1
|
Chen S, Huang Y, Liu Y, Jiang L, Chen Y. Shikonin promotes ferroptosis though NSUN2-mediated m 5C methylation modification of TFRC in acute myelocytic leukemia. Hematology 2025; 30:2495221. [PMID: 40270446 DOI: 10.1080/16078454.2025.2495221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/12/2025] [Indexed: 04/25/2025] Open
Abstract
Shikonin (SHK), extracted from the traditional Chinese herb Lithospermum erythrorhizon, demonstrates a wide range of pharmacological activities. This study aimed to explore the role and underlying mechanisms of the 5-methylcytosine (m5C) RNA methyltransferase NOL1/NOP2/SUN domain (NSUN)2 in acute myelocytic leukemia (AML). To assess cell viability and death, we employed Cell Counting Kit-8 and propidium iodide staining. Ferroptosis-related markers were evaluated using commercial kits and Western blot analysis. The m5C levels of ferroptosis-associated mRNAs were quantified by methylated RNA immunoprecipitation (MeRIP)-qPCR. The specific m5C sites on the transferrin receptor (TFRC) mRNA were identified through a dual-luciferase reporter assay, while the interaction between NSUN2 and TFRC was investigated using RNA immunoprecipitation (RIP). The role of SHK in vivo was explored using a xenografted tumor model. Our findings revealed that SHK significantly reduced cell viability and induced cell death and ferroptosis in HL-60 and NB4 cells. Notably, SHK treatment led to an upregulation of NSUN2 expression. Inhibition of NSUN2 reversed the effects of SHK, restoring cell viability and reducing cell death and ferroptosis. Mechanistically, NSUN2 enhanced TFRC expression via m5C-dependent methylation. Overexpression of NSUN2 similarly decreased cell viability and increased cell death and ferroptosis, effects that were mitigated upon silencing of TFRC. In vivo, SHK treatment effectively suppressed tumor growth in xenografted mice. In summary, our study demonstrated that SHK promoted cell death and ferroptosis in AML by modulating NSUN2-mediated m5C methylation of TFRC. These findings provided novel insights into potential therapeutic strategies for AML.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Mice
- Animals
- Methyltransferases/metabolism
- Methyltransferases/genetics
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Naphthoquinones/pharmacology
- Methylation
- Cell Line, Tumor
- HL-60 Cells
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Shuyu Chen
- School of Pharmacy, Zhangzhou Health Vocational College, Zhangzhou, People's Republic of China
| | - Yiqun Huang
- Department of Hematology, Zhangzhou Affiliated Hospital to Fujian Medical University, Zhangzhou, People's Republic of China
| | - Yinhao Liu
- Department of Diabetes Nephrology, Zhangzhou Hospital of Traditional Chinese Medicine, Zhangzhou, People's Republic of China
| | - Liuxuan Jiang
- The School of Clinical Medicine, Fujian Medical University, Zhangzhou, People's Republic of China
| | - Yuqing Chen
- School of Pharmacy, Zhangzhou Health Vocational College, Zhangzhou, People's Republic of China
| |
Collapse
|
2
|
Shi J, Cheng Y, Wang L, Xing W, Li Y, Sun X, Lv Y, Zhang Y, Li Y, Zhao W. SR-B1 deficiency suppresses progression in acute myeloid leukemia via ferroptosis and reverses resistance to venetoclax. Free Radic Biol Med 2025; 233:24-38. [PMID: 40122151 DOI: 10.1016/j.freeradbiomed.2025.03.031] [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: 01/21/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Increase of immature myeloid cells in the bone marrow drives the development of acute myeloid leukemia (AML). The study aimed to clarify the biological function and regulatory mechanism of scavenger receptor class B type 1 (SR-B1) in AML, mainly its effect on ferroptosis and the influences on leukemogenesis and resistance to venetoclax. In this study, we found that the SR-B1 deficiency directly reduced the invasion and promoted death of malignant cells in AML. Strikingly, SR-B1 deficiency could up-regulated the expression of ferroptosis-related proteins to facilitate the occurrence of ferroptosis in vivo, and could also down-regulated the expression of apoptosis related protein B-cell lymphoma-2 (BCL-2). And then, we confirmed SR-B1 inhibitor block lipid transport-1 (BLT-1) had a superior efficacy in AML cells and AML model mice. The study found that whether SR-B1 deficiency or BLT-1 treatment could cause iron deposition and the accumulation of lipid peroxides in vivo, thereby suppressing leukemogenesis through ferroptosis. Critically, we found that SR-B1 inhibitor BLT-1 could reverse drug-resistance of venetoclax to promote AML cells death via ferroptosis. Our finding identified that SR-B1 as a critical regulator of the proliferation in AML which could provide a promising therapeutic strategy against malignant myeloid leukemia cells and drug-resistance.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Animals
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Sulfonamides/pharmacology
- Mice
- Humans
- Drug Resistance, Neoplasm/genetics
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Scavenger Receptors, Class B/genetics
- Scavenger Receptors, Class B/deficiency
- Scavenger Receptors, Class B/metabolism
- Scavenger Receptors, Class B/antagonists & inhibitors
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Antineoplastic Agents/pharmacology
- Disease Progression
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
Collapse
Affiliation(s)
- Junfeng Shi
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yifeng Cheng
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Lixue Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wen Xing
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yudi Li
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiulin Sun
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yunpeng Lv
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yichuan Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yanming Li
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Wenhua Zhao
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
3
|
Li P, Wu Y, Deng Z, Samad A, Xi Y, Song J, Zhang Y, Li J, Zhou YA, Xiong Q, Wu C. Two novel SH3TC2 mutations predispose to Charcot-Marie-Tooth disease type 4C by mistargeting away from TFRC. Cell Signal 2025; 130:111669. [PMID: 39961410 DOI: 10.1016/j.cellsig.2025.111669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/08/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Charcot-Marie-Tooth disease type 4C (CMT4C) is an autosomal recessive form of demyelinating neuropathy caused by the biallelic pathogenic mutations in the SH3TC2 gene and characterized by progressive scoliosis, muscular atrophy, distal weakness, and reduced nerve conduction velocity. Here, we report two novel SH3TC2 mutations (c.452dupT and c.731 + 1G > T) from a proband with typical clinical manifestations of CMT4C. Splicing assay reveals the SH3TC2 c.731 + 1G > T mutation leads to a 58-nucleotide (nt) deletion from the downstream of exon 6 causing a frameshift and resulting in an early termination of protein expression. Protein expression assay indicates SH3TC2 c.452dupT mutant is degraded by both the nonsense mediated decay (NMD) and the ubiquitin-proteasome pathway. Moreover, our intracellular immunofluorescence, co-immunoprecipitation, liquid chromatography mass spectrometry and molecular docking describe that SH3TC2 interacts with the transferrin receptor protein 1 (TFRC) encoding a cell surface receptor playing a crucial role in mediating iron homeostasis. Interestingly, both the two novel SH3TC2 mutations present in our CMT4C patients are defective in the association with TFRC. Our study reveals the pathogenesis of these two novel SH3TC2 mutations and indicates that the SH3TC2-TFRC interaction is relevant for peripheral nerve pathophysiology, thus provides a novel insight into the pathophysiology of CMT4C neuropathy.
Collapse
Affiliation(s)
- Ping Li
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China.
| | - Ya Wu
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China
| | - Zhanjin Deng
- Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Abdus Samad
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Yuqian Xi
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China
| | - Jiawen Song
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China
| | - Yanping Zhang
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China
| | - Jianwei Li
- Precision Laboratory of Vascular Medicine, Shanxi Cardiovascular Hospital, Taiyuan, China
| | - Yong-An Zhou
- Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Qiuhong Xiong
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China.
| | - Changxin Wu
- Institutes of Biomedical Sciences, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education of China, the Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan, China.
| |
Collapse
|
4
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
5
|
Tsui KH, Li CJ, Lin LT. Melatonin supplementation attenuates cuproptosis and ferroptosis in aging cumulus and granulosa cells: potential for improving IVF outcomes in advanced maternal age. Reprod Biol Endocrinol 2024; 22:138. [PMID: 39516964 PMCID: PMC11545199 DOI: 10.1186/s12958-024-01311-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Advanced maternal age is associated with decreased oocyte quantity and quality and in vitro fertilization (IVF) success rates. This study aimed to investigate whether melatonin supplementation can improve IVF outcomes in women of advanced maternal age by modulating cuproptosis and ferroptosis. METHODS This prospective cohort study included 161 women aged 35-45 years undergoing IVF-frozen embryo transfer cycles. Participants were assigned to either melatonin (n = 86, 2 mg daily for ≥ 8 weeks) or control (n = 75) groups. Cumulus cells were analyzed for cuproptosis and ferroptosis-related gene expression. Additional experiments were conducted on the HGL5 human granulosa cell line to assess mitochondrial function and metabolic reprogramming. RESULTS Melatonin supplementation significantly improved IVF outcomes in women aged ≥ 38 years, increasing clinical pregnancy rates (46.0% vs. 20.3%, P < 0.01), ongoing pregnancy rates (36.5% vs. 15.3%, P < 0.01), and live birth rates (33.3% vs. 15.3%, P < 0.05). In cumulus cells from patients, gene expression analysis revealed that melatonin modulated cuproptosis and ferroptosis-related genes, including ATP7B and GPX4, with more pronounced effects in the ≥ 38 years group. This suggests melatonin enhances cellular resilience against oxidative stress and metal-induced toxicity in the ovarian microenvironment. In vitro studies using HGL5 cells showed melatonin reduced oxidative stress markers, improved mitochondrial function, restored expression of glycolysis and TCA cycle-related genes and modulated cuproptosis and ferroptosis-related gene expression. These findings provide mechanistic insight into melatonin's protective effects against regulated cell death in ovarian cells, potentially explaining the improved IVF outcomes observed. CONCLUSIONS Melatonin supplementation significantly improved IVF outcomes in women of advanced maternal age, particularly those ≥ 38 years old, likely by modulating cuproptosis and ferroptosis and enhancing mitochondrial function in cumulus and granulosa cells. These results suggest that melatonin could be a promising adjuvant therapy for improving IVF success rates in older women.
Collapse
Affiliation(s)
- Kuan-Hao Tsui
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, 112, Taiwan
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan
| | - Li-Te Lin
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, 813, Taiwan.
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung County, 90741, Taiwan.
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.
- Institute of Biopharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.
- Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, Taipei, 112, Taiwan.
| |
Collapse
|
6
|
Xing P, Zhou M, Sun J, Wang D, Huang W, An P. NAT10-mediated ac 4C acetylation of TFRC promotes sepsis-induced pulmonary injury through regulating ferroptosis. Mol Med 2024; 30:140. [PMID: 39251905 PMCID: PMC11382515 DOI: 10.1186/s10020-024-00912-w] [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: 05/09/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac4C) is mediated by the ac4C "writer", N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of NAT10 in SPI and the underlying mechanism. METHODS Twenty-three acute respiratory distress syndrome (ARDS) patients and 27 non-ARDS volunteers were recruited. A sepsis rat model was established. Reverse transcription-quantitative polymerase chain reaction was used to detect the expression of NAT10 and transferrin receptor (TFRC). Cell viability was detected by cell counting kit-8. The levels of Fe2+, glutathione, and malondialdehyde were assessed by commercial kits. Lipid reactive oxygen species production was measured by flow cytometric analysis. Western blot was used to detect ferroptosis-related protein levels. Haematoxylin & eosin staining was performed to observe the pulmonary pathological symptoms. RESULTS The results showed that NAT10 was increased in ARDS patients and lipopolysaccharide-treated human lung microvascular endothelial cell line-5a (HULEC-5a) cells. NAT10 inhibition increased cell viability and decreased ferroptosis in HULEC-5a cells. TFRC was a downstream regulatory target of NAT10-mediated ac4C acetylation. Overexpression of TFRC decreased cell viability and promoted ferroptosis. In in vivo study, NAT10 inhibition alleviated SPI. CONCLUSION NAT10-mediated ac4C acetylation of TFRC aggravated SPI through promoting ferroptosis.
Collapse
Affiliation(s)
- Pengcheng Xing
- Department of Emergency and Intensive Care Unit, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 222, West Three Road Aroud Lake, Nanhui New Town, Pudong New Area, Shanghai, 201306, China
| | - Minjie Zhou
- Department of Emergency and Intensive Care Unit, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 222, West Three Road Aroud Lake, Nanhui New Town, Pudong New Area, Shanghai, 201306, China
| | - Jian Sun
- Department of Emergency and Intensive Care Unit, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 222, West Three Road Aroud Lake, Nanhui New Town, Pudong New Area, Shanghai, 201306, China
| | - Donglian Wang
- Department of Emergency and Intensive Care Unit, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 222, West Three Road Aroud Lake, Nanhui New Town, Pudong New Area, Shanghai, 201306, China
| | - Weipeng Huang
- Department of Emergency and Intensive Care Unit, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 222, West Three Road Aroud Lake, Nanhui New Town, Pudong New Area, Shanghai, 201306, China
| | - Peng An
- Department of Emergency and Intensive Care Unit, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No. 222, West Three Road Aroud Lake, Nanhui New Town, Pudong New Area, Shanghai, 201306, China.
| |
Collapse
|
7
|
Long Z, Luo Y, Yu M, Wang X, Zeng L, Yang K. Targeting ferroptosis: a new therapeutic opportunity for kidney diseases. Front Immunol 2024; 15:1435139. [PMID: 39021564 PMCID: PMC11251909 DOI: 10.3389/fimmu.2024.1435139] [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: 05/19/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
Ferroptosis is a form of non-apoptotic regulated cell death (RCD) that depends on iron and is characterized by the accumulation of lipid peroxides to lethal levels. Ferroptosis involves multiple pathways including redox balance, iron regulation, mitochondrial function, and amino acid, lipid, and glycometabolism. Furthermore, various disease-related signaling pathways also play a role in regulating the process of iron oxidation. In recent years, with the emergence of the concept of ferroptosis and the in-depth study of its mechanisms, ferroptosis is closely associated with various biological conditions related to kidney diseases, including kidney organ development, aging, immunity, and cancer. This article reviews the development of the concept of ferroptosis, the mechanisms of ferroptosis (including GSH-GPX4, FSP1-CoQ1, DHODH-CoQ10, GCH1-BH4, and MBOAT1/2 pathways), and the latest research progress on its involvement in kidney diseases. It summarizes research on ferroptosis in kidney diseases within the frameworks of metabolism, reactive oxygen biology, and iron biology. The article introduces key regulatory factors and mechanisms of ferroptosis in kidney diseases, as well as important concepts and major open questions in ferroptosis and related natural compounds. It is hoped that in future research, further breakthroughs can be made in understanding the regulation mechanism of ferroptosis and utilizing ferroptosis to promote treatments for kidney diseases, such as acute kidney injury(AKI), chronic kidney disease (CKD), diabetic nephropathy(DN), and renal cell carcinoma. This paves the way for a new approach to research, prevent, and treat clinical kidney diseases.
Collapse
Affiliation(s)
- Zhiyong Long
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanfang Luo
- Department of Nephrology, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Min Yu
- Department of Physical Medicine and Rehabilitation, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Nephrology, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Liuting Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
8
|
Zhou J, Liu Y, Wei X, Yuan M, Zhang X, Qin L, Cui B, Li P, Zhang J, Feng Z, Jiang J, Yuan X, Xu R, Zhang Z, Zhang P, Zhang X, Yang Y. Glycnsisitin A: A promising bicyclic peptide against heart failure that facilitates TFRC-mediated uptake of iron in cardiomyocytes. Acta Pharm Sin B 2024; 14:3125-3139. [PMID: 39027250 PMCID: PMC11252382 DOI: 10.1016/j.apsb.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 07/20/2024] Open
Abstract
Zhigancao decoction is a traditional prescription for treating irregular pulse and palpitations in China. As the monarch drug of Zhigancao decoction, the bioactive molecules of licorice against heart diseases remain elusive. We established the HRESIMS-guided method leading to the isolation of three novel bicyclic peptides, glycnsisitins A-C (1-3), with distinctive C-C and C-O-C side-chain-to-side-chain linkages from the roots of Glycyrrhiza uralensis (Licorice). Glycnsisitin A demonstrated stronger cardioprotective activity than glycnsisitins B and C in an in vitro model of doxorubicin (DOX)-induced cardiomyocyte injury. Glycnsisitin A treatment not only reduced the mortality of heart failure (HF) mice in a dose-dependent manner but also significantly attenuated DOX-induced cardiac dysfunction and myocardial fibrosis. Gene set enrichment analysis (GSEA) of the differentially expressed genes indicated that the cardioprotective effect of glycnsisitin A was mainly attributed to its ability to maintain iron homeostasis in the myocardium. Mechanistically, glycnsisitin A interacted with transferrin and facilitated its binding to the transferrin receptor (TFRC), which caused increased uptake of iron in cardiomyocytes. These findings highlight the key role of bicyclic peptides as bioactive molecules of Zhigancao decoction for the treatment of HF, and glycnsisitin A constitutes a promising therapeutic agent for the treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | | | - Lingfeng Qin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bing Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jing Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ziming Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Jianshuang Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiang Yuan
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ruibing Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhimeng Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Peicheng Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiaowei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yanan Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
9
|
Liu ZY, Lin LC, Liu ZY, Yang JJ, Tao H. m6A epitranscriptomic and epigenetic crosstalk in cardiac fibrosis. Mol Ther 2024; 32:878-889. [PMID: 38311850 PMCID: PMC11163196 DOI: 10.1016/j.ymthe.2024.01.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/27/2023] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
Cardiac fibrosis, a crucial pathological characteristic of various cardiac diseases, presents a significant treatment challenge. It involves the deposition of the extracellular matrix (ECM) and is influenced by genetic and epigenetic factors. Prior investigations have predominantly centered on delineating the substantial influence of epigenetic and epitranscriptomic mechanisms in driving the progression of fibrosis. Recent studies have illuminated additional avenues for modulating the progression of fibrosis, offering potential solutions to the challenging issues surrounding fibrosis treatment. In the context of cardiac fibrosis, an intricate interplay exists between m6A epitranscriptomic and epigenetics. This interplay governs various pathophysiological processes: mitochondrial dysfunction, mitochondrial fission, oxidative stress, autophagy, apoptosis, pyroptosis, ferroptosis, cell fate switching, and cell differentiation, all of which affect the advancement of cardiac fibrosis. In this comprehensive review, we meticulously analyze pertinent studies, emphasizing the interplay between m6A epitranscriptomics and partial epigenetics (including histone modifications and noncoding RNA), aiming to provide novel insights for cardiac fibrosis treatment.
Collapse
Affiliation(s)
- Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Zhen-Yu Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P.R. China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China.
| |
Collapse
|
10
|
Liu Y, Liu D, Liu Y, Fu B, Ji S, Wang R, Yan F, Wang H, Zhao D, Yang W, Wang J, Tang L. Comprehensive Proteomics Analysis Reveals Dynamic Phenotypes of Tumor-Associated Macrophages and Their Precursor Cells in Tumor Progression. J Proteome Res 2024; 23:822-833. [PMID: 38173118 DOI: 10.1021/acs.jproteome.3c00725] [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: 01/05/2024]
Abstract
Tumor-associated macrophages (TAMs) are key regulators in tumor progression, but the precise role of bone marrow-derived monocytes (Mons) as TAM precursors and their dynamic phenotypes regulated by the tumor microenvironment (TME) remain unclear. Here, we developed an optimized microproteomics workflow to analyze low-cell-number mouse myeloid cells. We sorted TAMs and their corresponding Mons (1 × 105 per sample) from individual melanoma mouse models at both the early and late stages. We established the protein expression profiles for these cells by mass spectrometry. Subsequently, we analyzed the dynamics phenotypes of TAMs and identified a characteristic protein expression profile characterized by upregulated cholesterol metabolism and downregulated immune responses during tumor progression. Moreover, we found the downregulation of both STAT5 and PYCARD expression not only in late-stage TAMs but also in late-stage Mons, indicating a loss of the ability to induce inflammatory responses prior to Mons infiltration into TME. Taken together, our study provides valuable insights into the progression-dependent transitions between TAMs and their precursor cells, as well as the cross-organ communications of tumor and bone marrow.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Di Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yuchen Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Bin Fu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Shuhui Ji
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ruixuan Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fang Yan
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Huan Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Dianyuan Zhao
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Wenting Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jian Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Li Tang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
11
|
Lv Y, Shao Y, Jiang C, Wang Y, Li Y, Li Y, Duan X, Dong S, Lin J, Zhang H, Shan H. Quantitative proteomics based on TMT revealed the response of PK15 cells infected PEDV wild strain. Microb Pathog 2024; 186:106503. [PMID: 38142905 DOI: 10.1016/j.micpath.2023.106503] [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: 08/12/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023]
Abstract
Porcine epidemic diarrhea (PED), caused by porcine epidemic diarrhea virus (PEDV), is an acute and highly contagious enteric disease with a high mortality rate in suckling piglets. Identification of proteins associated with PEDV infection may provide insights into the pathogenesis of this viral disease. In this study, we employed tandem mass tag (TMT) quantitative protein analysis to investigate proteomic changes in PK15 cells following PEDV infection, and differential protein expression profiles were obtained at 0 h, 24 h, and 48 h post-infection. Overall, a total of 6330 proteins were identified. Applying criteria for fold change >1.5 < 0.67 and p-values <0.05 resulted in the identification of 59 up-regulated proteins and 103 down-regulated proteins that exhibited significant alterations in the H24 group compared to the H0 group. The H48 group demonstrated significant upregulation of 110 proteins and downregulation of 144 proteins compared to the H0 group; additionally, there were also 10 upregulated and 30 downregulated proteins in the H48 group when compared to the H24 group. These differentially expressed proteins (DEPs) were involved in immune response regulation, signal transduction, lipid transport and metabolism processes as well as cell apoptosis pathways. Based on these DEPs, we propose that PEDV may disrupt signal transduction pathways along with lipid transport and metabolism processes leading to maximal viral replication, it may also trigger inflammatory cascades accordingly. These findings could provide valuable information for elucidating specific pathogenesis related to PEDV infection while contributing towards developing new antiviral strategies.
Collapse
Affiliation(s)
- Yuting Lv
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yu Shao
- Gansu Agricultural University, Lanzhou, Gansu, China
| | - Chengyuan Jiang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yongming Wang
- Shandong Huahong Biological Engineering Co., LTD, Binzhou, Shandong, China
| | - Yingguang Li
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Yan Li
- Qingdao Animal Disease Prevention and Control Center, Qingdao, Shandong, China
| | - Xiaoxiao Duan
- Qingdao Animal Disease Prevention and Control Center, Qingdao, Shandong, China
| | - Shaoming Dong
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Jiaxu Lin
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| | - Hongliang Zhang
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China.
| | - Hu Shan
- Shandong Collaborative Innovation Center for Development of Veterinary Pharmaceuticals, College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong, China
| |
Collapse
|
12
|
Liu TY, Hu CC, Han CY, Mao SY, Zhang WX, Xu YM, Sun YJ, Jiang DB, Zhang XY, Zhang JX, Wang J, Qiao XP, Pan JY, Yang SY, Yang K. IGF2BP2 promotes colorectal cancer progression by upregulating the expression of TFRC and enhancing iron metabolism. Biol Direct 2023; 18:19. [PMID: 37088822 PMCID: PMC10122817 DOI: 10.1186/s13062-023-00373-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive system, ranking third for morbidity and mortality worldwide. At present, no effective control method is available for this cancer type. In tumor cells, especially iron metabolization, is necessary for its growth and proliferation. High levels of iron are an important feature to maintain tumor growth; however, the overall mechanism remains unclear. METHODS We used western blotting, immunohistochemistry (IHC) and real-time quantitative PCR to analyze the expression of IGF2BP2 in cell lines and tissues. Further, RNA-sequencing, RNA immunoprecipitation and methylated RNA immunoprecipitation experiments explored the specific binding of target genes. Moreover, the RNA stability assay was performed to determine the half-life of genes downstream of IGF2BP2. In addition, the Cell Counting Kit-8, colony formation assay, 5-ethynyl-2'-deoxyuridine assay and flow cytometry were used to evaluate the effects of IGF2BP2 on proliferation and iron metabolism. Lastly, the role of IGF2BP2 in promoting CRC growth was demonstrated in animal models. RESULTS We observed that IGF2BP2 is associated with iron homeostasis and that TFRC is a downstream target of IGF2BP2. Further, overexpression of TFRC can rescue the growth of IGF2BP2-knockdown CRC cells. Mechanistically, we determined that IGF2BP2 regulates TFRC methylation via METTL4, thereby regulating iron metabolism and promoting CRC growth. Furthermore, using animal models, we observed that IGF2BP2 promotes CRC growth. CONCLUSION IGF2BP2 regulates TFRC mRNA methylation via METTL4, thereby regulating iron metabolism and promoting CRC growth. Our study highlights the key roles of IGF2BP2 in CRC carcinogenesis and the iron transport pathways.
Collapse
Affiliation(s)
- Tian-Yue Liu
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chen-Chen Hu
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Chen-Ying Han
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Si-Yi Mao
- School of Basic Medicine, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Wen-Xin Zhang
- School of Basic Medicine, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yi-Ming Xu
- School of Basic Medicine, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yuan-Jie Sun
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Dong-Bo Jiang
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xi-Yang Zhang
- Military Medical Innovation Center, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jia-Xing Zhang
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jing Wang
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xu-Peng Qiao
- College of medical technology, Shaanxi University of traditional Chinese Medicine, Xianyang, Shaanxi, 712046, China
| | - Jing-Yu Pan
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shu-Ya Yang
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Kun Yang
- Department of Immunology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
- Department of Rheumatology and Immunology, Tangdu Hospital of the Air Force Medical University, Xi'an, Shaanxi, 710038, China.
| |
Collapse
|