1
|
Tang J, Huang Z, Yu P. The role of JPX in regulating FUS/SLC7A11 signaling pathway mediated ferroptosis in keloid fibroblasts and its potential in scar repair. Biochem Biophys Res Commun 2025; 763:151770. [PMID: 40233433 DOI: 10.1016/j.bbrc.2025.151770] [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: 03/26/2025] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Keloid scar, a fibrotic disease initiated by aberrant fibroblast proliferation, is influenced by ferroptosis. This investigation aims to elucidate the mechanism of lncRNA JPX regulating ferroptosis in keloid fibroblasts. METHODS AND RESULTS We procured 30 samples of keloid tissue and adjacent normal skin tissues from patients undergoing treatment for keloid scars, subsequently isolating fibroblasts from both the keloid lesions and unaffected portions. JPX expression levels in these lesion and keloid fibroblast samples were detected using qRT-PCR. We then validated the regulatory role of JPX on FUS and SLC7A11 through RNA immunoprecipitation and actinomycin D assays. Subsequently, we overexpressed or silenced JPX and/or SLC7A11 in keloid fibroblasts under conditions with or without ferroptosis inhibitor Fer-1, assessing cell viability, migration, invasion, extracellular matrix (ECM) markers via MTT, Transwell and Western blot assays, and evaluating cellular iron content, reactive oxygen species (ROS), malondialdehyde (MDA), and glutathione (GSH) levels as well as the expression of ferroptosis-related proteins to assess ferroptosis. JPX was up-regulated in keloid tissue and keloid fibroblasts. JPX promoted SLC7A11 stability and expression through FUS. JPX silence suppressed viability, migration, invasion and ECM production, yet facilitated ferroptosis of keloid fibroblasts, while these effects could be reversed by Fer-1 or SLC7A11 overexpression. CONCLUSION JPX regulates ferroptosis within fibroblast derived from scar tissue via the FUS/SLC7A11 pathway, demonstrating its potential utility in facilitating scar repair processes.
Collapse
Affiliation(s)
- Jianhui Tang
- Department of Medical Cosmetology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, People's Republic of China.
| | - Zhaoming Huang
- Department of Medical Cosmetology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, People's Republic of China
| | - Panpan Yu
- Department of Medical Cosmetology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei, 437000, People's Republic of China
| |
Collapse
|
2
|
Wang J, Song Y, Tan X, Wang T, Shi Y, Xu X, Du J, Yu Z, Song B. Targeting PIM1 by Bruceine D attenuates skin fibrosis via myofibroblast ferroptosis. Redox Biol 2025; 82:103619. [PMID: 40168881 PMCID: PMC11993190 DOI: 10.1016/j.redox.2025.103619] [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: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025] Open
Abstract
Skin pan-fibrosis diseases-such as hypertrophic scar (HS), keloid scar (KS), and systemic sclerosis (SSc)-pose significant threats to patients' health and quality of life. In this study, the authors conducted both in vivo and in vitro experiments and discovered that the serine/threonine kinase PIM1 is upregulated in the myofibroblasts of human HS, KS, and SSc tissues, as well as in various animal models of skin fibrosis. Overexpression of PIM1 enhanced the profibrotic phenotypes of human hypertrophic scar fibroblasts (HSFs), which serve as key effector cells in the pathogenesis of skin pan-fibrosis diseases. Through high-throughput screening and subsequent laboratory assays, we identified the small molecule Bruceine D (BD) as a direct binder of PIM1. BD promoted ferroptosis in HSFs by selectively suppressing the PIM1-KEAP1-NRF2 pathway through augmented degradation of PIM1. In various in vivo models-including a hypertrophic scar mouse model, a rabbit ear hypertrophic scar model, and a bleomycin (BLM)-induced skin fibrosis mouse model-BD effectively attenuated fibrotic phenotypes. Collectively, these findings demonstrate that PIM1 serves as a common biomarker and therapeutic target for skin pan-fibrosis diseases. BD mitigates skin fibrosis by activating ferroptosis via PIM1 inhibition, highlighting its great translational potential and high promise to be developed to a clinical drug in treating these conditions, especially those with abnormally elevated PIM1 expression.
Collapse
Affiliation(s)
- Jianzhang Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yajuan Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoying Tan
- Department of Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - Tong Wang
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Shi
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xingbo Xu
- Clinic for Cardiology and Pulmonology, University Medical Center Göttingen, Göttingen, 37075, Germany.
| | - Juan Du
- Department of Dermatology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Zhou Yu
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Baoqiang Song
- Department of Plastic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
Hennion N, Bedart C, Vandomber L, Gottrand F, Humez S, Chenivesse C, Desseyn JL, Gouyer V. Identification of early genes in the pathophysiology of fibrotic interstitial lung disease in a new model of pulmonary fibrosis. Cell Mol Life Sci 2025; 82:115. [PMID: 40074941 PMCID: PMC11904048 DOI: 10.1007/s00018-025-05620-0] [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: 09/19/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025]
Abstract
Some interstitial lung diseases involve pulmonary fibrosis, which is a process that is characterized by the excessive and abnormal accumulation of extracellular matrix in the pulmonary interalveolar space. Although the current anti-fibrotic therapy aims at slowing down the progression of pulmonary fibrosis, it does not reverse it, and many of the drugs that were identified in basic-research studies failed in clinical phases, mainly because of the lack of a model that can recapitulate the pathophysiological mechanisms of human pulmonary fibrosis. We developed a novel experimental model of pulmonary fibrosis induced by a cocktail of molecules on an air/liquid interface culture of mouse embryonic lung explants. Histological analyses revealed a pattern of usual interstitial pneumonia, the worst-prognosis form of pulmonary fibrosis. We performed a transcriptomics analysis at the single-cell level after the induction of fibrosis and before any histological signs of fibrosis could be observed. The results revealed increased expression of several gene families that are involved in early inflammation, fibrosis and iron homeostasis, as well as potential new genetic targets.
Collapse
Affiliation(s)
- Nathan Hennion
- Univ. Lille, Inserm, CHU Lille, Infinite U1286, Lille, F-59000, France
| | - Corentin Bedart
- Univ. Lille, Inserm, CHU Lille, Infinite U1286, Lille, F-59000, France
| | - Léonie Vandomber
- Univ. Lille, Inserm, CHU Lille, Infinite U1286, Lille, F-59000, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, Infinite U1286, Lille, F-59000, France
| | - Sarah Humez
- Univ. Lille, Department of Pathology, CHU Lille, Lille, F-59000, France
- Univ. Lille, CHU Lille, CNRS, Inserm, Institut Pasteur de Lille, UMR9020, UMR1277, Canther, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, F-59000, France
| | - Cécile Chenivesse
- Univ. Lille, CNRS, Inserm, CHU Lille, Centre de référence constitutif des maladies pulmonaires rares, U1019, UMR 9017, CIIL, Center for Infection and Immunity of Lille, Lille, F- 59000, France
| | - Jean-Luc Desseyn
- Univ. Lille, Inserm, CHU Lille, Infinite U1286, Lille, F-59000, France.
| | - Valérie Gouyer
- Univ. Lille, Inserm, CHU Lille, Infinite U1286, Lille, F-59000, France
| |
Collapse
|
4
|
Yan S, Zhao Y, Xu W, Zhang J, Zhang Y, Liu B, Li X, Ma Z, Yang Q. ADAM17/PTGS2 Facilitates Pulmonary Fibrosis by Regulating Ferroptosis. J Cell Mol Med 2025; 29:e70466. [PMID: 40077919 PMCID: PMC11903495 DOI: 10.1111/jcmm.70466] [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: 09/19/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterised by excessive deposition of extracellular matrix (ECM), resulting in high mortality rates. In this study, we provide evidence that ADAM17/PTGS2 plays a crucial role in inducing ferroptosis in fibroblasts, promoting PF. Initially, an assessment was made of ADAM17 protein levels in patients diagnosed with connective tissue diseases-interstitial lung diseases (CTD-ILD), using ELISA assays. Confirmation of the relationship between ADAM17 and fibrosis was achieved by stimulating cells with PMA or TAPI-1 (the ADAM17 inhibitor), in conjunction with the fibrosis-inducing factor, TGFβ1. To further explore the major downstream proteins of ADAM17 contributing to altered PF, we employed mRNA transcriptomics. To further investigate the role of ADAM17/PTGS2 in promoting ferroptosis and fibrosis, we employed western blot assays, immunofluorescence and transmission electron microscopy (TEM). Furthermore, the effects of the ADAM17/PTGS2/ferroptosis pathway in PF were verified using Adeno-associated virus (AAV)-mediated ADAM17 gene knockdown in mice. In CTD-ILD patients, ADAM17 expression was significantly elevated. Upon PMA stimulation, lung fibroblasts exhibited increased fibrosis-related proteins, and the combined stimulation of PMA and TGFβ1 synergistically promoted cellular fibrosis. Conversely, TAPI-1 alleviated fibrotic stimulation induced by TGFβ1. Transcriptomic analysis of lung fibroblast specimens overexpressing ADAM17 revealed significantly elevated PTGS2 expression levels. Knockdown and ferroptosis inhibition assays demonstrated that ADAM17 regulates ferroptosis in lung fibroblasts via PTGS2, ultimately inducing fibrosis. Furthermore, the deficiency of ADAM17 alleviated bleomycin-induced PF and inflammation in mice. These findings first verified that ADAM17/PTGS2/ferroptosis is a novel mechanism for regulating PF; it provides a new theoretical basis for further exploring the treatment of PF.
Collapse
Affiliation(s)
- Suyan Yan
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yaqi Zhao
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Xu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jin Zhang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ying Zhang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Baocheng Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xinya Li
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
5
|
Pei Z, Fan J, Tang M, Li Y. Ferroptosis: A New Strategy for the Treatment of Fibrotic Diseases. Adv Biol (Weinh) 2025; 9:e2400383. [PMID: 39377183 DOI: 10.1002/adbi.202400383] [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: 07/02/2024] [Revised: 09/07/2024] [Indexed: 10/09/2024]
Abstract
Ferroptosis is a new type of cell death characterized by iron dependence and the excessive accumulation of lipid reactive oxygen species (lipid ROS) that has gradually become better characterized. There is sufficient evidence indicating that ferroptosis is associated with a variety of human life activities and diseases, such as tumor suppression, ischemic organ injury, and degenerative disorders. Notably, ferroptosis is also involved in the initiation and development of fibrosis in various organs, including liver fibrosis, pulmonary fibrosis, renal fibrosis, and cardiac fibrosis, which is usually irreversible and refractory. Although a large number of patients with fibrosis urgently need to be treated, the current treatment options are still limited and unsatisfactory. Organ fibrosis involves a series of complex and orderly processes, such as parenchymal cell damage, recruitment of inflammatory cells and activation of fibroblasts, which ultimately leads to the accumulation of extracellular matrix (ECM) and the formation of fibrosis. An increasing number of studies have confirmed the close association between these pathological processes and ferroptosis. This review summarizes the role and function of ferroptosis in fibrosis and proposes several potential therapeutic strategies and pathways based on ferroptosis.
Collapse
Affiliation(s)
- Zhuo Pei
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang, 110044, China
| | - Maolin Tang
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Yuhong Li
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
6
|
Zha T, Zhang Z, Pan L, Peng L, Du Y, Wu P, Chen J, Xing W. Evaluating the Potential of Quantitative Susceptibility Mapping for Detecting Iron Deposition of Renal Fibrosis in a Rabbit Model. J Magn Reson Imaging 2025. [PMID: 39874142 DOI: 10.1002/jmri.29722] [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: 10/17/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND As ferroptosis is a key factor in renal fibrosis (RF), iron deposition monitoring may help evaluating RF. The capability of quantitative susceptibility mapping (QSM) for detecting iron deposition in RF remains uncertain. PURPOSE To investigate the potential of QSM to detect iron deposition in RF. STUDY TYPE Animal model. ANIMAL MODEL Eighty New Zealand rabbits were randomly divided into control (N = 10) and RF (N = 70) groups, consisting of baseline, 7, 14, 21, and 28 days (N = 12 in each), and longitudinal (N = 10) subgroups. RF was induced via unilateral renal arteria stenosis. FIELD STRENGTH/SEQUENCE 3 T, QSM with gradient echo, arterial spin labeling with gradient spin echo. ASSESSMENT Bilateral kidney QSM values (χ) in the cortex (χCO) and outer medulla (χOM) were evaluated with histopathology. STATISTICAL TESTS Analysis of variance, Kruskal-Wallis, Spearman's correlation, and the area under the receiver operating characteristic curve (AUC). P < 0.05 was significant. RESULTS In fibrotic kidneys, χCO decreased at 7 days ([-6.69 ± 0.98] × 10-2 ppm) and increased during 14-28 days ([-1.85 ± 2.11], [0.14 ± 0.58], and [1.99 ± 0.60] × 10-2 ppm, respectively), while the χOM had the opposite trend. Both significantly correlated with histopathology (|r| = 0.674-0.849). AUC of QSM for distinguishing RF degrees was 0.692-0.993. In contralateral kidneys, the χCO initially decreased ([-6.67 ± 0.84] × 10-2 ppm) then recovered to baseline ([-4.81 ± 0.89] × 10-2 ppm), while the χOM at 7-28 days ([2.58 ± 1.40], [2.25 ± 1.83], [2.49 ± 2.11], [2.43 ± 1.32] × 10-2 ppm, respectively) were significantly higher than baseline ([0.54 ± 0.18] × 10-2 ppm). DATA CONCLUSION Different iron deposition patterns were observed in RF with QSM values, suggesting the potential of QSM for iron deposition monitoring in RF. PLAIN LANGUAGE SUMMARY Renal fibrosis (RF) is a common outcome in most kidney diseases, leading to scarring and loss of kidney function. Increasing evidence suggests that abnormal iron metabolism plays an important role in RF. This study used a technique called quantitative susceptibility mapping (QSM) to measure kidney iron levels in rabbits with RF. Specifically, rabbits with advanced RF exhibited higher kidney iron concentrations, and moderate to strong correlations between QSM values and histopathology demonstrated that QSM could accurately detect changes in iron levels and assess RF severity. Overall, QSM shows promise as a tool for monitoring iron deposition in RF progression. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Tingting Zha
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Province Artificial Intelligence for Medical Images Engineering Research Center, Changzhou, Jiangsu, China
| | - Zhiping Zhang
- Department of Radiology, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Liang Pan
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Province Artificial Intelligence for Medical Images Engineering Research Center, Changzhou, Jiangsu, China
| | - Lei Peng
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Yanan Du
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Province Artificial Intelligence for Medical Images Engineering Research Center, Changzhou, Jiangsu, China
| | - Peng Wu
- Philips Healthcare, Shanghai, China
| | - Jie Chen
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Province Artificial Intelligence for Medical Images Engineering Research Center, Changzhou, Jiangsu, China
| | - Wei Xing
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Province Artificial Intelligence for Medical Images Engineering Research Center, Changzhou, Jiangsu, China
| |
Collapse
|
7
|
Khazaei M, Ardeshir RA. Protective effects of sulfated polysaccharides from Enteromorpha intestinalis on oxidative stress, liver iron overload and Ferroptosis in Zebra fish exposed to ethanol. Biomed Pharmacother 2024; 181:117715. [PMID: 39615168 DOI: 10.1016/j.biopha.2024.117715] [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: 08/29/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
The study investigates the protective effects of sulfated polysaccharides extracted from Enteromorpha intestinalis (EIP) against oxidative stress, liver iron overload, and ferroptosis in zebrafish exposed to ethanol, a model for alcohol-related liver disease (ALD). The extracted polysaccharides were characterized for sulfate and sugar content, molecular weight, and functional groups. Adult male zebrafish were divided into three groups: control, ethanol-exposed (EE) (0.2 % ethanol (v/v) in the water), and ethanol-exposed with EIP supplementation (1 % EIP incorporated into the basal diet) (EE+EIP) for 30 days. The study measured liver oxidative stress indexes, serum enzymological indexes, liver and serum lipid profiles, liver iron ion content, and expression of ferroptosis-related genes. Histological analysis was conducted to assess lipid accumulation and iron deposition in liver tissues. The findings indicate that EIP supplementation significantly mitigates ethanol-induced liver damage. Specifically, EIP reduced malondialdehyde levels, increased antioxidant enzyme and non-enzymatic antioxidant activity, and decreased iron ion accumulation and the area of iron granules in the liver tissue. Additionally, EIP treatment lowered lipids levels and aminotransferase enzyme activity in the serum. In the ALD model, EIP inhibited ethanol-induced ferroptosis by modulating the expression of key genes: it decreased the expression of transferrin (tf), transferrin receptor (tfr), ferroportin (fpn), and ferritin heavy chain (fth), while increasing the expression of glutathione peroxidase 4 (gpx4) and solute carrier family 7 member 11 (slc7a11). EIP has protective effects against ethanol-induced liver injury in zebrafish, offering a foundation for further research into its hepatoprotective action and potential application in preventing and treating ALD.
Collapse
Affiliation(s)
- Marziyeh Khazaei
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Rashid Alijani Ardeshir
- Marine Biotechnology Department, College of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
8
|
Zhao G, Hu Y. Mechanistic insights into intrauterine adhesions. Semin Immunopathol 2024; 47:3. [PMID: 39613882 DOI: 10.1007/s00281-024-01030-9] [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: 09/16/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
Intrauterine adhesions (IUA), also known as Asherman's syndrome, arise from damage to the basal layer of the endometrium, frequently caused by intrauterine interventions. This damage leads to nonregenerative healing of endometrium resulting in replacement by fibrous connective tissue, which bring about the adherence of opposing endometrium to render the uterine cavity and/or cervical canal partially or completely obliterated. IUA is a common cause of the refractory uterine infertility. Hysteroscopy is the gold standard for diagnosis of IUA. However, the method of accurately predicting the likelihood of achieving a live birth in the future remains established. Classical treatments have shown limited success, particularly in severe cases. Therefore, utilizing new research methods to deepen the understanding of the pathogenesis of IUA will facilitate the new treatment approaches to be found. In this article we briefly described the advances in the pathogenesis of IUA, with focus on inflammation and parenchymal cellular homeostasis disruption, defects in autophagy and the role of ferroptosis, and we also outlined the progress in IUA therapy.
Collapse
Affiliation(s)
- Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
9
|
Chen J, He Z, Xu W, Kang Y, Zhu F, Tang H, Wang J, Zhong F. Human umbilical cord mesenchymal stem cells restore chemotherapy-induced premature ovarian failure by inhibiting ferroptosis in vitro ovarian culture system. Reprod Biol Endocrinol 2024; 22:137. [PMID: 39511578 PMCID: PMC11542367 DOI: 10.1186/s12958-024-01310-x] [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: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown potential in repairing chemotherapy-induced premature ovarian failure (POF). However, challenges such as stem cell loss and immune phagocytosis post-transplantation hinder their application. Due to easy and safe handling, in vitro ovarian culture is widely available for drug screening, pathophysiological research, and in vitro fertilization. MSCs could exhibit therapeutic capacity for ovarian injury, and avoid stem cell loss and immune phagocytosis in vitro tissue culture system. Therefore, this study utilizes an in vitro ovarian culture system to investigate the reparative potential of human umbilical cord mesenchymal stem cells (hUCMSCs) and their mechanism. METHODS In this study, a chemotherapy-induced POF model was established by introducing cisplatin in vitro ovarian culture system. The reparative effects of hUCMSCs on damaged ovarian tissue were validated through Transwell chambers. Tissue histology examination, immunohistochemical staining, Western blotting, and RT-PCR were employed to evaluate the expression effects of hUCMSCs on ferroptosis and fibrosis-related genes during the process of repairing cisplatin-induced POF. RESULTS Cisplatin was found to activate ovarian follicles in vitro POF model. Transcriptomic sequencing analysis revealed that cisplatin could activate genes associated with ferroptosis. hUCMSCs alleviated cisplatin-induced POF by suppressing the expression of ferroptosis. Moreover, inhibiting ferroptosis by hUCMSCs also ameliorated ovarian hormone levels and reduced the expression of fibrosis-related factors α-SMA and COL-I in the ovaries. CONCLUSIONS This study confirms that cisplatin-induced ovarian damage via ferroptosis in vitro POF model, and hUCMSCs repair ovarian injury by inhibiting the ferroptosis pathway and suppressing fibrosis. This research contributes to evaluating the effectiveness of hUCMSCs in treating chemotherapy-induced POF by inhibiting ferroptosis in an in vitro ovarian culture system and provides a potential therapeutic strategy for chemotherapy-induced POF.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Zhuoying He
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Wenjuan Xu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China
| | - Yumiao Kang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Fengyu Zhu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China
| | - Heng Tang
- Wanbei Coal Electric Group General Hospital, Suzhou, Anhui Province, 234011, China.
| | - Jianye Wang
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China.
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China.
| |
Collapse
|
10
|
Zhang WY, Wen L, Du L, Liu TT, Sun Y, Chen YZ, Lu YX, Cheng XC, Sun HY, Xiao FJ, Wang LS. S-RBD-modified and miR-486-5p-engineered exosomes derived from mesenchymal stem cells suppress ferroptosis and alleviate radiation-induced lung injury and long-term pulmonary fibrosis. J Nanobiotechnology 2024; 22:662. [PMID: 39462403 PMCID: PMC11515248 DOI: 10.1186/s12951-024-02830-9] [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: 05/09/2024] [Accepted: 09/02/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Radiation-induced lung injury (RILI) is associated with alveolar epithelial cell death and secondary fibrosis in injured lung. Mesenchymal stem cell (MSC)-derived exosomes have regenerative effect against lung injury and the potential to intervene of RILI. However, their intervention efficacy is limited because they lack lung targeting characters and do not carry sufficient specific effectors. SARS-CoV-2 spike glycoprotein (SARS-CoV-2-S-RBD) binds angiotensin-converting enzyme 2 (ACE2) receptor and mediates interaction with host cells. MiR-486-5p is a multifunctional miRNA with angiogenic and antifibrotic potential and acts as an effector in MSC-derived exosomes. Ferroptosis is a form of cell death associated with radiation injury, its roles and mechanisms in RILI remain unclear. In this study, we developed an engineered MSC-derived exosomes with SARS-CoV-2-S-RBD- and miR-486-5p- modification and investigated their intervention effects on RIPF and action mechanisms via suppression of epithelial cell ferroptosis. RESULTS Adenovirus-mediated gene modification led to miR-486-5p overexpression in human umbilical cord MSC exosomes (p < 0.05), thereby constructing miR-486-5p engineered MSC exosomes (miR-486-MSC-Exo). MiR-486-MSC-Exo promoted the proliferation and migration of irradiated mouse lung epithelial (MLE-12) cells in vitro and inhibited RILI in vivo (all p < 0.05). MiR-486-MSC-Exo suppressed ferroptosis in MLE-12 cells, and an in vitro assay revealed that the expression of fibrosis-related genes is up-regulated following ferroptosis (both p < 0.05). MiR-486-MSC-Exo reversed the up-regulated expression of fibrosis-related genes induced by TGF-β1 in vitro and improved pathological fibrosis in RIPF mice in vivo (all p < 0.05). SARS-CoV-2-S-RBD-modified and miR-486-5p-engineered MSC exosomes (miR-486-RBD-MSC-Exo) were also constructed, and the distribution of DiR dye-labeled miR-486-RBD-MSC-Exo in hACE2CKI/CKI Sftpc-Cre+ mice demonstrated long-term retention in the lung (p < 0.05). MiR-486-RBD-MSC-Exo significantly improved the survival rate and pathological changes in hACE2CKI/CKI Sftpc-Cre+ RIPF mice (all p < 0.05). Furthermore, miR-486-MSC-Exo exerted anti-fibrotic effects via targeted SMAD2 inhibition and Akt phosphorylation activation (p < 0.05). CONCLUSIONS Engineered MSC exosomes with SARS-CoV-2-S-RBD- and miR-486-5p-modification were developed. MiR-486-RBD-MSC-Exo suppressed ferroptosis and fibrosis of MLE-12 cells in vitro, and alleviated RILI and long-term RIPF in ACE2 humanized mice in vivo. MiR-486-MSC-Exo exerted anti-fibrotic effects via SMAD2 inhibition and Akt activation. This study provides a potential approach for RIPF intervention.
Collapse
Affiliation(s)
- Wei-Yuan Zhang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Li Wen
- School of Nursing, Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Li Du
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Ting Ting Liu
- Department of Pulmonary and Critical Care Medicine, The Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Sun
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Yi-Zhu Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Yu-Xin Lu
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Xiao-Chen Cheng
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Hui-Yan Sun
- Yanda Medical Research Institute, Hebei Yanda Hospital, Langfang, 065201, China
| | - Feng-Jun Xiao
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Li-Sheng Wang
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266071, People's Republic of China.
- Laboratory of Molecular Diagnosis and Regenerative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
- School of Nursing, Jilin University, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
11
|
Wang M, Mo D, Zhang N, Yu H. Ferroptosis in diabetic cardiomyopathy: Advances in cardiac fibroblast-cardiomyocyte interactions. Heliyon 2024; 10:e35219. [PMID: 39165946 PMCID: PMC11334834 DOI: 10.1016/j.heliyon.2024.e35219] [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: 02/07/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common complication of diabetes, and its pathogenesis remains elusive. Ferroptosis, a process dependent on iron-mediated cell death, plays a crucial role in DCM via disrupted iron metabolism, lipid peroxidation, and weakened antioxidant defenses. Hyperglycemia, oxidative stress, and inflammation may exacerbate ferroptosis in diabetes. This review emphasizes the interaction between cardiac fibroblasts and cardiomyocytes in DCM, influencing ferroptosis occurrence. By exploring ferroptosis modulation for potential therapeutic targets, this article offers a fresh perspective on DCM treatment. The study systematically covers the interplay, mechanisms, and targeted drugs linked to ferroptosis in DCM development.
Collapse
Affiliation(s)
| | | | - Ning Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| |
Collapse
|
12
|
Viganò P, Caprara F, Giola F, Di Stefano G, Somigliana E, Vercellini P. Is retrograde menstruation a universal, recurrent, physiological phenomenon? A systematic review of the evidence in humans and non-human primates. Hum Reprod Open 2024; 2024:hoae045. [PMID: 39055487 PMCID: PMC11272177 DOI: 10.1093/hropen/hoae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
STUDY QUESTION What are the quantitative, qualitative, and temporal patterns of retrograde mentruation? SUMMARY ANSWER The extreme quantitative and qualitative heterogeneity of the available studies prevents the definitive conclusion that retrograde menstruation is a universal and consistent phenomenon during the reproductive period. WHAT IS KNOWN ALREADY Retrograde menstruation has been defined as a universal, physiological phenomenon that occurs similarly in about 90% of menstruators during the reproductive period. However, uncertainties still exist in terms of the event frequency, total amount, and cellular composition of retrograde menstruation and the differences between individuals with versus those without endometriosis. STUDY DESIGN SIZE DURATION Two systematic reviews were performed, one for human studies, and one for non-human primate studies. We retrieved studies from the PubMed and Embase databases published between 1 January 1980 and 1 November 2023. Studies published in the English language were included and identified using a combination of MeSH terms. References from relevant publications were systematically screened and further articles were identified using PubMed's 'similar articles' and 'cited by' functions. PARTICIPANTS/MATERIALS SETTING METHODS Results were reported in accordance with the PRISMA guidelines. Studies that did not report original data or provided a review of the field were excluded. Bias analysis was completed for each included human study by using the Newcastle-Ottawa scoring system. MAIN RESULTS AND THE ROLE OF CHANCE Fifteen studies were finally included in the human systematic review, mostly with limited sample sizes. The macroscopic visualization of blood in PF during menses was reported with a frequency ranging from 9% to 100%. A prevalence of endometrial cells detected in peritoneal fluid ranging from 8% to 75% was reported in the various studies. Controversial findings were reported in relation to patients with endometriosis. Retrograde menstruation has been evaluated cross-sectionally on single occasions, and no information is available on the course of the phenomenon within an entire cycle and between subsequent cycles. Two studies were included in the non-human primate systematic review; one of them showed that retrograde menstruation was observed more frequently in baboons with naturally occurring endometriosis (83%) than in those with a normal pelvis (51%). LIMITATIONS REASONS FOR CAUTION In humans, peritoneal fluid has often been collected at different cycle phases and not systematically during menstruation. The indication for laparoscopy was not always clear for all participants. A wide variety of methods were used to detect endometrial cells, including cytological staining, cell block analysis, immunocytochemistry, and various methods of cell culture. WIDER IMPLICATION OF THE FINDINGS The idea that almost all women experience retrograde menstruation regularly and similarly during their reproductive life is currently unsubstantiated. It is an academic notion accepted uncritically. Development of endometriosis may derive from differences in the frequency or severity of the event. STUDY FUNDING/COMPETING INTERESTS The review was partially funded by Italian Ministry of Health-Current Research IRCCS. P.Vi. serves as co-editor in Chief of Journal of Endometriosis and Uterine Disorders. E.S. serves as Editor in Chief of Human Reproduction Open and discloses research grants from Ferring, Ibsa, Gedeon Richter, and Theramex, and honoraria from Ibsa and Gedeon Richter. P.Ve. serves as Associate Editor for Human Reproduction Open; is a member of the Editorial Board of the Journal of Obstetrics and Gynaecology Canada, of the Italian Journal of Obstetrics and Gynaecology, and of the International Editorial Board of Acta Obstetricia et Gynecologica Scandinavica; has received royalties from Wolters Kluwer for chapters on endometriosis management in the clinical decision support resource UpToDate; and maintains both a public and private gynecological practice. All other authors declare they have no conflict of interest. REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Paola Viganò
- Infertility Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Caprara
- Department of Clinical Sciences and Community Health, Academic Center for Research on Adenomyosis and Endometriosis, Università degli Studi di Milano, Milan, Italy
| | - Francesca Giola
- Department of Clinical Sciences and Community Health, Academic Center for Research on Adenomyosis and Endometriosis, Università degli Studi di Milano, Milan, Italy
| | - Giorgia Di Stefano
- Infertility Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Edgardo Somigliana
- Infertility Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Academic Center for Research on Adenomyosis and Endometriosis, Università degli Studi di Milano, Milan, Italy
| | - Paolo Vercellini
- Infertility Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Academic Center for Research on Adenomyosis and Endometriosis, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
13
|
Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov 2024; 10:301. [PMID: 38914560 PMCID: PMC11196712 DOI: 10.1038/s41420-024-02078-0] [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: 03/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic interstitial lung disorder characterized by abnormal myofibroblast activation, accumulation of extracellular matrix (ECM), and thickening of fibrotic alveolar walls, resulting in deteriorated lung function. PF is initiated by dysregulated wound healing processes triggered by factors such as excessive inflammation, oxidative stress, and coronavirus disease (COVID-19). Despite advancements in understanding the disease's pathogenesis, effective preventive and therapeutic interventions are currently lacking. Ferroptosis, an iron-dependent regulated cell death (RCD) mechanism involving lipid peroxidation and glutathione (GSH) depletion, exhibits unique features distinct from other RCD forms (e.g., apoptosis, necrosis, and pyroptosis). Imbalance between reactive oxygen species (ROS) production and detoxification leads to ferroptosis, causing cellular dysfunction through lipid peroxidation, protein modifications, and DNA damage. Emerging evidence points to the crucial role of ferroptosis in PF progression, driving macrophage polarization, fibroblast proliferation, and ECM deposition, ultimately contributing to alveolar cell death and lung tissue scarring. This review provides a comprehensive overview of the latest findings on the involvement and signaling mechanisms of ferroptosis in PF pathogenesis, emphasizing potential novel anti-fibrotic therapeutic approaches targeting ferroptosis for PF management.
Collapse
Affiliation(s)
- Yixiang Hu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China
| | - Ying Huang
- Zhongshan Hospital of Traditional Chinese Medicine Afflilated to Guangzhou University of Chinese Medicine, Zhongshan, 528400, China
| | - Lijuan Zong
- Department of Rehabilitation Medicine, Zhongda Hospital of Southeast University, Nanjing, 210096, China
| | - Jiaxin Lin
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Xiang Liu
- Department of Clinical Pharmacy, The Affiliated Xiangtan Center Hospital of Hunan University, Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
14
|
Zaher A, Duchman B, Ivanovic M, Spitz DR, Furqan M, Allen BG, Petronek MS. Exploratory Analysis of Image-Guided Ionizing Radiation Delivery to Induce Long-Term Iron Accumulation and Ferritin Expression in a Lung Injury Model: Preliminary Results. Bioengineering (Basel) 2024; 11:182. [PMID: 38391668 PMCID: PMC10886280 DOI: 10.3390/bioengineering11020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/03/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Radiation therapy (RT) is an integral and commonly used therapeutic modality for primary lung cancer. However, radiation-induced lung injury (RILI) limits the irradiation dose used in the lung and is a significant source of morbidity. Disruptions in iron metabolism have been linked to radiation injury, but the underlying mechanisms remain unclear. PURPOSE To utilize a targeted radiation delivery approach to induce RILI for the development of a model system to study the role of radiation-induced iron accumulation in RILI. METHODS This study utilizes a Small Animal Radiation Research Platform (SARRP) to target the right lung with a 20 Gy dose while minimizing the dose delivered to the left lung and adjacent heart. Long-term pulmonary function was performed using RespiRate-x64image analysis. Normal-appearing lung volumes were calculated using a cone beam CT (CBCT) image thresholding approach in 3D Slicer software. Quantification of iron accumulation was performed spectrophotometrically using a ferrozine-based assay as well as histologically using Prussian blue and via Western blotting for ferritin heavy chain expression. RESULTS Mild fibrosis was seen histologically in the irradiated lung using hematoxylin and eosin-stained fixed tissue at 9 months, as well as using a scoring system from CBCT images, the Szapiel scoring system, and the highest fibrotic area metric. In contrast, no changes in breathing rate were observed, and median survival was not achieved up to 36 weeks following irradiation, consistent with mild lung fibrosis when only one lung was targeted. Our study provided preliminary evidence on increased iron content and ferritin heavy chain expression in the irradiated lung, thus warranting further investigation. CONCLUSIONS A targeted lung irradiation model may be a useful approach for studying the long-term pathological effects associated with iron accumulation and RILI following ionizing radiation.
Collapse
Affiliation(s)
- Amira Zaher
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Bryce Duchman
- Division of Pulmonary, Critical Care, Sleep Medicine & Physiology, UC San Diego Health, San Diego, CA 92093, USA
| | - Marina Ivanovic
- Department of Pathology and Laboratory Medicine, Loyola University Health System, Loyola University, Chicago, IL 60660, USA
| | - Douglas R Spitz
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Department of Internal Medicine Division of Hematology and Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Bryan G Allen
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Michael S Petronek
- Department of Radiation Oncology, Division of Free Radical and Radiation Biology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
15
|
Gensluckner S, Wernly B, Datz C, Aigner E. Iron, Oxidative Stress, and Metabolic Dysfunction-Associated Steatotic Liver Disease. Antioxidants (Basel) 2024; 13:208. [PMID: 38397806 PMCID: PMC10886327 DOI: 10.3390/antiox13020208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Excess free iron is a substrate for the formation of reactive oxygen species (ROS), thereby augmenting oxidative stress. Oxidative stress is a well-established cause of organ damage in the liver, the main site of iron storage. Ferroptosis, an iron-dependent mechanism of regulated cell death, has recently been gaining attention in the development of organ damage and the progression of liver disease. We therefore summarize the main mechanisms of iron metabolism, its close connection to oxidative stress and ferroptosis, and its particular relevance to disease mechanisms in metabolic-dysfunction-associated fatty liver disease and potential targets for therapy from a clinical perspective.
Collapse
Affiliation(s)
- Sophie Gensluckner
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
- Obesity Research Unit, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Bernhard Wernly
- Department of Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5110 Oberndorf, Austria; (B.W.); (C.D.)
| | - Christian Datz
- Department of Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5110 Oberndorf, Austria; (B.W.); (C.D.)
| | - Elmar Aigner
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
- Obesity Research Unit, Paracelsus Medical University, 5020 Salzburg, Austria
| |
Collapse
|
16
|
Liang Y, Qiu S, Zou Y, Luo L. Targeting ferroptosis with natural products in liver injury: new insights from molecular mechanisms to targeted therapies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155134. [PMID: 37863001 DOI: 10.1016/j.phymed.2023.155134] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Ferroptosis is a brand-new type of controlled cell death that is distinguished by its reliance on iron and the production of lipid peroxidation. The role of ferroptosis in damaging liver disorders has attracted a lot of attention in recent years. One effective strategy to reduce liver damage is to target ferroptosis. PURPOSE The purpose of this review is to clarify the connection between ferroptosis and liver damage and to look into the potential contribution of natural products to the clinical management of liver damage and the discovery of novel medications. METHODS To study the methods by which natural products operate on ferroptosis to cure liver damage and their main signaling pathways, we searched databases from the time of initial publication to August 2023 in PubMed, EMBASE, Web of Science, Ovid, ScienceDirect, and China National Knowledge Infrastructure. The liver illness that each natural product treats is categorized and summarized. It's interesting to note that several natural compounds, such Artemether, Fucoidan sulfate, Curcumin, etc., have the benefit of having many targets and multiple pathways of action. RESULTS We saw that in human samples or animal models of liver injury, ferroptosis indicators were activated, lipid peroxidation levels were elevated, and iron inhibitors had the ability to reduce liver damage. Liver damage can be treated with natural products by regulating ferroptosis. This is mostly accomplished through the modulation of Nrf2-related pathways (e.g., Conclusions and Astaxanthin), biological enzymes like GPX4 and the SIRT family (e.g., Chrysophanol and Decursin), and transcription factors like P53 (e.g., Artemether and Zeaxanthin). CONCLUSIONS This review proposes a promising path for the therapeutic therapy of liver damage by providing a theoretical foundation for the management of ferroptosis utilizing natural ingredients.
Collapse
Affiliation(s)
- Yongyi Liang
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Shaojun Qiu
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Youwen Zou
- The First Clinical College, Guangdong Medical University, Zhanjiang, 524023, Guangdong, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, Guangdong, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, 524023, Guangdong, China.
| |
Collapse
|
17
|
Wang Y, Hu J, Wu S, Fleishman JS, Li Y, Xu Y, Zou W, Wang J, Feng Y, Chen J, Wang H. Targeting epigenetic and posttranslational modifications regulating ferroptosis for the treatment of diseases. Signal Transduct Target Ther 2023; 8:449. [PMID: 38072908 PMCID: PMC10711040 DOI: 10.1038/s41392-023-01720-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/16/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jing Hu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300060, PR China
| | - Shuang Wu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, 430000, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yulin Li
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Yinshi Xu
- Department of Outpatient, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Wailong Zou
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yukuan Feng
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing, 100049, PR China.
| | - Hongquan Wang
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, PR China.
| |
Collapse
|
18
|
Wang C, Hua S, Song L. Ferroptosis in pulmonary fibrosis: an emerging therapeutic target. Front Physiol 2023; 14:1205771. [PMID: 37664432 PMCID: PMC10470006 DOI: 10.3389/fphys.2023.1205771] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/28/2023] [Indexed: 09/05/2023] Open
Abstract
In recent years, the role of ferroptosis in pulmonary fibrosis has garnered increasing interest as a potential therapeutic target. Pulmonary fibrosis is a pathological process characterized by the accumulation of extracellular matrix in affected lung tissues, and currently, there are no effective therapies for preventing or reversing the fibrotic lesions. Ferroptosis is a form of programmed cell death that is regulated by a network of enzymes and signaling pathways. Dysregulation of ferroptosis has been implicated in several diseases, including pulmonary fibrosis. The accumulation of lipid peroxides in the course of ferroptosis causes damage to cell membranes and other cellular components, leading ultimately to cell death. Relevant targets for therapeutic intervention in ferroptosis include key enzymes, such as glutathione peroxidase 4, transcription factors like nuclear factor erythroid 2-related factor 2, and iron chelation. This review provides an overview of the emerging role of ferroptosis in pulmonary fibrosis and highlights potential therapeutic targets in this pathway. Further research is needed to develop safe and effective approaches targeting ferroptosis in treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Chunyan Wang
- Department of General Practice, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Shucheng Hua
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Lei Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|