targeting ferroptosis in breast cancer treatment. This provides a theoretical basis and opens up a new pathway for research and the development of drugs for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wang Xiang
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| | - Yan Li
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Da Zhao
- 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, United States
- Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China
| | - Pawan Daga
- Department of Internal Medicine, University of Louisville, Louisville, KY, United States
| | - Charles C. Dai
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD, United States
| | - Kailin Yang
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yexing Yan
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic Laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | | | - Wei Xiao
- Department of Rheumatology, The First People’s Hospital Changde City, Changde, Hunan, China
| |
Collapse
|
108
|
Feng Y, Shi M, Zhang Y, Li X, Yan L, Xu J, Liu C, Li M, Bai F, Yuan F, Sun Y, Liu R, Zhao Y, Yang L, Zhang Y, Guo Y, Zhang J, Zhou R, Liu P. Protocatechuic acid relieves ferroptosis in hepatic lipotoxicity and steatosis via regulating NRF2 signaling pathway. Cell Biol Toxicol 2024; 40:104. [PMID: 39589556 PMCID: PMC11599353 DOI: 10.1007/s10565-024-09953-7] [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: 06/26/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Ferroptosis represents a newly programmed cell death, and the process is usually accompanied with iron-dependent lipid peroxidation. Importantly, ferroptosis is implicated in a myriad of diseases. Recent literature suggests a potential position of ferroptosis in the pathogenesis of metabolic dysfunction-associated fatty liver disease (MAFLD), the most widespread liver ailment worldwide. Intriguingly, several functional genes and metabolic pathways central to ferroptosis are regulated by nuclear factor erythroid-derived 2-like 2 (NRF2). In current work, we aim to identify protocatechuic acid (PCA), a primary metabolite of antioxidant polyphenols, as a potent NRF2 activator and ferroptosis inhibitor in the hepatic lipotoxicity and steatosis models. Herein, both NRF2+/+ and NRF2-/- cell lines and mice were used to analyze the importance of NRF2 in PCA function, and hepatic lipotoxicity and steatosis models were induced by palmitic acid and high-fat diet respectively. Our results indicated that ferroptosis was mitigated by PCA intervention in hepatic cells. Furthermore, PCA exhibited therapeutic efficacy against ferroptosis, as well as hepatic lipotoxicity and steatosis. The protective role of PCA was predominantly mediated through NRF2 activation, potentially elucidating a pivotal mechanism underlying PCA's therapeutic impact on MAFLD. Additionally, the augmented mitochondrial TCA cycle activity observed in hepatic lipotoxicity and steatosis models was ameliorated by PCA, in part via NRF2-dependent pathways, further bolstering PCA's anti-ferroptosis properties. Collectively, our findings underscore PCA's potential in alleviating hepatic ferroptosis, lipotoxicity and steatosis via inducing activation of NRF2 signaling pathway, offering a promising strategy for the therapy of MAFLD as well as related lipid metabolic disorders.
Collapse
Affiliation(s)
- Yetong Feng
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjiao Shi
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Yi Zhang
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Xinyan Li
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Liangwen Yan
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Jiayi Xu
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Chenyue Liu
- Department of Medical Image, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Miaomiao Li
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Fengyun Bai
- Shaanxi Dongtai Pharmaceutical Co., LTD, Xianyang, China
| | - Fenyue Yuan
- Shaanxi Dongtai Pharmaceutical Co., LTD, Xianyang, China
| | - Ying Sun
- Shaanxi Dongtai Pharmaceutical Co., LTD, Xianyang, China
| | - Rongrong Liu
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Yaping Zhao
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Lan Yang
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinggang Zhang
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Ying Guo
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China
| | - Jian Zhang
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Rui Zhou
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pengfei Liu
- International Joint Research Center On Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second, Xi'an, China.
- Key Laboratory of Environment and Genes Related To Diseases, Ministry of Education of China, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
109
|
Su H, Tan G, Guo W, Yu JS, Xu Z, Zhuang R, Xue H. Discovery of potential ferroptosis and osteoporosis biomarkers through TMT proteomics and bioinformatics analysis. Biomed Eng Online 2024; 23:120. [PMID: 39580392 PMCID: PMC11585094 DOI: 10.1186/s12938-024-01307-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 10/28/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Primary osteoporosis has increasingly emerged as a major issue affecting human health, with a complex specific pathogenic mechanism. As a research hotspot, ferroptosis plays a vital role in the pathogenesis of primary osteoporosis, aiming to explore the link and specific target genes between ferroptosis and primary osteoporosis. METHODS By utilizing TMT proteomics and bioinformatics analyses, we elucidated the linkages and key targets of the ferroptosis pathway in an ovariectomized osteoporotic rat model. Forty 12-week-old SD female rats were employed in the study, of which 20 female SD rats were ovariectomized as the OVX group and 20 female SD rats were employed as the SHAM group. At the end of the experiments, the femurs of the rats were excised for computed tomography tests and used for hematoxylin and eosin staining. Finally, we extracted bone tissue proteins for TMT proteomics analysis and protein blotting verification. RESULTS The proteomics results of the VX and SHAM groups showed that 133 proteins were significantly changed, of which 91 proteins were upregulated and 42 proteins were downregulated, including TXN, TMSB4X, TFRC, TF, RELA, PARP14, CP, CAPG, and ADIPOQ. The expression of key proteins in the bone tissues was detected by protein blotting. The expression of TFR1, TFRC and TF was upregulated, whereas the expression of Cp, TXN and BMP-2 was downregulated. CONCLUSIONS TMT proteomics and functional enrichment analyses in our study substantiated that in osteoporosis, disturbances in lipid metabolism lead to the emergence of oxidative stress with iron homeostasis imbalance.
Collapse
Affiliation(s)
- Hui Su
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Guoqing Tan
- Department of Spine and Spinal Cord, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - WenXuan Guo
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Department of Traumatology and Orthopedics, The First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jin Sheng Yu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Zhanwang Xu
- Department of Spine and Spinal Cord, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - RuJie Zhuang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Department of Orthopedics, Quzhou TCM Hospital at the Junction of Four Provinces Affiliated to Zhejiang Chinese Medical University, Quzhou, 324002, Zhejiang, China.
- Department of Traumatology and Orthopedics, The First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| | - Haipeng Xue
- Department of Spine and Spinal Cord, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China.
| |
Collapse
|
110
|
Sun WJ, An XD, Zhang YH, Tang SS, Sun YT, Kang XM, Jiang LL, Zhao XF, Gao Q, Ji HY, Lian FM. Autophagy-dependent ferroptosis may play a critical role in early stages of diabetic retinopathy. World J Diabetes 2024; 15:2189-2202. [PMID: 39582563 PMCID: PMC11580571 DOI: 10.4239/wjd.v15.i11.2189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/10/2024] [Accepted: 09/10/2024] [Indexed: 10/16/2024] Open
Abstract
Diabetic retinopathy (DR), as one of the most common and significant microvascular complications of diabetes mellitus (DM), continues to elude effective targeted treatment for vision loss despite ongoing enrichment of the understanding of its pathogenic mechanisms from perspectives such as inflammation and oxidative stress. Recent studies have indicated that characteristic neuroglial degeneration induced by DM occurs before the onset of apparent microvascular lesions. In order to comprehensively grasp the early-stage pathological changes of DR, the retinal neurovascular unit (NVU) will become a crucial focal point for future research into the occurrence and progression of DR. Based on existing evidence, ferroptosis, a form of cell death regulated by processes like ferritinophagy and chaperone-mediated autophagy, mediates apoptosis in retinal NVU components, including pericytes and ganglion cells. Autophagy-dependent ferroptosis-related factors, including BECN1 and FABP4, may serve as both biomarkers for DR occurrence and development and potentially crucial targets for future effective DR treatments. The aforementioned findings present novel perspectives for comprehending the mechanisms underlying the early-stage pathological alterations in DR and open up innovative avenues for investigating supplementary therapeutic strategies.
Collapse
Affiliation(s)
- Wen-Jie Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xue-Dong An
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Yue-Hong Zhang
- Department of Endocrinology, Fangshan Hospital of Beijing University of Chinese Medicine, Beijing 102400, China
| | - Shan-Shan Tang
- Department of Endocrinology, Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, China
| | - Yu-Ting Sun
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xiao-Min Kang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Lin-Lin Jiang
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Xue-Fei Zhao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Qing Gao
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Hang-Yu Ji
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| | - Feng-Mei Lian
- Department of Endocrinology, Guang’anmen Hospital, Beijing 100053, China
| |
Collapse
|
111
|
Yan P, Li X, He Y, Zhang Y, Wang Y, Liu J, Ren S, Wu D, Zhao Y, Ding L, Jia W, Lyu Y, Xiao D, Lin S, Lin Y. The synergistic protective effects of paeoniflorin and β-ecdysterone against cardiac hypertrophy through suppressing oxidative stress and ferroptosis. Cell Signal 2024; 125:111509. [PMID: 39549820 DOI: 10.1016/j.cellsig.2024.111509] [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: 06/23/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Exploring feasible drugs for the treatment of pathological cardiac hypertrophy has always been a focus of cardiovascular disease research. Paeoniflorin (PF) and β-Ecdysterone (β-Ecd) are the main active components of Paeonia lactiflora and Achyranthes bidentata, which can be used for the treatment of cardiovascular diseases, but their mechanism of action remains unclear. This study focused on oxidative stress and ferroptosis to investigate the protective effects of PF and β-Ecd on cardiac hypertrophy in primary cardiomyocytes and C57BL/6 mice, utilizing the integration of CCK8 assays, ROS detection, molecular docking, real-time quantitative PCR, western blot, immunofluorescence, etc. The result of combination indices demonstrated a significant synergistic protective effect of PF and β-Ecd on cardiac hypertrophy. Furthermore, in vitro and in vivo studies further showed that the combination of PF and β-Ecd could improve the abnormalities of cell surface area, ANP, β-MHC, MDA, SOD, calcium ion, mitochondrial membrane potential and so on induced by cardiac hypertrophy through the inhibition effects of oxidative stress and iron metabolism, which might be closely related to the impact on the Nrf2/HO-1 and SLC7A11/GPX4 pathways. Altogether, this work revealed the mechanism of the combination of PF and β-Ecd in the treatment of cardiac hypertrophy from the aspects of suppressing oxidative stress and ferroptosis, aiming to promote effective treatment of the disease and the clinical application of PF and β-Ecd.
Collapse
Affiliation(s)
- Peimei Yan
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Xue Li
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yuhui He
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yanyan Zhang
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yingwanqi Wang
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Jianing Liu
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Shan Ren
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Dingxiao Wu
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Yu Zhao
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Lin Ding
- Science and Technology Achievement Transformation Center, Qiqihar Medical University, Qiqihar 161006, China
| | - Weiwei Jia
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Ying Lyu
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China
| | - Dan Xiao
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150000, China
| | - Song Lin
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China; Heilongjiang Key Laboratory of Medicine and Food Resources and Metabolic Disease Prevention, Qiqihar Medical University, Qiqihar 161006, China.
| | - Yan Lin
- School of Basic Medicine, Qiqihar Medical University, Qiqihar 161006, China; Heilongjiang Key Laboratory of Medicine and Food Resources and Metabolic Disease Prevention, Qiqihar Medical University, Qiqihar 161006, China.
| |
Collapse
|
112
|
Ren M, Xu Q, Luan J, Ni Y, Xie B. Mir-509-3p targets SLC25A13 to regulate ferroptosis and protect retinal endothelial cells in diabetic retinopathy. Acta Diabetol 2024:10.1007/s00592-024-02400-3. [PMID: 39508857 DOI: 10.1007/s00592-024-02400-3] [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: 05/12/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024]
Abstract
AIMS Diabetic retinopathy (DR) is a major complication of diabetes that leads to vision impairment. The aim of this study was to investigate the regulatory role of miR-509-3p in DR, focusing on its interaction with SLC25A13 and its impact on retinal endothelial cell function, oxidative stress, apoptosis, and ferroptosis. METHODS HRVECs were cultured in high-glucose (HG) conditions to establish an in vitro DR model. miR-509-3p mimics and inhibitors were transfected into HRVECs to assess their effects on SLC25A13 expression, cell viability, apoptosis, reactive oxygen species (ROS) levels, and ferroptosis markers. A luciferase reporter assay and RNA immunoprecipitation were used to confirm the binding of miR-509-3p to SLC25A13 mRNA. For in vivo validation, agomiR-509-3p was injected into the vitreous of DR mice, and retinal thickness, pathological damage, and apoptosis were evaluated. Ferroptosis-related markers (GPX4, TlR4, ASCL4) were analyzed in HRVECs to explore the mechanism of miR-509-3p in regulating ferroptosis. RESULTS In vitro, miR-509-3p significantly decreased SLC25A13 expression, resulting in enhanced HRVEC viability, reduced apoptosis, and lower ROS levels under HG conditions. Overexpression of SLC25A13 reversed these protective effects, while miR-509-3p knockdown exacerbated oxidative stress and apoptosis. In vivo, agomiR-509-3p increased retinal thickness, reduced pathological damage, and decreased apoptosis in DR mice. Ferroptosis marker analysis revealed that miR-509-3p upregulated GPX4 expression and downregulated TlR4 and ASCL4, whereas SLC25A13 overexpression reversed these effects, further linking miR-509-3p to the regulation of ferroptosis. CONCLUSIONS miR-509-3p exerts a protective effect in DR by targeting SLC25A13, reducing oxidative stress, apoptosis, and ferroptosis in retinal endothelial cells. These findings highlight the potential of miR-509-3p as a therapeutic target for DR management.
Collapse
Affiliation(s)
- Meiqing Ren
- Department of Ophthalmology, School of medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Qian Xu
- Department of Ophthalmology, School of medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Jie Luan
- Department of Ophthalmology, School of medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yan Ni
- Department of Ophthalmology, School of medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Bo Xie
- The Diabetes Research Institute, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
113
|
Arbatskiy M, Balandin D, Akberdin I, Churov A. A Systems Biology Approach Towards a Comprehensive Understanding of Ferroptosis. Int J Mol Sci 2024; 25:11782. [PMID: 39519341 PMCID: PMC11546516 DOI: 10.3390/ijms252111782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Ferroptosis is a regulated cell death process characterized by iron ion catalysis and reactive oxygen species, leading to lipid peroxidation. This mechanism plays a crucial role in age-related diseases, including cancer and cardiovascular and neurological disorders. To better mimic iron-induced cell death, predict the effects of various elements, and identify drugs capable of regulating ferroptosis, it is essential to develop precise models of this process. Such drugs can be tested on cellular models. Systems biology offers a powerful approach to studying biological processes through modeling, which involves accumulating and analyzing comprehensive research data. Once a model is created, it allows for examining the system's response to various stimuli. Our goal is to develop a modular framework for ferroptosis, enabling the prediction and screening of compounds with geroprotective and antiferroptotic effects. For modeling and analysis, we utilized BioUML (Biological Universal Modeling Language), which supports key standards in systems biology, modular and visual modeling, rapid simulation, parameter estimation, and a variety of numerical methods. This combination fulfills the requirements for modeling complex biological systems. The integrated modular model was validated on diverse datasets, including original experimental data. This framework encompasses essential molecular genetic processes such as the Fenton reaction, iron metabolism, lipid synthesis, and the antioxidant system. We identified structural relationships between molecular agents within each module and compared them to our proposed system for regulating the initiation and progression of ferroptosis. Our research highlights that no current models comprehensively cover all regulatory mechanisms of ferroptosis. By integrating data on ferroptosis modules into an integrated modular model, we can enhance our understanding of its mechanisms and assist in the discovery of new treatment targets for age-related diseases. A computational model of ferroptosis was developed based on a modular modeling approach and included 73 differential equations and 93 species.
Collapse
Affiliation(s)
- Mikhail Arbatskiy
- Russian Clinical Research Center of Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia; (D.B.); (A.C.)
| | - Dmitriy Balandin
- Russian Clinical Research Center of Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia; (D.B.); (A.C.)
| | - Ilya Akberdin
- Department of Computational Biology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Alexey Churov
- Russian Clinical Research Center of Gerontology, Pirogov Russian National Research Medical University, Ministry of Healthcare of the Russian Federation, 129226 Moscow, Russia; (D.B.); (A.C.)
| |
Collapse
|
114
|
Peng W, Chung KB, Lawrence BP, O'Banion MK, Dirksen RT, Wojtovich AP, Onukwufor JO. DMT1 knockout abolishes ferroptosis induced mitochondrial dysfunction in C. elegans amyloid β proteotoxicity. Free Radic Biol Med 2024; 224:785-796. [PMID: 39317269 PMCID: PMC11568904 DOI: 10.1016/j.freeradbiomed.2024.09.034] [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: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 09/26/2024]
Abstract
Iron is critical for neuronal activity and metabolism, and iron dysregulation alters these functions in age-related neurodegenerative disorders, such as Alzheimer's disease (AD). AD is a chronic neurodegenerative disease characterized by progressive neuronal dysfunction, memory loss and decreased cognitive function. AD patients exhibit elevated iron levels in the brain compared to age-matched non-AD individuals. However, the degree to which iron overload contributes to AD pathogenesis is unclear. Here, we evaluated the involvement of ferroptosis, an iron-dependent cell death process, in mediating AD-like pathologies in C. elegans. Results showed that iron accumulation occurred prior to the loss of neuronal function as worms age. In addition, energetic imbalance was an early event in iron-induced loss of neuronal function. Furthermore, the loss of neuronal function was, in part, due to increased mitochondrial reactive oxygen species mediated oxidative damage, ultimately resulting in ferroptotic cell death. The mitochondrial redox environment and ferroptosis were modulated by pharmacologic processes that exacerbate or abolish iron accumulation both in wild-type worms and worms with increased levels of neuronal amyloid beta (Aβ). However, neuronal Aβ worms were more sensitive to ferroptosis-mediated neuronal loss, and this increased toxicity was ameliorated by limiting the uptake of ferrous iron through knockout of divalent metal transporter 1 (DMT1). In addition, DMT1 knockout completely suppressed phenotypic measures of Aβ toxicity with age. Overall, our findings suggest that iron-induced ferroptosis alters the mitochondrial redox environment to drive oxidative damage when neuronal Aβ is overexpressed. DMT1 knockout abolishes neuronal Aβ-associated pathologies by reducing neuronal iron uptake.
Collapse
Affiliation(s)
- Wilson Peng
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Kaitlin B Chung
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA, 14642
| | - M Kerry O'Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA, 14642
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Andrew P Wojtovich
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA; Department of Anesthesiology and Perioperative Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - John O Onukwufor
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA, 14642.
| |
Collapse
|
115
|
Zhuge R, Zhang L, Xue Q, Wang R, Xu J, Wang C, Meng C, Lu R, Yin F, Guo L. Ferritinophagy is involved in hexavalent chromium-induced ferroptosis in Sertoli cells. Toxicol Appl Pharmacol 2024; 492:117139. [PMID: 39486596 DOI: 10.1016/j.taap.2024.117139] [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: 08/10/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Hexavalent chromium [Cr(VI)] has significant adverse effects on the environment and human health, particularly on the male reproductive system. Previously, we observed ferroptosis and autophagy in rat testicular injury induced by Cr(VI). In the present study, we focused on the association between ferroptosis and autophagy in mouse Sertoli cells (TM4) exposed to concentrations of 2.5 μМ, 5 μМ, and 10 μМ Cr(VI). Cr(VI) exposure altered mitochondrial ultrastructure; increased intracellular iron, malondialdehyde, and reactive oxygen species (ROS) levels; decreased glutathione content; increased TfR1 protein expression; and decreased GPX4, FPN1, and SLC7A11 protein expression, ultimately resulting in ferroptosis. Additionally, we observed ferritinophagy, increased expression of BECLIN1, LC3B, and NCOA4, and decreased expression of FTH1 and P62. Inhibition of autophagy and ferritinophagy via 3-MA and small interfering RNA (siRNA)-mediated silencing of NCOA4 ameliorated changes in ferritinophagy- and ferroptosis-associated protein expression, and reduced ROS levels. Rats exposed to Cr(VI) exhibited atrophy of testicular seminiferous tubules, a reduction in germ and Sertoli cells, and the occurrence of ferritinophagy and ferroptosis in cells of the rat testes. These results indicate that ferroptosis, triggered by NCOA4-mediated ferritinophagy, is one of the mechanisms that contribute to Cr(VI)-induced damage in Sertoli cells.
Collapse
Affiliation(s)
- Ruijian Zhuge
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Le Zhang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Qian Xue
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Rui Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Jiayunzhu Xu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Chaofan Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Chunyang Meng
- Department of Spine Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Rifeng Lu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Fei Yin
- Department of Spine Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Li Guo
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
116
|
Liu S, Zhang L, Ding K, Zeng B, Li B, Zhou J, Li J, Wang J, Zhang H, Sun R, Su X. S. glabra exerts anti-lung cancer effects by inducing ferroptosis and anticancer immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155981. [PMID: 39260134 DOI: 10.1016/j.phymed.2024.155981] [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: 06/01/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Sarcandra glabra (S. glabra), a traditional Chinese medicine (TCM), has demonstrated significant anticancer activity; however, the underlying mechanisms have not yet been fully elucidated. PURPOSE This study aimed to investigate the effects of S. glabra on lung cancer and to explore its underlying mechanisms. METHODS The chemical profile of S. glabra was analyzed via ultrahigh-performance liquid chromatography coupled with mass spectrometry (UPLC-MS). The effects of S. glabra on the viability, proliferation, apoptosis, migration, and invasion of lung cancer cells were assessed via CCK8, colony formation, flow cytometry, scratch, and Transwell assays. In vivo anticancer activity was evaluated in an LLC mouse model. Proteomic analysis was performed to identify key molecules and pathways in S. glabra-treated LLC cells. The expression of ferroptotic proteins and associated cellular events were examined via western blotting, ROS production, iron accumulation, and lipid peroxidation assays. Immune modulation in tumor-bearing mice was evaluated by detecting immune cells and cytokines in the peripheral blood and tumor tissue. RESULTS Our analysis quantified 1997 chemical markers in S. glabra aqueous extracts. S. glabra inhibited the viability and proliferation of lung cancer cells and induced cell cycle arrest and apoptosis. Scratch and Transwell assays demonstrated that S. glabra suppressed the migration and invasion of lung cancer cells. Oral administration of S. glabra significantly inhibited tumor growth in LLC tumor-bearing mice. Proteomic analysis revealed that S. glabra upregulated the expression of the HMOX1 protein and activated the ferroptosis pathway. Consistent with these findings, we found that S. glabra triggered ferroptosis in lung cancer cells, as evidenced by the upregulation of HMOX1, downregulation of GPX4 and ferritin light chain proteins, iron accumulation, increased ROS production, and lipid peroxidation. Furthermore, S. glabra demonstrated immunostimulatory properties in LLC tumor-bearing mice, leading to increased populations of immune cells (NK cells) and elevated cytokine levels (IL-2). CONCLUSION This study is the first to demonstrate that S. glabra induces ferroptosis in lung cancer cells by regulating HMOX1, GPX4, and FTL. These findings provide a robust scientific basis for the clinical application of S. glabra in lung cancer treatment.
Collapse
Affiliation(s)
- Songyu Liu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Lu Zhang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Kai Ding
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Bin Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Bo Li
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Jinyi Zhou
- Department of Neurosurgery, The Third Affiliated Hospital of Kunming Medical University, Kunming 650118, China
| | - Jv Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Junliang Wang
- Scientific Research and Experimental Center, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Huijun Zhang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai 200040, China.
| | - Ruifen Sun
- School of Nursing, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Xiaosan Su
- Scientific Research and Experimental Center, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
117
|
Yin T, Zhang X, Xiong Y, Li B, Guo D, Sha Z, Lin X, Wu H. Exploring gut microbial metabolites as key players in inhibition of cancer progression: Mechanisms and therapeutic implications. Microbiol Res 2024; 288:127871. [PMID: 39137590 DOI: 10.1016/j.micres.2024.127871] [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: 05/16/2024] [Revised: 07/19/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
The gut microbiota plays a critical role in numerous biochemical processes essential for human health, such as metabolic regulation and immune system modulation. An increasing number of research suggests a strong association between the gut microbiota and carcinogenesis. The diverse metabolites produced by gut microbiota can modulate cellular gene expression, cell cycle dynamics, apoptosis, and immune system functions, thereby exerting a profound influence on cancer development and progression. A healthy gut microbiota promotes substance metabolism, stimulates immune responses, and thereby maintains the long-term homeostasis of the intestinal microenvironment. When the gut microbiota becomes imbalanced and disrupts the homeostasis of the intestinal microenvironment, the risk of various diseases increases. This review aims to elucidate the impact of gut microbial metabolites on cancer initiation and progression, focusing on short-chain fatty acids (SCFAs), polyamines (PAs), hydrogen sulfide (H2S), secondary bile acids (SBAs), and microbial tryptophan catabolites (MTCs). By detailing the roles and molecular mechanisms of these metabolites in cancer pathogenesis and therapy, this article sheds light on dual effects on the host at different concentrations of metabolites and offers new insights into cancer research.
Collapse
Affiliation(s)
- Tianxiang Yin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiang Zhang
- Medical School, Yan'an University, Yan'an 716000, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyuan Lin
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
118
|
Aydemir D, Öztürk K, Arslan FB, Çalis S, Ulusu NN. Gemcitabine-loaded chitosan nanoparticles enhanced apoptotic and ferroptotic response of gemcitabine treatment alone in the pancreatic cancer cells in vitro. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9051-9066. [PMID: 38884675 PMCID: PMC11522156 DOI: 10.1007/s00210-024-03193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/27/2024] [Indexed: 06/18/2024]
Abstract
Gemcitabine (GEM) is a first-line treatment for pancreatic ductal adenocarcinoma (PDAC) patients, causing side effects and poor overall survival. Eighty percent of patients often develop resistance rapidly to GEM. Developing therapeutic approaches and increasing sensitivity to gemcitabine in PDAC has become one of the challenges in cancer research. We synthesized GEM-loaded NPs prepared with a method that combines ultrasonication and ionotropic gelation to overcome GEM-related limitations in PDAC. CFPAC-1 cells were treated with increased concentrations of GEM, empty chitosan, and GEM-loaded NPs (0.66, 1.32, 2.64, 5.32 µg/ml) for up to 48 h. Empty chitosan NPs did not show toxicity on L929 cells. Antioxidant enzyme activities, including glucose 6-phosphate dehydrogenase (G6PD), 6-phosphogluconate dehydrogenase (6-PGD), glutathione reductase (GR), glutathione s-transferase (GST), and glutathione peroxidase (GPx), significantly reduced in GEM-loaded NPs compared to the GEM associated with increased oxidative stress, PPP, and glycolysis. Bcl-xL, NOXA/mcl-1, and Ca2+ levels significantly increased in GEM-loaded NP-administered cells compared to the GEM and control groups. In contrast, JNK, p38, STAT3, Akt, and CREB levels significantly decreased in the GEM-loaded NP group, addressing enhanced apoptotic response compared to the GEM alone. Increased ferroptosis activity in GEM-loaded NP-administered groups has been validated via decreased antioxidant enzyme activities, increased cytosolic Fe, Zn, Mg, and Mn levels, and reduced GPx activity compared to the GEM and control groups. For the first time in the literature, we showed biocompatible GEM-loaded NPs enhanced apoptotic and ferroptotic response in CFPAC-1 cells via downregulation of antioxidant, glycolysis, and PPP metabolism compared to the GEM alone.
Collapse
Affiliation(s)
- Duygu Aydemir
- School of Medicine, Department of Medical Biochemistry, Koc University, Istanbul, Turkey.
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
- Biochemistry Department, Koc University School of Medicine, Rumelifeneri Yolu, Sariyer, Istanbul, 34450, Turkey.
| | - Kıvılcım Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Fatma Betül Arslan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Çalis
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Nuriye Nuray Ulusu
- School of Medicine, Department of Medical Biochemistry, Koc University, Istanbul, Turkey.
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.
- Biochemistry Department, Koc University School of Medicine, Rumelifeneri Yolu, Sariyer, Istanbul, 34450, Turkey.
| |
Collapse
|
119
|
Liu Y, Bai Q, Pang N, Xue J. TCF12 induces ferroptosis by suppressing OTUB1-mediated SLC7A11 deubiquitination to promote cisplatin sensitivity in oral squamous cell carcinoma. Cell Biol Int 2024; 48:1649-1663. [PMID: 38946134 DOI: 10.1002/cbin.12211] [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: 02/25/2024] [Revised: 05/22/2024] [Accepted: 06/19/2024] [Indexed: 07/02/2024]
Abstract
Chemotherapy resistance is a major obstacle to effective cancer treatment, and promotion of ferroptosis can suppress cisplatin resistance in tumor cells. TCF12 plays a suppressive role in oral squamous cell carcinoma (OSCC), but whether it participates in the regulation of cisplatin resistance by modulating ferroptosis remains unclear. Here, we found that TCF12 expression was decreased in OSCC cells compared with normal oral cells, and it was reduced in cisplatin (DDP)-resistant OSCC cells compared with parental cells. Moreover, overexpression of TCF12 sensitized DDP-resistant cells to DDP by promoting ferroptosis. Intriguingly, silencing TCF12 reversed the promotion effect of the ferroptosis activator RSL3 on ferroptosis and DDP sensitivity, and overexpressing TCF12 antagonized the effect of the ferroptosis inhibitor liproxstatin-1 on ferroptosis and DDP resistance. Mechanically, TCF12 promoted ubiquitination of SLC7A11 and decreased SLC7A11 protein stability through transcriptional repression of OTUB1, thereby facilitating ferroptosis. Consistently, SLC7A11 overexpression neutralized the promotion effect of TCF12 on ferroptosis and DDP sensitivity. Additionally, upregulation of TCF12 hindered the growth of mouse OSCC xenografts and enhanced the DDP sensitivity of xenografts by inducing ferroptosis. In conclusion, TCF12 enhanced DDP sensitivity in OSCC cells by promoting ferroptosis, which was achieved through modulating SLC7A11 expression via transcriptional regulation of OTUB1.
Collapse
Affiliation(s)
- Yanchun Liu
- Department of Stomatology, Xianyang Hospital of Yan'an University, Xianyang, China
| | - Qin Bai
- Department of Stomatology, Xianyang Hospital of Yan'an University, Xianyang, China
| | - Nan Pang
- Department of Stomatology, Xianyang Hospital of Yan'an University, Xianyang, China
| | - Jun Xue
- Department of Stomatology, Xianyang Hospital of Yan'an University, Xianyang, China
| |
Collapse
|
120
|
Sun Z, Zhang X, Li M, Yang Q, Xiao X, Chen X, Liang W. Targeting ferroptosis in treating traumatic brain injury: Harnessing the power of traditional Chinese medicine. Biomed Pharmacother 2024; 180:117555. [PMID: 39413616 DOI: 10.1016/j.biopha.2024.117555] [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: 07/24/2024] [Revised: 09/20/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024] Open
Abstract
Traumatic brain injury (TBI) exhibits high prevalence and mortality, but current treatments remain suboptimal. Traditional Chinese medicine (TCM) has long been effectively used for TBI intervention. Moreover, the recently discovered iron-dependent cell death pathway, known as ferroptosis, characterized by lipid peroxidation, as a key target in TCM-based treatments for TBI. This review provides a comprehensive overview of the latest advancements in TCM strategies targeting ferroptosis in TBI therapy, covering natural product monomers, classic formulas, and acupuncture/moxibustion. The review also addresses current challenges and outlines future research directions to further advance the development and application of TBI management strategies.
Collapse
Affiliation(s)
- Zhongjie Sun
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiao Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Manrui Li
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Qiuyun Yang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiao Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University and the Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China.
| | - Xiameng Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| | - Weibo Liang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
121
|
Chen J, Yan L, Zhang Y, Liu X, Wei Y, Zhao Y, Li K, Shi Y, Liu H, Lai W, Tian L, Lin B. Maternal exposure to nanopolystyrene induces neurotoxicity in offspring through P53-mediated ferritinophagy and ferroptosis in the rat hippocampus. J Nanobiotechnology 2024; 22:651. [PMID: 39438901 PMCID: PMC11520165 DOI: 10.1186/s12951-024-02911-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
There are increasing concerns regarding the rapid expansion of polystyrene nanoplastics (PS-NPs), which could impact human health. Previous studies have shown that nanoplastics can be transferred from mothers to offspring through the placenta and breast milk, resulting in cognitive deficits in offspring. However, the neurotoxic effects of maternal exposure on offspring and its mechanisms remain unclear. In this study, PS-NPs (50 nm) were gavaged to female rats throughout gestation and lactation to establish an offspring exposure model to study the neurotoxicity and behavioral changes caused by PS-NPs on offspring. Neonatal rat hippocampal neuronal cells were used to investigate the pathways through which NPs induce neurodevelopmental toxicity in offspring rats, using iron inhibitors, autophagy inhibitors, reactive oxygen species (ROS) scroungers, P53 inhibitors, and NCOA4 inhibitors. We found that low PS-NPs dosages can cause ferroptosis in the hippocampus of the offspring, resulting in a decline in the cognitive, learning, and memory abilities of the offspring. PS-NPs induced NOCA4-mediated ferritinophagy and promoted ferroptosis by inciting ROS production to activate P53-mediated ferritinophagy. Furthermore, the levels of the antioxidant factors glutathione peroxidase 4 (GPX4) and glutathione (GSH), responsible for ferroptosis, were reduced. In summary, this study revealed that consumption of PS-NPs during gestation and lactation can cause ferroptosis and damage the hippocampus of offspring. Our results can serve as a basis for further research into the neurodevelopmental effects of nanoplastics in offspring.
Collapse
Affiliation(s)
- Jiang Chen
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Licheng Yan
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Yaping Zhang
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Xuan Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yizhe Wei
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yiming Zhao
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
- School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Kang Li
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Yue Shi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Huanliang Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Wenqing Lai
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China
| | - Lei Tian
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| | - Bencheng Lin
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, 300050, China.
| |
Collapse
|
122
|
Hou W, Zou Y, Li J, Jiang H, Li J, Wu J, Zhu S, Ding Y, Xu H, Jia F, Li X. Synergistic Therapy of Melanoma by Co-Delivery of Dacarbazine and Ferroptosis-Inducing Ursolic Acid Using Biomimetic Nanoparticles. ACS OMEGA 2024; 9:41532-41543. [PMID: 39398166 PMCID: PMC11465262 DOI: 10.1021/acsomega.4c05209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
Melanoma is one of the most aggressive types of cancer and is prone to metastasis, making current clinical treatment quite difficult. The usage of the first-line medication dacarbazine (DTIC) for melanoma is limited due to harsh side effects, limited water solubility, and a short half-life. To tackle these disadvantages, polylactic acid-hydroxyacetic acid copolymer nanoparticles (NPs) loaded with dacarbazine and ursolic acid (NPs) were fabricated, which were further encapsulated with a red blood cell membrane (RNPs). MTT, apoptosis assay, wound healing assay, colony formation assay, and immunohistochemistry were used to assess the antitumor effect of NPs and RNPs. Ferroptosis evaluation was implemented using GSH detection and the malondialdehyde assay. We found that RNPs exhibited stability and biosafety in vitro and in vivo and achieved superior anticancer ability against xenograft tumors compared with single agents and NPs, which indicated the synergistic and biomimetic efficacy. Furthermore, ferroptotic activity was observed in RNPs-treated tumor cells, and ferroptosis inhibition could partially rescue melanoma cells from RNPs-induced cell death. Collectively, this study evaluated the potential of RNPs as a novel biomimetic nanomedicine for synergistic melanoma therapy by eliciting ferroptosis in tumor cells with both anticancer activity and biosafety.
Collapse
Affiliation(s)
- Wenjun Hou
- Department
of Dermatology, Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing 210008, China
| | - Yifan Zou
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 101 Longmian Avenue, Nanjing 211166, China
- Department
of General Surgery, The First Affiliated
Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Jie Li
- Department
of Geriatric Gastroenterology, The First
Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Hui Jiang
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jinyu Li
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jie Wu
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Senlin Zhu
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Yan Ding
- Department
of Geriatric Gastroenterology, The First
Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Huae Xu
- Department
of Pharmaceutics, School of Pharmacy, Nanjing
Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Feng Jia
- Department
of Neurosurgery, Yancheng No. 1 People’s Hospital, The Affiliated Yancheng First Hospital of Nanjing
University Medical School, 66 Renmin South Road, Yancheng 224008, China
| | - Xiaolin Li
- Department
of Geriatric Gastroenterology, The First
Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
123
|
Xie H, Cao C, Shu D, Liu T, Zhang T. The important role of ferroptosis in inflammatory bowel disease. Front Med (Lausanne) 2024; 11:1449037. [PMID: 39434776 PMCID: PMC11491328 DOI: 10.3389/fmed.2024.1449037] [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: 06/24/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Ferroptosis is a type of regulated cell death that occurs due to the iron-dependent accumulation of lethal reactive oxygen species (ROS) from lipids. Ferroptosis is characterized by distinct morphological, biochemical, and genetic features that differentiate it from other regulated cell death (RCD) types, which include apoptosis, various necrosis types, and autophagy. Recent reports show that ferritin formation is correlated to many disorders, such as acute injury, infarction, inflammation, and cancer. Iron uptake disorders have also been associated with intestinal epithelial dysfunction, particularly inflammatory bowel disease (IBD). Studies of iron uptake disorders may provide new insights into the pathogenesis of IBD, thereby improving the efficacy of medical interventions. This review presents an overview of ferroptosis, elucidating its fundamental mechanisms and highlighting its significant involvement in IBD.
Collapse
Affiliation(s)
- Hanhan Xie
- The Second Affiliated Hospital of Chengdu Medical College, China Nation Nuclear Corporation 416 Hospital, Chengdu, China
| | - Chun Cao
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Dan Shu
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| | - Tong Liu
- The Third Affiliated Hospital of Chengdu Medical College, Chengdu Pidu District People’s Hospital, Chengdu, China
| | - Tao Zhang
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
124
|
Zakaria S, Ibrahim N, Abdo W, E El-Sisi A. JNK inhibitor and ferroptosis modulator as possible therapeutic modalities in Alzheimer disease (AD). Sci Rep 2024; 14:23293. [PMID: 39375359 PMCID: PMC11458622 DOI: 10.1038/s41598-024-73596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
Alzheimer disease (AD) is among the most prevalent neurodegenerative diseases globally, marked by cognitive and behavioral disruptions. Ferroptosis is a form of controlled cell death characterized by intracellular iron accumulation associated with lipid peroxide formation, which subsequently promotes AD initiation and progression. We hypothesized that targeting the ferroptosis pathway may help in AD management. Therefore, our study aimed to evaluate the potential neuroprotective effect of the antifungal Ciclopirox olamine (CPX-O) that acts through iron chelation. We employed CPX-O separately or in combination with the JNK inhibitor (SP600125) in a mice model of AlCl3-induced AD. Animals underwent examination for behavioral, biochemical, histological, and immunohistochemical findings. Our results revealed that AlCl3 was associated with disruptions in learning and memory parameters, neuronal degeneration in the hippocampus, increased immunoreactivity of amyloid-β and tau proteins, a significant rise in iron, nitric oxide (NO), malondialdehyde (MDA), JNK, and P53 levels, along with the significant decrease in glutathione peroxidase activity. Interestingly, the administration of CPX-O alone or in combination with SP600125 in the AlCl3-induced AD model caused an improvement in the previously described examination findings. Therefore, CPX-O may be a promising candidate for AD treatment, and future clinical trials will be required to confirm these preclinical findings.
Collapse
Affiliation(s)
- Sherin Zakaria
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Nashwa Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Alaa E El-Sisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, 31512, Egypt
| |
Collapse
|
125
|
Huang W, Guo Y, Qian Y, Liu X, Li G, Wang J, Yang X, Wu M, Fan Y, Luo H, Chen Y, Zhang L, Yang N, Liu Z, Liu Y. Ferroptosis-inducing compounds synergize with docetaxel to overcome chemoresistance in docetaxel-resistant non-small cell lung cancer cells. Eur J Med Chem 2024; 276:116670. [PMID: 39018922 DOI: 10.1016/j.ejmech.2024.116670] [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: 04/23/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Development of resistance to therapy-induced cell death is a major hurdle in the effective treatment of advanced solid tumors. Erastin and RSL3 were originally found to induce synthetic lethality by induction of a novel form of cell death termed ferroptosis. Emerging evidence suggests that ferroptosis inducers enhance chemosensitivity of classic therapeutic agents by triggering ferroptotic cell death. In this study we evaluated the effects of erastin and RSL3 on the resistance of docetaxel, doxorubicin, and cisplatin, and revealed a mechanism whereby these ferroptosis inducers augment docetaxel efficacy in non-small cell lung cancer by regulating redox signaling to promote ferroptosis. Transcriptome analysis revealed that combination treatment modulated not only p53 signaling pathway but also immune responses and several signaling pathways including MAPK, NF-κB and PI3K/Akt. Considering that glutathione peroxidase 4 (GPX4) serves as the main effector to protect cells from ferroptosis, this study identified three novel non-covalent GPX4 inhibitors with the aid of pharmacophore-based virtual screening. The new ferroptosis-inducing compounds synergized with docetaxel to increase the cytotoxicity by promoting ferroptotic cell death in docetaxel-resistant A549/DTX cells. Collectively, the induction of ferroptosis contributed to docetaxel-induced cytotoxic effects and overcame drug resistance in A549/DTX cells. Ferroptosis has a great potential to become a new approach to attenuate resistance to some classic therapeutic drugs in cancer patients.
Collapse
Affiliation(s)
- Wei Huang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China.
| | - Yi Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Yazhi Qian
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Xiaoang Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Gaoxiang Li
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China; Medical College, Tibet University, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - Jun Wang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Xiaozhou Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Mo Wu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Ying Fan
- Medical College, Tibet University, Lhasa, Tibet Autonomous Region, 850000, PR China
| | - Haojun Luo
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Yuzhu Chen
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Nan Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China.
| | - Yanyong Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, 100005, PR China; Medical College, Tibet University, Lhasa, Tibet Autonomous Region, 850000, PR China.
| |
Collapse
|
126
|
Muluh TA, Fu Q, Ai X, Wang C, Chen W, Zheng X, Wang W, Wang M, Shu XS, Ying Y. Targeting Ferroptosis as an Advance Strategy in Cancer Therapy. Antioxid Redox Signal 2024; 41:616-636. [PMID: 38959114 DOI: 10.1089/ars.2024.0608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Significance: This study innovates by systematically integrating the molecular mechanisms of iron death and its application in cancer therapy. By deeply analyzing the interaction between iron death and the tumor microenvironment, the study provides a new theoretical basis for cancer treatment and directions for developing more effective treatment strategies. In addition, the study points to critical issues and barriers that need to be addressed in future research, providing valuable insights into the use of iron death in clinical translation. Recent Advances: These findings are expected to drive further advances in cancer treatment, bringing patients more treatment options and hope. Through this paper, we see the great potential of iron death in cancer treatment and look forward to more research results being translated into clinical applications in the future to contribute to the fight against cancer. Critical Issues: In today's society, cancer is still one of the major diseases threatening human health. Despite advances in existing treatments, cancer recurrence and drug resistance remain a severe problem. These problems increase the difficulty of treatment and bring a substantial physical and mental burden to patients. Therefore, finding new treatment strategies to overcome these challenges has become significant. Future Directions: The study delved into the molecular basis of iron death in tumor biology. It proposed a conceptual framework to account for the interaction of iron death with the tumor immune microenvironment, guide treatment selection, predict efficacy, explore combination therapies, and identify new therapeutic targets to overcome cancer resistance to standard treatments, peeving a path for future research and clinical translation of ferroptosis as a potential strategy in cancer therapy. Antioxid. Redox Signal. 41, 616-636. [Figure: see text].
Collapse
Affiliation(s)
- Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qianqian Fu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiaojiao Ai
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Changfeng Wang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Wei Chen
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xiangyi Zheng
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Wei Wang
- Shanghai Waker Bioscience Co., Ltd., Shanghai, China
| | - Maolin Wang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xing-Sheng Shu
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Ying Ying
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
127
|
Liu Y, He F, Chen L, Zhang Y, Zhang H, Xiao J, Meng Q. Imidazolyl Lipids Enhanced LNP Endosomal Escape for Ferroptosis RNAi Treatment of Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402362. [PMID: 38829038 DOI: 10.1002/smll.202402362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/25/2024] [Indexed: 06/05/2024]
Abstract
Treatments for cancer that incorporate small interfering RNA (siRNA) to target iron-dependent ferroptosis are thought to be highly promising. However, creating a reliable and clinically feasible siRNA delivery system continues to be a major obstacle in the field of cancer treatment. Here, three imidazole-based ionizable lipid nanoparticles (LNPs) with pH-sensitive effects are rationally designed and synthesized for siRNA delivery. LNPs formulated with the top-performing lipid (O12-D3-I3) encapsulating FVII siRNA (FVII@O-LNP) elicited greater gene silencing than those with the benchmark Onpattro lipid DLin-MC3-DMA (MC3) due to its stronger endosomal escape. Moreover, Fc-siRNA@O-LNPs encapsulated with ferrocene (Fc) and SLC7A11/Nrf2-targeted siRNA is formulated. The outcomes demonstrate optimal safety profiles and a significant anti-tumor effect by inducing long-lasting and efficient ferroptosis through a synergistic action in vivo. In summary, this work shows that imidazolyl lipid-prepared LNPs are efficient delivery vehicles for cancer therapy and ferroptosis-targeting siRNA administration, both of which have extensive clinical application potential.
Collapse
Affiliation(s)
- Yuanyuan Liu
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| | - Fengyang He
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| | - Longming Chen
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| | - Yahan Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| | - Han Zhang
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| | - Junhai Xiao
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| | - Qingbin Meng
- State Key Laboratory of National Security Specially Needed Medicines, Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing, 100850, China
| |
Collapse
|
128
|
Zhao M, Shen Z, Zheng Z, Xu Y, Zhang J, Liu J, Peng S, Wan J, Qin JJ, Wang M. Cardiomyocyte LGR6 alleviates ferroptosis in diabetic cardiomyopathy via regulating mitochondrial biogenesis. Metabolism 2024; 159:155979. [PMID: 39038735 DOI: 10.1016/j.metabol.2024.155979] [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: 05/19/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
AIMS The majority of people with diabetes are susceptible to cardiac dysfunction and heart failure, and conventional drug therapy cannot correct the progression of diabetic cardiomyopathy. We assessed the potential role and therapeutic value of LGR6 (G protein-coupled receptor containing leucine-rich repeats 6) in diabetic cardiomyopathy. METHODS AND RESULTS Type 2 diabetes models were established using high-fat diet/streptozotocin-induced diabetes in mice. LGR6 knockout mice were generated. Recombinant adeno-associated virus serotype 9 carrying LGR6 under the cardiac troponin T promoter was injected into diabetic mice. Cardiomyocytes incubated with high glucose (HG) were used to imitate diabetic cardiomyopathy in vitro. The molecular mechanism was explored through RNA sequencing and a chromatin immunoprecipitation assay. We found that LGR6 expression was upregulated in diabetic hearts and HL1 cardiomyocytes treated with HG. The LGR6 knockout aggravated, but cardiomyocyte-specific LGR6 overexpression ameliorated, cardiac dysfunction and remodeling in diabetic mice. Mechanistically, in vivo and in vitro experiments revealed that LGR6 deletion aggravated, whereas LGR6 overexpression alleviated, ferroptosis and disrupted mitochondrial biogenesis by regulating STAT3/Pgc1a signaling. STAT3 inhibition and Pgc1a activation abrogated LGR6 knockout-induced mitochondrial dysfunction and ferroptosis in diabetic mice. In addition, LGR6 activation by recombinant RSPO3 treatment ameliorated cardiac dysfunction, ferroptosis and mitochondrial dysfunction in diabetic mice. CONCLUSIONS We identified a previously undescribed signaling pathway of the LGR6-STAT3-Pgc1a axis that plays a critical role in ferroptosis and mitochondrial disorders during diabetic cardiomyopathy and provides an option for treatment of diabetic hearts.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zican Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Juan-Juan Qin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute, Wuhan University, Wuhan, China; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
129
|
Guo J, Su K, Wang L, Feng B, You X, Deng M, Toh WS, Wu J, Cheng B, Xia J. Poly( p-coumaric acid) nanoparticles alleviate temporomandibular joint osteoarthritis by inhibiting chondrocyte ferroptosis. Bioact Mater 2024; 40:212-226. [PMID: 38973989 PMCID: PMC11224931 DOI: 10.1016/j.bioactmat.2024.06.007] [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: 03/28/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Oxidative stress and inflammation are key drivers of osteoarthritis (OA) pathogenesis and disease progression. Herein we report the synthesis of poly(p-coumaric) nanoparticles (PCA NPs) from p-courmaic acid (p-CA), a naturally occurring phytophenolic acid, to be a multifunctional and drug-free therapeutic for temporomandibular joint osteoarthritis (TMJOA). Compared to hyaluronic acid (HA) that is clinically given as viscosupplementation, PCA NPs exhibited long-term efficacy, superior anti-oxidant and anti-inflammatory properties in alleviating TMJOA and repairing the TMJ cartilage and subchondral bone in a rat model of TMJOA. Notably, TMJ repair mediated by PCA NPs could be attributed to their anti-oxidant and anti-inflammatory properties in enhancing cell proliferation and matrix synthesis, while reducing inflammation, oxidative stress, matrix degradation, and chondrocyte ferroptosis. Overall, our study demonstrates a multifunctional nanoparticle, synthesized from natural p-coumaric acid, that is stable and possess potent antioxidant, anti-inflammatory properties and ferroptosis inhibition, beneficial for treatment of TMJOA.
Collapse
Affiliation(s)
- Jiaxin Guo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Kai Su
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Liying Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Bingyu Feng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Xinru You
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Miao Deng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Wei Seong Toh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- Faculty of Dentistry, National University of Singapore, 119085, Singapore
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511455, PR China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| |
Collapse
|
130
|
Zhang M, Liu M, Yang L, Liu Y, Niu H, Yu Y, Zhang Y, Yang J, Tang P, Shao Z, Xing L, Wang H. Increased ferroptosis of erythrocytes is associated with myelodysplastic syndromes. Ann Hematol 2024; 103:4009-4020. [PMID: 39177794 DOI: 10.1007/s00277-024-05946-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
Anemia is the most common symptom in patients with myelodysplastic syndromes (MDS). Programmed cell death of erythrocytes is one of the contributing factors to anemia. Ferroptosis is a newly identified form of iron-dependent cell death. The aim of this study is to investigate whether anemia in MDS patients is associated with ferroptosis of nucleated erythrocytes(NEs).We detected lipid peroxidation levels, Fe2+ contents, cell death rates, glutathione (GSH) and malondialdehyde (MDA) levels in bone marrow CD235a+ NEs of MDS patients. Expression levels of ferroptosis-related molecules (ACSL4, GPX4, and SLC7A11) were evaluated through qRT-PCR and Western Blotting. Correlation between these markers and clinical parameters were analyzed. To further substantiate that the mode of cell death with CD235a+ NEs of MDS patients was attributed to the ferroptosis pathway, we applied Fer-1 to inhibit ferroptosis. Cell viability was assessed using CCK8, and changes in ferroptosis-related indicators were simultaneously evaluated. We discover that the ferroptosis level of bone marrow NEs in MDS patients was increased, which is related to anemia and iron overload. Ferroptosis might be one of the causes of anemia in MDS patients.
Collapse
Affiliation(s)
- Mengying Zhang
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Mengyuan Liu
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Liyan Yang
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Yumei Liu
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Haiyue Niu
- Department of Hematology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yating Yu
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Yue Zhang
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Jinyue Yang
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Pu Tang
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Zonghong Shao
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China
| | - Limin Xing
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China.
| | - Huaquan Wang
- Department of Hematology, General Hospital, Tianjin Medical University, 154 Anshandao, Heping District, Tianjin, 300052, China.
| |
Collapse
|
131
|
Zhou X, Lin L. Mechanisms and therapeutic target of anti-tumour treatment-related Ferroptosis: How to improve cancer therapy? Biomed Pharmacother 2024; 179:117323. [PMID: 39208665 DOI: 10.1016/j.biopha.2024.117323] [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: 07/21/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Recently, increased attention has been focused on the regulatory mechanism and potential clinical application of ferroptosis in cancer cells, especially therapy-related ferroptosis. However, the mechanism of treatment-related ferroptosis and the application prospects and strategies for future treatment still require further clarification. This review highlights the molecular relationships between different clinical antitumour drugs, including commonly used chemotherapy drugs, radiation therapy and vitamins, and ferroptosis. This review also proposes strategies for future treatments that involve ferroptosis, with an aim to develop a new strategy for the transformative potential of the emerging field of ferroptosis to improve cancer therapy.
Collapse
Affiliation(s)
- Xiangyu Zhou
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Lin Lin
- Department of General Surgery, Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
132
|
Elsakka EGE, Midan HM, Abulsoud AI, Fathi D, Abdelmaksoud NM, Abdel Mageed SS, Zaki MB, Abd-Elmawla MA, Rizk NI, Elrebehy MA, Abdelghany TM, Elesawy AE, Shahin RK, El Tabaa MM, Mohammed OA, Abdel-Reheim MA, Elballal MS, Doghish AS. Emerging insights: miRNA modulation of ferroptosis pathways in lung cancer. Exp Cell Res 2024; 442:114272. [PMID: 39362302 DOI: 10.1016/j.yexcr.2024.114272] [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: 07/22/2024] [Revised: 08/26/2024] [Accepted: 09/29/2024] [Indexed: 10/05/2024]
Abstract
The newly discovered programmed iron-dependent necrosis, ferroptosis, is a novel pathway that is controlled by iron-dependent lipid peroxidation and cellular redox changes. It can be triggered intrinsically by low antioxidant enzyme activity or extrinsically by blocking amino acid transporters or activating iron transporters. The induction of ferroptosis involves the activation of specific proteins, suppression of transporters, and increased endoplasmic reticulum (ER) stress (a condition in which the ER, a crucial organelle involved in protein folding and processing, becomes overwhelmed by an accumulation of misfolded or unfolded proteins. This situation disrupts the normal functioning of the ER, leading to a cellular stress response known as the unfolded protein response), leading to lipid peroxidation byproduct accumulation and toxic reactive oxygen species (ROS), which are highly reactive molecules derived from diatomic oxygen and include various forms such as superoxide (O₂⁻), hydroxyl radicals (•OH), and hydrogen peroxide (H₂O₂). Ferroptosis is closely associated with signaling molecules in lung cancer, including epidermal growth factor receptor (EGFR), mitogen-activated protein kinase (MAPK), hypoxia-inducible factor 1-alpha (HIF-1α), and P53, and is regulated by epigenetic factors such as microRNAs (miRNAs). miRNAs are small non-coding RNA molecules that regulate gene expression by binding to target messenger RNAs (mRNAs), leading to translational repression or degradation. Several miRNAs have been found to modulate ferroptosis by targeting key genes involved in iron metabolism, lipid peroxidation, and antioxidant defense pathways. The research on ferroptosis has expanded to target its role in lung cancer treatment and resistance prevention. This review encapsulates the significance of ferroptosis in lung cancer. Understanding the mechanisms and implications of ferroptosis in lung cancer cells may lead to targeted therapies exploiting cancer cell vulnerabilities to ferroptosis Also, improving treatment outcomes, and overcoming resistance.
Collapse
Affiliation(s)
- Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Heba M Midan
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Doaa Fathi
- Biochemistry Department, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Nourhan M Abdelmaksoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia, 32897, Egypt
| | - Mai A Abd-Elmawla
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nehal I Rizk
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Tamer M Abdelghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University, 3 Cairo-Belbeis Desert Road, P.O. Box 3020, El Salam, 11785, Cairo, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Reem K Shahin
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang, Republic of Korea
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
133
|
Li D, Zhang Z, Wang L. Emerging role of tumor microenvironmental nutrients and metabolic molecules in ferroptosis: Mechanisms and clinical implications. Biomed Pharmacother 2024; 179:117406. [PMID: 39255738 DOI: 10.1016/j.biopha.2024.117406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
In recent years, ferroptosis has gradually attracted increasing attention because of its important role in tumors. Ferroptosis resistance is an important cause of tumor metastasis, recurrence and drug resistance. Exploring the initiating factors and specific mechanisms of ferroptosis has become a key strategy to block tumor progression and improve drug sensitivity. As the external space in direct contact with tumor cells, the tumor microenvironment has a great impact on the biological function of tumor cells. The relationships between abnormal environmental characteristics (hypoxia, lactic acid accumulation, etc.) in the microenvironment and ferroptosis of tumor cells has not been fully characterized. This review focuses on the characteristics of the tumor microenvironment and summarizes the mechanisms of ferroptosis under different environmental factors, aiming to provide new insights for subsequent targeted therapy. Moreover, considering the presence of anticancer drugs in the microenvironment, we further summarize the mechanisms of ferroptosis to provide new strategies for the sensitization of tumor cells to drugs.
Collapse
Affiliation(s)
- Dongyu Li
- Department of VIP In-Patient Ward, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Lei Wang
- Department of Vascular and Thyroid Surgery, the First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
134
|
Liu Y, Yang P, Wang J, Peng W, Zhao J, Wang Z. MiRNA Regulates Ferroptosis in Cardiovascular and Cerebrovascular Diseases. DNA Cell Biol 2024; 43:492-509. [PMID: 39417991 DOI: 10.1089/dna.2024.0135] [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: 10/19/2024] Open
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) significantly contribute to global mortality and morbidity due to their complex pathogenesis involving multiple biological processes. Ferroptosis is an important physiological process in CCVDs, manifested by an abnormal increase in intracellular iron concentration. MiRNAs, a key class of noncoding RNA molecules, are crucial in regulating CCVDs through pathways like glutathione-glutathione peroxidase 4, glutamate/cystine transport, iron metabolism, lipid metabolism, and other oxidative stress pathways. This article summarizes the progress of miRNAs' regulation on CCVDs, aiming to provide insights for the diagnosis and treatment of CCVDs.
Collapse
Affiliation(s)
- Yiman Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Peijuan Yang
- Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Jingjing Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| | - Wu Peng
- Hengyang Maternal and Child Health Hospital, Hengyang, China
| | - Jinli Zhao
- Emergency Department, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
135
|
Zhao H, Dong Q, Hua H, Wu H, Ao L. Contemporary insights and prospects on ferroptosis in rheumatoid arthritis management. Front Immunol 2024; 15:1455607. [PMID: 39381004 PMCID: PMC11458427 DOI: 10.3389/fimmu.2024.1455607] [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: 06/27/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized primarily by persistent synovial inflammation and joint destruction. In recent years, ferroptosis, as a novel form of cell death, has garnered widespread attention due to its critical role in various diseases. This review explores the potential mechanisms of ferroptosis in RA and its relationship with the pathogenesis of RA, systematically analyzing the regulatory role of ferroptosis in synovial cells, chondrocytes, and immune cells. We emphasize the evaluation of ferroptosis-related pathways and their potential as therapeutic targets, including the development and application of inhibitors and activators. Although ferroptosis shows some promise in RA treatment, its dual role and safety issues in clinical application still require in-depth study. Future research should focus on elucidating the specific mechanisms of ferroptosis in RA pathology and developing more effective and safer therapeutic strategies to provide new treatment options for RA patients.
Collapse
Affiliation(s)
| | | | | | | | - Limei Ao
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Huhhot, Inner Mongolia, China
| |
Collapse
|
136
|
Zheng D, Liu T, Yu S, Liu Z, Wang J, Wang Y. Antimalarial Mechanisms and Resistance Status of Artemisinin and Its Derivatives. Trop Med Infect Dis 2024; 9:223. [PMID: 39330912 PMCID: PMC11435542 DOI: 10.3390/tropicalmed9090223] [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: 07/28/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Artemisinin is an endoperoxide sesquiterpene lactone isolated from Artemisia annua and is often used to treat malaria. Artemisinin's peroxide bridge is the key structure behind its antimalarial action. Scientists have created dihydroartemisinin, artemether, artesunate, and other derivatives preserving artemisinin's peroxide bridge to increase its clinical utility value. Artemisinin compounds exhibit excellent efficacy, quick action, and minimal toxicity in malaria treatment and have greatly contributed to malaria control. With the wide and unreasonable application of artemisinin-based medicines, malaria parasites have developed artemisinin resistance, making malaria prevention and control increasingly challenging. Artemisinin-resistant Plasmodium strains have been found in many countries and regions. The mechanisms of antimalarials and artemisinin resistance are not well understood, making malaria prevention and control a serious challenge. Understanding the antimalarial and resistance mechanisms of artemisinin drugs helps develop novel antimalarials and guides the rational application of antimalarials to avoid the spread of resistance, which is conducive to malaria control and elimination efforts. This review will discuss the antimalarial mechanisms and resistance status of artemisinin and its derivatives, which will provide a reference for avoiding drug resistance and the research and development of new antimalarial drugs.
Collapse
Affiliation(s)
- Dan Zheng
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Zhilong Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (D.Z.); (T.L.); (S.Y.); (Z.L.); (J.W.)
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| |
Collapse
|
137
|
Huang YQ, Peng K, Yan J, Chen HL, Jiang PY, Du YF, Ling X, Zhang SL, Wu J. The Participation of Ferroptosis in Fibrosis of the Heart and Kidney Tissues in Dahl Salt-Sensitive Hypertensive Rats. Am J Hypertens 2024; 37:784-791. [PMID: 38850192 DOI: 10.1093/ajh/hpae076] [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: 02/23/2024] [Revised: 04/29/2024] [Accepted: 05/20/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Salt-sensitive hypertension is often more prone to induce damage to target organs such as the heart and kidneys. Abundant recent studies have demonstrated a close association between ferroptosis and cardiovascular diseases. Therefore, we hypothesize that ferroptosis may be closely associated with organ damage in salt-sensitive hypertension. This study aimed to investigate whether ferroptosis is involved in the occurrence and development of myocardial fibrosis and renal fibrosis in salt-sensitive hypertensive rats. METHODS Ten 7-week-old male Dahl salt-sensitive (Dahl-SS) rats were adaptively fed for 1 week, then randomly divided into two groups and fed either a normal diet (0.3% NaCl, normal diet group) or a high-salt diet (8% NaCl, high-salt diet group) for 8 weeks. Blood pressure of the rats was observed, and analysis of the hearts and kidneys of Dahl-SS rats was conducted via hematoxylin-eosin (HE) staining, Masson staining, Prussian blue staining, transmission electron microscopy, tissue iron content detection, malondialdehyde content detection, immunofluorescence, and Western blot. RESULTS Compared to the normal diet group, rats in the high-salt diet group had increases in systolic blood pressure and diastolic blood pressure (P < 0.05); collagen fiber accumulation was observed in the heart and kidney tissues (P < 0.01), accompanied by alterations in mitochondrial ultrastructure, reduced mitochondrial volume, and increased density of the mitochondrial double membrane. Additionally, there were significant increases in both iron content and malondialdehyde levels (P < 0.05). Immunofluorescence and Western blot results both indicated significant downregulation (P < 0.05) of xCT and GPX4 proteins associated with ferroptosis in the high-salt diet group. CONCLUSIONS Ferroptosis is involved in the damage and fibrosis of the heart and kidney tissues in salt-sensitive hypertensive rats.
Collapse
Affiliation(s)
- Ya-Qi Huang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Kuang Peng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Jun Yan
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui-Lin Chen
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Pei-Yong Jiang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Ya-Fang Du
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Xiang Ling
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Si-Liang Zhang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Jie Wu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| |
Collapse
|
138
|
Lv J, Meng X, Li Y, Zhang R, Zhao Y, Yang X, Wang F, Wang X. Enhanced computed tomography radiomics predicts solute carrier family 7, member 11 in head and neck squamous cell carcinoma. Front Genet 2024; 15:1418578. [PMID: 39350768 PMCID: PMC11439659 DOI: 10.3389/fgene.2024.1418578] [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: 04/16/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Traditional prognostic indicators for head and neck squamous cell carcinoma (HNSCC), such as clinicopathological features, human papillomavirus status, and imaging examinations, often lack precision in guiding medical therapy. Therefore, discovering novel tumor biomarkers that can accurately assess prognosis and aid in personalized medical treatment for HNSCC is critical. Solute carrier family 7, member 11 (SLC7A11), is implicated in ferroptosis, and various malignant tumor therapies regulate its expression. However, the mechanisms regulating SLC7A11 expression, the transporter activity, and its specific role in controlling ferroptosis in cancer cells remain unknown. Thus, in this study, we aimed to develop an improved computed tomography (CT) radiomics model that could predict SLC7A11 expression in patients with HNSCC. Methods We used patient genomic data and corresponding augmented CT images for prognostic analysis and building models. Further, we investigated the potential molecular mechanisms underlying SLC7A11 expression in the immune microenvironment. Our radiomics model successfully predicted SLC7A11 mRNA expression in HNSCC tissues and elucidated its association with relevant genes and prognostic outcomes. Results SLC7A11 expression level was high within tumor tissues and was connected to the infiltration of eosinophil, CD8+ T-cell, and macrophages, which was associated with poor overall survival. Our models demonstrated robust predictive power. The distribution of radiomics scores (RAD scores) within the training and validation sets was markedly different between the high- and low-expression groups of SLC7A11. Conclusion SLC7A11 is likely an important factor in the prognosis of HNSCC. SLC7A11 expression can be predicted effectively and reliably by radiomics models based on enhanced CT.
Collapse
Affiliation(s)
- Jilian Lv
- Department of Oral and Maxillofacial Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiangze Meng
- Department of Oromaxillofacial Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanyuan Li
- Department of Oral and Maxillofacial Surgery, Lishui Central Hospital, Lishui Hospital of Zhejiang University, Lishui, China
| | - Rui Zhang
- Department of Oral and Maxillofacial Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yuan Zhao
- Department of Oral and Maxillofacial Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xi Yang
- Department of Oromaxillofacial Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinbin Wang
- Department of Oral and Maxillofacial Surgery, Lishui Central Hospital, Lishui Hospital of Zhejiang University, Lishui, China
| |
Collapse
|
139
|
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
|
140
|
Mo M, Pan L, Deng L, Liang M, Xia N, Liang Y. Iron Overload Induces Hepatic Ferroptosis and Insulin Resistance by Inhibiting the Jak2/stat3/slc7a11 Signaling Pathway. Cell Biochem Biophys 2024; 82:2079-2094. [PMID: 38801513 DOI: 10.1007/s12013-024-01315-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Recent studies showed that patients with iron overload had increased risk of insulin resistance or diabetes. Ferroptosis is a new type of cell death mainly caused by iron-dependent oxidative damage. In the present study, we investigated potential mechanisms of iron overload induced hepatic ferroptosis and insulin resistance through in vivo and in vitro experiments. In vivo, the mice models of iron overload were established by intraperitoneal injection of iron dextran. The changes of body weight, serum ferritin and blood glucose were measured. Hematoxylin-eosin (HE) and Perl's stainings were used to observe the pathological changes and iron deposition in the liver of mice. In vitro, HepG2 cells were treated with ferric ammonium citrate (FAC, 9 mmol/L, 24 h) to establish the cell models of iron overload. The labile iron pool, cell viability, glucose consumption and glycogen contents were measured. The ultrastructure of mitochondria was observed by transmission electron microscope (TEM). The malondialdehyde (MDA) and glutathione (GSH) kits were used to detect lipid peroxidation in liver tissues of mice and HepG2 cells. RT-PCR and Western blot were used to detect the mRNA and protein expression levels of ferroptosis factors and JAK2/STAT3 signaling pathway. In this study, we used the iron chelator deferasirox in mice and HepG2 cells. Iron overload caused weight loss, elevated serum ferritin, fasting blood glucose, fasting insulin, HOMA-IR, impaired glucose tolerance, and decreased insulin sensitivity in mice. HE staining and Perls staining showed clumps of iron deposition in the liver of iron overload mice. Iron overload could reduce the glucose consumption, increase MDA contents of HepG2 cells, while reduce glycogen and GSH contents in liver tissues of mice and HepG2 cells. TEM showed deletion of mitochondrial ridge and rupture of outer membrane in HepG2 cells with iron overload. Iron chelator deferasirox could significantly improve the above indicators, which might be related to the activation of JAK2/STAT3/SLC7A11 signaling pathway and hepatic ferroptosis. Iron overload could induce hepatic ferroptosis and insulin resistance by inhibiting the JAK2/STAT3/SLC7A11 signaling pathway, and the iron chelator deferasirox might improve hepatic insulin resistance induced by iron overload.
Collapse
Affiliation(s)
- Manqiu Mo
- Geriatric Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ling Pan
- Department of Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ling Deng
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Min Liang
- Geriatric Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ning Xia
- Geriatric Department of Endocrinology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Yuzhen Liang
- Department of Endocrinology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
141
|
Na X, Li L, Liu D, He J, Zhang L, Zhou Y. Natural products targeting ferroptosis pathways in cancer therapy (Review). Oncol Rep 2024; 52:123. [PMID: 39054952 PMCID: PMC11292301 DOI: 10.3892/or.2024.8782] [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: 04/14/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Ferroptosis inducers (FIN) have a key role in cancer therapy and provide novel and innovative treatment strategies. Although many researchers have performed FIN screening of synthetic compounds, studies on the identification of FIN from natural products are limited, particularly in the field of drug development and combination therapy. In this review, this gap was addressed by comprehensively summarizing recent studies on ferroptosis. The causes of ferroptosis were categorized into driving and defensive factors, elucidating key pathways and targets. Next, through summarizing research on natural products that induce ferroptosis, the study elaborated in detail on the natural products that have FIN functions. Their discovery and development were also described and insight for clinical drug development was provided. In addition, the mechanisms of action were analyzed and potential combination therapies, resistance reversal and structural enhancements were presented. By highlighting the potential of natural products in inducing ferroptosis for cancer treatment, this review may serve as a reference for utilizing these compounds against cancer. It not only showed the significance of natural products but may also promote further investigation into their therapeutic effects, thus encouraging research in this field.
Collapse
Affiliation(s)
- Xin Na
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Lin Li
- Yunnan Cancer Hospital (Third Affiliated Hospital of Kunming Medical University), Kunming, Yunnan 650118, P.R. China
| | - Dongmei Liu
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jiaqi He
- The First Clinical Medical College of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ling Zhang
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yiping Zhou
- School of Pharmaceutical Sciences & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
142
|
Bernkop-Schnürch AD, Huber K, Clauser A, Cziferszky M, Leitner D, Talasz H, Hermann M, Hohloch S, Gust R, Kircher B. Design, synthesis, and biological evaluation of novel halogenated chlorido[N,N'-bis(salicylidene)-1,2-bis(3-methoxyphenyl)ethylenediamine]iron(III) complexes as anticancer agents. J Biol Inorg Chem 2024; 29:583-599. [PMID: 39133326 PMCID: PMC11390779 DOI: 10.1007/s00775-024-02067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024]
Abstract
Iron(III) complexes based on N,N´-bis(salicylidene)ethylenediamine (salene) scaffolds have demonstrated promising anticancer features like induction of ferroptosis, an iron dependent cell death. Since poor cellular uptake limits their therapeutical potential, this study aimed to enhance the lipophilic character of chlorido[N,N'-bis(salicylidene)-1,2-bis(3-methoxyphenyl)ethylenediamine]iron(III) complexes by introducing lipophilicity improving ligands such as fluorine (X1), chlorine (X2) and bromine (X3) in 5-position in the salicylidene moieties. After detailed characterization the binding to nucleophiles, logP values and cellular uptake were determined. The complexes were further evaluated regarding their biological activity on MDA-MB 231 mammary carcinoma, the non-tumorous SV-80 fibroblast, HS-5 stroma and MCF-10A mammary gland cell lines. Stability of the complexes in aqueous and biological environments was proven by the lack of interactions with amino acids and glutathione. Cellular uptake was positively correlated with the logP values, indicating that higher lipophilicity enhanced cellular uptake. The complexes induced strong antiproliferative and antimetabolic effects on MDA-MB 231 cells, but were inactive on all non-malignant cells tested. Generation of mitochondrial reactive oxygen species, increase of lipid peroxidation and induction of both ferroptosis and necroptosis were identified as mechanisms of action. In conclusion, halogenation of chlorido[N,N'-bis(salicylidene)-1,2-bis(3-methoxyphenyl)ethylenediamine]iron(III) complexes raises their lipophilic character resulting in improved cellular uptake.
Collapse
Affiliation(s)
- Astrid Dagmar Bernkop-Schnürch
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Klaus Huber
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Armida Clauser
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
- Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology), Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Monika Cziferszky
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Daniel Leitner
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Heribert Talasz
- Biocenter, Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Stephan Hohloch
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Ronald Gust
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Brigitte Kircher
- Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology), Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria.
| |
Collapse
|
143
|
Zhong YJ, Liu LL, Zhao Y, Feng Z, Liu Y. Elucidating the molecular mechanisms behind the therapeutic impact of median nerve stimulation on cognitive dysfunction post-traumatic brain injury. Exp Gerontol 2024; 194:112500. [PMID: 38901771 DOI: 10.1016/j.exger.2024.112500] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE Ferroptosis represents a form of regulated cellular death dependent upon iron and lipid peroxidation derivatives, holding considerable implications for cerebral and neurologic pathologies. In the present study, we endeavored to elucidate the molecular mechanisms governing ferroptosis and appraise the therapeutic value of electrical stimulation of median nerve in addressing cognitive impairments following traumatic brain injury (TBI), employing a rodent model. METHODS In this study, we established a rat model to investigate the cognitive impairments resulting from TBI, followed by the application of median nerve stimulation (MNS). Initially, rats received an intraperitoneal injection of Erastin (2 mg/kg) prior to undergoing MNS. After 24 h of MNS treatment, the rats were subjected to an open field test to evaluate their cognitive and motor functions. Subsequently, we conducted biochemical assays to measure the serum levels of GSH, MDA and SOD. The structural integrity and cellular morphology of hippocampal tissue were examined through H&E staining, Nissl staining and transmission electron microscopy. Additionally, we assessed the expression levels of proteins crucial for neuronal health and function in the hippocampus, including VEGF, SLC7A11, GPX4, Nrf2, α-syn, NEUN and PSD95. RESULTS Compared to the control group, rats in the stimulation group demonstrated enhanced mobility, reduced levels of tissue damage, a decrease in MDA concentration, and increased levels of GSH and SOD. Additionally, there was a significant upregulation in the expression of proteins critical for cellular defense and neuronal health, including GPX4, SLC7A11, Nrf2, VEGF, α-syn, NEUN, and PSD95 proteins. Conversely, rats in the Erastin group demonstrated decreased mobility, exacerbated pathological tissue damage, elevated MDA concentration, and decreased levels of GSH and SOD. There was also a notable decrease in the expression of GPX4, SLC7CA11, Nrf2, and VEGF proteins. The expression levels of α-syn, NEUN, and PSD95 were similarly diminished in the Erastin group. Each of these findings was statistically significant, indicating that MNS exerts neuroprotective effect in the hippocampal tissue of rats with TBI by inhibiting the ferroptosis pathway. CONCLUSION (1) MNS may enhance the cognitive and behavioral performance of rats after TBI; (2) MNS can play a neuroprotective role by promoting the expression of nerve injury-related proteins, alleviating oxidative stress and ferroptosis process; (3) MNS may inhibit ferroptosis of neuronal cells by activating Nrf2/ GPX4 signaling pathway, thereby improving cognitive impairment in TBI rats.
Collapse
Affiliation(s)
- Ying-Jun Zhong
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Ling-Ling Liu
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yue Zhao
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China.
| | - Yuan Liu
- Department of Orthopedics, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China.
| |
Collapse
|
144
|
Delgado-Martín S, Martínez-Ruiz A. The role of ferroptosis as a regulator of oxidative stress in the pathogenesis of ischemic stroke. FEBS Lett 2024; 598:2160-2173. [PMID: 38676284 DOI: 10.1002/1873-3468.14894] [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: 11/10/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024]
Abstract
Ferroptosis is a unique form of cell death that was first described in 2012 and plays a significant role in various diseases, including neurodegenerative conditions. It depends on a dysregulation of cellular iron metabolism, which increases free, redox-active, iron that can trigger Fenton reactions, generating hydroxyl radicals that damage cells through oxidative stress and lipid peroxidation. Lipid peroxides, resulting mainly from unsaturated fatty acids, damage cells by disrupting membrane integrity and propagating cell death signals. Moreover, lipid peroxide degradation products can further affect cellular components such as DNA, proteins, and amines. In ischemic stroke, where blood flow to the brain is restricted, there is increased iron absorption, oxidative stress, and compromised blood-brain barrier integrity. Imbalances in iron-transport and -storage proteins increase lipid oxidation and contribute to neuronal damage, thus pointing to the possibility of brain cells, especially neurons, dying from ferroptosis. Here, we review the evidence showing a role of ferroptosis in ischemic stroke, both in recent studies directly assessing this type of cell death, as well as in previous studies showing evidence that can now be revisited with our new knowledge on ferroptosis mechanisms. We also review the efforts made to target ferroptosis in ischemic stroke as a possible treatment to mitigate cellular damage and death.
Collapse
Affiliation(s)
- Susana Delgado-Martín
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Spain
| |
Collapse
|
145
|
Zhang F, Wan J, Zhong J, Mo J. ANK1 inhibits malignant progression of osteosarcoma by promoting ferroptosis. BMC Cancer 2024; 24:1075. [PMID: 39217322 PMCID: PMC11365275 DOI: 10.1186/s12885-024-12836-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE Osteosarcoma (OS) is a primary bone tumor with high malignancy and poor prognosis. Ferroptosis plays a crucial role in OS. This study aimed to evaluate the effects of Ankyrin 1 (ANK1) on OS and to investigate its specific mechanisms. METHODS Microarray datasets related to "osteosarcoma" were selected for this study. Relevant hub genes in OS were identified through bioinformatics analysis. Transfected U-2OS and MG-63 cells were used for in vitro experiments. The effects of ANK1 overexpression on cell viability, migration, and invasion were determined through CCK-8, wound healing, and transwell assays. An OS mouse model was established for the in vivo experiments. Hematoxylin-eosin staining and immunohistochemistry were conducted to observe the histological effects of ANK1 overexpression on mouse tumors. TUNEL staining was performed to evaluate apoptosis in mouse. RESULTS There were 159 common differentially expressed genes in the GSE16088 and GSE19276 datasets. The hub genes ANK1, AHSP, GYPB, GYPA, KEL, and ALAS2 were identified. Pan-cancer analysis verified that ANK1 was closely associated with cancer prognosis and immune infiltration. Furthermore, ANK1 overexpression inhibited the proliferation, migration, and invasion of OS cells and promoted ferroptosis, while ferroptosis inhibitor (fer-1) weakened these effects. Moreover, ANK1 overexpression suppressed tumor growth, promoted apoptosis, reduced the number of Ki67 positive cells, and elevated the number of caspase-3 positive cells in vivo. CONCLUSIONS ANK1 is a prognosis biomarker of OS that can alleviate the progression of OS by promoting ferroptosis.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China
| | - Junming Wan
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628, Zhenyuan Road, Guangming District, Shenzhen City, 518107, Guangdong Province, China
| | - Jinghua Zhong
- Department of Medical oncology, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City City, 341000, Jiangxi Province, China
| | - Jianwen Mo
- Department of Orthopaedics, First Affiliated Hospital of Gannan Medical University, No. 23, Qingnian Road, Zhanggong District, Ganzhou City, 341000, Jiangxi Province, China.
| |
Collapse
|
146
|
Jiang M, Wu S, Xie K, Zhou G, Zhou W, Bao P. The significance of ferroptosis in renal diseases and its therapeutic potential. Heliyon 2024; 10:e35882. [PMID: 39220983 PMCID: PMC11363859 DOI: 10.1016/j.heliyon.2024.e35882] [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: 01/13/2024] [Revised: 04/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Kidney diseases are significant global public health concern, with increasing prevalence and substantial economic impact. Developing novel therapeutic approaches are essential for delaying disease progression and improving patient quality of life. Cell death signifying the termination of cellular life, could facilitate appropriate bodily development and internal homeostasis. Recently, regulated cell death (RCD) forms such as ferroptosis, characterized by iron-dependent lipid peroxidation, has garnered attention in diverse renal diseases and other pathological conditions. This review offers a comprehensive examination of ferroptosis, encompassing an analysis of the involvement of iron and lipid metabolism, the System Xc - /glutathione/glutathione peroxidase 4 signaling, and additional associated pathways. Meanwhile, the review delves into the potential of targeting ferroptosis as a therapeutic approach in the management of acute kidney injury (AKI), chronic kidney disease (CKD), diabetic nephropathy, and renal tumors. Furthermore, it emphasizes the significance of ferroptosis in the transition from AKI to CKD and further accentuates the potential for repurposing drug and utilizing traditional medicine in targeting ferroptosis-related pathways for clinical applications. The integrated review provides valuable insights into the role of ferroptosis in kidney diseases and highlights the potential for targeting ferroptosis as a therapeutic strategy.
Collapse
Affiliation(s)
- Mingzhu Jiang
- The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Shujun Wu
- The Yangzhou School of Clinical Medicine of Dalian Medical University, Yangzhou, China
| | - Kun Xie
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Gang Zhou
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Wei Zhou
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, China
| | - Ping Bao
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
147
|
Li J, Chen D, Suo J, Li J, Zhang Y, Wang Y, Deng Z, Zhang Q, Ma B. Triptolide induced spermatogenesis dysfunction via ferroptosis activation by promoting K63-linked GPX4 polyubiquitination in spermatocytes. Chem Biol Interact 2024; 399:111130. [PMID: 38960301 DOI: 10.1016/j.cbi.2024.111130] [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: 05/13/2024] [Revised: 06/19/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
Triptolide (TP) is a major bioactive compound derived from Tripterygium wilfordii Hook. F. (TwHF) known for its medicinal properties, but it also exhibits potential toxic effects. It has been demonstrated to induce severe male reproductive toxicity, yet the precise mechanism behind this remains unclear, which limits its broad clinical application. This study aimed to investigate the mechanisms underlying testicular damage and spermatogenesis dysfunction induced by TP in mice, using both mouse models and the spermatocyte-derived cell line GC-2spd. In the present study, it was found that TP displayed significant testicular microstructure damaged and spermatogenesis defects including lower concentration and abnormal morphology by promoting ROS formation, MDA production and restraining GSH level, glutathione peroxidase 4 (GPX4) expression in vivo. Furthermore, Ferrostatin-1 (FER-1), a ferroptosis inhibitor, was found to significantly reduce the accumulation of lipid peroxidation, alleviate testicular microstructural damage, and enhance spermatogenic function in mice. Besides, notably decreased cell viability, collapsed mitochondrial membrane potential, and elevated DNA damage were observed in vitro. The above-mentioned phenomenon could be reversed by pre-treatment of FER-1, indicating that ferroptosis participated in the TP-mediated spermatogenesis dysfunction. Mechanistically, TP could enhance GPX4 ubiquitin degradation via triggering K63-linked polyubiquitination of GPX4, thereby stimulating ferroptosis in spermatocytes. Functionally, GPX4 deletion intensified ferroptosis and exacerbated DNA damage in GC-2 cells, while GPX4 overexpression mitigated ferroptosis induced by TP. Overall, these findings for the first time indicated a vital role of ferroptosis in TP induced-testicular injury and spermatogenic dysfunction through promoting GPX4 K63-linked polyubiquitination, which hopefully offers a potential therapeutic avenue for TP-related male reproductive damage. In addition, this study also provides a theoretical foundation for the improved clinical application of TP or TwHF in the future.
Collapse
Affiliation(s)
- Jiaqi Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Dezhi Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Jialiang Suo
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Jiaqi Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yimu Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Yu Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Zhewen Deng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
148
|
Hong G, Chen W, Gong M, Wu Y, Shu G, Xiao Y, Zhang T, ShuXiong X. KAT7 suppresses tumorigenesis in clear cell renal cell carcinoma (ccRCC) by regulating cell cycle and ferroptosis sensitivity. Exp Cell Res 2024; 441:114149. [PMID: 38960363 DOI: 10.1016/j.yexcr.2024.114149] [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: 04/26/2024] [Revised: 06/23/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most aggressive malignancies in the urological system, known for its high immunogenicity. However, its pathogenesis remains unclear. This study utilized bioinformatics algorithms and in vitro experiments to investigate the role of KAT7 in ccRCC. The results indicate that KAT7 is significantly downregulated in ccRCC tissues and cell lines, which is linked to distant metastasis and unfavorable outcomes in ccRCC patients. Overexpression of KAT7 in vitro notably decreased the proliferation, migration, and invasion of renal cancer cells and inhibited Epithelial-Mesenchymal Transition (EMT). Additionally, Gene Set Enrichment Analysis (GSEA) demonstrated that KAT7-related gene functions are associated with cell cycle and ferroptosis transcription factors. Treatment with a KAT7 acetylation inhibitor in ccRCC cell lines reversed the S phase arrest caused by KAT7 overexpression. Similarly, ferroptosis inhibitors alleviated ferroptosis induced by overexpressed KAT7. In conclusion, the findings suggest that KAT7 acts as a tumor suppressor in ccRCC by modulating the cell cycle and ferroptosis sensitivity, underscoring its potential as a therapeutic target and prognostic biomarker for renal cell carcinoma patients.
Collapse
Affiliation(s)
- GuangYi Hong
- Guizhou University Medicine College, Guiyang, 550025, Guizhou Province, China
| | - Wei Chen
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - MaoDi Gong
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - YiKun Wu
- Guizhou University Medicine College, Guiyang, 550025, Guizhou Province, China
| | - GuoFeng Shu
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - Yu Xiao
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China
| | - Tao Zhang
- Guizhou University Medicine College, Guiyang, 550025, Guizhou Province, China
| | - Xu ShuXiong
- Department of Urology, Guizhou Provincial People's Hospital, No.83, East Zhongshan Road, Guiyang, Guizhou, China.
| |
Collapse
|
149
|
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
|
150
|
Peng W, Chung KB, Lawrence BP, O’Banion MK, Dirksen RT, Wojtovich AP, Onukwufor JO. DMT1 knockout abolishes ferroptosis induced mitochondrial dysfunction in C. elegans amyloid β proteotoxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607074. [PMID: 39149382 PMCID: PMC11326247 DOI: 10.1101/2024.08.08.607074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Iron is critical for neuronal activity and metabolism, and iron dysregulation alters these functions in age-related neurodegenerative disorders, such as Alzheimer's disease (AD). AD is a chronic neurodegenerative disease characterized by progressive neuronal dysfunction, memory loss and decreased cognitive function. AD patients exhibit elevated iron levels in the brain compared to age-matched non-AD individuals. However, the degree to which iron overload contributes to AD pathogenesis is unclear. Here, we evaluated the involvement of ferroptosis, an iron-dependent cell death process, in mediating AD-like pathologies in C. elegans. Results showed that iron accumulation occurred prior to the loss of neuronal function as worms age. In addition, energetic imbalance was an early event in iron-induced loss of neuronal function. Furthermore, the loss of neuronal function was, in part, due to increased mitochondrial reactive oxygen species mediated oxidative damage, ultimately resulting in ferroptotic cell death. The mitochondrial redox environment and ferroptosis were modulated by pharmacologic processes that exacerbate or abolish iron accumulation both in wild-type worms and worms with increased levels of neuronal amyloid beta (Aβ). However, neuronal Aβ worms were more sensitive to ferroptosis-mediated neuronal loss, and this increased toxicity was ameliorated by limiting the uptake of ferrous iron through knockout of divalent metal transporter 1 (DMT1). In addition, DMT1 knockout completely suppressed phenotypic measures of Aβ toxicity with age. Overall, our findings suggest that iron-induced ferroptosis alters the mitochondrial redox environment to drive oxidative damage when neuronal Aβ is overexpressed. DMT1 knockout abolishes neuronal Aβ-associated pathologies by reducing neuronal iron uptake.
Collapse
Affiliation(s)
- Wilson Peng
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester NY, 14642 USA
| | - Kaitlin B Chung
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester NY, 14642 USA
| | - B Paige Lawrence
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA 14642
| | - M Kerry O’Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA 14642
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester NY, 14642 USA
| | - Andrew P Wojtovich
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester NY, 14642 USA
- Department of Anesthesiology and Perioperative Medicine, University of Rochester School of Medicine and Dentistry, Rochester NY, 14642 USA
| | - John O Onukwufor
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester NY, 14642 USA
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA 14642
| |
Collapse
|