101
|
Li Z, Jiang W, Chu H, Ge J, Wang X, Jiang J, Xiao Q, Meng Q, Hao W, Wei X. Exploration of potential mechanism of interleukin-33 up-regulation caused by 1,4-naphthoquinone black carbon in RAW264.7 cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 835:155357. [PMID: 35452731 DOI: 10.1016/j.scitotenv.2022.155357] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/30/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND As air pollution has been paid more attention to by public in recent years, effects and mechanism in particulate matter-triggered health problems become a focus of research. Lysosomes and mitochondria play an important role in regulation of inflammation. Interleukin-33 (IL-33) has been proved to promote inflammation in our previous studies. In this research, macrophage cell line RAW264.7 was used to explore the potential mechanism of upregulation of IL-33 induced by 1,4-naphthoquinone black carbon (1,4-NQ-BC), and to explore changes of lysosomes and mitochondria during the process. RESULTS 50 μg/mL 1,4-NQ-BC exposure for 24 h dramatically increased expression of IL-33 in RAW264.7 cells. Lysosomal membrane permeability was damaged by 1,4-NQ-BC treatment, and higher mitochondrial membrane potential and ROS level were induced by 1,4-NQ-BC. The results of proteomics suggested that expression of ferritin light chain was increased after cells were challenged with 1,4-NQ-BC, and it was verified by Western blot. Meanwhile, expressions of p62 and LC3B-II were increased by 50 μg/mL 1,4-NQ-BC in RAW264.7 cells. Ultimately, expression of IL-33 could return to same level as control in cells treated with 50 μg/mL 1,4-NQ-BC and 50 μM deferoxamine combined. CONCLUSIONS 1,4-NQ-BC induces IL-33 upregulation in RAW264.7 cells, and it is responsible for higher lysosomal membrane permeability and ROS level, lower mitochondrial membrane potential, and inhibition of autophagy. Ferritin light chain possibly plays an important role in the upregulation of IL-33 evoked by 1,4-NQ-BC.
Collapse
Affiliation(s)
- Zekang Li
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Wanyu Jiang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, PR China; Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, PR China
| | - Jianhong Ge
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Xiaoyun Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Jianjun Jiang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Qianqian Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Qinghe Meng
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, PR China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, PR China.
| |
Collapse
|
102
|
Sharawy N, Imam AAA, Aboulhoda BE, Khalifa MM, Morcos GNB, Abd Algaleel WA, Moustafa PE, Abdelbaset MA, Shoukry T. Iron dyshomeostasis and time-course changes in iron-uptake systems and ferritin level in relation to pro-inflammatory microglia polarization in sepsis-induced encephalopathy. Front Physiol 2022; 13:953206. [PMID: 36035473 PMCID: PMC9413069 DOI: 10.3389/fphys.2022.953206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Encephalopathy is a frequent and lethal consequence of sepsis. Recently, a growing body of evidence has provided important insights into the role of iron dyshomeostasis in the context of inflammation. The molecular mechanisms underlying iron dyshomeostasis and its relationship with macrophage phenotypes are largely unknown. Here, we aimed to characterize the changes in iron-transporter and storage proteins and the microglia phenotype that occur during the course of sepsis, as well as their relationship with sepsis-induced encephalopathy. We used a cecal ligation and puncture (CLP) murine model that closely resembles sepsis-induced encephalopathy. Rats were subjected to CLP or sham laparotomy, then were neurologically assessed at 6 h, 24 h, and 3 days after sepsis induction. The serum and brain were collected for subsequent biochemical, histological, and immunohistochemical assessment. Here, an iron excess was observed at time points that followed the pro-inflammatory macrophage polarization in CLP-induced encephalopathy. Our results revealed that the upregulation of non-transferrin-bound iron uptake (NTBI) and ferritin reduction appeared to be partially responsible for the excess free iron detected within the brain tissues. We further demonstrated that the microglia were shifted toward the pro-inflammatory phenotype, leading to persistent neuro-inflammation and neuronal damage after CLP. Taken together, these findings led us to conclude that sepsis increased the susceptibility of the brain to the iron burden via the upregulation of NTBI and the reduction of ferritin, which was concomitantly and correlatively associated with dominance of pro-inflammatory microglia and could explain the neurological dysfunction observed during sepsis.
Collapse
Affiliation(s)
- Nivin Sharawy
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmad Abdel-Aliem Imam
- Preclinical Sciences, College of Osteopathic Medicine, William Carey University, Hattiesburg, MS, United States
- Faculty of Medicine, Cairo University, Cairo, Egypt
- *Correspondence: Ahmad Abdel-Aliem Imam, ; Basma Emad Aboulhoda,
| | - Basma Emad Aboulhoda
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo, Egypt
- *Correspondence: Ahmad Abdel-Aliem Imam, ; Basma Emad Aboulhoda,
| | - Mohamed Mansour Khalifa
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Human Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - George N. B. Morcos
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Basic Medical Sciences, Faculty of Medicine, King Salman International University, El-Tor, Egypt
| | | | | | | | - Tarek Shoukry
- Department of Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
103
|
Wen H, Hun M, Zhao M, Han P, He Q. Serum ferritin as a crucial biomarker in the diagnosis and prognosis of intravenous immunoglobulin resistance and coronary artery lesions in Kawasaki disease: A systematic review and meta-analysis. Front Med (Lausanne) 2022; 9:941739. [PMID: 36035423 PMCID: PMC9399505 DOI: 10.3389/fmed.2022.941739] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/11/2022] [Indexed: 12/15/2022] Open
Abstract
Background Early identification and treatment are paramount for intravenous immunoglobulin (IVIG) resistance and coronary artery lesions (CALs) in patients with Kawasaki disease (KD). Unfortunately, there is no single crucial biomarker to identify these patients in a timely manner, which makes KD the most common cause of acquired heart disease in children in developed countries. Recently, many studies have focused on the association between serum ferritin (SF), IVIG resistance, and CALs in KD. We thus performed a systematic review and meta-analysis to ascertain the diagnostic and prognostic values of SF in predicting IVIG resistance and CALs in KD in the acute phase. Methods The pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and area under the receiver operating characteristic curve (AUC) were extracted from the data to evaluate the SF levels in KD. The hazard ratios (HRs) of related risk factors and their corresponding 95% confidence intervals (CIs) were applied to compute the pooled assessments of the outcomes. Results A total of 11 eligible articles were included in this meta-analysis, including twenty studies for diagnosis and five studies for prognosis. In terms of diagnostic values, SF could identify KD patients in the overall studies with a relatively high pooled sensitivity, specificity, PLR, NLR, DOR, and AUC of 0.76 (95% CI: 0.69-0.82), 0.82 (95% CI: 0.76-0.88), 4.33 (95% CI: 3.07-6.11), 0.29 (95% CI: 0.22-0.38), 15.0 (95% CI: 9.00-25.00), and 0.86 (95% CI: 0.83-0.89), respectively. In studies comparing KD patients and controls, there were a relatively high pooled sensitivity, specificity, PLR, NLR, DOR, and AUC of 0.79 (95% CI: 0.72-0.84), 0.84 (95% CI: 0.79-0.91), 4.61 (95% CI: 3.27-6.51), 0.26 (95% CI: 0.20-0.34), 20.82 (95% CI: 11.83-36.64), and 0.89 (95% CI: 0.86-0.91), respectively. For the prognostic values, we found poor survival outcomes based on KD patients (HR = 1.31, 95% CI: 1.07-1.59, P = 0.008). Conclusion Our meta-analysis suggests that SF may be used as a workable and critical biomarker for the diagnosis and prognosis of IVIG resistance and CALs in patients with KD. We also propose that maintaining the dynamic balance between iron, SF, and ferroptosis will be an important therapeutic strategy to reduce the morbidity of CALs. Systematic review registration [https://www.crd.york.ac.uk/prospero/], identifier [CRD42022279157].
Collapse
Affiliation(s)
- Huai Wen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Marady Hun
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Phanna Han
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qingnan He
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
104
|
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: 13] [Impact Index Per Article: 4.3] [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
|
105
|
David S, Jhelum P, Ryan F, Jeong SY, Kroner A. Dysregulation of Iron Homeostasis in the Central Nervous System and the Role of Ferroptosis in Neurodegenerative Disorders. Antioxid Redox Signal 2022; 37:150-170. [PMID: 34569265 DOI: 10.1089/ars.2021.0218] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Iron accumulation occurs in the central nervous system (CNS) in a variety of neurological conditions as diverse as spinal cord injury, stroke, multiple sclerosis, Parkinson's disease, and others. Iron is a redox-active metal that gives rise to damaging free radicals if its intracellular levels are not controlled or if it is not properly sequestered within cells. The accumulation of iron occurs due to dysregulation of mechanisms that control cellular iron homeostasis. Recent Advances: The molecular mechanisms that regulate cellular iron homeostasis have been revealed in much detail in the past three decades, and new advances continue to be made. Understanding which aspects of iron homeostasis are dysregulated in different conditions will provide insights into the causes of iron accumulation and iron-mediated tissue damage. Recent advances in iron-dependent lipid peroxidation leading to cell death, called ferroptosis, has provided useful insights that are highly relevant for the lipid-rich environment of the CNS. Critical Issues: This review examines the mechanisms that control normal cellular iron homeostasis, the dysregulation of these mechanisms in neurological disorders, and more recent work on how iron can induce tissue damage via ferroptosis. Future Directions: Quick and reliable tests are needed to determine if and when ferroptosis contributes to the pathogenesis of neurological disorders. In addition, there is need to develop better druggable agents to scavenge lipid radicals and reduce CNS damage for neurological conditions for which there are currently few effective treatments. Antioxid. Redox Signal. 37, 150-170.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Priya Jhelum
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Fari Ryan
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Suh Young Jeong
- Department of Molecular & Medical Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
106
|
Liu X, Zhang Y, Wu X, Xu F, Ma H, Wu M, Xia Y. Targeting Ferroptosis Pathway to Combat Therapy Resistance and Metastasis of Cancer. Front Pharmacol 2022; 13:909821. [PMID: 35847022 PMCID: PMC9280276 DOI: 10.3389/fphar.2022.909821] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis is an iron-dependent regulated form of cell death caused by excessive lipid peroxidation. This form of cell death differed from known forms of cell death in morphological and biochemical features such as apoptosis, necrosis, and autophagy. Cancer cells require higher levels of iron to survive, which makes them highly susceptible to ferroptosis. Therefore, it was found to be closely related to the progression, treatment response, and metastasis of various cancer types. Numerous studies have found that the ferroptosis pathway is closely related to drug resistance and metastasis of cancer. Some cancer cells reduce their susceptibility to ferroptosis by downregulating the ferroptosis pathway, resulting in resistance to anticancer therapy. Induction of ferroptosis restores the sensitivity of drug-resistant cancer cells to standard treatments. Cancer cells that are resistant to conventional therapies or have a high propensity to metastasize might be particularly susceptible to ferroptosis. Some biological processes and cellular components, such as epithelial–mesenchymal transition (EMT) and noncoding RNAs, can influence cancer metastasis by regulating ferroptosis. Therefore, targeting ferroptosis may help suppress cancer metastasis. Those progresses revealed the importance of ferroptosis in cancer, In order to provide the detailed molecular mechanisms of ferroptosis in regulating therapy resistance and metastasis and strategies to overcome these barriers are not fully understood, we described the key molecular mechanisms of ferroptosis and its interaction with signaling pathways related to therapy resistance and metastasis. Furthermore, we summarized strategies for reversing resistance to targeted therapy, chemotherapy, radiotherapy, and immunotherapy and inhibiting cancer metastasis by modulating ferroptosis. Understanding the comprehensive regulatory mechanisms and signaling pathways of ferroptosis in cancer can provide new insights to enhance the efficacy of anticancer drugs, overcome drug resistance, and inhibit cancer metastasis.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yiqian Zhang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| | - Fuyan Xu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Ma
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengling Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
- *Correspondence: Yong Xia,
| |
Collapse
|
107
|
Liu Y, Cao X, He C, Guo X, Cai H, Aierken A, Hua J, Peng S. Effects of Ferroptosis on Male Reproduction. Int J Mol Sci 2022; 23:ijms23137139. [PMID: 35806144 PMCID: PMC9267104 DOI: 10.3390/ijms23137139] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is a relatively novel form of regulated cell death that was discovered in 2012. With the increasing research related to the mechanisms of ferroptosis, previous studies have demonstrated that the inactive of the intracellular antioxidant system and iron overload can result in the accumulation of reactive oxygen species (ROS), which can ultimately cause lipid peroxidation in the various cell types of the body. ROS accumulation can cause sperm damage by attacking the plasma membrane and damaging DNA. Acute ferroptosis causes oxidative damage to sperm DNA and testicular oxidative stress, thereby causing male reproductive dysfunction. This review aims to discuss the metabolic network of ferroptosis, summarize and analyze the relationship between male reproductive diseases caused by iron overload as well as lipid peroxidation, and provide a novel direction for the research and prevention of various male reproductive diseases.
Collapse
|
108
|
Abstract
Subcellular compartmentalization is a defining feature of all cells. In prokaryotes, compartmentalization is generally achieved via protein-based strategies. The two main classes of microbial protein compartments are bacterial microcompartments and encapsulin nanocompartments. Encapsulins self-assemble into proteinaceous shells with diameters between 24 and 42 nm and are defined by the viral HK97-fold of their shell protein. Encapsulins have the ability to encapsulate dedicated cargo proteins, including ferritin-like proteins, peroxidases, and desulfurases. Encapsulation is mediated by targeting sequences present in all cargo proteins. Encapsulins are found in many bacterial and archaeal phyla and have been suggested to play roles in iron storage, stress resistance, sulfur metabolism, and natural product biosynthesis. Phylogenetic analyses indicate that they share a common ancestor with viral capsid proteins. Many pathogens encode encapsulins, and recent evidence suggests that they may contribute toward pathogenicity. The existing information on encapsulin structure, biochemistry, biological function, and biomedical relevance is reviewed here.
Collapse
Affiliation(s)
- Tobias W. Giessen
- Departments of Biomedical Engineering and Biological Chemistry, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
109
|
Liu M, Yang C, Chu Q, Wang J, Wang Q, Kong F, Sun G. High serum levels of ferritin may predict poor survival and walking ability for patients with hip fractures: a propensity score matching study. Biomark Med 2022; 16:857-866. [PMID: 35704298 DOI: 10.2217/bmm-2022-0160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To identify the relationship between ferritin and outcomes for patients with hip fractures. Patients & methods: All patients with hip fractures presenting between May 2017 and January 2021 were included. Univariate and multivariate analyses were performed to determine the risk factors for 1-year survival. Propensity score matching (PSM) was performed for groups divided by ferritin levels. Results: A total of 165 patients were included of whom 28 died during the first year after surgery. Ferritin levels differed significantly between groups divided by 1-year survival. High ferritin (≥308.5 ng/ml) was related to poor 1-year survival and 6-month and 1-year independent walking rate. Conclusion: High ferritin (≥308.5 ng/ml) may predict poor survival and free-walking abilities after surgery for patients with hip fractures.
Collapse
Affiliation(s)
- Mingchong Liu
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Chensong Yang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qining Chu
- Emergency Trauma Center, Nanyang Second General Hospital, No 66, East Jianshe Road, Nanyang, 473000, China
| | - Jiansong Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qidong Wang
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fanyu Kong
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Guixin Sun
- Department of Traumatic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
110
|
Liu T, Li L, Cheng C, He B, Jiang T. Emerging prospects of protein/peptide-based nanoassemblies for drug delivery and vaccine development. NANO RESEARCH 2022; 15:7267-7285. [PMID: 35692441 PMCID: PMC9166156 DOI: 10.1007/s12274-022-4385-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 05/09/2023]
Abstract
Proteins have been widely used in the biomedical field because of their well-defined architecture, accurate molecular weight, excellent biocompatibility and biodegradability, and easy-to-functionalization. Inspired by the wisdom of nature, increasing proteins/peptides that possess self-assembling capabilities have been explored and designed to generate nanoassemblies with unique structure and function, including spatially organized conformation, passive and active targeting, stimuli-responsiveness, and high stability. These characteristics make protein/peptide-based nanoassembly an ideal platform for drug delivery and vaccine development. In this review, we focus on recent advances in subsistent protein/peptide-based nanoassemblies, including protein nanocages, virus-like particles, self-assemblable natural proteins, and self-assemblable artificial peptides. The origin and characteristics of various protein/peptide-based assemblies and their applications in drug delivery and vaccine development are summarized. In the end, the prospects and challenges are discussed for the further development of protein/peptide-based nanoassemblies.
Collapse
Affiliation(s)
- Taiyu Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Lu Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Cheng Cheng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| |
Collapse
|
111
|
Li X, Liang J, Qu L, Liu S, Qin A, Liu H, Wang T, Li W, Zou W. Exploring the role of ferroptosis in the doxorubicin-induced chronic cardiotoxicity using a murine model. Chem Biol Interact 2022; 363:110008. [PMID: 35667395 DOI: 10.1016/j.cbi.2022.110008] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/28/2022] [Accepted: 06/01/2022] [Indexed: 11/03/2022]
Abstract
Doxorubicin (DOX) is an effective antitumor drug; however, but its clinical application is seriously limited by the cardiotoxicity induced by its use. Recent studies have found that ferroptosis is an important mechanism underlying DOX-induced cardiotoxicity. However, existing studies are based on DOX-induced acute or subacute cardiotoxicity model. Therefore, we established a murine model of DOX-induced chronic cardiotoxicity using the clinically relevant cumulative dose, to evaluate the potential molecular mechanism underlying ferroptosis of cardiomycocytes. Male C57 mice were received intraperitoneal injections of DOX at a dose of 3 mg/kg body weight, once a week for 12 weeks. We dynamically analysed echocardiographic findings, serum myocardial enzyme levels, haematological indexes and cardiac histopathological changes. The results showed that, after receiving a cumulative DOX dose of 15 mg/kg, the mice developed anaemia and the function and structure of the heart changed significantly with an increase in the cumulative DOX dose. Importantly, with a cumulative DOX dose of 36 mg/kg, iron overload occurred in the heart tissue. Moreover, RNA-sequencing analysis and experimental verification revealed that ferropotosis is the underlying mechanism of DOX-induced chronic cardiotoxicity. Our results showed that DOX inhibits Slc7a11 in system-Xc, resulting in the reduction of GSH synthesis to prevent GPX4 from scavenging lipid peroxides. In addition, DOX induced the occurrence of ferroptosis via down-regulating Nrf2 expression to inhibit HO-1 and GPx4 levels. Our study provides a new perspective for evaluating the pathophysiology of DOX-induced chronic cardiotoxicity in the future, and developing new potential therapeutic strategies for the prevention and treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaofen Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jiyi Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Liping Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Sili Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Anquan Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Honglin Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Tao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
112
|
Wang J, Sun Q, Wang G, Wang H, Liu H. The effects of blunt snout bream (Megalobrama amblycephala) IL-6 trans-signaling on immunity and iron metabolism via JAK/STAT3 pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 131:104372. [PMID: 35217123 DOI: 10.1016/j.dci.2022.104372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 06/14/2023]
Abstract
Interleukin-6 (IL-6) is a pleiotropic inflammatory cytokine, which plays a dual role in mammalian inflammation through both classical signaling (IL-6 binds to IL-6 receptor/IL-6R) and trans-signaling (IL-6 binds to soluble IL-6R). However, the function of IL-6, especially the regulatory mechanism of IL-6 trans-signaling in immunity and iron metabolism remains largely unclear in teleost. Here, L8824 cells (Ctenopharyngodon idella hepatic cells) were stimulated with blunt snout bream (Megalobrama amblycephala) IL-6 combination with sIL-6R protein (rmaIL-6+rmasIL-6R/maIL-6 trans-signaling) or STAT3 inhibitor (c188-9), and RNA-sequencing, global transcriptional analyses. The enrichment analysis of GO and KEGG showed that maIL-6 trans-signaling is mainly involved in stress and inflammation response, and the activation of STAT3 is mainly related to cell proliferation, apoptosis and immune regulation. Furthermore, after treated L8824 cells with JAK2 inhibitors, it was found that the induction of IL-6 trans-signaling on the selected immune-related genes could be inhibited. These results implied that in early stage after rmaIL-6+rmasIL-6R treatment, the maIL-6 trans-signaling played an important role in the immune regulation through the JAK2/STAT3 pathway. By extending the rmaIL-6+rmasIL-6R treatment time, it was found that maIL-6 trans-signaling could promote the expression of iron metabolism related genes (ft, tf, tfr1, hamp and fpn1) in L8824 cells, indicating that maIL-6 trans-signaling may be involved in iron metabolism in the non-acute immune phase. Finally, after treated L8824 cells with JAK2 and STAT3 inhibitors, it was found that only tf and fpn1 were regulated by maIL-6 trans-signaling through the JAK2/STAT3 pathway. These findings provided novel insights into IL-6 trans-signaling regulatory mechanism in teleost, enriching our knowledge of fish immunity and iron metabolism.
Collapse
Affiliation(s)
- Jixiu Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Qianhui Sun
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Guowen Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Huanling Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Hong Liu
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affair / Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China.
| |
Collapse
|
113
|
Girelli D, Busti F, Brissot P, Cabantchik I, Muckenthaler MU, Porto G. Hemochromatosis classification: update and recommendations by the BIOIRON Society. Blood 2022; 139:3018-3029. [PMID: 34601591 PMCID: PMC11022970 DOI: 10.1182/blood.2021011338] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022] Open
Abstract
Hemochromatosis (HC) is a genetically heterogeneous disorder in which uncontrolled intestinal iron absorption may lead to progressive iron overload (IO) responsible for disabling and life-threatening complications such as arthritis, diabetes, heart failure, hepatic cirrhosis, and hepatocellular carcinoma. The recent advances in the knowledge of pathophysiology and molecular basis of iron metabolism have highlighted that HC is caused by mutations in at least 5 genes, resulting in insufficient hepcidin production or, rarely, resistance to hepcidin action. This has led to an HC classification based on different molecular subtypes, mainly reflecting successive gene discovery. This scheme was difficult to adopt in clinical practice and therefore needs revision. Here we present recommendations for unambiguous HC classification developed by a working group of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society), including both clinicians and basic scientists during a meeting in Heidelberg, Germany. We propose to deemphasize the use of the molecular subtype criteria in favor of a classification addressing both clinical issues and molecular complexity. Ferroportin disease (former type 4a) has been excluded because of its distinct phenotype. The novel classification aims to be of practical help whenever a detailed molecular characterization of HC is not readily available.
Collapse
Affiliation(s)
- Domenico Girelli
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Pierre Brissot
- INSERM, Univ-Rennes, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1241, Institut NuMeCan, Rennes, France
| | - Ioav Cabantchik
- Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
| | - Martina U. Muckenthaler
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
| | - Graça Porto
- Institute for Molecular and Cell Biology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Clinical Hematology, Santo António Hospital, Porto University, Porto, Portugal
| | - on behalf of the Nomenclature Committee of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society)
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
- INSERM, Univ-Rennes, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1241, Institut NuMeCan, Rennes, France
- Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
- Institute for Molecular and Cell Biology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Clinical Hematology, Santo António Hospital, Porto University, Porto, Portugal
| |
Collapse
|
114
|
Labra-Muñoz JA, de Reuver A, Koeleman F, Huber M, van der Zant HSJ. Ferritin-Based Single-Electron Devices. Biomolecules 2022; 12:biom12050705. [PMID: 35625632 PMCID: PMC9138424 DOI: 10.3390/biom12050705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 11/26/2022] Open
Abstract
We report on the fabrication of single-electron devices based on horse-spleen ferritin particles. At low temperatures the current vs. voltage characteristics are stable, enabling the acquisition of reproducible data that establishes the Coulomb blockade as the main transport mechanism through them. Excellent agreement between the experimental data and the Coulomb blockade theory is demonstrated. Single-electron charge transport in ferritin, thus, establishes a route for further characterization of their, e.g., magnetic, properties down to the single-particle level, with prospects for electronic and medical applications.
Collapse
Affiliation(s)
- Jacqueline A. Labra-Muñoz
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2300 RA Leiden, The Netherlands;
- Kavli Institute of Nanoscience, Delft University of Technology, Orentzweg 1, 2628 CJ Delft, The Netherlands; (A.d.R.); (F.K.)
- Correspondence: (J.A.L.-M.); (H.S.J.v.d.Z.)
| | - Arie de Reuver
- Kavli Institute of Nanoscience, Delft University of Technology, Orentzweg 1, 2628 CJ Delft, The Netherlands; (A.d.R.); (F.K.)
| | - Friso Koeleman
- Kavli Institute of Nanoscience, Delft University of Technology, Orentzweg 1, 2628 CJ Delft, The Netherlands; (A.d.R.); (F.K.)
| | - Martina Huber
- Department of Physics, Huygens-Kamerlingh Onnes Laboratory, Leiden University, Niels Bohrweg 2, 2300 RA Leiden, The Netherlands;
| | - Herre S. J. van der Zant
- Kavli Institute of Nanoscience, Delft University of Technology, Orentzweg 1, 2628 CJ Delft, The Netherlands; (A.d.R.); (F.K.)
- Correspondence: (J.A.L.-M.); (H.S.J.v.d.Z.)
| |
Collapse
|
115
|
Supplementation with High or Low Iron Reduces Colitis Severity in an AOM/DSS Mouse Model. Nutrients 2022; 14:nu14102033. [PMID: 35631174 PMCID: PMC9147005 DOI: 10.3390/nu14102033] [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: 04/07/2022] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/17/2022] Open
Abstract
The relationship between colitis-associated colorectal cancer (CAC) and the dysregulation of iron metabolism has been implicated. However, studies on the influence of dietary iron deficiency on the incidence of CAC are limited. This study investigated the effects of dietary iron deficiency and dietary non-heme iron on CAC development in an azoxymethane/dextran sodium sulfate (AOM/DSS) mouse model. The four-week-old mice were divided into the following groups: iron control (IC; 35 ppm iron/kg) + normal (NOR), IC + AOM/DSS, iron deficient (ID; <5 ppm iron/kg diet) + AOM/DSS, and iron overload (IOL; approximately 2000 ppm iron/kg) + AOM/DSS. The mice were fed the respective diets for 13 weeks, and the AOM/DSS model was established at week five. FTH1 expression increased in the mice’s colons in the IC + AOM/DSS group compared with that observed in the ID and IOL + AOM/DSS groups. The reduced number of colonic tumors in the ID + AOM/DSS and IOL + AOM/DSS groups was accompanied by the downregulated expression of cell proliferation regulators (PCNA, cyclin D1, and c-Myc). Iron overload inhibited the increase in the expression of NF-κB and its downstream inflammatory cytokines (IL-6, TNFα, iNOS, COX2, and IL-1β), likely due to the elevated expression of antioxidant genes (SOD1, TXN, GPX1, GPX4, CAT, HMOX1, and NQO1). ID + AOM/DSS may hinder tumor development in the AOM/DSS model by inhibiting the PI3K/AKT pathway by increasing the expression of Ndrg1. Our study suggests that ID and IOL diets suppress AOM/DSS-induced tumors and that long-term iron deficiency or overload may negate CAC progression.
Collapse
|
116
|
Kuno S, Fujita H, Tanaka Y, Ogra Y, Iwai K. Iron-induced NCOA4 condensation regulates ferritin fate and iron homeostasis. EMBO Rep 2022; 23:e54278. [PMID: 35318808 PMCID: PMC9066066 DOI: 10.15252/embr.202154278] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022] Open
Abstract
Iron is not only essential but also a toxic trace element. Under iron repletion, ferritin maintains cellular iron homeostasis by storing iron to avoid iron toxicity. Under iron depletion, the ferritin-specific autophagy adaptor NCOA4 delivers ferritin to lysosomes via macroautophagy to enable cells to use stored iron. Here, we show that NCOA4 also plays crucial roles in the regulation of ferritin fate under iron repletion. NCOA4 forms insoluble condensates via multivalent interactions generated by the binding of iron to its intrinsically disordered region. This sequesters NCOA4 away from ferritin and allows ferritin accumulation in the early phase of iron repletion. Under prolonged iron repletion, NCOA4 condensates can deliver ferritin to lysosomes via a TAX1BP1-dependent non-canonical autophagy pathway, thereby preventing relative iron deficiency due to excessive iron storage and reduced iron uptake. Together, these observations suggest that the NCOA4-ferritin axis modulates intracellular iron homeostasis in accordance with cellular iron availability.
Collapse
Affiliation(s)
- Sota Kuno
- Department of Molecular and Cellular PhysiologyGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroaki Fujita
- Department of Molecular and Cellular PhysiologyGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Yu‐ki Tanaka
- Laboratory of Toxicology and Environmental HealthGraduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Yasumitsu Ogra
- Laboratory of Toxicology and Environmental HealthGraduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Kazuhiro Iwai
- Department of Molecular and Cellular PhysiologyGraduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
117
|
Light and heavy ferritin chain expression in the liver and kidneys of Wistar rats: aging, sex differences, and impact of gonadectomy. Arh Hig Rada Toksikol 2022; 73:48-61. [PMID: 35390238 PMCID: PMC8999590 DOI: 10.2478/aiht-2022-73-3621] [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: 12/01/2021] [Accepted: 03/01/2022] [Indexed: 12/02/2022] Open
Abstract
Ferritin is the main intracellular storage of iron. Animal studies show that female liver and kidney express more ferritin and accumulate more iron than male. However, no study so far has investigated sex and age differences in light (FtL) and heavy (FtH) ferritin chain expression. To address this, we relied on specific antibodies and immunochemical methods to analyse the expression of both ferritin chains in the liver and kidney of 3-month and 2-year-old male and female Wistar rats. To see how sex hormones may affect expression we also studied adult animals gonadectomised at the age of 10 weeks. FtL and FtH were more expressed in both organs of female rats, while gonadectomy increased the expression in males and decreased it in females, which suggests that it is stimulated by female and inhibited by male steroid hormones. Normal kidney ferritin distribution and change with aging warrant more attention in studies of (patho) physiological and toxicological processes.
Collapse
|
118
|
Song Y, Wang X, Bu X, Huang Q, Qiao F, Chen X, Shi Q, Qin J, Chen L. A Comparation Between Different Iron Sources on Growth Performance, Iron Utilization, Antioxidant Capacity and Non-specific Immunity in Eriocheir sinensis. Anim Feed Sci Technol 2022. [DOI: 10.1016/j.anifeedsci.2022.115300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
119
|
Distinct structural characteristics define a new subfamily of Mycoplasma ferritin. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.03.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
120
|
Hanke N, Rami A. Cerebral ischemia induced iron deposit, ferritin accumulation, nuclear receptor coactivator 4-depletion and ferroptosis. Curr Neurovasc Res 2022; 19:47-60. [PMID: 35319371 DOI: 10.2174/1567202619666220321120954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The neuronal death upon cerebral ischemia shares not only characteristics of necrosis, apoptosis and autophagy, but exhibits also biochemical and morphological characteristics of ferroptosis. Ferroptosis is a regulated form of cell death which is considered to be an oxidative iron-dependent process. It is now commonly accepted that iron and free radicals are considered to cause lipid peroxidation as well as the oxidation of proteins and nucleic acids, leading to increased membrane and enzymatic dysfunction, and finally contributing to cell death. Although ferroptosis was first described in cancer cells, emerging evidence now links mechanisms of ferroptosis to many different diseases, including cerebral ischemia. METHODS The objective of this study was to identify the ferroptosis key players and the underlying biochemical pathways leading to cell death upon focal cerebral ischemia in mice by using immunofluorescence, Western blotting, histochemistry and densitometry. RESULTS In this study, we demonstrated that cerebral ischemia induced iron-deposition, down-regulated dramatically the expression of the glutathione peroxidase 4 (GPX4), decreased the expression of the nuclear receptor coactivator 4 (NCOA4) and induced inappropriate accumulation of ferritin in the ischemic brain. This supports the hypothesis that an ischemic insult may induce ferroptosis through inhibition of GPX4. CONCLUSION We conclude that iron excess following cerebral ischemia leads to cell death despite activation of compensatory mechanisms for iron homeostasis, as illustrated by the accumulation of ferritins. These data emphasize the presence of a cellular mechanism that allows neuronal cells to handle restriction in iron overload.
Collapse
Affiliation(s)
- Nora Hanke
- Institut für Experimentelle Neurobiologie (Anatomie II), Klinikum der Johann Wolfgang von Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| | - Abdelhaq Rami
- Institut für Experimentelle Neurobiologie (Anatomie II), Klinikum der Johann Wolfgang von Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| |
Collapse
|
121
|
Bonilla DA, Moreno Y, Petro JL, Forero DA, Vargas-Molina S, Odriozola-Martínez A, Orozco CA, Stout JR, Rawson ES, Kreider RB. A Bioinformatics-Assisted Review on Iron Metabolism and Immune System to Identify Potential Biomarkers of Exercise Stress-Induced Immunosuppression. Biomedicines 2022; 10:724. [PMID: 35327526 PMCID: PMC8945881 DOI: 10.3390/biomedicines10030724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/01/2022] [Accepted: 03/09/2022] [Indexed: 02/01/2023] Open
Abstract
The immune function is closely related to iron (Fe) homeostasis and allostasis. The aim of this bioinformatics-assisted review was twofold; (i) to update the current knowledge of Fe metabolism and its relationship to the immune system, and (ii) to perform a prediction analysis of regulatory network hubs that might serve as potential biomarkers during stress-induced immunosuppression. Several literature and bioinformatics databases/repositories were utilized to review Fe metabolism and complement the molecular description of prioritized proteins. The Search Tool for the Retrieval of Interacting Genes (STRING) was used to build a protein-protein interactions network for subsequent network topology analysis. Importantly, Fe is a sensitive double-edged sword where two extremes of its nutritional status may have harmful effects on innate and adaptive immunity. We identified clearly connected important hubs that belong to two clusters: (i) presentation of peptide antigens to the immune system with the involvement of redox reactions of Fe, heme, and Fe trafficking/transport; and (ii) ubiquitination, endocytosis, and degradation processes of proteins related to Fe metabolism in immune cells (e.g., macrophages). The identified potential biomarkers were in agreement with the current experimental evidence, are included in several immunological/biomarkers databases, and/or are emerging genetic markers for different stressful conditions. Although further validation is warranted, this hybrid method (human-machine collaboration) to extract meaningful biological applications using available data in literature and bioinformatics tools should be highlighted.
Collapse
Affiliation(s)
- Diego A. Bonilla
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogota 110311, Colombia; (Y.M.); (J.L.P.)
- Research Group in Biochemistry and Molecular Biology, Faculty of Science and Education, Universidad Distrital Francisco José de Caldas, Bogota 110311, Colombia
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
- Sport Genomics Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
| | - Yurany Moreno
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogota 110311, Colombia; (Y.M.); (J.L.P.)
- Research Group in Biochemistry and Molecular Biology, Faculty of Science and Education, Universidad Distrital Francisco José de Caldas, Bogota 110311, Colombia
| | - Jorge L. Petro
- Research Division, Dynamical Business & Science Society—DBSS International SAS, Bogota 110311, Colombia; (Y.M.); (J.L.P.)
- Research Group in Physical Activity, Sports and Health Sciences (GICAFS), Universidad de Córdoba, Montería 230002, Colombia
| | - Diego A. Forero
- Health and Sport Sciences Research Group, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia; (D.A.F.); (C.A.O.)
| | - Salvador Vargas-Molina
- Faculty of Sport Sciences, EADE-University of Wales Trinity Saint David, 29018 Málaga, Spain;
| | - Adrián Odriozola-Martínez
- Sport Genomics Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain;
- kDNA Genomics, Joxe Mari Korta Research Center, University of the Basque Country UPV/EHU, 20018 Donostia, Spain
| | - Carlos A. Orozco
- Health and Sport Sciences Research Group, School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá 111221, Colombia; (D.A.F.); (C.A.O.)
| | - Jeffrey R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, University of Central Florida, Orlando, FL 32816, USA;
| | - Eric S. Rawson
- Department of Health, Nutrition and Exercise Science, Messiah University, Mechanicsburg, PA 17055, USA;
| | - Richard B. Kreider
- Exercise & Sport Nutrition Laboratory, Human Clinical Research Facility, Department of Health & Kinesiology, Texas A&M University, College Station, TX 77843, USA;
| |
Collapse
|
122
|
Upregulation of Nrf2 signaling and suppression of ferroptosis and NF-κB pathway by leonurine attenuate iron overload-induced hepatotoxicity. Chem Biol Interact 2022; 356:109875. [PMID: 35247364 DOI: 10.1016/j.cbi.2022.109875] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/08/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022]
Abstract
Hepatotoxicity is a major health concern that associates the iron overload diseases including hemochromatosis, sickle cell anemia, and thalassemia. Induction of ferroptosis, oxidative stress, and inflammation substantially mediates the iron-evoked hepatotoxicity. The current work investigated the potential protective effect of the natural alkaloid leonurine against the iron-induced hepatotoxicity and elucidated the underlining molecular mechanisms. Male Wistar rats were treated with iron only (30 mg/kg every other day over a ten-day period via intraperitoneal injection) or with iron and leonurine (leonurine: 100 mg/kg/day per oral via gastric gavage for 10 days) to establish the iron-overload model. Liver and blood specimens were then collected and subjected to molecular, biochemical, and histopathological investigations. The results revealed the ability of leonuirne to suppress the iron-induced ferroptosis as reflected by modulation of the ferroptotic biomarkers glutathione peroxidase 4, cyclooxygenase-2, liver iron content, lipid hydroperoxides, and the leakage of the liver intracellular enzymes. Leonurine alleviated the iron-induced oxidative damage and inflammatory response in the liver tissues as indicated by decreased levels of DNA oxidation, lipid peroxidation, and the pro-inflammatory cytokines. In the same context, it improved the antioxidant potential of the liver tissues and ameliorated the iorn-induced histopathological abnormalities. Mechanistically, leonurine enhanced nuclear translocation of the antioxidant transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) and increased protein levels of its downstream targets NAD(P)H-quinone oxidoreductase 1 and heme oxygenase-1. Additionally, it suppressed the nuclear translocation of the inflammatory transcription factor nuclear factor kappa B (NF-κB) and downregulated its downstream pro-inflammatory cytokines tumor necrosis factor-alpha and interleukin-1 beta. The study highlights the hepatoprotective activity of leonurine against the iron-evoked hepatotoxicity that is potentially mediated through modulation of Nrf2 and NF-κB signaling.
Collapse
|
123
|
Singh N, Bhatla SC. Heme oxygenase-nitric oxide crosstalk-mediated iron homeostasis in plants under oxidative stress. Free Radic Biol Med 2022; 182:192-205. [PMID: 35247570 DOI: 10.1016/j.freeradbiomed.2022.02.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 12/22/2022]
Abstract
Plant growth under abiotic stress conditions significantly enhances intracellular generation of reactive oxygen species (ROS). Oxidative status of plant cells is directly affected by the modulation of iron homeostasis. Among mammals and plants, heme oxygenase-1 (HO-1) is a well-known antioxidant enzyme. It catalyzes oxygenation of heme, thereby producing Fe2+, CO and biliverdin as byproducts. The antioxidant potential of HO-1 is primarily due to its catalytic reaction byproducts. Biliverdin and bilirubin possess conjugated π-electrons which escalate the ability of these biomolecules to scavenge free radicals. CO also enhances the ROS scavenging ability of plants cells by upregulating catalase and peroxidase activity. Enhanced expression of HO-1 in plants under oxidative stress accompanies sequestration of iron in specialized iron storage proteins localized in plastids and mitochondria, namely ferritin for Fe3+ storage and frataxin for storage of Fe-S clusters, respectively. Nitric oxide (NO) crosstalks with HO-1 at multiple levels, more so in plants under oxidative stress, in order to maintain intracellular iron status. Formation of dinitrosyl-iron complexes (DNICs) significantly prevents Fenton reaction during oxidative stress. DNICs also release NO upon dissociation in target cells over long distance in plants. They also function as antioxidants against superoxide anions and lipidic free radicals. A number of NO-modulated transcription factors also facilitate iron homeostasis in plant cells. Plants facing oxidative stress exhibit modulation of lateral root formation by HO-1 through NO and auxin-dependent pathways. The present review provides an in-depth analysis of the structure-function relationship of HO-1 in plants and mammals, correlating them with their adaptive mechanisms of survival under stress.
Collapse
Affiliation(s)
- Neha Singh
- Department of Botany, Gargi College, University of Delhi, India.
| | - Satish C Bhatla
- Laboratory of Plant Physiology and Biochemistry, Department of Botany, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
124
|
Karmi O, Rowland L, King SD, Manrique-Acevedo C, Cabantchik IZ, Nechushtai R, Mittler R. The [2Fe-2S] protein CISD2 plays a key role in preventing iron accumulation in cardiomyocytes. FEBS Lett 2022; 596:747-761. [PMID: 34997963 DOI: 10.1002/1873-3468.14277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/11/2021] [Accepted: 12/30/2021] [Indexed: 11/08/2022]
Abstract
Considered a key aging gene, CISD2, encoding CDGSH iron-sulfur domain-containing protein 2, plays a central role in regulating calcium homeostasis, preventing mitochondrial dysfunction, and the activation of autophagy and apoptosis in different cells. Here, we show that cardiomyocytes from CISD2-null mice accumulate high levels of iron and contain high levels of transferrin receptor and ferritin. Using proteomics and transmission electron microscopy, we further show that the lack of CISD2 induces several features of the aging process in young mice, but other features are not induced. Taken together, our findings suggest that CISD2 protects cardiomyocytes from overaccumulation of iron, which is common in aging hearts and can contribute to the pathogenesis of heart failure.
Collapse
Affiliation(s)
- Ola Karmi
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Israel
| | - Linda Rowland
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Skylar D King
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Ioav Z Cabantchik
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Israel
| | - Rachel Nechushtai
- The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Israel
| | - Ron Mittler
- Department of Surgery, University of Missouri School of Medicine, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- The Division of Plant Sciences and Interdisciplinary Plant Group, College of Agriculture, Food and Natural Resources, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
125
|
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
|
126
|
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
|
127
|
Heme oxygenase-1, carbon monoxide, and malaria – The interplay of chemistry and biology. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
128
|
Liang Q, Zhao Q, Hao X, Wang J, Ma C, Xi X, Kang W. The Effect of Flammulina velutipes Polysaccharide on Immunization Analyzed by Intestinal Flora and Proteomics. Front Nutr 2022; 9:841230. [PMID: 35155543 PMCID: PMC8832141 DOI: 10.3389/fnut.2022.841230] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
Proteomics and intestinal flora were used to determine the mechanism of immune modulatory effects of Flammulina velutipes polysaccharide on immunosuppressed mice. The results showed that compared with the model group, F. velutipes polysaccharide could increase thymus and spleen indices and improve thymus tissue structure in mice; IL-2 and IL-4 contents were significantly increased and IL-6 and TNF-α contents were significantly decreased; serum acid phosphatase (ACP), lactate dehydrogenase (LDH) and total antioxidant capacity (T-AOC) activities were increased (P < 0.05); in the liver, superoxide dismutase (SOD) and catalase (CAT) activities were increased (P < 0.001), while malondialdehyde (MDA) content was decreased (P < 0.001). Proteomics discovered that F. velutipes polysaccharides may exert immune modulatory effects by participating in signaling pathways such as immune diseases, transport and catabolism, phagosomes and influenza A, regulating the immune-related proteins Transferrin receptor protein 1 (TFRC) and Radical S-adenosyl methionine domain-containing protein 2 (RSAD2), etc. Gut microbial studies showed that F. velutipes polysaccharides could increase the abundance of intestinal flora and improve the flora structure. Compared to the model group, the content of short-chain fatty acids (SCFAs) and the relative abundance of SCFA-producers Bacteroides and Alloprevotella were increased in the F. velutipes polysaccharide administration group, while Lachnospiraceae_NK4A136_group and f_Lachnospiraceae_Unclassified decreased in relative abundance. Thus, F. velutipes polysaccharide may play an immunomodulatory role by regulating the intestinal environment and improving the balance of flora.
Collapse
Affiliation(s)
- Qiongxin Liang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Qingchun Zhao
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Xuting Hao
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
| | - Jinmei Wang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng, China
- *Correspondence: Changyang Ma
| | - Xuefeng Xi
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- College of Physical Education, Henan University, Kaifeng, China
- Xuefeng Xi
| | - Wenyi Kang
- National R&D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Functional Food Engineering Technology Research Center, Kaifeng, China
- Joint International Research Laboratory of Food & Medicine Resource Function, Kaifeng, China
- Wenyi Kang
| |
Collapse
|
129
|
Moreira AC, Silva T, Mesquita G, Gomes AC, Bento CM, Neves JV, Rodrigues DF, Rodrigues PN, Almeida AA, Santambrogio P, Gomes MS. H-Ferritin Produced by Myeloid Cells Is Released to the Circulation and Plays a Major Role in Liver Iron Distribution during Infection. Int J Mol Sci 2021; 23:ijms23010269. [PMID: 35008695 PMCID: PMC8745395 DOI: 10.3390/ijms23010269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 11/16/2022] Open
Abstract
During infections, the host redistributes iron in order to starve pathogens from this nutrient. Several proteins are involved in iron absorption, transport, and storage. Ferritin is the most important iron storage protein. It is composed of variable proportions of two peptides, the L- and H-ferritins (FTL and FTH). We previously showed that macrophages increase their expression of FTH1 when they are infected in vitro with Mycobacterium avium, without a significant increase in FTL. In this work, we investigated the role of macrophage FTH1 in M. avium infection in vivo. We found that mice deficient in FTH1 in myeloid cells are more resistant to M. avium infection, presenting lower bacterial loads and lower levels of proinflammatory cytokines than wild-type littermates, due to the lower levels of available iron in the tissues. Importantly, we also found that FTH1 produced by myeloid cells in response to infection may be found in circulation and that it plays a key role in iron redistribution. Specifically, in the absence of FTH1 in myeloid cells, increased expression of ferroportin is observed in liver granulomas and increased iron accumulation occurs in hepatocytes. These results highlight the importance of FTH1 expression in myeloid cells for iron redistribution during infection.
Collapse
Affiliation(s)
- Ana C. Moreira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Tânia Silva
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Gonçalo Mesquita
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Ana Cordeiro Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Clara M. Bento
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, 4200-135 Porto, Portugal
| | - João V. Neves
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Daniela F. Rodrigues
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Pedro N. Rodrigues
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Agostinho A. Almeida
- LAQV/REQUIMTE, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal;
| | - Paolo Santambrogio
- Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Maria Salomé Gomes
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (A.C.M.); (T.S.); (A.C.G.); (C.M.B.); (J.V.N.); (D.F.R.); (P.N.R.)
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Correspondence:
| |
Collapse
|
130
|
Fox B, Roberts G, Atkinson E, Rigsby P, Ball C. International collaborative study to evaluate and calibrate two recombinant L chain Ferritin preparations for use as a WHO International Standard. Clin Chem Lab Med 2021; 60:370-378. [PMID: 34939377 DOI: 10.1515/cclm-2021-1139] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/05/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVES To evaluate and calibrate two candidate preparations for the 4th International Standard for Ferritin (Human, Recombinant) (codes: 19/118 and 19/162) against the 3rd International Standard for Ferritin (Human, Recombinant) (code: 94/572), and three serum commutability samples in an international collaborative study involving 12 laboratories in nine countries. METHODS Eleven of the 12 participating laboratories performed Ferritin quantitation using automated assay platforms and one laboratory used a manual ELISA kit. RESULTS There was better overall agreement between all laboratories and between assay methods for the potency of preparation 19/118 than for preparation 19/162. The overall geometric mean potency (from all methods) of the candidate 4th International Standard, 19/118, was 10.5 µg/ampoule, with inter-laboratory variability, expressed as % geometric coefficient of variation (GCV), of 4.7%. Accelerated stability studies have predicted both 19/118 and 19/162 to be very stable for long term storage at -20 °C. CONCLUSIONS The candidate 4th International Standard for Ferritin (Human, Recombinant) (19/118) has been shown to be immunologically similar to the 3rd International Standard for Ferritin (Human, Recombinant) (94/572). It was recommended to and accepted by the WHO Expert Committee on Biological Standardization that 19/118 be established as the 4th International Standard for Ferritin (Human, Recombinant) with an assigned potency of 10.5 µg/ampoule and expanded uncertainty limits 10.2-10.8 µg/ampoule (95% confidence; k=2.23).
Collapse
Affiliation(s)
- Bernard Fox
- Biotherapeutics Division, National Institute for Biological Standards and Control, Hertfordshire, UK
| | - Graham Roberts
- Biotherapeutics Division, National Institute for Biological Standards and Control, Hertfordshire, UK
| | - Eleanor Atkinson
- Division of Analytical and Biological Sciences, National Institute for Biological Standards and Control, Hertfordshire, UK
| | - Peter Rigsby
- Division of Analytical and Biological Sciences, National Institute for Biological Standards and Control, Hertfordshire, UK
| | - Christina Ball
- Biotherapeutics Division, National Institute for Biological Standards and Control, Hertfordshire, UK
| |
Collapse
|
131
|
Sloan MA, Aghabi D, Harding CR. Orchestrating a heist: uptake and storage of metals by apicomplexan parasites. MICROBIOLOGY (READING, ENGLAND) 2021; 167. [PMID: 34898419 PMCID: PMC7612242 DOI: 10.1099/mic.0.001114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Megan A Sloan
- Wellcome Centre for Integrative Parasitology, Institute for Infection, Immunity and Inflammation, University of Glasgow, UK
| | - Dana Aghabi
- Wellcome Centre for Integrative Parasitology, Institute for Infection, Immunity and Inflammation, University of Glasgow, UK
| | | |
Collapse
|
132
|
Kim Y, Roe JH, Park JH, Cho YJ, Lee KL. Regulation of iron homeostasis by peroxide-sensitive CatR, a Fur-family regulator in Streptomyces coelicolor. J Microbiol 2021; 59:1083-1091. [PMID: 34865197 DOI: 10.1007/s12275-021-1457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/08/2021] [Accepted: 10/20/2021] [Indexed: 11/30/2022]
Abstract
CatR, a peroxide-sensing transcriptional repressor of Fur family, can de-repress the transcription of the catA gene encoding catalase upon peroxide stress in Streptomyces coelicolor. Since CatR-regulated genes other than catA and its own gene catR have not been identified in detail, the understanding of the role of CatR regulon is very limited. In this study, we performed transcriptomic analysis to identify genes influenced by both ΔcatR mutation and hydrogen peroxide treatment. Through ChIP-qPCR and other analyses, a new consensus sequence was found in CatR-responsive promoter region of catR gene and catA operon for direct regulation. In addition, vtlA (SCO2027) and SCO4983 were identified as new members of the CatR regulon. Expression levels of iron uptake genes were reduced by hydrogen peroxide and a DmdR1 binding sequence was identified in promoters of these genes. The increase in free iron by hydrogen peroxide was thought to suppress the iron import system by DmdR1. A putative exporter protein VtlA regulated by CatR appeared to reduce intracellular iron to prevent oxidative stress. The name vtlA (VIT1-like transporter) was proposed for iron homeostasis related gene SCO2027.
Collapse
Affiliation(s)
- Yeonbum Kim
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jung-Hye Roe
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Joo-Hong Park
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yong-Joon Cho
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,The Research Institute of Basic Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Kang-Lok Lee
- Department of Biology Education, IALS, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
133
|
Alfarouk KO, Alhoufie STS, Hifny A, Schwartz L, Alqahtani AS, Ahmed SBM, Alqahtani AM, Alqahtani SS, Muddathir AK, Ali H, Bashir AHH, Ibrahim ME, Greco MR, Cardone RA, Harguindey S, Reshkin SJ. Of mitochondrion and COVID-19. J Enzyme Inhib Med Chem 2021; 36:1258-1267. [PMID: 34107824 PMCID: PMC8205080 DOI: 10.1080/14756366.2021.1937144] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/20/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023] Open
Abstract
COVID-19, a pandemic disease caused by a viral infection, is associated with a high mortality rate. Most of the signs and symptoms, e.g. cytokine storm, electrolytes imbalances, thromboembolism, etc., are related to mitochondrial dysfunction. Therefore, targeting mitochondrion will represent a more rational treatment of COVID-19. The current work outlines how COVID-19's signs and symptoms are related to the mitochondrion. Proper understanding of the underlying causes might enhance the opportunity to treat COVID-19.
Collapse
Affiliation(s)
- Khalid Omer Alfarouk
- Research Center, Zamzam University College, Khartoum, Sudan
- Department of Evolutionary Pharmacology and Tumor Metabolism, Hala Alfarouk Cancer Center, Khartoum, Sudan
- Al-Ghad International College for Applied Medical Sciences, Al-Madinah Al-Munwarah, Saudi Arabia
| | - Sari T. S. Alhoufie
- Medical Laboratories Technology Department, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Munwarah, Saudi Arabia
| | | | | | - Ali S. Alqahtani
- College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | | | - Ali M. Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Saad S. Alqahtani
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | | | - Heyam Ali
- Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Adil H. H. Bashir
- Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | | | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| | - Rosa A. Cardone
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| | | | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
134
|
|
135
|
Massai L, Ciambellotti S, Cosottini L, Messori L, Turano P, Pratesi A. Direct detection of iron clusters in L ferritins through ESI-MS experiments. Dalton Trans 2021; 50:16464-16467. [PMID: 34729572 DOI: 10.1039/d1dt03106f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human cytoplasmic ferritins are heteropolymers of H and L subunits containing a catalytic ferroxidase center and a nucleation site for iron biomineralization, respectively. Here, ESI-MS successfully detected labile metal-protein interactions revealing the formation of tetra- and octa-iron clusters bound to L subunits, as previously underscored by X-ray crystallography.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Silvia Ciambellotti
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy. .,Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy
| | - Lucrezia Cosottini
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy. .,Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy
| | - Luigi Messori
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy.
| | - Paola Turano
- Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 3-13, 50019 Sesto Fiorentino, FI, Italy. .,Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy. .,Consorzio Interuniversitario Risonanze Magnetiche di Metallo Proteine (C.I.R.M.M.P.), Via Luigi Sacconi 6, 50019 Sesto Fiorentino, FI, Italy
| | - Alessandro Pratesi
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy
| |
Collapse
|
136
|
Sudharson S, Kalic T, Hafner C, Breiteneder H. Newly defined allergens in the WHO/IUIS Allergen Nomenclature Database during 01/2019-03/2021. Allergy 2021; 76:3359-3373. [PMID: 34310736 PMCID: PMC9290965 DOI: 10.1111/all.15021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 01/03/2023]
Abstract
The WHO/IUIS Allergen Nomenclature Database (http://allergen.org) provides up‐to‐date expert‐reviewed data on newly discovered allergens and their unambiguous nomenclature to allergen researchers worldwide. This review discusses the 106 allergens that were accepted by the Allergen Nomenclature Sub‐Committee between 01/2019 and 03/2021. Information about protein family membership, patient cohorts, and assays used for allergen characterization is summarized. A first allergenic fungal triosephosphate isomerase, Asp t 36, was discovered in Aspergillus terreus. Plant allergens contained 1 contact, 38 respiratory, and 16 food allergens. Can s 4 from Indian hemp was identified as the first allergenic oxygen‐evolving enhancer protein 2 and Cic a 1 from chickpeas as the first allergenic group 4 late embryogenesis abundant protein. Among the animal allergens were 19 respiratory, 28 food, and 3 venom allergens. Important discoveries include Rap v 2, an allergenic paramyosin in molluscs, and Sal s 4 and Pan h 4, allergenic fish tropomyosins. Paramyosins and tropomyosins were previously known mainly as arthropod allergens. Collagens from barramundi, Lat c 6, and salmon, Sal s 6, were the first members from the collagen superfamily added to the database. In summary, the addition of 106 new allergens to the previously listed 930 allergens reflects the continuous linear growth of the allergen database. In addition, 17 newly described allergen sources were included.
Collapse
Affiliation(s)
- Srinidhi Sudharson
- Department of Dermatology University Hospital St. Poelten Karl Landsteiner University of Health Sciences St. Poelten Austria
- Division of Medical Biotechnology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Tanja Kalic
- Department of Dermatology University Hospital St. Poelten Karl Landsteiner University of Health Sciences St. Poelten Austria
- Division of Medical Biotechnology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Christine Hafner
- Department of Dermatology University Hospital St. Poelten Karl Landsteiner University of Health Sciences St. Poelten Austria
| | - Heimo Breiteneder
- Division of Medical Biotechnology Department of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| |
Collapse
|
137
|
Fin56-induced ferroptosis is supported by autophagy-mediated GPX4 degradation and functions synergistically with mTOR inhibition to kill bladder cancer cells. Cell Death Dis 2021; 12:1028. [PMID: 34716292 PMCID: PMC8556316 DOI: 10.1038/s41419-021-04306-2] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/10/2021] [Accepted: 10/13/2021] [Indexed: 01/07/2023]
Abstract
Ferroptosis is a form of regulated cell death that emerges to be relevant for therapy-resistant and dedifferentiating cancers. Although several lines of evidence suggest that ferroptosis is a type of autophagy-dependent cell death, the underlying molecular mechanisms remain unclear. Fin56, a type 3 ferroptosis inducer, triggers ferroptosis by promoting glutathione peroxidase 4 (GPX4) protein degradation via a not fully understood pathway. Here, we determined that Fin56 induces ferroptosis and autophagy in bladder cancer cells and that Fin56-triggered ferroptosis mechanistically depends on the autophagic machinery. Furthermore, we found that autophagy inhibition at different stages attenuates Fin56-induced oxidative stress and GPX4 degradation. Moreover, we investigated the effects of Fin56 in combination with Torin 2, a potent mTOR inhibitor used to activate autophagy, on cell viability. We found that Fin56 synergizes with Torin 2 in cytotoxicity against bladder cancer cells. Collectively, our findings not only support the concept that ferroptosis is a type of autophagy-dependent cell death but imply that the combined application of ferroptosis inducers and mTOR inhibitors is a promising approach to improve therapeutic options in the treatment of bladder cancer.
Collapse
|
138
|
Bossoni L, Hegeman-Kleinn I, van Duinen SG, Bulk M, Vroegindeweij LHP, Langendonk JG, Hirschler L, Webb A, van der Weerd L. Off-resonance saturation as an MRI method to quantify mineral- iron in the post-mortem brain. Magn Reson Med 2021; 87:1276-1288. [PMID: 34655092 PMCID: PMC9293166 DOI: 10.1002/mrm.29041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022]
Abstract
Purpose To employ an off‐resonance saturation method to measure the mineral‐iron pool in the postmortem brain, which is an endogenous contrast agent that can give information on cellular iron status. Methods An off‐resonance saturation acquisition protocol was implemented on a 7 Tesla preclinical scanner, and the contrast maps were fitted to an established analytical model. The method was validated by correlation and Bland‐Altman analysis on a ferritin‐containing phantom. Mineral‐iron maps were obtained from postmortem tissue of patients with neurological diseases characterized by brain iron accumulation, that is, Alzheimer disease, Huntington disease, and aceruloplasminemia, and validated with histology. Transverse relaxation rate and magnetic susceptibility values were used for comparison. Results In postmortem tissue, the mineral‐iron contrast colocalizes with histological iron staining in all the cases. Iron concentrations obtained via the off‐resonance saturation method are in agreement with literature. Conclusions Off‐resonance saturation is an effective way to detect iron in gray matter structures and partially mitigate for the presence of myelin. If a reference region with little iron is available in the tissue, the method can produce quantitative iron maps. This method is applicable in the study of diseases characterized by brain iron accumulation and can complement existing iron‐sensitive parametric methods.
Collapse
Affiliation(s)
- Lucia Bossoni
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sjoerd G van Duinen
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein Bulk
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Neurology, Alzheimer Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lena H P Vroegindeweij
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Porphyria Center Rotterdam, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Janneke G Langendonk
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Porphyria Center Rotterdam, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Lydiane Hirschler
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew Webb
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Louise van der Weerd
- C. J. Gorter Center for High field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
139
|
Pujol-Carrion N, Gonzalez-Alfonso A, Puig S, de la Torre-Ruiz MA. Both human and soya bean ferritins highly improve the accumulation of bioavailable iron and contribute to extend the chronological life in budding yeast. Microb Biotechnol 2021; 15:1525-1541. [PMID: 34644442 PMCID: PMC9049602 DOI: 10.1111/1751-7915.13939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/02/2022] Open
Abstract
Ferritin proteins have an enormous capacity to store iron in cells. In search for the best conditions to accumulate and store bioavailable iron, we made use of a double mutant null for the monothiol glutaredoxins GRX3 and GRX4. The strain grx3grx4 accumulates high iron concentrations in the cytoplasm, making the metal easily available for ferritin chelation. Here, we perform a comparative study between human (L and H) and soya bean ferritins (H1 and H2) function in the eukaryotic system Saccharomyces cerevisiae. We demonstrate that the four human and soya bean ferritin chains are successfully expressed in our model system. Upon coexpression of either both human or soya bean ferritin chains, respiratory conditions along with iron supplementation led us to obtain the maximum yields of iron stored in yeast described to date. Human and soya bean ferritin chains are functional and present equivalent properties as promoters of cell survival in iron overload conditions. The best system revealed that the four human and soya bean ferritins possess a novel function as anti‐ageing proteins in conditions of iron excess. In this respect, both ferritin chains with oxidoreductase capacity (human‐H and soya bean‐H2) bear the highest capacity to extend life suggesting the possibility of an evolutionary conservation.
Collapse
Affiliation(s)
- Nuria Pujol-Carrion
- Cell Signalling in Yeast Unit, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLleida), University of Lleida, Lleida, 25198, Spain
| | - Alma Gonzalez-Alfonso
- Cell Signalling in Yeast Unit, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLleida), University of Lleida, Lleida, 25198, Spain
| | - Sergi Puig
- Departamento de Biotecnología, Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Paterna, Valencia, E-46980, Spain
| | - Maria Angeles de la Torre-Ruiz
- Cell Signalling in Yeast Unit, Department of Basic Medical Sciences, Institut de Recerca Biomèdica de Lleida (IRBLleida), University of Lleida, Lleida, 25198, Spain
| |
Collapse
|
140
|
Høiseth G, Hilberg T, Trydal T, Husa A, Vindenes V, Bogstrand ST. The alcohol marker phosphatidylethanol is closely related to AST, GGT, ferritin and HDL-C. Basic Clin Pharmacol Toxicol 2021; 130:182-190. [PMID: 34591374 DOI: 10.1111/bcpt.13662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the quantitative relation between common clinical chemical analyses and ethanol use, measured by a combination of the two alcohol markers phosphatidylethanol (PEth) and carbohydrate-deficient transferrin (CDT). METHODS Results of PEth and CDT in whole blood and serum, respectively, were included, together with information on 10 different commonly measured clinical chemical analytes, as well as age and sex. PEth was analysed by UPC2 -MS/MS and CDT was measured by capillary electrophoresis. RESULTS Samples from 4873 patients were included. The strongest relation to alcohol consumption as measured by PEth, when correcting for age and sex, was found for HDL-C (standardized β = 0.472, p < 0.001), AST (standardized β = 0.372, p < 0.001), ferritin (standardized β = 0.332, p < 0.001) and GGT (standardized β = 0.325, p < 0.001). The relation to PEth was weak for total cholesterol, TG and ALP. No relation was found for Hb and LDL-C. CONCLUSIONS When using PEth as a marker for alcohol consumption, this study demonstrated the quantitative relation to commonly used test as AST or GGT, but also an important relation to ferritin or HDL-C. In clinical practice, elevated levels of these clinical chemical analytes should initiate further work-up on possibly harmful alcohol use.
Collapse
Affiliation(s)
- Gudrun Høiseth
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway.,Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | | | - Vigdis Vindenes
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway.,Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Stig Tore Bogstrand
- Department of Forensic Sciences, Oslo University Hospital, Oslo, Norway.,Faculty of Medicine, Institute of Health and Society, University of Oslo, Oslo, Norway
| |
Collapse
|
141
|
Zhang L, Zhang J, Jin Y, Yao G, Zhao H, Qiao P, Wu S. Nrf2 Is a Potential Modulator for Orchestrating Iron Homeostasis and Redox Balance in Cancer Cells. Front Cell Dev Biol 2021; 9:728172. [PMID: 34589492 PMCID: PMC8473703 DOI: 10.3389/fcell.2021.728172] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
Iron is an essential trace mineral element in almost all living cells and organisms. However, cellular iron metabolism pathways are disturbed in most cancer cell types. Cancer cells have a high demand of iron. To maintain rapid growth and proliferation, cancer cells absorb large amounts of iron by altering expression of iron metabolism related proteins. However, iron can catalyze the production of reactive oxygen species (ROS) through Fenton reaction. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is an important player in the resistance to oxidative damage by inducing the transcription of antioxidant genes. Aberrant activation of Nrf2 is observed in most cancer cell types. It has been revealed that the over-activation of Nrf2 promotes cell proliferation, suppresses cell apoptosis, enhances the self-renewal capability of cancer stem cells, and even increases the chemoresistance and radioresistance of cancer cells. Recently, several genes involving cellular iron homeostasis are identified under the control of Nrf2. Since cancer cells require amounts of iron and Nrf2 plays pivotal roles in oxidative defense and iron metabolism, it is highly probable that Nrf2 is a potential modulator orchestrating iron homeostasis and redox balance in cancer cells. In this hypothesis, we summarize the recent findings of the role of iron and Nrf2 in cancer cells and demonstrate how Nrf2 balances the oxidative stress induced by iron through regulating antioxidant enzymes and iron metabolism. This hypothesis provides new insights into the role of Nrf2 in cancer progression. Since ferroptosis is dependent on lipid peroxide and iron accumulation, Nrf2 inhibition may dramatically increase sensitivity to ferroptosis. The combination of Nrf2 inhibitors with ferroptosis inducers may exert greater efficacy on cancer therapy.
Collapse
Affiliation(s)
- Lingyan Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jian Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yuanqing Jin
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Gang Yao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hai Zhao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Penghai Qiao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Shuguang Wu
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
142
|
Yamaguchi Y, Zampino M, Tanaka T, Bandinelli S, Moaddel R, Fantoni G, Candia J, Ferrucci L, Semba RD. The Plasma Proteome Fingerprint Associated with Circulating Carotenoids and Retinol in Older Adults. J Nutr 2021; 152:40-48. [PMID: 34550359 PMCID: PMC8754576 DOI: 10.1093/jn/nxab340] [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: 05/12/2021] [Revised: 06/23/2021] [Accepted: 09/16/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Although diets rich in carotenoids are associated with reduced risks of cardiovascular disease, age-related macular degeneration, disability, and other adverse aging outcomes, the underlying biological mechanisms are not fully elucidated. OBJECTIVES To characterize the plasma proteome fingerprint associated with circulating carotenoid and retinol concentrations in older adults. METHODS In 728 adults ≥65 y participating in the Invecchiare in Chianti (InCHIANTI) Study, plasma α-carotene, β-carotene, β-cryptoxanthin, lutein, zeaxanthin, and lycopene were measured using HPLC. The SOMAscan assay was used to measure 1301 plasma proteins. Multivariable linear regression models were used to examine the relationship of individual carotenoids and retinol with plasma proteins. A false discovery rate approach was used to deal with multiple comparisons using a q-value < 0.05. RESULTS Plasma β-carotene, β-cryptoxanthin, lutein, zeaxanthin, and lycopene were associated with 85, 39, 4, 2, and 5 plasma proteins, respectively, in multivariable linear regression models adjusting for potential confounders (q < 0.05). No proteins were associated with α-carotene or retinol. Two or more carotenoids were positively associated with ferritin, 6-phosphogluconate dehydrogenase (decarboxylating), hepcidin, thrombospondin-2, and choline/ethanolamine kinase. The proteins associated with circulating carotenoids were related to energy metabolism, sirtuin signaling, inflammation and oxidative stress, iron metabolism, proteostasis, innate immunity, and longevity. CONCLUSIONS The plasma proteomic fingerprint associated with elevated circulating carotenoids in older adults provides insight into the mechanisms underlying the protective role of carotenoids on health.
Collapse
Affiliation(s)
| | - Marta Zampino
- National Institutes on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Toshiko Tanaka
- National Institutes on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | - Ruin Moaddel
- National Institutes on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Giovanna Fantoni
- National Institutes on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Julián Candia
- National Institutes on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- National Institutes on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Center for a Livable Future, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
143
|
TRIB2 desensitizes ferroptosis via βTrCP-mediated TFRC ubiquitiantion in liver cancer cells. Cell Death Discov 2021; 7:196. [PMID: 34315867 PMCID: PMC8316344 DOI: 10.1038/s41420-021-00574-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/07/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Tribbles homolog 2 (TRIB2) is known to boost liver tumorigenesis via regulating Ubiquitin (Ub) proteasome system (UPS). At least two ways are involved, i.e., acts as an adaptor protein to modulate ubiquitination functions of certain ubiquitin E3 ligases (E3s) and reduces global Ub levels via increasing the proteolysis activity of proteasome. Recently, we have identified the role of TRIB2 to relieve oxidative damage via reducing the availability of Ub that is essential for the ubiquitination and subsequent degradation of Glutathione peroxidase 4 (GPX4). Although GPX4 is a critical antioxidant factor to protect against ferroptosis, the exact evidence showing that TRIB2 desensitizes ferroptosis is lacking. Also, whether such function is via E3 remains unclear. Here, we demonstrated that deletion of TRIB2 sensitized ferroptosis via lifting labile iron in liver cancer cells. By contrast, overexpression of TRIB2 led to the opposite outcome. We further demonstrated that transferrin receptor (TFRC) was required for TRIB2 to desensitize the cells to ferroptosis. Without TFRC, the labile iron pool could not be reduced by overexpressing TRIB2. We also found that beta-transducin repeat containing E3 ubiqutin protein ligase (βTrCP) was a genuine E3 for the ubiquitination of TFRC, and TRIB2 was unable to decline labile iron level once upon βTrCP was knocked out. In addition, we confirmed that the opposite effects on ferroptosis and ferroptosis-associated lipid reactive oxygen species (ROS) generation resulted from knockout and overexpression of TRIB2 were all indispensible of TFRC and βTrCP. Finally, we demonstrated that TRIB2 exclusively manipulated RSL3- and erastin-induced-ferroptosis independent of GPX4 and glutathione (GSH). In conclusion, we elucidated a novel role of TRIB2 to desensitize ferroptosis via E3 βTrCP, by which facilitates TFRC ubiquitiation and finally decreases labile iron in liver cancer cells.
Collapse
|
144
|
Ajoolabady A, Aslkhodapasandhokmabad H, Libby P, Tuomilehto J, Lip GYH, Penninger JM, Richardson DR, Tang D, Zhou H, Wang S, Klionsky DJ, Kroemer G, Ren J. Ferritinophagy and ferroptosis in the management of metabolic diseases. Trends Endocrinol Metab 2021; 32:444-462. [PMID: 34006412 DOI: 10.1016/j.tem.2021.04.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/07/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a form of regulated cell death modality associated with disturbed iron-homeostasis and unrestricted lipid peroxidation. Ample evidence has depicted an essential role for ferroptosis as either the cause or consequence for human diseases, denoting the likely therapeutic promises for targeting ferroptosis in the preservation of human health. Ferritinophagy, a selective form of autophagy, contributes to the initiation of ferroptosis through degradation of ferritin, which triggers labile iron overload (IO), lipid peroxidation, membrane damage, and cell death. In this review, we will delineate the role of ferritinophagy in ferroptosis, and its underlying regulatory mechanisms, to unveil the therapeutic value of ferritinophagy as a target in the combat of ferroptosis to manage metabolic diseases.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | | | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jaakko Tuomilehto
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland; Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gregory Y H Lip
- University of Liverpool Institute of Ageing and Chronic Disease, Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria; Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, Medical Foundation Building (K25), 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 and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hao Zhou
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, 100853, China
| | - Shuyi Wang
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; School of Medicine Shanghai University, Shanghai 200444, China.
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
145
|
Prajapati M, Conboy HL, Hojyo S, Fukada T, Budnik B, Bartnikas TB. Biliary excretion of excess iron in mice requires hepatocyte iron import by Slc39a14. J Biol Chem 2021; 297:100835. [PMID: 34051234 PMCID: PMC8214222 DOI: 10.1016/j.jbc.2021.100835] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/19/2022] Open
Abstract
Iron is essential for erythropoiesis and other biological processes, but is toxic in excess. Dietary absorption of iron is a highly regulated process and is a major determinant of body iron levels. Iron excretion, however, is considered a passive, unregulated process, and the underlying pathways are unknown. Here we investigated the role of metal transporters SLC39A14 and SLC30A10 in biliary iron excretion. While SLC39A14 imports manganese into the liver and other organs under physiological conditions, it imports iron under conditions of iron excess. SLC30A10 exports manganese from hepatocytes into the bile. We hypothesized that biliary excretion of excess iron would be impaired by SLC39A14 and SLC30A10 deficiency. We therefore analyzed biliary iron excretion in Slc39a14-and Slc30a10-deficient mice raised on iron-sufficient and -rich diets. Bile was collected surgically from the mice, then analyzed with nonheme iron assays, mass spectrometry, ELISAs, and an electrophoretic assay for iron-loaded ferritin. Our results support a model in which biliary excretion of excess iron requires iron import into hepatocytes by SLC39A14, followed by iron export into the bile predominantly as ferritin, with iron export occurring independently of SLC30A10. To our knowledge, this is the first report of a molecular determinant of mammalian iron excretion and can serve as basis for future investigations into mechanisms of iron excretion and relevance to iron homeostasis.
Collapse
Affiliation(s)
- Milankumar Prajapati
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Heather L Conboy
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA
| | - Shintaro Hojyo
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Graduate School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Toshiyuki Fukada
- Department of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Bogdan Budnik
- Mass Spectrometry and Proteomics Resource Laboratory, Faculty of Arts and Sciences, Division of Science, Harvard University, Cambridge, Massachusetts, USA
| | - Thomas B Bartnikas
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island, USA.
| |
Collapse
|
146
|
Wang S, Cheng M, Peng P, Lou Y, Zhang A, Liu P. Iron Released after Cryo-Thermal Therapy Induced M1 Macrophage Polarization, Promoting the Differentiation of CD4 + T Cells into CTLs. Int J Mol Sci 2021; 22:ijms22137010. [PMID: 34209797 PMCID: PMC8268875 DOI: 10.3390/ijms22137010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages play critical roles in both innate and adaptive immunity and are known for their high plasticity in response to various external signals. Macrophages are involved in regulating systematic iron homeostasis and they sequester iron by phagocytotic activity, which triggers M1 macrophage polarization and typically exerts antitumor effects. We previously developed a novel cryo-thermal therapy that can induce the mass release of tumor antigens and damage-associated molecular patterns (DAMPs), promoting M1 macrophage polarization. However, that study did not examine whether iron released after cryo-thermal therapy induced M1 macrophage polarization; this question still needed to be addressed. We hypothesized that cryo-thermal therapy would cause the release of a large quantity of iron to augment M1 macrophage polarization due to the disruption of tumor cells and blood vessels, which would further enhance antitumor immunity. In this study, we investigated iron released in primary tumors, the level of iron in splenic macrophages after cryo-thermal therapy and the effect of iron on macrophage polarization and CD4+ T cell differentiation in metastatic 4T1 murine mammary carcinoma. We found that a large amount of iron was released after cryo-thermal therapy and could be taken up by splenic macrophages, which further promoted M1 macrophage polarization by inhibiting ERK phosphorylation. Moreover, iron promoted DC maturation, which was possibly mediated by iron-induced M1 macrophages. In addition, iron-induced M1 macrophages and mature DCs promoted the differentiation of CD4+ T cells into the CD4 cytolytic T lymphocytes (CTL) subset and inhibited differentiation into Th2 and Th17 cells. This study explains the role of iron in cryo-thermal therapy-induced antitumor immunity from a new perspective.
Collapse
Affiliation(s)
- Shicheng Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.W.); (M.C.); (P.P.); (Y.L.); (A.Z.)
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Man Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.W.); (M.C.); (P.P.); (Y.L.); (A.Z.)
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Peng Peng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.W.); (M.C.); (P.P.); (Y.L.); (A.Z.)
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yue Lou
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.W.); (M.C.); (P.P.); (Y.L.); (A.Z.)
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Aili Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.W.); (M.C.); (P.P.); (Y.L.); (A.Z.)
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ping Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.W.); (M.C.); (P.P.); (Y.L.); (A.Z.)
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: ; Tel.: +86-021-62933231
| |
Collapse
|
147
|
Kron NS, Fieber LA. Co-expression analysis identifies neuro-inflammation as a driver of sensory neuron aging in Aplysia californica. PLoS One 2021; 16:e0252647. [PMID: 34116561 PMCID: PMC8195618 DOI: 10.1371/journal.pone.0252647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 05/20/2021] [Indexed: 01/08/2023] Open
Abstract
Aging of the nervous system is typified by depressed metabolism, compromised proteostasis, and increased inflammation that results in cognitive impairment. Differential expression analysis is a popular technique for exploring the molecular underpinnings of neural aging, but technical drawbacks of the methodology often obscure larger expression patterns. Co-expression analysis offers a robust alternative that allows for identification of networks of genes and their putative central regulators. In an effort to expand upon previous work exploring neural aging in the marine model Aplysia californica, we used weighted gene correlation network analysis to identify co-expression networks in a targeted set of aging sensory neurons in these animals. We identified twelve modules, six of which were strongly positively or negatively associated with aging. Kyoto Encyclopedia of Genes analysis and investigation of central module transcripts identified signatures of metabolic impairment, increased reactive oxygen species, compromised proteostasis, disrupted signaling, and increased inflammation. Although modules with immune character were identified, there was no correlation between genes in Aplysia that increased in expression with aging and the orthologous genes in oyster displaying long-term increases in expression after a virus-like challenge. This suggests anti-viral response is not a driver of Aplysia sensory neuron aging.
Collapse
Affiliation(s)
- N. S. Kron
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, United States of America
| | - L. A. Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, United States of America
| |
Collapse
|
148
|
Mazgaj R, Lipiński P, Edison ES, Bednarz A, Staroń R, Haberkiewicz O, Lenartowicz M, Smuda E, Jończy A, Starzyński RR. Marginally reduced maternal hepatic and splenic ferroportin under severe nutritional iron deficiency in pregnancy maintains systemic iron supply. Am J Hematol 2021; 96:659-670. [PMID: 33684239 PMCID: PMC8251567 DOI: 10.1002/ajh.26152] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
The demand for iron is high in pregnancy to meet the increased requirements for erythropoiesis. Even pregnant females with initially iron‐replete stores develop iron‐deficiency anemia, due to inadequate iron absorption. In anemic females, the maternal iron supply is dedicated to maintaining iron metabolism in the fetus and placenta. Here, using a mouse model of iron deficiency in pregnancy, we show that iron recycled from senescent erythrocytes becomes a predominant source of this microelement that can be transferred to the placenta in females with depleted iron stores. Ferroportin is a key protein in the molecular machinery of cellular iron egress. We demonstrate that under iron deficiency in pregnancy, levels of ferroportin are greatly reduced in the duodenum, placenta and fetal liver, but not in maternal liver macrophages and in the spleen. Although low expression of both maternal and fetal hepcidin predicted ferroportin up‐regulation in examined locations, its final expression level was very likely correlated with tissue iron status. Our results argue that iron released into the circulation of anemic females is taken up by the placenta, as evidenced by high expression of iron importers on syncytiotrophoblasts. Then, a substantial decrease in levels of ferroportin on the basolateral side of syncytiotrophoblasts, may be responsible for the reduced transfer of iron to the fetus. As attested by the lowest decrease in iron content among analyzed tissues, some part is retained in the placenta. These findings confirm the key role played by ferroportin in tuning iron turnover in iron‐deficient pregnant mouse females and their fetuses.
Collapse
Affiliation(s)
- Rafał Mazgaj
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences Magdalenka Poland
| | - Paweł Lipiński
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences Magdalenka Poland
| | | | - Aleksandra Bednarz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research Jagiellonian University Kraków Poland
| | - Robert Staroń
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences Magdalenka Poland
| | - Olga Haberkiewicz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research Jagiellonian University Kraków Poland
| | - Małgorzata Lenartowicz
- Department of Genetics and Evolution, Institute of Zoology and Biomedical Research Jagiellonian University Kraków Poland
| | - Ewa Smuda
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences Magdalenka Poland
| | - Aneta Jończy
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences Magdalenka Poland
| | - Rafał R. Starzyński
- Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences Magdalenka Poland
| |
Collapse
|
149
|
Pogorelova TN, Gunko VO, Nikashina AA, Mikhelson AA, Botasheva TL, Kaushanskaya LV. Violation of the balance of free metals and metal-containing proteins in amniotic fluid in placental insufficiency. Klin Lab Diagn 2021; 66:266-270. [PMID: 34047511 DOI: 10.51620/0869-2084-2021-66-5-266-270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The content of metal ions and proteins containing or binding these metals in amniotic fluid during different periods of physiological pregnancy and placental insufficiency (PI) was studied. The content of zinc, copper, magnesium, iron, zinc-α-2-glycoprotein, ferritin, ceruloplasmin and the activity of Ca2+, Mg2+-ATPase were estimated using spectrophotometric methods, immunoturbometric and enzyme immunoassay methods. It was found that in PI in both trimesters there is a decrease in the content of zinc, copper, iron and an increase in the level of copper. The indices of ceruloplasmin, ferritin, Ca2+, Mg2+-ATPase in PI are lower, and zinc-α-2-glycoprotein is higher than in similar periods of physiological gestation. A close correlation of different directions has been established between the level of metals and the corresponding proteins. The revealed violations obviously play a certain pathogenetic role in the development of PI, and the indicators of the ratio between metals can serve as markers for predicting the state of newborns.
Collapse
|
150
|
Kontoghiorghes GJ, Kolnagou A, Demetriou T, Neocleous M, Kontoghiorghe CN. New Era in the Treatment of Iron Deficiency Anaemia Using Trimaltol Iron and Other Lipophilic Iron Chelator Complexes: Historical Perspectives of Discovery and Future Applications. Int J Mol Sci 2021; 22:ijms22115546. [PMID: 34074010 PMCID: PMC8197347 DOI: 10.3390/ijms22115546] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
The trimaltol iron complex (International Non-proprietary Name: ferric maltol) was originally designed, synthesised, and screened in vitro and in vivo in 1980–1981 by Kontoghiorghes G.J. following his discovery of the novel alpha-ketohydroxyheteroaromatic (KHP) class of iron chelators (1978–1981), which were intended for clinical use, including the treatment of iron deficiency anaemia (IDA). Iron deficiency anaemia is a global health problem affecting about one-third of the world’s population. Many (and different) ferrous and ferric iron complex formulations are widely available and sold worldwide over the counter for the treatment of IDA. Almost all such complexes suffer from instability in the acidic environment of the stomach and competition from other dietary molecules or drugs. Natural and synthetic lipophilic KHP chelators, including maltol, have been shown in in vitro and in vivo studies to form stable iron complexes, to transfer iron across cell membranes, and to increase iron absorption in animals. Trimaltol iron, sold as Feraccru or Accrufer, was recently approved for clinical use in IDA patients in many countries, including the USA and in EU countries, and was shown to be effective and safe, with a better therapeutic index in comparison to other iron formulations. Similar properties of increased iron absorption were also shown by lipophilic iron complexes of 8-hydroxyquinoline, tropolone, 2-hydroxy-4-methoxypyridine-1-oxide, and related analogues. The interactions of the KHP iron complexes with natural chelators, drugs, metal ions, proteins, and other molecules appear to affect the pharmacological and metabolic effects of both iron and the KHP chelators. A new era in the treatment of IDA and other possible clinical applications, such as theranostic and anticancer formulations and metal radiotracers in diagnostic medicine, are envisaged from the introduction of maltol, KHP, and similar lipophilic chelators.
Collapse
|