1
|
Li B, Zhang B, Cheng Z, Lou Y, Chen S. Nanomaterials targeting iron homeostasis: a promising strategy for cancer treatment. Front Bioeng Biotechnol 2025; 13:1511197. [PMID: 40144390 PMCID: PMC11937013 DOI: 10.3389/fbioe.2025.1511197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Iron is essential for vital cellular processes, including DNA synthesis, repair, and proliferation, necessitating enhanced iron uptake and intracellular accumulation. Tumor cells, in particular, exhibit a pronounced elevation in iron uptake to sustain their continuous proliferation, migration and invasion. This elevated iron acquisition is facilitated predominantly through the upregulation of transferrin receptors, which are closely associated with tumorigenesis and tumor progression. Incorporating transferrin into drug delivery systems has been shown to enhance cytotoxic effects in drug-sensitive cancer cells, offering a potential method to surpass the limitations of current cancer therapies. Intracellular iron predominantly exists as ferritin heavy chain (FTH), ferritin light chain (FTL), and labile iron pool (LIP). The innovation of nanocarriers incorporating iron chelating agents has attracted considerable interest. Iron chelators such as Deferoxamine (DFO), Deferasirox (DFX), and Dp44mT have demonstrated significant promise in cancer treatment by inducing iron deficiency within tumor cells. This review explores recent advancements in nanotechnology aimed at targeting iron metabolism in cancer cells and discusses their potential applications in cancer treatment strategies.
Collapse
Affiliation(s)
- Bin Li
- Institute of Urology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Bing Zhang
- Department of Urology, Rushan Hospital of Traditional Chinese Medicine, Weihai, China
| | - Ziyue Cheng
- Institute of Urology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yantao Lou
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Shuqiu Chen
- Institute of Urology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
2
|
Chen P, Li B, Lu Z, Xu Q, Zheng H, Jiang S, Jiang L, Zheng X. PCBP2 as an intrinsic agi ng factor regulates the senescence of hBMSCs through the ROS-FGF2 signaling axis. eLife 2025; 13:RP92419. [PMID: 40053388 PMCID: PMC11888601 DOI: 10.7554/elife.92419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
Background It has been reported that loss of PCBP2 led to increased reactive oxygen species (ROS) production and accelerated cell aging. Knockdown of PCBP2 in HCT116 cells leads to significant downregulation of fibroblast growth factor 2 (FGF2). Here, we tried to elucidate the intrinsic factors and potential mechanisms of bone marrow mesenchymal stromal cells (BMSCs) aging from the interactions among PCBP2, ROS, and FGF2. Methods Unlabeled quantitative proteomics were performed to show differentially expressed proteins in the replicative senescent human bone marrow mesenchymal stromal cells (RS-hBMSCs). ROS and FGF2 were detected in the loss-and-gain cell function experiments of PCBP2. The functional recovery experiments were performed to verify whether PCBP2 regulates cell function through ROS/FGF2-dependent ways. Results PCBP2 expression was significantly lower in P10-hBMSCs. Knocking down the expression of PCBP2 inhibited the proliferation while accentuated the apoptosis and cell arrest of RS-hBMSCs. PCBP2 silence could increase the production of ROS. On the contrary, overexpression of PCBP2 increased the viability of both P3-hBMSCs and P10-hBMSCs significantly. Meanwhile, overexpression of PCBP2 led to significantly reduced expression of FGF2. Overexpression of FGF2 significantly offset the effect of PCBP2 overexpression in P10-hBMSCs, leading to decreased cell proliferation, increased apoptosis, and reduced G0/G1 phase ratio of the cells. Conclusions This study initially elucidates that PCBP2 as an intrinsic aging factor regulates the replicative senescence of hBMSCs through the ROS-FGF2 signaling axis. Funding This study was supported by the National Natural Science Foundation of China (82172474).
Collapse
Affiliation(s)
- Pengbo Chen
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Bo Li
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Zeyu Lu
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Qingyin Xu
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Huoliang Zheng
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Shengdan Jiang
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Leisheng Jiang
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Xinfeng Zheng
- Spine Center, Xinhua Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
3
|
Shi S, Zhu C, Shi S, Li X, Muhammad I, Xu Q, Li X, Zhao Z, Liu H, Fu G, Song M, Huang X, Wang F, Cai J. Human spindle-shaped urine-derived stem cell exosomes alleviate severe fatty liver ischemia-reperfusion injury by inhibiting ferroptosis via GPX4. Stem Cell Res Ther 2025; 16:81. [PMID: 39985001 PMCID: PMC11846247 DOI: 10.1186/s13287-025-04202-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/29/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Severe hepatic steatosis can exacerbate Ischemia-reperfusion injury (IRI), potentially leading to early graft dysfunction and primary non-function. In this study, we investigated the heterogeneity of different subpopulations of Urine-derived stem cells (USCs) to explore the most suitable cell subtype for treating severe steatotic liver IRI. METHODS This study utilized scRNA-seq and Bulk RNA-seq to investigate the transcriptional heterogeneity between Spindle-shaped USCs (SS-USCs) and Rice-shaped USCs (RS-USCs). Additionally, rat fatty Liver transplantation (LT) model, mouse fatty liver IRI model, and Steatotic Hepatocyte Hypoxia-Reoxygenation (SHP-HR) model were constructed. Extracellular vesicles derived from SS-USCs and RS-USCs were isolated and subjected to mass spectrometry analysis. The therapeutic effects of Spindle-shaped USCs Exosomes (SS-USCs-Exo) and Rice-shaped USCs Exosomes (RS-USCs-Exo) were explored, elucidating their potential mechanisms in inhibiting ferroptosis and alleviating IRI. RESULTS Multiple omics analyses confirmed that SS-USCs possess strong tissue repair and antioxidant capabilities, while RS-USCs have the potential to differentiate towards specific directions such as the kidney, nervous system, and skeletal system, particularly showing great application potential in renal system reconstruction. Further experiments demonstrated in vivo and in vitro models confirming that SS-USCs and SS-USCs-Exo significantly inhibit ferroptosis and alleviate severe fatty liver IRI, whereas the effects of RS-USCs/RS-USCs-Exo are less pronounced. Analysis comparing the proteomic differences between SS-USCs-Exo and RS-USCs-Exo revealed that SS-USCs-Exo primarily inhibit ferroptosis and improve cellular viability by secreting exosomes containing Glutathione Peroxidase 4 (GPX4) protein. This highlights the most suitable cell subtype for treating severe fatty liver IRI. CONCLUSIONS SS-USCs possess strong tissue repair and antioxidant capabilities, primarily alleviating ferroptosis in the donor liver of fatty liver through the presence of GPX4 protein in their exosomes. This highlights SS-USCs as the most appropriate cell subtype for treating severe fatty liver IRI.
Collapse
Affiliation(s)
- Shangheng Shi
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cunle Zhu
- Organ Transplant Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shangxuan Shi
- Shanghai Institute for Advanced Immunochemical Studies, Shanghai Tech University, Shanghai, China
| | - Xinqiang Li
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Imran Muhammad
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qingguo Xu
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinwei Li
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ziyin Zhao
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huan Liu
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangming Fu
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Meiying Song
- Department of Immunology, Medical College of Qingdao University, Qingdao, China
| | - Xijian Huang
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Feng Wang
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China.
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jinzhen Cai
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China.
- Institute of Transplantation Science, Affiliated Hospital of Qingdao University, Qingdao, China.
- Organ Transplant Center, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
4
|
Jin R, Dai Y, Wang Z, Hu Q, Zhang C, Gao H, Yan Q. Unraveling Ferroptosis: A New Frontier in Combating Renal Fibrosis and CKD Progression. BIOLOGY 2024; 14:12. [PMID: 39857243 PMCID: PMC11763183 DOI: 10.3390/biology14010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025]
Abstract
Chronic kidney disease (CKD) is a global health concern caused by conditions such as hypertension, diabetes, hyperlipidemia, and chronic nephritis, leading to structural and functional kidney injury. Kidney fibrosis is a common outcome of CKD progression, with abnormal fatty acid oxidation (FAO) disrupting renal energy homeostasis and leading to functional impairments. This results in maladaptive repair mechanisms and the secretion of profibrotic factors, and exacerbates renal fibrosis. Understanding the molecular mechanisms of renal fibrosis is crucial for delaying CKD progression. Ferroptosis is a type of discovered an iron-dependent lipid peroxidation-regulated cell death. Notably, Ferroptosis contributes to tissue and organ fibrosis, which is correlated with the degree of renal fibrosis. This study aims to clarify the complex mechanisms of ferroptosis in renal parenchymal cells and explore how ferroptosis intervention may help alleviate renal fibrosis, particularly by addressing the gap in CKD mechanisms related to abnormal lipid metabolism under the ferroptosis context. The goal is to provide a new theoretical basis for clinically delaying CKD progression.
Collapse
Affiliation(s)
- Rui Jin
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Dai
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Wang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qinyang Hu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongyu Gao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (R.J.); (Y.D.); (Z.W.); (Q.H.); (C.Z.)
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Provincial Clinical Medical Research Center for Nephropathy, Enshi 445000, China
| |
Collapse
|
5
|
Teh MR, Armitage AE, Drakesmith H. Why cells need iron: a compendium of iron utilisation. Trends Endocrinol Metab 2024; 35:1026-1049. [PMID: 38760200 PMCID: PMC11616622 DOI: 10.1016/j.tem.2024.04.015] [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: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/19/2024]
Abstract
Iron deficiency is globally prevalent, causing an array of developmental, haematological, immunological, neurological, and cardiometabolic impairments, and is associated with symptoms ranging from chronic fatigue to hair loss. Within cells, iron is utilised in a variety of ways by hundreds of different proteins. Here, we review links between molecular activities regulated by iron and the pathophysiological effects of iron deficiency. We identify specific enzyme groups, biochemical pathways, cellular functions, and cell lineages that are particularly iron dependent. We provide examples of how iron deprivation influences multiple key systems and tissues, including immunity, hormone synthesis, and cholesterol metabolism. We propose that greater mechanistic understanding of how cellular iron influences physiological processes may lead to new therapeutic opportunities across a range of diseases.
Collapse
Affiliation(s)
- Megan R Teh
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew E Armitage
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hal Drakesmith
- MRC Translational Immune Discovery Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
6
|
Majerníková N, Marmolejo-Garza A, Salinas CS, Luu MDA, Zhang Y, Trombetta-Lima M, Tomin T, Birner-Gruenberger R, Lehtonen Š, Koistinaho J, Wolters JC, Ayton S, den Dunnen WFA, Dolga AM. The link between amyloid β and ferroptosis pathway in Alzheimer's disease progression. Cell Death Dis 2024; 15:782. [PMID: 39468028 PMCID: PMC11519607 DOI: 10.1038/s41419-024-07152-0] [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: 06/03/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
Alzheimer's disease (AD) affects millions of people worldwide and represents the most prevalent form of dementia. Treatment strategies aiming to interfere with the formation of amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs), the two major AD hallmarks, have shown modest or no effect. Recent evidence suggests that ferroptosis, a type of programmed cell death caused by iron accumulation and lipid peroxidation, contributes to AD pathogenesis. The existing link between ferroptosis and AD has been largely based on cell culture and animal studies, while evidence from human brain tissue is limited. Here we evaluate if Aβ is associated with ferroptosis pathways in post-mortem human brain tissue and whether ferroptosis inhibition could attenuate Aβ-related effects in human brain organoids. Performing positive pixel density scoring on immunohistochemically stained post-mortem Brodmann Area 17 sections revealed that the progression of AD pathology was accompanied by decreased expression of nuclear receptor co-activator 4 and glutathione peroxidase 4 in the grey matter. Differentiating between white and grey matter, allowed for a more precise understanding of the disease's impact on different brain regions. In addition, ferroptosis inhibition prevented Aβ pathology, decreased lipid peroxidation and restored iron storage in human AD iPSCs-derived brain cortical organoids at day 50 of differentiation. Differential gene expression analysis of RNAseq of AD organoids compared to isogenic controls indicated activation of the ferroptotic pathway. This was also supported by results from untargeted proteomic analysis revealing significant changes between AD and isogenic brain organoids. Determining the causality between the development of Aβ plaques and the deregulation of molecular pathways involved in ferroptosis is crucial for developing potential therapeutic interventions.
Collapse
Affiliation(s)
- Naďa Majerníková
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research, Research School of Behavioural and Cognitive Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Alejandro Marmolejo-Garza
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells and Systems, Molecular Neurobiology Section, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Casandra Salinas Salinas
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
| | - Minh D A Luu
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
| | - Yuequ Zhang
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
| | - Marina Trombetta-Lima
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Tamara Tomin
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Ruth Birner-Gruenberger
- Institute of Chemical Technologies and Analytics, Faculty of Technical Chemistry, Technische Universität Wien, Vienna, Austria
| | - Šárka Lehtonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Justina C Wolters
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, Faculty of Medical Sciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Scott Ayton
- The Florey Neuroscience Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research, Research School of Behavioural and Cognitive Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Research School of Behavioural and Cognitive Neuroscience, University of Groningen, Groningen, The Netherlands.
- Department of Pathology and Medical Biology, Research Institute Brain and Cognition, Molecular Neuroscience and Aging Research, Research School of Behavioural and Cognitive Neuroscience, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
7
|
Pall AE, Bond S, Bailey DK, Stoj CS, Deschamps I, Huggins P, Parsons J, Bradbury MJ, Kosman DJ, Stemmler TL. ATH434, a promising iron-targeting compound for treating iron regulation disorders. Metallomics 2024; 16:mfae044. [PMID: 39317669 DOI: 10.1093/mtomcs/mfae044] [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: 05/29/2024] [Accepted: 09/22/2024] [Indexed: 09/26/2024]
Abstract
Cytotoxic accumulation of loosely bound mitochondrial Fe2+ is a hallmark of Friedreich's Ataxia (FA), a rare and fatal neuromuscular disorder with limited therapeutic options. There are no clinically approved medications targeting excess Fe2+ associated with FA or the neurological disorders Parkinson's disease and Multiple System Atrophy. Traditional iron-chelating drugs clinically approved for systemic iron overload that target ferritin-stored Fe3+ for urinary excretion demonstrated limited efficacy in FA and exacerbated ataxia. Poor treatment outcomes reflect inadequate binding to excess toxic Fe2+ or exceptionally high affinities (i.e. ≤10-31) for non-pathologic Fe3+ that disrupts intrinsic iron homeostasis. To understand previous treatment failures and identify beneficial factors for Fe2+-targeted therapeutics, we compared traditional Fe3+ chelators deferiprone (DFP) and deferasirox (DFX) with additional iron-binding compounds including ATH434, DMOG, and IOX3. ATH434 and DFX had moderate Fe2+ binding affinities (Kd's of 1-4 µM), similar to endogenous iron chaperones, while the remaining had weaker divalent metal interactions. These compounds had low/moderate affinities for Fe3+(0.46-9.59 µM) relative to DFX and DFP. While all compounds coordinated iron using molecular oxygen and/or nitrogen ligands, thermodynamic analyses suggest ATH434 completes Fe2+ coordination using H2O. ATH434 significantly stabilized bound Fe2+ from ligand-induced autooxidation, reducing reactive oxygen species (ROS) production, whereas DFP and DFX promoted production. The comparable affinity of ATH434 for Fe2+ and Fe3+ position it to sequester excess Fe2+ and facilitate drug-to-protein iron metal exchange, mimicking natural endogenous iron binding proteins, at a reduced risk of autooxidation-induced ROS generation or perturbation of cellular iron stores.
Collapse
Affiliation(s)
- Ashley E Pall
- De partment of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Silas Bond
- Alterity Therapeutics Limited, Melbourne, 3000, Australia
| | - Danielle K Bailey
- Department of Biochemistry, University of Buffalo, Buffalo, NY14203, USA
| | - Christopher S Stoj
- Department of Biochemistry, Chemistry and Physics, Niagara University, Lewiston, NY 14109, USA
| | - Isabel Deschamps
- Department of Biochemistry, Chemistry and Physics, Niagara University, Lewiston, NY 14109, USA
| | - Penny Huggins
- Alterity Therapeutics Limited, Melbourne, 3000, Australia
| | - Jack Parsons
- Alterity Therapeutics Limited, Melbourne, 3000, Australia
| | | | - Daniel J Kosman
- Department of Biochemistry, University of Buffalo, Buffalo, NY14203, USA
| | - Timothy L Stemmler
- De partment of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
8
|
Condeles AL, da Silva GS, Hernandes MBB, Toledo Junior JC. Insights on the endogenous labile iron pool binding properties. Biometals 2024; 37:1065-1077. [PMID: 38691278 DOI: 10.1007/s10534-024-00591-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/18/2024] [Indexed: 05/03/2024]
Abstract
Under normal physiological conditions, the endogenous Labile Iron Pool (LIP) constitutes a ubiquitous, dynamic, tightly regulated reservoir of cellular ferrous iron. Furthermore, LIP is loaded into new apo-iron proteins, a process akin to the activity of metallochaperones. Despite such importance on iron metabolism, the LIP identity and binding properties have remained elusive. We hypothesized that LIP binds to cell constituents (generically denoted C) and forms an iron complex termed CLIP. Combining this binding model with the established Calcein (CA) methodology for assessing cytosolic LIP, we have formulated an equation featuring two experimentally quantifiable parameters (the concentrations of the cytosolic free CA and CA and LIP complex termed CALIP) and three unknown parameters (the total concentrations of LIP and C and their thermodynamic affinity constant Kd). The fittings of cytosolic CALIP × CA concentrations data encompassing a few cellular models to this equation with floating unknown parameters were successful. The computed adjusted total LIP (LIPT) and C (CT) concentrations fall within the sub-to-low micromolar range while the computed Kd was in the 10-2 µM range for all cell types. Thus, LIP binds and has high affinity to cellular constituents found in low concentrations and has remarkably similar properties across different cell types, shedding fresh light on the properties of endogenous LIP within cells.
Collapse
Affiliation(s)
- André Luís Condeles
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Gabriel Simonetti da Silva
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - Maria Beatriz Braghetto Hernandes
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-901, Brazil
| | - José Carlos Toledo Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-901, Brazil.
| |
Collapse
|
9
|
Gawargi FI, Mishra PK. MMP9 drives ferroptosis by regulating GPX4 and iron signaling. iScience 2024; 27:110622. [PMID: 39252956 PMCID: PMC11382059 DOI: 10.1016/j.isci.2024.110622] [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: 12/12/2023] [Revised: 02/08/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Ferroptosis, defined by the suppression of glutathione peroxidase-4 (GPX4) and iron overload, is a distinctive form of regulated cell death. Our in-depth research identifies matrix metalloproteinase-9 (MMP9) as a critical modulator of ferroptosis through its influence on GPX4 and iron homeostasis. Employing an innovative MMP9 construct without collagenase activity, we reveal that active MMP9 interacts with GPX4 and glutathione reductase, reducing GPX4 expression and activity. Furthermore, MMP9 suppresses key transcription factors (SP1, CREB1, NRF2, FOXO3, and ATF4), alongside GPX1 and ferroptosis suppressor protein-1 (FSP1), thereby disrupting the cellular redox balance. MMP9 regulates iron metabolism by modulating iron import, storage, and export via a network of protein interactions. LC-MS/MS has identified 83 proteins that interact with MMP9 at subcellular levels, implicating them in ferroptosis regulation. Integrated pathway analysis (IPA) highlights MMP9's extensive influence on ferroptosis pathways, underscoring its potential as a therapeutic target in conditions with altered redox homeostasis and iron metabolism.
Collapse
Affiliation(s)
- Flobater I Gawargi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
10
|
Qu T, Cha L, Liu H, Tian L, Hu X, Zou H, Feng Y, Sun C, Cao J, Guo W, Qiu F, Zhou B. Circ_0005397 inhibits ferroptosis of pancreatic cancer cells by up-regulating PCBP2 through KAT6A/H3K9Ac. FASEB J 2024; 38:e70028. [PMID: 39235355 DOI: 10.1096/fj.202401151r] [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: 05/23/2024] [Revised: 07/08/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
Pancreatic cancer is a highly aggressive and lethal carcinoma. Circular RNAs (circRNAs) serve key regulatory functions in pancreatic cancer. Ferroptosis was induced by erastin treatment and analyzed by examining malondialdehyde (MDA), iron, Fe2+ and glutathione (GSH). C11-BODIPY 581/591 was used to stain cells for analyzing lipid peroxidation. RNA immunoprecipitation, pull-down and chromatin immunoprecipitation assays were applied to evaluate intermolecular interaction. Mice received subcutaneous injection of pancreatic cancer cells as a model of subcutaneous tumor for in vivo tests. Circ_0005397 was abundantly expressed in pancreatic cancer, and its upregulation was associated with low survival of patients with pancreatic cancer. Circ_0005397 expression was induced by EIF4A3. PCBP2 was highly expressed in pancreatic cancer, and circ_0005397 and PCBP2 were positively correlated in patients with pancreatic cancer. Circ_0005397 knockdown sensitized pancreatic carcinoma cells to ferroptosis via downregulating PCBP2. Circ_0005397 promoted PCBP2 transcription via facilitating the binding of KAT6A and H3K9ac to PCBP2 promoter. Silencing of circ_0005397 reduced tumor growth by enhancing erastin-induced ferroptosis in vivo. EIF4A3-induced circ_0005397 inhibited erastin-induced ferroptosis in pancreatic cancer by promoting PCBP2 expression through KAT6A and H3K9ac.
Collapse
Affiliation(s)
- Tengfei Qu
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lichao Cha
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongliang Liu
- Department of Hepatobiliary Surgery, Qingdao Women's and Children's Hospital, Qingdao, China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao Hu
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hao Zou
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujie Feng
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuandong Sun
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingyu Cao
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weidong Guo
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fabo Qiu
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Retroperitoneal Tumor Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Lai Y, Gao FF, Ge RT, Liu R, Ma S, Liu X. Metal ions overloading and cell death. Cell Biol Toxicol 2024; 40:72. [PMID: 39162885 PMCID: PMC11335907 DOI: 10.1007/s10565-024-09910-4] [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: 04/10/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Cell death maintains cell morphology and homeostasis during development by removing damaged or obsolete cells. The concentration of metal ions whithin cells is regulated by various intracellular transporters and repositories to maintain dynamic balance. External or internal stimuli might increase the concentration of metal ions, which results in ions overloading. Abnormal accumulation of large amounts of metal ions can lead to disruption of various signaling in the cell, which in turn can produce toxic effects and lead to the occurrence of different types of cell deaths. In order to further study the occurrence and development of metal ions overloading induced cell death, this paper reviewed the regulation of Ca2+, Fe3+, Cu2+ and Zn2+ metal ions, and the internal mechanism of cell death induced by overloading. Furthermore, we found that different metal ions possess a synergistic and competitive relationship in the regulation of cell death. And the enhanced level of oxidative stress was present in all the processes of cell death due to metal ions overloading, which possibly due to the combination of factors. Therefore, this review offers a theoretical foundation for the investigation of the toxic effects of metal ions, and presents innovative insights for targeted regulation and therapeutic intervention. HIGHLIGHTS: • Metal ions overloading disrupts homeostasis, which in turn affects the regulation of cell death. • Metal ions overloading can cause cell death via reactive oxygen species (ROS). • Different metal ions have synergistic and competitive relationships for regulating cell death.
Collapse
Affiliation(s)
- Yun Lai
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Fen Fen Gao
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Ruo Ting Ge
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Rui Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
| | - Xiaodong Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou Medical University, Wenzhou, China.
- Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
12
|
da Silva GS, Hernandes MBB, Toledo Junior JC. The Ubiquity of the Reaction of the Labile Iron Pool That Attenuates Peroxynitrite-Dependent Oxidation Intracellularly. Biomolecules 2024; 14:871. [PMID: 39062585 PMCID: PMC11274960 DOI: 10.3390/biom14070871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Although the labile iron pool (LIP) biochemical identity remains a topic of debate, it serves as a universal homeostatically regulated and essential cellular iron source. The LIP plays crucial cellular roles, being the source of iron that is loaded into nascent apo-iron proteins, a process akin to protein post-translational modification, and implicated in the programmed cell death mechanism known as ferroptosis. The LIP is also recognized for its reactivity with chelators, nitric oxide, and peroxides. Our recent investigations in a macrophage cell line revealed a reaction of the LIP with the oxidant peroxynitrite. In contrast to the LIP's pro-oxidant interaction with hydrogen peroxide, this reaction is rapid and attenuates the peroxynitrite oxidative impact. In this study, we demonstrate the existence and antioxidant characteristic of the LIP and peroxynitrite reaction in various cell types. Beyond its potential role as a ubiquitous complementary or substitute protection system against peroxynitrite for cells, the LIP and peroxynitrite reaction may influence cellular iron homeostasis and ferroptosis by changing the LIP redox state and LIP binding properties and reactivity.
Collapse
Affiliation(s)
| | | | - José Carlos Toledo Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| |
Collapse
|
13
|
Jiang L, Zheng H, Ishida M, Lyu Q, Akatsuka S, Motooka Y, Sato K, Sekido Y, Nakamura K, Tanaka H, Ishikawa K, Kajiyama H, Mizuno M, Hori M, Toyokuni S. Elaborate cooperation of poly(rC)-binding proteins 1/2 and glutathione in ferroptosis induced by plasma-activated Ringer's lactate. Free Radic Biol Med 2024; 214:28-41. [PMID: 38325565 DOI: 10.1016/j.freeradbiomed.2024.02.001] [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: 12/28/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Reactive species are involved in various aspects of neoplastic diseases, including carcinogenesis, cancer-specific metabolism and therapeutics. Non-thermal plasma (NTP) can directly provide reactive species, by integrating atmospheric and interjacent molecules as substrates, to represent a handy strategy to load oxidative stress in situ. NTP causes apoptosis and/or ferroptosis specifically in cancer cells of various types. Plasma-activated Ringer's lactate (PAL) is another modality at the preclinical stage as cancer therapeutics, based on more stable reactive species. PAL specifically kills malignant mesothelioma (MM) cells, employing lysosomal ·NO as a switch from autophagy to ferroptosis. However, the entire molecular mechanisms have not been elucidated yet. Here we studied cytosolic iron regulations in MM and other cancer cells in response to PAL exposure. We discovered that cells with higher catalytic Fe(II) are more susceptible to PAL-induced ferroptosis. PAL caused a cytosolic catalytic Fe(II)-associated pathology through iron chaperones, poly (rC)-binding proteins (PCBP)1/2, inducing a disturbance in glutathione-regulated iron homeostasis. PCBP1/NCOA4-mediated ferritinophagy started at a later phase, further increasing cytosolic catalytic Fe(II), ending in ferroptosis. In contrast, PCBP2 after PAL exposure contributed to iron loading to mitochondria, leading to mitochondrial dysfunction. Therapeutic effect of PAL was successfully applied to an orthotopic MM xenograft model in mice. In conclusion, PAL can selectively sensitize MM cells to ferroptosis by remodeling cytoplasmic iron homeostasis, where glutathione and PCBPs play distinct roles, resulting in lethal ferritinophagy and mitochondrial dysfunction. Our findings indicate the clinical application of PAL as a ferroptosis-inducer and the potential of PCBPs as novel targets in cancer therapeutics.
Collapse
Affiliation(s)
- Li Jiang
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Hao Zheng
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Moe Ishida
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Qinying Lyu
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shinya Akatsuka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yashiro Motooka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Kotaro Sato
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan; Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Yoshitaka Sekido
- Division of Cancer Biology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, 464-8681, Japan
| | - Kae Nakamura
- Center for Low-temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8603, Japan; Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Hiromasa Tanaka
- Center for Low-temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8603, Japan; Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, 65 Tsurumai-cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Kenji Ishikawa
- Center for Low-temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8603, Japan
| | - Hiroaki Kajiyama
- Center for Low-temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8603, Japan; Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masaaki Mizuno
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, 65 Tsurumai-cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Masaru Hori
- Center for Low-temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8603, Japan
| | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8603, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Furo-Cho, Chikusa-ku, Nagoya, 464-8603, Japan.
| |
Collapse
|
14
|
Halon-Golabek M, Flis DJ, Zischka H, Akdogan B, Wieckowski MR, Antosiewicz J, Ziolkowski W. Amyotrophic lateral sclerosis associated disturbance of iron metabolism is blunted by swim training-role of AKT signaling pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167014. [PMID: 38171451 DOI: 10.1016/j.bbadis.2023.167014] [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: 08/25/2023] [Revised: 12/08/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024]
Abstract
Swim training has increased the life span of the transgenic animal model of amyotrophic lateral sclerosis (ALS). Conversely, the progress of the disease is associated with the impairment of iron metabolism and insulin signaling. We used transgenic hmSOD1 G93A (ALS model) and non-transgenic mice in the present study. The study was performed on the muscles taken from trained (ONSET and TERMINAL) and untrained animals at three stages of the disease: BEFORE, ONSET, and TERMINAL. In order to study the molecular mechanism of changes in iron metabolism, we used SH-SY5Y and C2C12 cell lines expression vector pcDNA3.1 and transiently transfected with specific siRNAs. The progress of ALS resulted in decreased P-Akt/Akt ratio, which is associated with increased proteins responsible for iron storage ferritin L, ferritin H, PCBP1, and skeletal muscle iron at ONSET. Conversely, proteins responsible for iron export- TAU significantly decrease. The training partially reverses changes in proteins responsible for iron metabolism. AKT silencing in the SH-SY5Y cell line decreased PCBP2 and ferroportin and increased ferritin L, H, PCBP1, TAU, transferrin receptor 1, and APP. Moreover, silencing APP led to an increase in ferritin L and H. Our data suggest that swim training in the mice ALS model is associated with significant changes in iron metabolism related to AKT activity. Down-regulation of AKT mainly upregulates proteins involved in iron import and storage but decreases proteins involved in iron export.
Collapse
Affiliation(s)
- Małgorzata Halon-Golabek
- Department of Physiotherapy, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Damian Jozef Flis
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Institute of Toxicology and Environmental Hygiene, Technical University Munich, School of Medicine, Munich, Germany
| | - Banu Akdogan
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Mariusz Roman Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Jedrzej Antosiewicz
- Department of Bioenergetics and Physiology of Exercise, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland.
| | - Wiesław Ziolkowski
- Department of Rehabilitation Medicine, Faculty of Health Sciences Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
15
|
Wang Y, Protchenko O, Huber KD, Shakoury-Elizeh M, Ghosh MC, Philpott CC. The iron chaperone poly(rC)-binding protein 1 regulates iron efflux through intestinal ferroportin in mice. Blood 2023; 142:1658-1671. [PMID: 37624904 PMCID: PMC10656723 DOI: 10.1182/blood.2023020504] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/28/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Iron is an essential nutrient required by all cells but used primarily for red blood cell production. Because humans have no effective mechanism for ridding the body of excess iron, the absorption of dietary iron must be precisely regulated. The critical site of regulation is the transfer of iron from the absorptive enterocyte to the portal circulation via the sole iron efflux transporter, ferroportin. Here, we report that poly(rC)-binding protein 1 (PCBP1), the major cytosolic iron chaperone, is necessary for the regulation of iron flux through ferroportin in the intestine of mice. Mice lacking PCBP1 in the intestinal epithelium exhibit low levels of enterocyte iron, poor retention of dietary iron in enterocyte ferritin, and excess efflux of iron through ferroportin. Excess iron efflux occurred despite lower levels of ferroportin protein in enterocytes and upregulation of the iron regulatory hormone hepcidin. PCBP1 deletion and the resulting unregulated dietary iron absorption led to poor growth, severe anemia on a low-iron diet, and liver oxidative stress with iron loading on a high-iron diet. Ex vivo culture of PCBP1-depleted enteroids demonstrated no defects in hepcidin-mediated ferroportin turnover. However, measurement of kinetically labile iron pools in enteroids competent or blocked for iron efflux indicated that PCBP1 functioned to bind and retain cytosolic iron and limit its availability for ferroportin-mediated efflux. Thus, PCBP1 coordinates enterocyte iron and reduces the concentration of unchaperoned "free" iron to a low level that is necessary for hepcidin-mediated regulation of ferroportin activity.
Collapse
Affiliation(s)
- Yubo Wang
- Genetics and Metabolism Section, Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Olga Protchenko
- Genetics and Metabolism Section, Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Kari D. Huber
- Genetics and Metabolism Section, Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Minoo Shakoury-Elizeh
- Genetics and Metabolism Section, Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Manik C. Ghosh
- Section on Human Iron Metabolism, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Caroline C. Philpott
- Genetics and Metabolism Section, Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
16
|
Zheng X, Zhang C. The Regulation of Ferroptosis by Noncoding RNAs. Int J Mol Sci 2023; 24:13336. [PMID: 37686142 PMCID: PMC10488123 DOI: 10.3390/ijms241713336] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
As a novel form of regulated cell death, ferroptosis is characterized by intracellular iron and lipid peroxide accumulation, which is different from other regulated cell death forms morphologically, biochemically, and immunologically. Ferroptosis is regulated by iron metabolism, lipid metabolism, and antioxidant defense systems as well as various transcription factors and related signal pathways. Emerging evidence has highlighted that ferroptosis is associated with many physiological and pathological processes, including cancer, neurodegeneration diseases, cardiovascular diseases, and ischemia/reperfusion injury. Noncoding RNAs are a group of functional RNA molecules that are not translated into proteins, which can regulate gene expression in various manners. An increasing number of studies have shown that noncoding RNAs, especially miRNAs, lncRNAs, and circRNAs, can interfere with the progression of ferroptosis by modulating ferroptosis-related genes or proteins directly or indirectly. In this review, we summarize the basic mechanisms and regulations of ferroptosis and focus on the recent studies on the mechanism for different types of ncRNAs to regulate ferroptosis in different physiological and pathological conditions, which will deepen our understanding of ferroptosis regulation by noncoding RNAs and provide new insights into employing noncoding RNAs in ferroptosis-associated therapeutic strategies.
Collapse
Affiliation(s)
| | - Cen Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350108, China;
| |
Collapse
|
17
|
Yanatori I, Nishina S, Kishi F, Hino K. Newly uncovered biochemical and functional aspects of ferritin. FASEB J 2023; 37:e23095. [PMID: 37440196 DOI: 10.1096/fj.202300918r] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023]
Abstract
Iron homeostasis is strictly regulated at both the systemic and cellular levels by complex mechanisms because of its indispensability and toxicity. Among the various iron-regulatory proteins, ferritin is the earliest discovered regulator of iron metabolism and is a molecule that safely retains excess intracellular iron in the cores of its shells. Two types of ferritin, cytosolic ferritin and mitochondrial ferritin (FTMT), have been identified in a range of organisms from plants to humans. FTMT was identified approximately 60 years after the discovery of cytosolic ferritin. Cytosolic ferritin expression is regulated in an iron-responsive manner. Recently, the molecular mechanisms of iron-dependent degradation of cytosolic ferritin or its secretion into serum have been clarified. FTMT, which shares a high degree of sequence homology with cytosolic ferritin, has distinct functions and is regulated in different ways from cytosolic ferritin. Although knowledge of the physiological role of FTMT is still incomplete, recent studies have shed light on the function and regulation of FTMT. The accumulating biological evidence of both ferritins has made it possible to deepen our knowledge about iron metabolism and its significance in diseases. In this review, we discuss the biological properties of both ferritins, focusing on their newly uncovered behaviors.
Collapse
Affiliation(s)
- Izumi Yanatori
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sohji Nishina
- Department of Gastroenterology, Kawasaki Medical School, Kurashiki, Japan
| | - Fumio Kishi
- Kenjinkai Healthcare Corporation, Sanyo-Onoda, Japan
| | - Keisuke Hino
- Department of Gastroenterology, Kawasaki Medical School, Kurashiki, Japan
- Digestive Disease Center, Shunan Memorial Hospital, Kudamatsu, Japan
| |
Collapse
|
18
|
Cheng X, Zhang J, Xiao Y, Wang Z, He J, Ke M, Liu S, Wang Q, Zhang L. Mitochondrial Regulation of Ferroptosis in Cancer Therapy. Int J Mol Sci 2023; 24:10037. [PMID: 37373183 DOI: 10.3390/ijms241210037] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Ferroptosis, characterized by glutamate overload, glutathione depletion, and cysteine/cystine deprivation during iron- and oxidative-damage-dependent cell death, is a particular mode of regulated cell death. It is expected to effectively treat cancer through its tumor-suppressor function, as mitochondria are the intracellular energy factory and a binding site of reactive oxygen species production, closely related to ferroptosis. This review summarizes relevant research on the mechanisms of ferroptosis, highlights mitochondria's role in it, and collects and classifies the inducers of ferroptosis. A deeper understanding of the relationship between ferroptosis and mitochondrial function may provide new strategies for tumor treatment and drug development based on ferroptosis.
Collapse
Affiliation(s)
- Xiaoxia Cheng
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Jiale Zhang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Yichen Xiao
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Zhihang Wang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Jin He
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Mengquan Ke
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Sijie Liu
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Qun Wang
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| | - Lei Zhang
- School of Basic Medical Science, Henan University, Kaifeng 475004, China
| |
Collapse
|
19
|
Bayır H, Dixon SJ, Tyurina YY, Kellum JA, Kagan VE. Ferroptotic mechanisms and therapeutic targeting of iron metabolism and lipid peroxidation in the kidney. Nat Rev Nephrol 2023; 19:315-336. [PMID: 36922653 DOI: 10.1038/s41581-023-00689-x] [Citation(s) in RCA: 131] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/17/2023]
Abstract
Ferroptosis is a mechanism of regulated necrotic cell death characterized by iron-dependent, lipid peroxidation-driven membrane destruction that can be inhibited by glutathione peroxidase 4. Morphologically, it is characterized by cellular, organelle and cytoplasmic swelling and the loss of plasma membrane integrity, with the release of intracellular components. Ferroptosis is triggered in cells with dysregulated iron and thiol redox metabolism, whereby the initial robust but selective accumulation of hydroperoxy polyunsaturated fatty acid-containing phospholipids is further propagated through enzymatic and non-enzymatic secondary mechanisms, leading to formation of oxidatively truncated electrophilic species and their adducts with proteins. Thus, ferroptosis is dependent on the convergence of iron, thiol and lipid metabolic pathways. The kidney is particularly susceptible to redox imbalance. A growing body of evidence has linked ferroptosis to acute kidney injury in the context of diverse stimuli, such as ischaemia-reperfusion, sepsis or toxins, and to chronic kidney disease, suggesting that ferroptosis may represent a novel therapeutic target for kidney disease. However, further work is needed to address gaps in our understanding of the triggers, execution and spreading mechanisms of ferroptosis.
Collapse
Affiliation(s)
- Hülya Bayır
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John A Kellum
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Departments of Environmental Health, Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Philpott CC, Protchenko O, Wang Y, Novoa-Aponte L, Leon-Torres A, Grounds S, Tietgens AJ. Iron-tracking strategies: Chaperones capture iron in the cytosolic labile iron pool. Front Mol Biosci 2023; 10:1127690. [PMID: 36818045 PMCID: PMC9932599 DOI: 10.3389/fmolb.2023.1127690] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Cells express hundreds of iron-dependent enzymes that rely on the iron cofactors heme, iron-sulfur clusters, and mono-or di-nuclear iron centers for activity. Cells require systems for both the assembly and the distribution of iron cofactors to their cognate enzymes. Proteins involved in the binding and trafficking of iron ions in the cytosol, called cytosolic iron chaperones, have been identified and characterized in mammalian cells. The first identified iron chaperone, poly C-binding protein 1 (PCBP1), has also been studied in mice using genetic models of conditional deletion in tissues specialized for iron handling. Studies of iron trafficking in mouse tissues have necessitated the development of new approaches, which have revealed new roles for PCBP1 in the management of cytosolic iron. These approaches can be applied to investigate use of other nutrient metals in mammals.
Collapse
|
21
|
Fuhrmann DC, Becker S, Brüne B. Mitochondrial ferritin expression in human macrophages is facilitated by thrombin-mediated cleavage under hypoxia. FEBS Lett 2023; 597:276-287. [PMID: 36416578 DOI: 10.1002/1873-3468.14545] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/26/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022]
Abstract
Ferritins are iron storage proteins, which maintain cellular iron homeostasis. Among these proteins, the ferritin heavy chain is well characterized, but the regulatory principles of mitochondrial ferritin (FTMT) remain elusive. FTMT appears to be cleaved from a 27 kDa to a 22 kDa form. In human macrophages, FTMT increased under hypoxia in a hypoxia-inducible factor 2-dependent manner. Occurrence of FTMT resulted from cleavage by thrombin, which was supplied by serum. Inhibition of thrombin as well as serum removal decreased FTMT, while supplementation of thrombin under serum-deprived conditions restored its expression. Besides hypoxia, thrombin facilitated FTMT expression after treatment with the ferroptosis inducer RSL3 and the pro-inflammatory stimulus lipopolysaccharide. This study provides insights into the regulation of FTMT under hypoxia and identifies thrombin as a FTMT maturation-associated peptidase.
Collapse
Affiliation(s)
- Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site, Frankfurt, Germany
| | - Sabrina Becker
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Germany.,German Cancer Consortium (DKTK), Partner Site, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| |
Collapse
|
22
|
Martinelli M, Aguilar G, Lee DS, Kromer A, Nguyen N, Wilkins BJ, Akimova T, Beier UH, Ghanem LR. The poly(C)-binding protein Pcbp2 is essential for CD4 + T cell activation and proliferation. iScience 2022; 26:105860. [PMID: 36632062 PMCID: PMC9826892 DOI: 10.1016/j.isci.2022.105860] [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: 06/16/2022] [Revised: 11/16/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
The RNA-binding protein Pcbp2 is widely expressed in the innate and adaptive immune systems and is essential for mouse development. To determine whether Pcbp2 is required for CD4+ T cell development and function, we derived mice with conditional Pcbp2 deletion in CD4+ T cells and assessed their overall phenotype and proliferative responses to activating stimuli. We found that Pcbp2 is essential for T conventional cell (Tconv) proliferation, working through regulation of co-stimulatory signaling. Pcbp2 deficiency in the CD4+ lineage did not impact Treg abundance in vivo or function in vitro. In addition, our data demonstrate a clear association between Pcbp2 control of Runx1 exon 6 splicing in CD4+ T cells and a specific role for Pcbp2 in the maintenance of peripheral CD4+ lymphocyte population size. Last, we show that Pcbp2 function is required for optimal in vivo Tconv cell activation in a T cell adoptive transfer colitis model system.
Collapse
Affiliation(s)
- Massimo Martinelli
- Division of Gastroenterology, Hepatology and Nutrition Division, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA,Department of Translational Medical Science, Section of Pediatrics, University of Naples “Federico II”, Naples 80131, Italy
| | - Gabrielle Aguilar
- Division of Gastroenterology, Hepatology and Nutrition Division, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - David S.M. Lee
- Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA,Institute for Biomedical Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew Kromer
- Division of Gastroenterology, Hepatology and Nutrition Division, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nhu Nguyen
- Division of Gastroenterology, Hepatology and Nutrition Division, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Benjamin J. Wilkins
- Division of Anatomic Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA,Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tatiana Akimova
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ulf H. Beier
- Division of Nephrology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Louis R. Ghanem
- Division of Gastroenterology, Hepatology and Nutrition Division, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA,Corresponding author
| |
Collapse
|
23
|
Kawabata T. Iron-Induced Oxidative Stress in Human Diseases. Cells 2022; 11:cells11142152. [PMID: 35883594 PMCID: PMC9324531 DOI: 10.3390/cells11142152] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Iron is responsible for the regulation of several cell functions. However, iron ions are catalytic and dangerous for cells, so the cells sequester such redox-active irons in the transport and storage proteins. In systemic iron overload and local pathological conditions, redox-active iron increases in the human body and induces oxidative stress through the formation of reactive oxygen species. Non-transferrin bound iron is a candidate for the redox-active iron in extracellular space. Cells take iron by the uptake machinery such as transferrin receptor and divalent metal transporter 1. These irons are delivered to places where they are needed by poly(rC)-binding proteins 1/2 and excess irons are stored in ferritin or released out of the cell by ferroportin 1. We can imagine transit iron pool in the cell from iron import to the export. Since the iron in the transit pool is another candidate for the redox-active iron, the size of the pool may be kept minimally. When a large amount of iron enters cells and overflows the capacity of iron binding proteins, the iron behaves as a redox-active iron in the cell. This review focuses on redox-active iron in extracellular and intracellular spaces through a biophysical and chemical point of view.
Collapse
Affiliation(s)
- Teruyuki Kawabata
- Department of Applied Physics, Postgraduate School of Science, Okayama University of Science, Okayama 700-0005, Japan
| |
Collapse
|
24
|
Kotla NK, Dutta P, Parimi S, Das NK. The Role of Ferritin in Health and Disease: Recent Advances and Understandings. Metabolites 2022; 12:metabo12070609. [PMID: 35888733 PMCID: PMC9320524 DOI: 10.3390/metabo12070609] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
Systemic iron homeostasis needs to be tightly controlled, as both deficiency and excess iron cause major global health concerns, such as iron deficiency anemia, hemochromatosis, etc. In mammals, sufficient dietary acquisition is critical for fulfilling the systemic iron requirement. New questions are emerging about whether and how cellular iron transport pathways integrate with the iron storage mechanism. Ferritin is the intracellular iron storage protein that stores surplus iron after all the cellular needs are fulfilled and releases it in the face of an acute demand. Currently, there is a surge in interest in ferritin research after the discovery of novel pathways like ferritinophagy and ferroptosis. This review emphasizes the most recent ferritin-related discoveries and their impact on systemic iron regulation.
Collapse
|
25
|
Ou M, Jiang Y, Ji Y, Zhou Q, Du Z, Zhu H, Zhou Z. Role and Mechanism of Ferroptosis in Neurological Diseases. Mol Metab 2022; 61:101502. [PMID: 35447365 PMCID: PMC9170779 DOI: 10.1016/j.molmet.2022.101502] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 02/08/2023] Open
Abstract
Background Ferroptosis, as a new form of cell death, is different from other cell deaths such as autophagy or senescence. Ferroptosis involves in the pathophysiological progress of several diseases, including cancers, cardiovascular diseases, nervous system diseases, and kidney damage. Since oxidative stress and iron deposition are the broad pathological features of neurological diseases, the role of ferroptosis in neurological diseases has been widely explored. Scope of review Ferroptosis is mainly characterized by changes in iron homeostasis, iron-dependent lipid peroxidation, and glutamate toxicity accumulation, of which can be specifically reversed by ferroptosis inducers or inhibitors. The ferroptosis is mainly regulated by the metabolism of iron, lipids and amino acids through System Xc−, voltage-dependent anion channels, p53, p62-Keap1-Nrf2, mevalonate and other pathways. This review also focus on the regulatory pathways of ferroptosis and its research progress in neurological diseases. Major conclusions The current researches of ferroptosis in neurological diseases mostly focus on the key pathways of ferroptosis. At the same time, ferroptosis was found playing a bidirectional regulation role in neurological diseases. Therefore, the specific regulatory mechanisms of ferroptosis in neurological diseases still need to be further explored to provide new perspectives for the application of ferroptosis in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Mengmeng Ou
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Ying Jiang
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Yingying Ji
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Qin Zhou
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Zhiqiang Du
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Haohao Zhu
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| | - Zhenhe Zhou
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| |
Collapse
|
26
|
Ferritinophagy and α-Synuclein: Pharmacological Targeting of Autophagy to Restore Iron Regulation in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23042378. [PMID: 35216492 PMCID: PMC8878351 DOI: 10.3390/ijms23042378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
A major hallmark of Parkinson’s disease (PD) is the fatal destruction of dopaminergic neurons within the substantia nigra pars compacta. This event is preceded by the formation of Lewy bodies, which are cytoplasmic inclusions composed of α-synuclein protein aggregates. A triad contribution of α-synuclein aggregation, iron accumulation, and mitochondrial dysfunction plague nigral neurons, yet the events underlying iron accumulation are poorly understood. Elevated intracellular iron concentrations up-regulate ferritin expression, an iron storage protein that provides cytoprotection against redox stress. The lysosomal degradation pathway, autophagy, can release iron from ferritin stores to facilitate its trafficking in a process termed ferritinophagy. Aggregated α-synuclein inhibits SNARE protein complexes and destabilizes microtubules to halt vesicular trafficking systems, including that of autophagy effectively. The scope of this review is to describe the physiological and pathological relationship between iron regulation and α-synuclein, providing a detailed understanding of iron metabolism within nigral neurons. The underlying mechanisms of autophagy and ferritinophagy are explored in the context of PD, identifying potential therapeutic targets for future investigation.
Collapse
|
27
|
Liu WJ, Pan PY, Sun Y, Wang JB, Zhou H, Xie X, Duan ZY, Dong HY, Chen WN, Zhang LD, Wang C. Deferoxamine Counteracts Cisplatin Resistance in A549 Lung Adenocarcinoma Cells by Increasing Vulnerability to Glutamine Deprivation-Induced Cell Death. Front Oncol 2022; 11:794735. [PMID: 35127502 PMCID: PMC8810525 DOI: 10.3389/fonc.2021.794735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/30/2021] [Indexed: 02/06/2023] Open
Abstract
Glutamine, like glucose, is a major nutrient consumed by cancer cells, yet these cells undergo glutamine starvation in the cores of tumors, forcing them to evolve adaptive metabolic responses. Pharmacologically targeting glutamine metabolism or withdrawal has been exploited for therapeutic purposes, but does not always induce cancer cell death. The mechanism by which cancer cells adapt to resist glutamine starvation in cisplatin-resistant non-small-cell lung cancer (NSCLC) also remains uncertain. Here, we report the potential metabolic vulnerabilities of A549/DDP (drug-resistant human lung adenocarcinoma cell lines) cells, which were more easily killed by the iron chelator deferoxamine (DFO) during glutamine deprivation than their parental cisplatin-sensitive A549 cells. We demonstrate that phenotype resistance to cisplatin is accompanied by adaptive responses during glutamine deprivation partly via higher levels of autophagic activity and apoptosis resistance characteristics. Moreover, this adaptation could be explained by sustained glucose instead of glutamine-dominant complex II-dependent oxidative phosphorylation (OXPHOS). Further investigation revealed that cisplatin-resistant cells sustain OXPHOS partly via iron metabolism reprogramming during glutamine deprivation. This reprogramming might be responsible for mitochondrial iron-sulfur [Fe-S] cluster biogenesis, which has become an “Achilles’ heel,” rendering cancer cells vulnerable to DFO-induced autophagic cell death and apoptosis through c-Jun N-terminal kinase (JNK) signaling. Finally, in vivo studies using xenograft mouse models also confirmed the growth-slowing effect of DFO. In summary, we have elucidated the adaptive responses of cisplatin-resistant NSCLC cells, which balanced stability and plasticity to overcome metabolic reprogramming and permitted them to survive under stress induced by chemotherapy or glutamine starvation. In addition, for the first time, we show that suppressing the growth of cisplatin-resistant NSCLC cells via iron chelator-induced autophagic cell death and apoptosis was possible with DFO treatment. These findings provide a solid basis for targeting mitochondria iron metabolism in cisplatin-resistant NSCLC for therapeutic purposes, and it is plausible to consider that DFO facilitates in the improvement of treatment responses in cisplatin-resistant NSCLC patients.
Collapse
Affiliation(s)
- Wen-Jun Liu
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China.,Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Peng-Yu Pan
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Ye Sun
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, Shenyang, China
| | - Jian-Bo Wang
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine (TCM) Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Huan Zhou
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xin Xie
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Zhi-Yuan Duan
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Han-Yu Dong
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wen-Na Chen
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine (TCM) Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Li-de Zhang
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Chun Wang
- Department of Cell Biology, College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
28
|
Mechanistic Insights Expatiating the Redox-Active-Metal-Mediated Neuronal Degeneration in Parkinson's Disease. Int J Mol Sci 2022; 23:ijms23020678. [PMID: 35054862 PMCID: PMC8776156 DOI: 10.3390/ijms23020678] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 02/06/2023] Open
Abstract
Parkinson’s disease (PD) is a complicated and incapacitating neurodegenerative malady that emanates following the dopaminergic (DArgic) nerve cell deprivation in the substantia nigra pars compacta (SN-PC). The etiopathogenesis of PD is still abstruse. Howbeit, PD is hypothesized to be precipitated by an amalgamation of genetic mutations and exposure to environmental toxins. The aggregation of α-synucelin within the Lewy bodies (LBs), escalated oxidative stress (OS), autophagy-lysosome system impairment, ubiquitin-proteasome system (UPS) impairment, mitochondrial abnormality, programmed cell death, and neuroinflammation are regarded as imperative events that actively participate in PD pathogenesis. The central nervous system (CNS) relies heavily on redox-active metals, particularly iron (Fe) and copper (Cu), in order to modulate pivotal operations, for instance, myelin generation, synthesis of neurotransmitters, synaptic signaling, and conveyance of oxygen (O2). The duo, namely, Fe and Cu, following their inordinate exposure, are viable of permeating across the blood–brain barrier (BBB) and moving inside the brain, thereby culminating in the escalated OS (through a reactive oxygen species (ROS)-reliant pathway), α-synuclein aggregation within the LBs, and lipid peroxidation, which consequently results in the destruction of DArgic nerve cells and facilitates PD emanation. This review delineates the metabolism of Fe and Cu in the CNS, their role and disrupted balance in PD. An in-depth investigation was carried out by utilizing the existing publications obtained from prestigious medical databases employing particular keywords mentioned in the current paper. Moreover, we also focus on decoding the role of metal complexes and chelators in PD treatment. Conclusively, metal chelators hold the aptitude to elicit the scavenging of mobile/fluctuating metal ions, which in turn culminates in the suppression of ROS generation, and thereby prelude the evolution of PD.
Collapse
|
29
|
Jadhav S, Protchenko O, Li F, Baratz E, Shakoury-Elizeh M, Maschek A, Cox J, Philpott CC. Mitochondrial dysfunction in mouse livers depleted of iron chaperone PCBP1. Free Radic Biol Med 2021; 175:18-27. [PMID: 34455040 PMCID: PMC9137418 DOI: 10.1016/j.freeradbiomed.2021.08.232] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/05/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
Iron is an essential nutrient that forms cofactors required for the activity of hundreds of cellular proteins. However, iron can be toxic and must be precisely managed. Poly r(C) binding protein 1 (PCBP1) is an essential, multifunctional protein that binds both iron and nucleic acids, regulating the fate of both. As an iron chaperone, PCBP1 binds cytosolic iron and delivers it to iron enzymes for activation and to ferritin for storage. Mice deleted for PCBP1 in the liver exhibit dysregulated iron balance, with lower levels of liver iron stores and iron enzymes, but higher levels of chemically-reactive iron. Unchaperoned iron triggers the formation of reactive oxygen species, leading to lipid peroxidation and ferroptotic cell death. Hepatic PCBP1 deletion produces chronic liver disease in mice, with steatosis, triglyceride accumulation, and elevated plasma ALT levels. Human and mouse models of fatty liver disease are associated with mitochondrial dysfunction. Here we show that, although deletion of PCBP1 does not affect mitochondrial iron balance, it does affect mitochondrial function. PCBP1 deletion affected mitochondrial morphology and reduced levels of respiratory complexes II and IV, oxygen consumption, and ATP production. Depletion of mitochondrial lipids cardiolipin and coenzyme Q, along with reduction of mitochondrial oxygen consumption, were the first manifestations of mitochondrial dysfunction. Although dietary supplementation with vitamin E ameliorated the liver disease in mice with hepatic PCBP1 deletion, supplementation with coenzyme Q was required to fully restore mitochondrial lipids and function. In conclusion, our studies indicate that mitochondrial function can be restored in livers subjected to ongoing oxidative damage from unchaperoned iron by supplementation with coenzyme Q, a mitochondrial lipid essential for respiration that also functions as a lipophilic radical-trapping agent.
Collapse
Affiliation(s)
| | - Olga Protchenko
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD, USA
| | - Fengmin Li
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD, USA
| | - Ethan Baratz
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD, USA
| | | | - Alan Maschek
- Dept. of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - James Cox
- Dept. of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | |
Collapse
|
30
|
Condeles AL, Toledo Junior JC. The Labile Iron Pool Reacts Rapidly and Catalytically with Peroxynitrite. Biomolecules 2021; 11:1331. [PMID: 34572543 PMCID: PMC8466499 DOI: 10.3390/biom11091331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022] Open
Abstract
While investigating peroxynitrite-dependent oxidation in murine RAW 264.7 macrophage cells, we observed that removal of the Labile Iron Pool (LIP) by chelation increases the intracellular oxidation of the fluorescent indicator H2DCF, so we concluded that the LIP reacts with peroxynitrite and decreases the yield of peroxynitrite-derived oxidants. This was a paradigm-shifting finding in LIP biochemistry and raised many questions. In this follow-up study, we address fundamental properties of the interaction between the LIP and peroxynitrite by using the same cellular model and fluorescence methodology. We have identified that the reaction between the LIP and peroxynitrite has catalytic characteristics, and we have estimated that the rate constant of the reaction is in the range of 106 to 107 M-1s-1. Together, these observations suggest that the LIP represents a constitutive peroxynitrite reductase system in RAW 264.7 cells.
Collapse
Affiliation(s)
| | - José Carlos Toledo Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil;
| |
Collapse
|
31
|
Dietz JV, Fox JL, Khalimonchuk O. Down the Iron Path: Mitochondrial Iron Homeostasis and Beyond. Cells 2021; 10:cells10092198. [PMID: 34571846 PMCID: PMC8468894 DOI: 10.3390/cells10092198] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cellular iron homeostasis and mitochondrial iron homeostasis are interdependent. Mitochondria must import iron to form iron–sulfur clusters and heme, and to incorporate these cofactors along with iron ions into mitochondrial proteins that support essential functions, including cellular respiration. In turn, mitochondria supply the cell with heme and enable the biogenesis of cytosolic and nuclear proteins containing iron–sulfur clusters. Impairment in cellular or mitochondrial iron homeostasis is deleterious and can result in numerous human diseases. Due to its reactivity, iron is stored and trafficked through the body, intracellularly, and within mitochondria via carefully orchestrated processes. Here, we focus on describing the processes of and components involved in mitochondrial iron trafficking and storage, as well as mitochondrial iron–sulfur cluster biogenesis and heme biosynthesis. Recent findings and the most pressing topics for future research are highlighted.
Collapse
Affiliation(s)
- Jonathan V. Dietz
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA;
| | - Jennifer L. Fox
- Department of Chemistry and Biochemistry, College of Charleston, Charleston, SC 29424, USA;
| | - Oleh Khalimonchuk
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA;
- Nebraska Redox Biology Center, University of Nebraska, Lincoln, NE 68588, USA
- Fred and Pamela Buffett Cancer Center, Omaha, NE 68198, USA
- Correspondence:
| |
Collapse
|
32
|
Kosman DJ. A holistic view of mammalian (vertebrate) cellular iron uptake. Metallomics 2021; 12:1323-1334. [PMID: 32766655 DOI: 10.1039/d0mt00065e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cell iron uptake in mammals is commonly distinguished by whether the iron is presented to the cell as transferrin-bound or not: TBI or NTBI. This generic perspective conflates TBI with canonical transferrin receptor, endosomal iron uptake, and NTBI with uptake supported by a plasma membrane-localized divalent metal ion transporter, most often identified as DMT1. In fact, iron uptake by mammalian cells is far more nuanced than this somewhat proscribed view suggests. This view fails to accommodate the substantial role that ZIP8 and ZIP14 play in iron uptake, while adhering to the traditional premise that a relatively high endosomal [H+] is thermodynamically required for release of iron from holo-Tf. The canonical view of iron uptake also does not encompass the fact that plasma membrane electron transport - PMET - has long been linked to cell iron uptake. In fact, the known mammalian metallo-reductases - Dcytb and the STEAP proteins - are members of this cohort of cytochrome-dependent oxido-reductases that shuttle reducing equivalents across the plasma membrane. A not commonly appreciated fact is the reduction potential of ferric iron in holo-Tf is accessible to cytoplasmic reducing equivalents - reduced pyridine and flavin mono- and di-nucleotides and dihydroascorbic acid. This allows for the reductive release of Fe2+ at the extracellular surface of the PM and subsequent transport into the cytoplasm by a neutral pH transporter - a ZIP protein. What this perspective emphasizes is that there are two TfR-dependent uptake pathways, one which does and one which does not involve clathrin-dependent, endolysosomal trafficking. This raises the question as to the selective advantage of having two Tf, TfR-dependent routes of iron accumulation. This review of canonical and non-canonical iron uptake uses cerebral iron trafficking as a point of discussion, a focus that encourages inclusion also of the importance of ferritin as a circulating 'chaperone' of ferric iron.
Collapse
Affiliation(s)
- Daniel J Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of Buffalo, Suite 4102, 995 Main St., Buffalo, NY 14203, USA.
| |
Collapse
|
33
|
Ladha FA, Thakar K, Pettinato AM, Legere N, Ghahremani S, Cohn R, Romano R, Meredith E, Chen YS, Hinson JT. Actinin BioID reveals sarcomere crosstalk with oxidative metabolism through interactions with IGF2BP2. Cell Rep 2021; 36:109512. [PMID: 34380038 PMCID: PMC8447243 DOI: 10.1016/j.celrep.2021.109512] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 05/16/2021] [Accepted: 07/21/2021] [Indexed: 01/13/2023] Open
Abstract
Actinins are strain-sensing actin cross-linkers that are ubiquitously expressed and harbor mutations in human diseases. We utilize CRISPR, pluripotent stem cells, and BioID to study actinin interactomes in human cardiomyocytes. We identify 324 actinin proximity partners, including those that are dependent on sarcomere assembly. We confirm 19 known interactors and identify a network of RNA-binding proteins, including those with RNA localization functions. In vivo and biochemical interaction studies support that IGF2BP2 localizes electron transport chain transcripts to actinin neighborhoods through interactions between its K homology (KH) domain and actinin’s rod domain. We combine alanine scanning mutagenesis and metabolic assays to disrupt and functionally interrogate actinin-IGF2BP2 interactions, which reveal an essential role in metabolic responses to pathological sarcomere activation using a hypertrophic cardiomyopathy model. This study expands our functional knowledge of actinin, uncovers sarcomere interaction partners, and reveals sarcomere crosstalk with IGF2BP2 for metabolic adaptation relevant to human disease. Ladha et al. combine BioID, human cardiomyocytes, and CRISPR-Cas9 to interrogate the actinin interactome. This reveals 324 actinin proximity partners, including RNA-binding proteins that bind transcripts encoding ETC functional components. Among these RNA-binding proteins, IGF2BP2 directly binds actinin, and actinin-IGF2BP2 interactions regulate ETC transcript localization and metabolic adaptation to sarcomere function.
Collapse
Affiliation(s)
- Feria A Ladha
- University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ketan Thakar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | | | - Nicholas Legere
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | | | - Rachel Cohn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Robert Romano
- University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Emily Meredith
- University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Yu-Sheng Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - J Travis Hinson
- University of Connecticut Health Center, Farmington, CT 06030, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Cardiology Center, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
34
|
Itoh A, Adams D, Huang W, Wu Y, Kachapati K, Bednar KJ, Leung PSC, Zhang W, Flavell RA, Gershwin ME, Ridgway WM. Enoxacin Up-Regulates MicroRNA Biogenesis and Down-Regulates Cytotoxic CD8 T-Cell Function in Autoimmune Cholangitis. Hepatology 2021; 74:835-846. [PMID: 33462854 DOI: 10.1002/hep.31724] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/01/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Primary biliary cholangitis (PBC) is a prototypical organ-specific autoimmune disease that is mediated by autoreactive T-cell attack and destruction of cholangiocytes. Despite the clear role of autoimmunity in PBC, immune-directed therapies have failed to halt PBC, including biologic therapies effective in other autoimmune diseases. MicroRNA (miRNA) dysregulation is implicated in the pathogenesis (PBC). In the dominant-negative TGF-β receptor type II (dnTGFβRII) mouse model of PBC, autoreactive CD8 T cells play a major pathogenic role and demonstrate a striking pattern of miRNA down-regulation. Enoxacin is a small molecule fluoroquinolone that enhances miRNA biogenesis, partly by stabilizing the interaction of transactivation response RNA-binding protein with Argonaute (Ago) 2. APPROACH AND RESULTS We hypothesized that correcting aberrant T-cell miRNA expression with enoxacin in dnTGFβRII mice could modulate autoreactive T-cell function and prevent PBC. Here, we show that liver-infiltrating dnTGFβRII CD8 T cells have significantly decreased levels of the miRNA biogenesis molecules prolyl 4-hydroxylase subunit alpha 1 (P4HA1) and Ago2 along with significantly increased levels of granzyme B and perforin. Enoxacin treatment significantly up-regulated miRNAs in dnTGFβRII CD8 T cells and effectively treated autoimmune cholangitis in dnTGFβRII mice. Enoxacin treatment directly altered T cells both ex vivo and in vitro, resulting in altered memory subset numbers, decreased proliferation, and decreased interferon-γ production. Enoxacin significantly decreased CD8 T-cell expression of the transcription factor, Runx3, and significantly decreased perforin expression at both the mRNA and protein levels. CONCLUSIONS Enoxacin increases miRNA expression in dnTGFβRII CD8 T cells, reduces CD8 T-cell pathogenicity, and effectively halted progression of autoimmune biliary disease. Targeting the miRNA pathway is a therapeutic approach to autoimmunity that corrects pathological miRNA abnormalities in autoreactive T cells.
Collapse
Affiliation(s)
- Arata Itoh
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - David Adams
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Wenting Huang
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Yuehong Wu
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Kritika Kachapati
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Kyle J Bednar
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH
| | - Patrick S C Leung
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California, DavisDavisCA
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California, DavisDavisCA
| | - Richard A Flavell
- Department of Internal MedicineYale University School of MedicineNew HavenCT
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California, DavisDavisCA
| | - William M Ridgway
- Division of Immunology, Allergy and RheumatologyUniversity of Cincinnati College of MedicineCincinnatiOH.,Division of Rheumatology, Allergy and Clinical ImmunologyUniversity of California, DavisDavisCA
| |
Collapse
|
35
|
Pálfi P, Bakacsy L, Kovács H, Szepesi Á. Hypusination, a Metabolic Posttranslational Modification of eIF5A in Plants during Development and Environmental Stress Responses. PLANTS 2021; 10:plants10071261. [PMID: 34206171 PMCID: PMC8309165 DOI: 10.3390/plants10071261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/15/2021] [Accepted: 06/21/2021] [Indexed: 12/25/2022]
Abstract
Hypusination is a unique posttranslational modification of eIF5A, a eukaryotic translation factor. Hypusine is a rare amino acid synthesized in this process and is mediated by two enzymes, deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase (DOHH). Despite the essential participation of this conserved eIF5A protein in plant development and stress responses, our knowledge of its proper function is limited. In this review, we demonstrate the main findings regarding how eIF5A and hypusination could contribute to plant-specific responses in growth and stress-related processes. Our aim is to briefly discuss the plant-specific details of hypusination and decipher those signal pathways which can be effectively modified by this process. The diverse functions of eIF5A isoforms are also discussed in this review.
Collapse
|
36
|
The iron chaperone and nucleic acid-binding activities of poly(rC)-binding protein 1 are separable and independently essential. Proc Natl Acad Sci U S A 2021; 118:2104666118. [PMID: 34161287 DOI: 10.1073/pnas.2104666118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Poly(rC)-binding protein (PCBP1) is a multifunctional adaptor protein that can coordinate single-stranded nucleic acids and iron-glutathione complexes, altering the processing and transfer of these ligands through interactions with other proteins. Multiple phenotypes are ascribed to cells lacking PCBP1, but the relative contribution of RNA, DNA, or iron chaperone activity is not consistently clear. Here, we report the identification of amino acid residues required for iron coordination on each structural domain of PCBP1 and confirm the requirement of iron coordination for binding target proteins BolA2 and ferritin. We further construct PCBP1 variants that lack either nucleic acid- or iron-binding activity and examine their functions in human cells and mouse tissues depleted of endogenous PCBP1. We find that these activities are separable and independently confer essential functions. While iron chaperone activity controls cell cycle progression and suppression of DNA damage, RNA/DNA-binding activity maintains cell viability in both cultured cell and mouse models. The coevolution of RNA/DNA binding and iron chaperone activities on a single protein may prove advantageous for nucleic acid processing that depends on enzymes with iron cofactors.
Collapse
|
37
|
Yuan C, Chen M, Cai X. Advances in poly(rC)-binding protein 2: Structure, molecular function, and roles in cancer. Biomed Pharmacother 2021; 139:111719. [PMID: 34233389 DOI: 10.1016/j.biopha.2021.111719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 02/08/2023] Open
Abstract
Poly(rC)-binding protein 2 (PCBP2) is an RNA-binding protein that is characterized by its ability to interact with poly(C) with high affinity in a sequence-specific manner. PCBP2 contains three K homology domains, which are consensus RNA-binding domains that play a role in recognizing and combining with RNA and DNA. The specific structure and localization of PCBP2 lay the foundation for its multiple roles in transcriptional, posttranscriptional, and translational processes, even in iron metabolism. Numerous studies have indicated that PCBP2 expression is increased in many cancer types. PCBP2 is considered as an oncogene that promotes tumorigenesis, development of cancer cells, and metastasis. Here, we summarized the current evidence regarding PCBP2 in the proliferation, migration, invasion of cancer cells, and drug resistance, aiming to clarify the molecular mechanisms of PCBP2 in cancer. Results from this review suggest that an in-depth study of PCBP2 in cancer may provide novel biomarkers for prognostic or therapeutic purposes.
Collapse
Affiliation(s)
- Chendong Yuan
- Department of Vascular Surgery, Zhuji Affiliated Hospital of Shaoxing University, Zhuji, Zhejiang 311800, China.
| | - Mingxiang Chen
- Department of Cardiovascular surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, Yubei 401120, China.
| | - Xiaolu Cai
- Department of Oncological Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
38
|
Wei Z, Hao C, Huangfu J, Srinivasagan R, Zhang X, Fan X. Aging lens epithelium is susceptible to ferroptosis. Free Radic Biol Med 2021; 167:94-108. [PMID: 33722625 PMCID: PMC8096685 DOI: 10.1016/j.freeradbiomed.2021.02.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/29/2021] [Accepted: 02/07/2021] [Indexed: 12/11/2022]
Abstract
Age-related cataracts (ARC) are the primary cause of blindness worldwide, and oxidative stress is considered the central pathogenesis of age-related cataractogenesis. Interestingly, ample evidence suggests that there is no remarkable apoptosis present in aged and cataractous human lenses despite the profound disruption of redox homeostasis, raising an essential question regarding the existence of other cell death mechanisms. Here we sought to explore the lens epithelial cell's (LEC) susceptibility to ferroptosis after documentation has concluded that aged and cataractous human lenses manifest with increased reactive oxygen species (ROS) formation, elevated lipid peroxidation, and accumulative intracellular redox-active iron, constituting the three hallmarks of ferroptosis during aging and cataractogenesis. Here we show that very low concentrations of system Xc- inhibitor Erastin (0.5 μM) and glutathione peroxidase 4 (GPX4) inhibitor RSL3 (0.1 μM) can drastically induce human LEC (FHL124) ferroptosis in vitro and mouse lens epithelium ferroptosis ex vivo. Depletion of intracellular glutathione (GSH) in human LECs and mouse lens epithelium significantly sensitizes ferroptosis, particularly under RSL3 challenge. Intriguingly, both human LECs and the mouse lens epithelium demonstrate an age-related sensitization of ferroptosis. Transcriptome analysis indicates that clusters of genes are up-or down-regulated in aged LECs, impacting cellular redox and iron homeostases, such as downregulation of both cystine/glutamate antiporter subunits SLC7A11 and SLC3A2 and iron exporter ferroportin (SLC40A1). Here, for the first time, we are suggesting that LECs are highly susceptible to ferroptosis. Moreover, aged and cataractous human lenses may possess more pro-ferroptotic criteria than any other organ in the human body.
Collapse
Affiliation(s)
- Zongbo Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Caili Hao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Jingru Huangfu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia; Department of Ophthalmology, Chongqing Medical University, Chongqing, China
| | - Ramkumar Srinivasagan
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
| | - Xiang Zhang
- Genomics, Epigenomics and Sequencing Core, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Xingjun Fan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia.
| |
Collapse
|
39
|
Protchenko O, Baratz E, Jadhav S, Li F, Shakoury-Elizeh M, Gavrilova O, Ghosh MC, Cox JE, Maschek JA, Tyurin VA, Tyurina YY, Bayir H, Aron AT, Chang CJ, Kagan VE, Philpott CC. Iron Chaperone Poly rC Binding Protein 1 Protects Mouse Liver From Lipid Peroxidation and Steatosis. Hepatology 2021; 73:1176-1193. [PMID: 32438524 PMCID: PMC8364740 DOI: 10.1002/hep.31328] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/06/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Iron is essential yet also highly chemically reactive and potentially toxic. The mechanisms that allow cells to use iron safely are not clear; defects in iron management are a causative factor in the cell-death pathway known as ferroptosis. Poly rC binding protein 1 (PCBP1) is a multifunctional protein that serves as a cytosolic iron chaperone, binding and transferring iron to recipient proteins in mammalian cells. Although PCBP1 distributes iron in cells, its role in managing iron in mammalian tissues remains open for study. The liver is highly specialized for iron uptake, utilization, storage, and secretion. APPROACH AND RESULTS Mice lacking PCBP1 in hepatocytes exhibited defects in liver iron homeostasis with low levels of liver iron, reduced activity of iron enzymes, and misregulation of the cell-autonomous iron regulatory system. These mice spontaneously developed liver disease with hepatic steatosis, inflammation, and degeneration. Transcriptome analysis indicated activation of lipid biosynthetic and oxidative-stress response pathways, including the antiferroptotic mediator, glutathione peroxidase type 4. Although PCBP1-deleted livers were iron deficient, dietary iron supplementation did not prevent steatosis; instead, dietary iron restriction and antioxidant therapy with vitamin E prevented liver disease. PCBP1-deleted hepatocytes exhibited increased labile iron and production of reactive oxygen species (ROS), were hypersensitive to iron and pro-oxidants, and accumulated oxidatively damaged lipids because of the reactivity of unchaperoned iron. CONCLUSIONS Unchaperoned iron in PCBP1-deleted mouse hepatocytes leads to production of ROS, resulting in lipid peroxidation (LPO) and steatosis in the absence of iron overload. The iron chaperone activity of PCBP1 is therefore critical for limiting the toxicity of cytosolic iron and may be a key factor in preventing the LPO that triggers the ferroptotic cell-death pathway.
Collapse
Affiliation(s)
| | - Ethan Baratz
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD
| | | | - Fengmin Li
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD
| | | | | | - Manik C. Ghosh
- Section on Human Iron Metabolism, NICHD, NIH, Bethesda, MD
| | - James E. Cox
- Deparment of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - J. Alan Maschek
- Deparment of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT
| | - Vladimir A. Tyurin
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Yulia Y. Tyurina
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Hülya Bayir
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Allegra T. Aron
- Department of Chemistry, University of California, Berkeley, CA
| | | | - Valerian E. Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | | |
Collapse
|
40
|
The influence of overnight orthokeratology on ocular surface and dry eye-related cytokines IL-17A, IL-6, and PGE2 in children. Cont Lens Anterior Eye 2021; 44:81-88. [DOI: 10.1016/j.clae.2020.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/19/2022]
|
41
|
Zhu H, Aarons J, Peng Q. High spin polarized Fe2 cluster combined with vicinal nonmetallic sites for catalytic ammonia synthesis from a theoretical perspective. Inorg Chem Front 2021. [DOI: 10.1039/d1qi01083b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Compared to other Fen (n > 2) clusters, Fe2 cluster catalysts combined with vicinal nonmetallic sites are expected to be an ideal catalyst for ammonia synthesis with a lower N–H formation (0.47 eV) and N–N dissociation (0.50 eV) energy barrier at the same time.
Collapse
Affiliation(s)
- Hongdan Zhu
- State Key Laboratory and Institute of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Jolyon Aarons
- Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Qian Peng
- State Key Laboratory and Institute of Elemento-Organic Chemistry and Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
42
|
Hsu MY, Mina E, Roetto A, Porporato PE. Iron: An Essential Element of Cancer Metabolism. Cells 2020; 9:cells9122591. [PMID: 33287315 PMCID: PMC7761773 DOI: 10.3390/cells9122591] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cells undergo considerable metabolic changes to foster uncontrolled proliferation in a hostile environment characterized by nutrient deprivation, poor vascularization and immune infiltration. While metabolic reprogramming has been recognized as a hallmark of cancer, the role of micronutrients in shaping these adaptations remains scarcely investigated. In particular, the broad electron-transferring abilities of iron make it a versatile cofactor that is involved in a myriad of biochemical reactions vital to cellular homeostasis, including cell respiration and DNA replication. In cancer patients, systemic iron metabolism is commonly altered. Moreover, cancer cells deploy diverse mechanisms to increase iron bioavailability to fuel tumor growth. Although iron itself can readily participate in redox reactions enabling vital processes, its reactivity also gives rise to reactive oxygen species (ROS). Hence, cancer cells further rely on antioxidant mechanisms to withstand such stress. The present review provides an overview of the common alterations of iron metabolism occurring in cancer and the mechanisms through which iron promotes tumor growth.
Collapse
Affiliation(s)
- Myriam Y. Hsu
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy; (M.Y.H.); (E.M.)
| | - Erica Mina
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy; (M.Y.H.); (E.M.)
| | - Antonella Roetto
- Department of Clinical and Biological Science, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy
- Correspondence: (A.R.); (P.E.P.)
| | - Paolo E. Porporato
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy; (M.Y.H.); (E.M.)
- Correspondence: (A.R.); (P.E.P.)
| |
Collapse
|
43
|
Bae DH, Gholam Azad M, Kalinowski DS, Lane DJR, Jansson PJ, Richardson DR. Ascorbate and Tumor Cell Iron Metabolism: The Evolving Story and Its Link to Pathology. Antioxid Redox Signal 2020; 33:816-838. [PMID: 31672021 DOI: 10.1089/ars.2019.7903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Vitamin C or ascorbate (Asc) is a water-soluble vitamin and an antioxidant that is involved in many crucial biological functions. Asc's ability to reduce metals makes it an essential enzyme cofactor. Recent Advances: The ability of Asc to act as a reductant also plays an important part in its overall role in iron metabolism, where Asc induces both nontransferrin-bound iron and transferrin-bound iron uptake at physiological concentrations (∼50 μM). Moreover, Asc has emerged to play an important role in multiple diseases and its effects at pharmacological doses could be important for their treatment. Critical Issues: Asc's role as a regulator of cellular iron metabolism, along with its cytotoxic effects and different roles at pharmacological concentrations, makes it a candidate as an anticancer agent. Ever since the controversy regarding the studies from the Mayo Clinic was finally explained, there has been a renewed interest in using Asc as a therapeutic approach toward cancer due to its minimal side effects. Numerous studies have been able to demonstrate the anticancer activity of Asc through selective oxidative stress toward cancer cells via H2O2 generation at pharmacological concentrations. Studies have demonstrated that Asc's cytotoxic mechanism at concentrations (>1 mM) has been associated with decreased cellular iron uptake. Future Directions: Recent studies have also suggested other mechanisms, such as Asc's effects on autophagy, polyamine metabolism, and the cell cycle. Clearly, more has yet to be discovered about Asc's mechanism of action to facilitate safe and effective treatment options for cancer and other diseases.
Collapse
Affiliation(s)
- Dong-Hun Bae
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Mahan Gholam Azad
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Danuta S Kalinowski
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Darius J R Lane
- The Florey Institute of Neuroscience and Mental Health, Melbourne Dementia Research Centre, The University of Melbourne, Parkville, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney, Sydney, Australia.,Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Japan
| |
Collapse
|
44
|
Solyman M, Brayton KA, Shaw DK, Omsland A, McGeehan S, Scoles GA, Noh SM. Predicted iron metabolism genes in hard ticks and their response to iron reduction in Dermacentor andersoni cells. Ticks Tick Borne Dis 2020; 12:101584. [PMID: 33059171 DOI: 10.1016/j.ttbdis.2020.101584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/28/2022]
Abstract
For most organisms, iron is an essential nutrient due to its role in fundamental cellular processes. Insufficient iron causes sub-optimal metabolism with potential effects on viability, while high levels of iron are toxic due to the formation of oxidative radicals, which damage cellular components. Many molecules and processes employed in iron uptake, storage, transport and metabolism are conserved, however significant knowledge gaps remain regarding these processes in ticks due to their unique physiology. In this study, we first identified and sequenced 13 genes likely to be involved in iron metabolism in Dermacentor andersoni cells. We then developed a method to reduce iron levels in D. andersoni cells using the iron chelator 2,2'-bipyridyl and measured the transcriptional response of these genes to iron reduction. The genes include a putative transferrin receptor, divalent metal transporter 1, duodenal cytochrome b, zinc/iron transporters zip7, zip13, zip14, mitoferrin, ferrochelatase, iron regulatory protein 1, ferritin1, ferritin2, transferrin and poly r(C)-binding protein. Overall, the transcriptional response of the target genes to iron reduction was modest. The most marked changes were a decrease in ferritin2, which transports iron through the tick hemolymph, the mitochondrial iron transporter mitoferrin, and the mitochondrial enzyme ferrochelatase. Iron regulatory protein1 was the only gene with an overall increase in transcript in response to reduced iron levels. This work lays the foundation for an improved understanding of iron metabolism in ticks which may provide molecular targets for the development of novel tick control methods and aid in the understanding of tick-pathogen interactions.
Collapse
Affiliation(s)
- Muna Solyman
- Department of Veterinary Microbiology and Pathology, P. O. Box 647040, Washington State University, Pullman, Wash. 99164, USA.
| | - Kelly A Brayton
- Department of Veterinary Microbiology and Pathology, P. O. Box 647040, Washington State University, Pullman, Wash. 99164, USA.
| | - Dana K Shaw
- Department of Veterinary Microbiology and Pathology, P. O. Box 647040, Washington State University, Pullman, Wash. 99164, USA.
| | - Anders Omsland
- The Paul G. Allen School for Global Animal Health, 1155 College Ave., Washington State University, Pullman, Wash. 99164-70403, USA.
| | - Steven McGeehan
- Analytical Sciences Laboratory, University of Idaho, Moscow, ID 83844-2293, USA.
| | - Glen A Scoles
- Animal Diseases Research Unit, USDA-ARS, 3003 ADBF, Pullman, Wash. 99164-6630, USA.
| | - Susan M Noh
- Animal Diseases Research Unit, USDA-ARS, 3003 ADBF, Pullman, Wash. 99164-6630, USA.
| |
Collapse
|
45
|
Philpott CC, Patel SJ, Protchenko O. Management versus miscues in the cytosolic labile iron pool: The varied functions of iron chaperones. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118830. [PMID: 32835748 DOI: 10.1016/j.bbamcr.2020.118830] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 01/01/2023]
Abstract
Iron-containing proteins rely on the incorporation of a set of iron cofactors for activity. The cofactors must be synthesized or assembled from raw materials located within the cell. The chemical nature of this pool of raw material - referred to as the labile iron pool - has become clearer with the identification of micro- and macro-molecules that coordinate iron within the cell. These molecules function as a buffer system for the management of intracellular iron and are the focus of this review, with emphasis on the major iron chaperone protein coordinating the labile iron pool: poly C-binding protein 1.
Collapse
Affiliation(s)
| | - Sarju J Patel
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD, USA
| | - Olga Protchenko
- Genetics and Metabolism Section, NIDDK, NIH, Bethesda, MD, USA
| |
Collapse
|
46
|
Fuhrmann DC, Mondorf A, Beifuß J, Jung M, Brüne B. Hypoxia inhibits ferritinophagy, increases mitochondrial ferritin, and protects from ferroptosis. Redox Biol 2020; 36:101670. [PMID: 32810738 PMCID: PMC7452134 DOI: 10.1016/j.redox.2020.101670] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
Abstract
Cellular iron, at the physiological level, is essential to maintain several metabolic pathways, while an excess of free iron may cause oxidative damage and/or provoke cell death. Consequently, iron homeostasis has to be tightly controlled. Under hypoxia these regulatory mechanisms for human macrophages are not well understood. Hypoxic primary human macrophages reduced intracellular free iron and increased ferritin expression, including mitochondrial ferritin (FTMT), to store iron. In parallel, nuclear receptor coactivator 4 (NCOA4), a master regulator of ferritinophagy, decreased and was proven to directly regulate FTMT expression. Reduced NCOA4 expression resulted from a lower rate of hypoxic NCOA4 transcription combined with a micro RNA 6862-5p-dependent degradation of NCOA4 mRNA, the latter being regulated by c-jun N-terminal kinase (JNK). Pharmacological inhibition of JNK under hypoxia increased NCOA4 and prevented FTMT induction. FTMT and ferritin heavy chain (FTH) cooperated to protect macrophages from RSL-3-induced ferroptosis under hypoxia as this form of cell death is linked to iron metabolism. In contrast, in HT1080 fibrosarcome cells, which are sensitive to ferroptosis, NCOA4 and FTMT are not regulated. Our study helps to understand mechanisms of hypoxic FTMT regulation and to link ferritinophagy and macrophage sensitivity to ferroptosis. Hypoxia decreases NCOA4 transcription in primary human macrophages. NCOA4 mRNA is a target of miR-6862-5p. Lowering NCOA4 increases FTMT abundance under hypoxia. FTMT and FTH protect from ferroptosis. Tumor cells lack the hypoxic decrease of NCOA4 and fail to stabilize FTMT.
Collapse
Affiliation(s)
- Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Antonia Mondorf
- Department of Internal Medicine 1, University Hospital Frankfurt, Germany
| | - Josefine Beifuß
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany; Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany; Branch for Translational Medicine and Pharmacology TMP of the Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt, Germany.
| |
Collapse
|
47
|
Bae DH, Lane DJR, Siafakas AR, Sutak R, Paluncic J, Huang MLH, Jansson PJ, Rahmanto YS, Richardson DR. Acireductone dioxygenase 1 (ADI1) is regulated by cellular iron by a mechanism involving the iron chaperone, PCBP1, with PCBP2 acting as a potential co-chaperone. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165844. [PMID: 32480040 DOI: 10.1016/j.bbadis.2020.165844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022]
Abstract
The iron-containing protein, acireductone dioxygenase 1 (ADI1), is a dioxygenase important for polyamine synthesis and proliferation. Using differential proteomics, the studies herein demonstrated that ADI1 was significantly down-regulated by cellular iron depletion. This is important, since ADI1 contains a non-heme, iron-binding site critical for its activity. Examination of multiple human cell-types demonstrated a significant decrease in ADI1 mRNA and protein after incubation with iron chelators. The decrease in ADI1 after iron depletion was reversible upon incubation of cells with the iron salt, ferric ammonium citrate (FAC). A significant decrease in ADI1 mRNA levels was observed after 14 h of iron depletion. In contrast, the chelator-mediated reduction in ADI1 protein occurred earlier after 10 h of iron depletion, suggesting additional post-transcriptional regulation. The proteasome inhibitor, MG-132, prevented the iron chelator-mediated decrease in ADI1 expression, while the lysosomotropic agent, chloroquine, had no effect. These results suggest an iron-dependent, proteasome-mediated, degradation mechanism. Poly r(C)-binding protein (PCBPs) 1 and 2 act as iron delivery chaperones to other iron-containing dioxygenases and were shown herein for the first time to be regulated by iron levels. Silencing of PCBP1, but not PCBP2, led to loss of ADI1 expression. Confocal microscopy co-localization studies and proximity ligation assays both demonstrated decreased interaction of ADI1 with PCBP1 and PCBP2 under conditions of iron depletion using DFO. These data indicate PCBP1 and PCBP2 interact with ADI1, but only PCBP1 plays a role in ADI1 expression. In fact, PCBP2 appeared to play an accessory role, being involved as a potential co-chaperone.
Collapse
Affiliation(s)
- Dong-Hun Bae
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Darius J R Lane
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia; Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Aritee R Siafakas
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Robert Sutak
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia; BIOCEV - Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University, Průmyslová 595, 252 50 Vestec, Czech Republic
| | - Jasmina Paluncic
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Michael L H Huang
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia; Cancer Drug Resistance Program, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Yohan Suryo Rahmanto
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Centre for Cancer Cell Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia.
| |
Collapse
|
48
|
Nguyen TQ, Kim JE, Brawley HN, Lindahl PA. Chromatographic detection of low-molecular-mass metal complexes in the cytosol of Saccharomyces cerevisiae. Metallomics 2020; 12:1094-1105. [PMID: 32301942 PMCID: PMC7497409 DOI: 10.1039/c9mt00312f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Fluorescence-based chelators are commonly used to probe labile low-molecular-mass (LMM) metal pools in the cytosol of eukaryotic cells, but such chelators destroy the complexes of interest during detection. The objective of this study was to use chromatography to directly detect such complexes. Towards this end, 47 batches of cytosol were isolated from fermenting S. cerevisiae yeast cells and passed through a 10 kDa cut-off membrane. The metal contents of the cytosol and resulting flow-through solution (FTS) were determined. FTSs were applied to a size-exclusion LC column located in an anaerobic refrigerated glove box. The LC system was coupled to an online inductively-coupled-plasma mass spectrometer (ICP-MS) for detection of individual metals. Iron-detected chromatograms of cytosolic FTSs from WT cells exhibited 2-4 major species with apparent masses between 500-1300 Da. Increasing the iron concentration in the growth medium 40-fold increased the overall intensity of these peaks. Approximately 3 LMM cytosolic copper complexes with apparent masses between 300-1300 Da were also detected; their LC intensities were weak, but these increased with increasing concentrations of copper in the growth medium. Observed higher-mass copper-detected peaks were tentatively assigned to copper-bound metallothioneins Cup1 and Crs5. FTSs from strains in which Cup1 or the Cox17 copper chaperone were deleted altered the distribution of LMM copper complexes. LMM zinc- and manganese-detected species were also present in cytosol, albeit at low concentrations. Supplementing the growth medium with zinc increased the intensity of the zinc peak assigned to Crs5 but the intensities of LMM zinc complexes were unaffected. Phosphorus-detected chromatograms were dominated by peaks at apparent masses 400-800 Da, with minor peaks at 1000-1500 Da in some batches. Sulfur chromatograms contained a low-intensity peak that comigrated with a glutathione standard; quantification suggested a GSH concentration in the cytosol of ca. 13 mM. A second LMM sulfur peak that migrated at an apparent mass of 100 Da was also evident.
Collapse
Affiliation(s)
- Trang Q Nguyen
- Department of Chemistry, Texas A&M University, College Station, TX 77843-3255, USA.
| | | | | | | |
Collapse
|
49
|
O'Doherty C, O'Sullivan F, Henry M, Meleady P, Clynes M, Horgan K, Keenan J, Murphy R. LC-MS proteomic profiling of Caco-2 human intestinal cells exposed to the copper-chelating agent, triethylenetetramine: A preliminary study. Biochem Biophys Res Commun 2020; 524:847-852. [PMID: 32046857 DOI: 10.1016/j.bbrc.2020.01.138] [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: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/01/2022]
Abstract
Homeostasis of metal micronutrients such as copper is tightly regulated to ensure deficiency does not occur while restricting damage resulting from excess accumulation. Using LC-MS the effect on the proteome of intestinal Caco-2 cells of exposure to the chelator triethylenetetramine (TETA) was investigated. Continuous exposure of TETA at 25 μM to Caco-2 cells caused decreased cell yields and morphological changes. These effects were reversed when cells were no longer exposed to TETA. Quantitative proteomic analysis identified 957 mostly low-fold differentially expressed proteins, 41 of these returned towards control Caco-2 expression following recovery. Proteins exhibiting this "reciprocal" behaviour included upregulated deoxyhypusine hydroxylase (DOHH, 15.69- fold), a protein essential for eIF-5A factor hypsuination, a post translational modification responsible for eIF-5A maturation, which in turn is responsible for translation elongation. Exposure to TETA also resulted in 87 proteins, the expression of which was stable and remained differentially expressed following recovery. This study helps to elucidate the stable and transient proteomic effects of TETA exposure in intestinal cells.
Collapse
Affiliation(s)
- Charles O'Doherty
- National Institute for Cellular Biotechnology and SSPC-SFI. Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin, D09 NR58, Ireland.
| | - Finbarr O'Sullivan
- National Institute for Cellular Biotechnology and SSPC-SFI. Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin, D09 NR58, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology and SSPC-SFI. Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin, D09 NR58, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology and SSPC-SFI. Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin, D09 NR58, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology and SSPC-SFI. Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin, D09 NR58, Ireland
| | - Karina Horgan
- Alltech Ireland, European Bioscience Centre, Summerhill Rd, Sarney, Dunboyne, Co. Meath, Ireland
| | - Joanne Keenan
- National Institute for Cellular Biotechnology and SSPC-SFI. Centre for Pharmaceuticals, Dublin City University, Glasnevin, Dublin, D09 NR58, Ireland
| | - Richard Murphy
- Alltech Ireland, European Bioscience Centre, Summerhill Rd, Sarney, Dunboyne, Co. Meath, Ireland
| |
Collapse
|
50
|
Bayır H, Anthonymuthu TS, Tyurina YY, Patel SJ, Amoscato AA, Lamade AM, Yang Q, Vladimirov GK, Philpott CC, Kagan VE. Achieving Life through Death: Redox Biology of Lipid Peroxidation in Ferroptosis. Cell Chem Biol 2020; 27:387-408. [PMID: 32275865 DOI: 10.1016/j.chembiol.2020.03.014] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/02/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022]
Abstract
Redox balance is essential for normal brain, hence dis-coordinated oxidative reactions leading to neuronal death, including programs of regulated death, are commonly viewed as an inevitable pathogenic penalty for acute neuro-injury and neurodegenerative diseases. Ferroptosis is one of these programs triggered by dyshomeostasis of three metabolic pillars: iron, thiols, and polyunsaturated phospholipids. This review focuses on: (1) lipid peroxidation (LPO) as the major instrument of cell demise, (2) iron as its catalytic mechanism, and (3) thiols as regulators of pro-ferroptotic signals, hydroperoxy lipids. Given the central role of LPO, we discuss the engagement of selective and specific enzymatic pathways versus random free radical chemical reactions in the context of the phospholipid substrates, their biosynthesis, intracellular location, and related oxygenating machinery as participants in ferroptotic cascades. These concepts are discussed in the light of emerging neuro-therapeutic approaches controlling intracellular production of pro-ferroptotic phospholipid signals and their non-cell-autonomous spreading, leading to ferroptosis-associated necroinflammation.
Collapse
Affiliation(s)
- Hülya Bayır
- Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Tamil S Anthonymuthu
- Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sarju J Patel
- Genetics and Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Andrew A Amoscato
- Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Andrew M Lamade
- Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Qin Yang
- Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Georgy K Vladimirov
- Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Caroline C Philpott
- Genetics and Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD 20892, USA
| | - Valerian E Kagan
- Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA; Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA 15213, USA; Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia.
| |
Collapse
|