1
|
Fan W, Zheng J, Jiang F. Analysis of ferroptosis-related genes in cerebral ischemic stroke via immune infiltration and single-cell RNA-sequencing. BMC Med Genomics 2025; 18:31. [PMID: 39934808 PMCID: PMC11817822 DOI: 10.1186/s12920-025-02098-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/30/2025] [Indexed: 02/13/2025] Open
Abstract
Ischemic stroke (IS) represents a harmful neurological disorder with limited treatment options. Ferroptosis accounts for the iron-dependent, nonapoptotic cell death pattern, which shows the feature of fatal lipid ROS accumulation. Nonetheless, ferroptosis-related biomarkers for identifying IS early are currently lacking. The present study focused on investigating the possible ferroptosis-related biomarkers for IS and analyzing their effects on immune infiltration. Altogether five hub differentially expressed ferroptosis-related genes (DEFRGs) were identified from the relevant databases. Additionally, single-cell RNA-sequencing (seq) analysis was conducted for the comprehensive mapping of cell populations based on the IS database. These five hub DEFRGs were analyzed using gene set enrichment analysis, miRNA prediction, and single-cell RNA-seq analysis. A transient middle cerebral artery occlusion mouse model was constructed. We also adopted bioinformatics methods combined with western blot, changes to mitochondria, hematoxylin & eosin staining, Nissl staining, ROS fluorescence staining, immunohistochemistry, and quantitative real-time polymerase chain reaction (qRT-PCR) to show the involvement of ferroptosis in IS progression. The results revealed that nuclear factor erythroid-derived 2-like 2 (Nfe2l2) was the potential candidate biomarker for IS diagnosis, and ferroptosis may be suppressed via the Nfe2l2/HO-1 pathway. Thus, drug targeting Nfe2l2 can shed novel lights on IS treatment.
Collapse
Affiliation(s)
- Wei Fan
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jinhua Zheng
- Department of Anatomy, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Fangchao Jiang
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, Henan, China.
| |
Collapse
|
2
|
Arosio P, Cairo G, Bou-Abdallah F. A Brief History of Ferritin, an Ancient and Versatile Protein. Int J Mol Sci 2024; 26:206. [PMID: 39796064 PMCID: PMC11719527 DOI: 10.3390/ijms26010206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Ferritin, a highly conserved iron storage protein, is among the earliest proteins that have been purified, named, and characterized due to its unique properties that continue to captivate researchers. Ferritin is composed of 24 subunits that form an almost spherical shell delimiting a cavity where thousands of iron atoms can be stored in a nontoxic ferric form, thereby preventing cytosolic iron from catalyzing oxidative stress. Mitochondrial and extracellular ferritin have also been described and characterized, with the latter being associated with several signaling functions. In addition, serum ferritin serves as a reliable indicator of both iron stores and inflammatory conditions. First identified and purified through crystallization in 1937, ferritin has since drawn significant attention for its critical role in iron metabolism and regulation. Its unique structural features have recently been exploited for many diverse biological and technological applications. To date, more than 40,000 publications have explored this remarkable protein. Here, we present a historical overview, tracing its journey from discovery to current applications and highlighting the evolution of biochemical techniques developed for its structure-function characterization over the past eight decades.
Collapse
Affiliation(s)
- Paolo Arosio
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| | - Fadi Bou-Abdallah
- Department of Chemistry, State University of New York at Potsdam, Potsdam, NY 13676, USA;
| |
Collapse
|
3
|
Lv C, Larbi A, Li C, Liang J, Wu G, Shao Q, Quan Q. Decoding the influence of semen collection processes on goat sperm quality from a perspective of seminal plasma proteomics. J Proteomics 2024; 298:105141. [PMID: 38408605 DOI: 10.1016/j.jprot.2024.105141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
This study aims to assess the impact of semen collection methods on goat semen quality and seminal plasma (SP) proteomes. Semen was collected by artificial vagina (AV) or electro-ejaculator (EE) and semen parameters were evaluated. Tandem mass tag coupled with liquid chromatography-tandem mass spectrometry was used to screen SP differentially abundant proteins (DAPs) between EE and AV. PRM was used to confirm the reliability of the data. In contrast to EE, a lower volume, higher progressive motility and concentration were observed in AV. No differences were found in total motility, membrane integrity, acrosome integrity, and ROS production between EE and AV. In total, 1692 proteins were identified in SP, including 210 DAPs. Among them, 120 and 90 proteins were down-regulated and up-regulated in AV compared to EE, respectively. The GO annotation showed that DAPs are mainly localized in the membrane, involved in deference responses to bacterium, RNA processing, and related to oxidoreductase activity. KEGG demonstrated tight associations of DAPs with specific amino acids, carbon metabolism, citrate cycle, and propanoate metabolism. In conclusion, this study provides valuable insights into the effects of semen collection on goat semen quality and explores the potential action mechanism based on the modification of SP proteomes. SIGNIFICANCE OF THE STUDY: The quality of fresh semen directly influences the results of artificial insemination and semen cryopreservation in livestock. This study represents the first attempt to evaluate the impact of semen collection methods including electroejaculation and artificial vagina on sperm quality and seminal plasma proteomes in goat. The results of this study demonstrated that semen collection methods directly impacted the quality of goat semen. Then, the proteomic strategy was used to explore the potential action mechanism of semen collection methods on sperm. Some differentially abundant proteins that potentially influence semen quality were identified. Furthermore, this study suggests the possibility of utilizing specific proteins as predictive markers for goat semen quality.
Collapse
Affiliation(s)
- Chunrong Lv
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Panlong District, Kunming City, Yunnan Province, China
| | - Allai Larbi
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Laboratory of Sustainable Agriculture Management, Higher School of Technology Sidi Bennour, Chouaib Doukkali University El Jadida, Morocco
| | - Chunyan Li
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Panlong District, Kunming City, Yunnan Province, China
| | - Jiangchong Liang
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Panlong District, Kunming City, Yunnan Province, China
| | - Guoquan Wu
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Panlong District, Kunming City, Yunnan Province, China
| | - Qingyong Shao
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Panlong District, Kunming City, Yunnan Province, China
| | - Quobo Quan
- Yunnan Animal Science and Veterinary Institute, Jindian, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Engineering Research Center of Animal Genetic Resource Conservation and Germplasm Enhancement, Panlong District, Kunming City, Yunnan Province, China; Yunnan Provincial Genebank of Livestock and Poultry Genetic Resources, Panlong District, Kunming City, Yunnan Province, China.
| |
Collapse
|
4
|
Han T, Liao X, Guo Z, Chen JY, He C, Lu Z. Deciphering temporal gene expression dynamics in multiple coral species exposed to heat stress: Implications for predicting resilience. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:169021. [PMID: 38061659 DOI: 10.1016/j.scitotenv.2023.169021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 01/18/2024]
Abstract
Coral reefs are facing unprecedented threats due to global climate change, particularly elevated sea surface temperatures causing coral bleaching. Understanding coral responses at the molecular level is crucial for predicting their resilience and developing effective conservation strategies. In this study, we conducted a comprehensive gene expression analysis of four coral species to investigate their long-term molecular response to heat stress. We identified distinct gene expression patterns among the coral species, with laminar corals exhibiting a stronger response compared to branching corals. Heat shock proteins (HSPs) showed an overall decreasing expression trend, indicating the high energy cost associated with sustaining elevated HSP levels during prolonged heat stress. Peroxidases and oxidoreductases involved in oxidative stress response demonstrated significant upregulation, highlighting their role in maintaining cellular redox balance. Differential expression of genes related to calcium homeostasis and bioluminescence suggested distinct mechanisms for coping with heat stress among the coral species. Furthermore, the impact of heat stress on coral biomineralization varied, with downregulation of carbonic anhydrase and skeletal organic matrix proteins indicating reduced capacity for biomineralization in the later stages of heat stress. Our findings provide insights into the molecular mechanisms underlying coral responses to heat stress and highlight the importance of considering species-specific responses in assessing coral resilience. The identified biomarkers may serve as indicators of heat stress and contribute to early detection of coral bleaching events. These findings contribute to our understanding of coral resilience and provide a basis for future research aimed at enhancing coral survival in the face of climate change.
Collapse
Affiliation(s)
- Tingyu Han
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xin Liao
- Guangxi Key Lab of Mangrove Conservation and Utilization, Guangxi Mangrove Research Center, Beihai 536000, China
| | - Zhuojun Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - J-Y Chen
- Nanjing Institute of Paleontology and Geology, Nanjing 210008, China
| | - Chunpeng He
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zuhong Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
5
|
Tian X, Wang Y, Yuan M, Zheng W, Zuo H, Zhang X, Song H. Heme Oxygenase-1-Modified BMMSCs Activate AMPK-Nrf2-FTH1 to Reduce Severe Steatotic Liver Ischemia-Reperfusion Injury. Dig Dis Sci 2023; 68:4196-4211. [PMID: 37707747 PMCID: PMC10570260 DOI: 10.1007/s10620-023-08102-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 08/28/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is an important cause of graft dysfunction post-liver transplantation, where donor liver with severe steatosis is more sensitive to IRI. Liver IRI involves ferroptosis and can be alleviated by heme oxygenase-1-modified bone marrow mesenchymal stem cells (HO-1/BMMSCs). AIMS To explore the role and mechanism of HO-1/BMMSCs in severe steatotic liver IRI. METHODS A severe steatotic liver IRI rat model and a hypoxia/reoxygenation (H/R) of severe steatosis hepatocyte model were established. Liver and hepatocyte damage was evaluated via liver histopathology and cell activity. Ferroptosis was evaluated through ferroptosis indexes. Nuclear factor erythroid 2-related factor 2 (Nrf2) was knocked down in severe steatotic hepatocytes. The role of Nrf2 and AMPK in HO-1/BMMSC inhibition of ferroptosis was examined using the AMP-activated protein kinase (AMPK) pathway inhibitor Compound C. RESULTS The HO-1/BMMSCs alleviated severe steatotic liver IRI and ferroptosis. HO-1/BMMSCs promoted ferritin heavy chain 1(FTH1), Nrf2, and phosphorylated (p)-AMPK expression in the H/R severe steatotic hepatocytes. Nrf2 knockdown decreased FTH1 expression levels but did not significantly affect p-AMPK expression levels. The protective effect of HO-1/BMMSCs against H/R injury in severe steatotic hepatocytes and the inhibitory effect on ferroptosis were reduced. Compound C decreased p-AMPK, Nrf2, and FTH1 expression levels, weakened the HO-1/BMMSC protective effect against severe steatotic liver IRI and H/R-injured severe steatotic hepatocytes, and reduced the inhibition of ferroptosis. CONCLUSIONS Ferroptosis was involved in HO-1/BMMSC reduction of severe steatotic liver IRI. HO-1/BMMSCs protected against severe steatotic liver IRI by inhibiting ferroptosis through the AMPK-Nrf2-FTH1 pathway. HO-1/BMMSCs activate AMPK, which activates Nrf2, promotes its nuclear transcription, then promotes the expression of its downstream protein FTH1, thereby inhibiting ferroptosis and attenuating severe steatotic liver IRI in rats. Glu: glutamic acid; Cys: cystine; GSH: glutathione; GPX4: glutathione peroxidase 4; HO-1/BMMSCs: HO-1-modified BMMSCs; Fer-1: ferrostatin-1; DFO: deferoxamine; FTH1: ferritin heavy chain1; p-AMPK: phosphorylated AMP-activated protein kinase; Nrf2: nuclear factor erythroid 2-related factor 2; IRI: ischemia-reperfusion injury; MCD: methionine-choline deficiency.
Collapse
Affiliation(s)
- Xiaorong Tian
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yuxin Wang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Mengshu Yuan
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, People's Republic of China
- NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, People's Republic of China
| | - Huaiwen Zuo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xinru Zhang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, People's Republic of China.
- Tianjin Key Laboratory of Organ Transplantation, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.
| |
Collapse
|
6
|
Shakya A, McKee NW, Dodson M, Chapman E, Zhang DD. Anti-Ferroptotic Effects of Nrf2: Beyond the Antioxidant Response. Mol Cells 2023; 46:165-175. [PMID: 36994475 PMCID: PMC10070163 DOI: 10.14348/molcells.2023.0005] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 03/31/2023] Open
Abstract
The transcription factor Nrf2 was originally identified as a master regulator of redox homeostasis, as it governs the expression of a battery of genes involved in mitigating oxidative and electrophilic stress. However, the central role of Nrf2 in dictating multiple facets of the cellular stress response has defined the Nrf2 pathway as a general mediator of cell survival. Recent studies have indicated that Nrf2 regulates the expression of genes controlling ferroptosis, an ironand lipid peroxidation-dependent form of cell death. While Nrf2 was initially thought to have anti-ferroptotic function primarily through regulation of the antioxidant response, accumulating evidence has indicated that Nrf2 also exerts anti-ferroptotic effects via regulation of key aspects of iron and lipid metabolism. In this review, we will explore the emerging role of Nrf2 in mediating iron homeostasis and lipid peroxidation, where several Nrf2 target genes have been identified that encode critical proteins involved in these pathways. A better understanding of the mechanistic relationship between Nrf2 and ferroptosis, including how genetic and/or pharmacological manipulation of Nrf2 affect the ferroptotic response, should facilitate the development of new therapies that can be used to treat ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Aryatara Shakya
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Nicholas W. McKee
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
7
|
Cheng H, Wang P, Wang N, Dong W, Chen Z, Wu M, Wang Z, Yu Z, Guan D, Wang L, Zhao R. Neuroprotection of NRF2 against Ferroptosis after Traumatic Brain Injury in Mice. Antioxidants (Basel) 2023; 12:731. [PMID: 36978979 PMCID: PMC10044792 DOI: 10.3390/antiox12030731] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Ferroptosis and iron-related redox imbalance aggravate traumatic brain injury (TBI) outcomes. NRF2 is the predominant transcription factor regulating oxidative stress and neuroinflammation in TBI, but its role in iron-induced post-TBI damage is unclear. We investigated ferroptotic neuronal damage in the injured cortex and observed neurological deficits post-TBI. These were ameliorated by the iron chelator deferoxamine (DFO) in wild-type mice. In Nrf2-knockout (Nrf2-/-) mice, more sever ferroptosis and neurological deficits were detected. Dimethyl fumarate (DMF)-mediated NRF2 activation alleviated neural dysfunction in TBI mice, partly due to TBI-induced ferroptosis mitigation. Additionally, FTH-FTL and FSP1 protein levels, associated with iron metabolism and the ferroptotic redox balance, were highly NRF2-dependent post-TBI. Thus, NRF2 is neuroprotective against TBI-induced ferroptosis through both the xCT-GPX4- and FTH-FTL-determined free iron level and the FSP1-regulated redox status. This yields insights into the neuroprotective role of NRF2 in TBI-induced neuronal damage and its potential use in TBI treatment.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- School of Forensic Medicine, Kunming Medical University, Kunming 650500, China
| | - Pengfei Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ning Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Wenwen Dong
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ziyuan Chen
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Mingzhe Wu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ziwei Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Ziqi Yu
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Dawei Guan
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Linlin Wang
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| | - Rui Zhao
- Department of Forensic Pathology, China Medical University School of Forensic Medicine, Shenyang 110122, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Collaborative Laboratory of Intelligentized Forensic Science, Shenyang 110034, China
| |
Collapse
|
8
|
Gehrer CM, Mitterstiller AM, Grubwieser P, Meyron-Holtz EG, Weiss G, Nairz M. Advances in Ferritin Physiology and Possible Implications in Bacterial Infection. Int J Mol Sci 2023; 24:4659. [PMID: 36902088 PMCID: PMC10003477 DOI: 10.3390/ijms24054659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/17/2023] [Accepted: 02/25/2023] [Indexed: 03/04/2023] Open
Abstract
Due to its advantageous redox properties, iron plays an important role in the metabolism of nearly all life. However, these properties are not only a boon but also the bane of such life forms. Since labile iron results in the generation of reactive oxygen species by Fenton chemistry, iron is stored in a relatively safe form inside of ferritin. Despite the fact that the iron storage protein ferritin has been extensively researched, many of its physiological functions are hitherto unresolved. However, research regarding ferritin's functions is gaining momentum. For example, recent major discoveries on its secretion and distribution mechanisms have been made as well as the paradigm-changing finding of intracellular compartmentalization of ferritin via interaction with nuclear receptor coactivator 4 (NCOA4). In this review, we discuss established knowledge as well as these new findings and the implications they may have for host-pathogen interaction during bacterial infection.
Collapse
Affiliation(s)
- Clemens M. Gehrer
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anna-Maria Mitterstiller
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Philipp Grubwieser
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Esther G. Meyron-Holtz
- Laboratory of Molecular Nutrition, Faculty of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Günter Weiss
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Manfred Nairz
- Department of Internal Medicine II, Infectious Diseases, Immunology, Rheumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
9
|
Tsuji Y. Optimization of Biotinylated RNA or DNA Pull-Down Assays for Detection of Binding Proteins: Examples of IRP1, IRP2, HuR, AUF1, and Nrf2. Int J Mol Sci 2023; 24:3604. [PMID: 36835018 PMCID: PMC9965622 DOI: 10.3390/ijms24043604] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Investigation of RNA- and DNA-binding proteins to a defined regulatory sequence, such as an AU-rich RNA and a DNA enhancer element, is important for understanding gene regulation through their interactions. For in vitro binding studies, an electrophoretic mobility shift assay (EMSA) was widely used in the past. In line with the trend toward using non-radioactive materials in various bioassays, end-labeled biotinylated RNA and DNA oligonucleotides can be more practical probes to study protein-RNA and protein-DNA interactions; thereby, the binding complexes can be pulled down with streptavidin-conjugated resins and identified by Western blotting. However, setting up RNA and DNA pull-down assays with biotinylated probes in optimum protein binding conditions remains challenging. Here, we demonstrate the step-by step optimization of pull-down for IRP (iron-responsive-element-binding protein) with a 5'-biotinylated stem-loop IRE (iron-responsive element) RNA, HuR, and AUF1 with an AU-rich RNA element and Nrf2 binding to an antioxidant-responsive element (ARE) enhancer in the human ferritin H gene. This study was designed to address key technical questions in RNA and DNA pull-down assays: (1) how much RNA and DNA probes we should use; (2) what binding buffer and cell lysis buffer we can use; (3) how to verify the specific interaction; (4) what streptavidin resin (agarose or magnetic beads) works; and (5) what Western blotting results we can expect from varying to optimum conditions. We anticipate that our optimized pull-down conditions can be applicable to other RNA- and DNA-binding proteins along with emerging non-coding small RNA-binding proteins for their in vitro characterization.
Collapse
Affiliation(s)
- Yoshiaki Tsuji
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, NC 27695, USA
| |
Collapse
|
10
|
Differential Diagnosis of Hyperferritinemia in Critically Ill Patients. J Clin Med 2022; 12:jcm12010192. [PMID: 36614993 PMCID: PMC9821140 DOI: 10.3390/jcm12010192] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/16/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Elevated serum ferritin is a common condition in critically ill patients. It is well known that hyperferritinemia constitutes a good biomarker for hemophagocytic lymphohistiocytosis (HLH) in critically ill patients. However, further differential diagnoses of hyperferritinemia in adult critically ill patients remain poorly investigated. We sought to systematically investigate hyperferritinemia in adult critically ill patients without HLH. METHODS In this secondary analysis of a retrospective observational study, patients ≥18 years admitted to at least one adult intensive care unit at Charité-Universitätsmedizin Berlin between January 2006 and August 2018, and with hyperferritinemia of ≥500 μg/L were included. Patients with HLH were excluded. All patients were categorized into non-sepsis, sepsis, and septic shock. They were also classified into 17 disease groups, based on their ICD-10 codes, and pre-existing immunosuppression was determined. Uni- and multivariable linear regression analyses were performed in all patients. RESULTS A total of 2583 patients were analyzed. Multivariable linear regression analysis revealed positive associations of maximum SOFA score, sepsis or septic shock, liver disease (except hepatitis), and hematological malignancy with maximum ferritin. T/NK cell lymphoma, acute myeloblastic leukemia, Kaposi's sarcoma, acute or subacute liver failure, and hepatic veno-occlusive disease were positively associated with maximum ferritin in post-hoc multivariable linear regression analysis. CONCLUSIONS Sepsis or septic shock, liver disease (except hepatitis) and hematological malignancy are important differential diagnoses in hyperferritinemic adult critically ill patients without HLH. Together with HLH, they complete the quartet of important differential diagnoses of hyperferritinemia in adult critically ill patients. As these conditions are also related to HLH, it is important to apply HLH-2004 criteria for exclusion of HLH in hyperferritinemic patients. Hyperferritinemic critically ill patients without HLH require quick investigation of differential diagnoses.
Collapse
|
11
|
Xie CH, Chen LW, Lin CL, Hu CC, Chien CH. Serum Uric Acid but Not Ferritin Level Is Associated with Hepatic Fibrosis in Lean Subjects with Metabolic Dysfunction-Associated Fatty Liver Disease: A Community-Based Study. J Pers Med 2022; 12:jpm12122009. [PMID: 36556230 PMCID: PMC9782820 DOI: 10.3390/jpm12122009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Elevated serum ferritin and uric acid levels are common in patients with fatty liver disease. This study assessed the association between serum ferritin and uric acid levels and liver fibrosis in subjects with lean metabolic dysfunction-associated fatty liver disease (MAFLD). This cross-sectional study used data from a community screening examination for metabolic syndrome from December 2018 to September 2019 at Keelung Chang Gung Memorial Hospital. Subjects with lean MAFLD were defined as those with a body mass index (BMI) < 23 kg/m2 and hepatic steatosis according to the MAFLD criteria. A total of 182 lean subjects were included and were divided into lean MAFLD and lean healthy groups. Serum ferritin and uric acid concentrations were positively correlated with liver fibrosis, regardless of whether FIB-4, APRI, or NFS were used as references. Univariate logistic regression analysis showed that age and uric acid were associated with advanced liver fibrosis. After adjusting for potential confounders, only uric acid level was statistically significant in predicting the advanced liver fibrosis (OR = 6.907 (1.111−42.94), p = 0.038) in the lean MAFLD group. We found that an elevated serum uric acid level is an independent factor associated with advanced liver fibrosis in lean MAFLD subjects by noninvasive fibrosis scores.
Collapse
Affiliation(s)
- Cheng-Han Xie
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
| | - Li-Wei Chen
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
- Community Medicine Research Center, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
- Correspondence: ; Tel.: +886-2-24313131 (ext. 6203); Fax: +886-2-24335342
| | - Chih-Lang Lin
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
- Community Medicine Research Center, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
| | - Ching-Chih Hu
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
- Community Medicine Research Center, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
| | - Cheng-Hung Chien
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
- Community Medicine Research Center, Chang-Gung Memorial Hospital and University, Keelung 204, Taiwan
| |
Collapse
|
12
|
Li S, Wang R, Wang Y, Liu Y, Qiao Y, Li P, Chen J, Pan S, Feng Q, Liu Z, Liu D. Ferroptosis: A new insight for treatment of acute kidney injury. Front Pharmacol 2022; 13:1065867. [PMID: 36467031 PMCID: PMC9714487 DOI: 10.3389/fphar.2022.1065867] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/07/2022] [Indexed: 09/16/2023] Open
Abstract
Acute kidney injury (AKI), one of the most prevalent clinical diseases with a high incidence rate worldwide, is characterized by a rapid deterioration of renal function and further triggers the accumulation of metabolic waste and toxins, leading to complications and dysfunction of other organs. Multiple pathogenic factors, such as rhabdomyolysis, infection, nephrotoxic medications, and ischemia-reperfusion injury, contribute to the onset and progression of AKI. However, the detailed mechanism remains unclear. Ferroptosis, a recently identified mechanism of nonapoptotic cell death, is iron-dependent and caused by lipid peroxide accumulation in cells. A variety of studies have demonstrated that ferroptosis plays a significant role in AKI development, in contrast to other forms of cell death, such as apoptosis, necroptosis, and pyroptosis. In this review, we systemically summarized the definition, primary biochemical mechanisms, key regulators and associated pharmacological research progress of ferroptosis in AKI. We further discussed its therapeutic potential for the prevention of AKI, in the hope of providing a useful reference for further basic and clinical studies.
Collapse
Affiliation(s)
- Shiyang Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Rui Wang
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yixue Wang
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yong Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Yingjin Qiao
- Blood Purification Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peipei Li
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Jingfang Chen
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shaokang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Qi Feng
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dongwei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Research Institute of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| |
Collapse
|
13
|
Sudarev VV, Dolotova SM, Bukhalovich SM, Bazhenov SV, Ryzhykau YL, Uversky VN, Bondarev NA, Osipov SD, Mikhailov AE, Kuklina DD, Murugova TN, Manukhov IV, Rogachev AV, Gordeliy VI, Gushchin IY, Kuklin AI, Vlasov AV. Ferritin self-assembly, structure, function, and biotechnological applications. Int J Biol Macromol 2022; 224:319-343. [DOI: 10.1016/j.ijbiomac.2022.10.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
|
14
|
Silva RCMC, Vasconcelos LR, Travassos LH. The different facets of heme-oxygenase 1 in innate and adaptive immunity. Cell Biochem Biophys 2022; 80:609-631. [PMID: 36018440 DOI: 10.1007/s12013-022-01087-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 07/20/2022] [Indexed: 11/26/2022]
Abstract
Heme oxygenase (HO) enzymes are responsible for the main oxidative step in heme degradation, generating equimolar amounts of free iron, biliverdin and carbon monoxide. HO-1 is induced as a crucial stress response protein, playing protective roles in physiologic and pathological conditions, due to its antioxidant, anti-apoptotic and anti-inflammatory effects. The mechanisms behind HO-1-mediated protection are being explored by different studies, affecting cell fate through multiple ways, such as reduction in intracellular levels of heme and ROS, transcriptional regulation, and through its byproducts generation. In this review we focus on the interplay between HO-1 and immune-related signaling pathways, which culminate in the activation of transcription factors important in immune responses and inflammation. We also discuss the dual interaction of HO-1 and inflammatory mediators that govern resolution and tissue damage. We highlight the dichotomy of HO-1 in innate and adaptive immune cells development and activation in different disease contexts. Finally, we address different known anti-inflammatory pharmaceuticals that are now being described to modulate HO-1, and the possible contribution of HO-1 in their anti-inflammatory effects.
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Luiz Ricardo Vasconcelos
- Cellular Signaling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Leonardo Holanda Travassos
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
15
|
Li F, Wang H, Chen H, Guo J, Dang X, Ru Y, Wang H. Mechanism of Ferroptosis and Its Role in Spinal Cord Injury. Front Neurol 2022; 13:926780. [PMID: 35756929 PMCID: PMC9218271 DOI: 10.3389/fneur.2022.926780] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a non-necrotic form of regulated cell death (RCD) that is primarily characterized by iron-dependent membrane lipid peroxidation and is regulated by cysteine transport, glutathione synthesis, and glutathione peroxidase 4 function as well as other proteins including ferroptosis suppressor protein 1. It has been found that ferroptosis played an important role in many diseases, such as neurodegenerative diseases and ischemia-reperfusion injury. Spinal cord injury (SCI), especially traumatic SCI, is an urgent problem worldwide due to its high morbidity and mortality, as well as the destruction of functions of the human body. Various RCDs, including ferroptosis, are found in SCI. Different from necrosis, since RCD is a form of cell death regulated by various molecular mechanisms in cells, the study of the role played by RCD in SCI will contribute to a deeper understanding of the pathophysiological process, as well as the treatment and functional recovery. The present review mainly introduces the main mechanism of ferroptosis and its role in SCI, so as to provide a new idea for further exploration.
Collapse
Affiliation(s)
- Fei Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haifan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Chen
- Basic Medical Science Academy, The Air Force Medical University, Xi'an, China
| | - Jianing Guo
- Basic Medical Science Academy, The Air Force Medical University, Xi'an, China
| | - Xiaoqian Dang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Ru
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Basic Medical Science Academy, The Air Force Medical University, Xi'an, China
| | - Haoyu Wang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
16
|
Banga R, Banga V, Eltalla A, Shahin L, Parag S, Naim M, Iyer E, Kumrah N, Zacharias B, Nathanson L, Beljanski V. Effects of autophagy modulators tamoxifen and chloroquine on the expression profiles of long non-coding RNAs in MIAMI cells exposed to IFNγ. PLoS One 2022; 17:e0266179. [PMID: 35446871 PMCID: PMC9022845 DOI: 10.1371/journal.pone.0266179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/15/2022] [Indexed: 11/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) can be utilized clinically for treatment of conditions that result from excessive inflammation. In a pro-inflammatory environment, MSCs adopt an anti-inflammatory phenotype resulting in immunomodulation. A sub-type of MSCs referred to as “marrow-isolated adult multilineage inducible” (MIAMI) cells, which were isolated from bone marrow, were utilized to show that the addition of autophagy modulators, tamoxifen (TX) or chloroquine (CQ), can alter how MIAMI cells respond to IFNγ exposure in vitro resulting in an increased immunoregulatory capacity of the MIAMI cells. Molecularly, it was also shown that TX and CQ each alter both the levels of immunomodulatory genes and microRNAs which target such genes. However, the role of other non-coding RNAs (ncRNAs) such as long non-coding RNAs (lncRNAs) in regulating the response of MSCs to inflammation has been poorly studied. Here, we utilized transcriptomics and data mining to analyze the putative roles of various differentially regulated lncRNAs in MIAMI cells exposed to IFNγ with (or without) TX or CQ. The aim of this study was to investigate how the addition of TX and CQ alters lncRNA levels and evaluate how such changes could alter previously observed TX- and CQ-driven changes to the immunomodulatory properties of MIAMI cells. Data analysis revealed 693 long intergenic non-coding RNAS (lincRNAs), 480 pseudogenes, and 642 antisense RNAs that were differentially regulated with IFNγ, IFNγ+TX and IFNγ+CQ treatments. Further analysis of these RNA species based on the existing literature data revealed 6 antisense RNAs, 2 pseudogenes, and 5 lincRNAs that have the potential to modulate MIAMI cell’s response to IFNγ treatment. Functional analysis of these genomic species based on current literature linking inflammatory response and ncRNAs indicated their potential for regulation of several key pro- and anti-inflammatory responses, including NFκB signaling, cytokine secretion and auto-immune responses. Overall, this work found potential involvement of multiple pro-and anti-inflammatory pathways and molecules in modulating MIAMI cells’ response to inflammation.
Collapse
Affiliation(s)
- Rajkaran Banga
- Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, Florida
| | - Veerkaran Banga
- Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, Florida
| | - Amr Eltalla
- Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, Florida
| | - Lauren Shahin
- Dr Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, Florida
| | - Sonam Parag
- Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, Florida
| | - Maha Naim
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, Davie, Florida
| | - Easha Iyer
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, Davie, Florida
| | - Neha Kumrah
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, Davie, Florida
| | - Brian Zacharias
- Department of Biological Sciences, Halmos College of Arts and Sciences, Nova Southeastern University, Fort Lauderdale, Davie, Florida
| | - Lubov Nathanson
- Dr Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, Florida
- Institute for Neuroimmune Medicine, Dr Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, Florida
| | - Vladimir Beljanski
- Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, Florida
- Cell Therapy Institute, Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, Florida
- * E-mail:
| |
Collapse
|
17
|
Zhao Y, Liu L, Liu JB, Wu CY, Duan DY, Cheng TY. Cloning, expression, and function of ferritins in the tick Haemaphysalis flava. Ticks Tick Borne Dis 2021; 13:101892. [PMID: 34942560 DOI: 10.1016/j.ttbdis.2021.101892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 10/19/2022]
Abstract
The full-length cDNA of two ferritins of Haemaphysalis flava were cloned after which recombinant Hf-FER1 and Hf-FER2 were expressed and their function was analyzed. In addition, RNA interference (RNAi) based on the injection of Hf-fer1 or Hf-fer2 dsRNA into fully engorged female ticks was performed. The cDNA encoding Hf-FER1 is 834 bp in length. It contains an iron-responsive element in the 5' untranslated region and encodes 174 amino acid residues. The full-length cDNA of Hf-FER2 contains 696 bp and encodes 199 amino acids, including a putative signal peptide sequence. Hf-FER1 and Hf-FER2 both have the ferroxidase iron center and the ferrihydrite nucleation center. The evolutionary relationship of Hf-FER1 and Hf-FER2 was established, and the predicted quaternary structures were assembled as typical spherical shells composed of 24 subunits which was demonstrated by nature PAGE. Real-time PCR showed that Hf-fer1 and Hf-fer2 were expressed in all developmental stages, with the highest expression in fully engorged females. The expression of Hf-fer1 and Hf-fer2 were relatively high in unfed larvae. Hf-fer1 was expressed in all tissues and was especially abundant in the salivary glands of fully engorged females. In contrast, the highest levels of Hf-fer2 were found in the midgut of fully engorged females, and no expression was found in the salivary glands of this life stage. Both recombinant Hf-FER1 and Hf-FER2 had iron-binding capabilities. Silencing of both Hf-fer1 and Hf-fer2 affected fecundity. Compared to the control, the percentage of ticks that laid eggs in the Hf-fer1 and Hf-fer2 RNAi groups was 73.3% and 66.7%, respectively. The silenced ticks that laid eggs had lower egg weight to body weight ratios, and the eggs had abnormal morphologies. The hatchability of eggs with normal morphology in the Hf-fer1 and Hf-fer2 silenced groups was 47.8% and 22.8%, respectively, which was significantly different from the control group (P < 0.005). These findings indicate that Hf-FER1 and Hf-FER2 play important roles in the iron storage of H. flava.
Collapse
Affiliation(s)
- Yu Zhao
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China; College of Animal Science and Veterinary Medicine, Xinyang Agriculture and Forestry University, Xinyang 464000, Henan, China
| | - Lei Liu
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Jin-Bao Liu
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Cong-Ying Wu
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - De-Yong Duan
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Tian-Yin Cheng
- Research Center for Parasites & Vectors, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China.
| |
Collapse
|
18
|
Duarte TL, Talbot NP, Drakesmith H. NRF2 and Hypoxia-Inducible Factors: Key Players in the Redox Control of Systemic Iron Homeostasis. Antioxid Redox Signal 2021; 35:433-452. [PMID: 32791852 DOI: 10.1089/ars.2020.8148] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Oxygen metabolism and iron homeostasis are closely linked. Iron facilitates the oxygen-carrying capacity of blood, and its deficiency causes anemia. Conversely, excess free iron is detrimental for stimulating the formation of reactive oxygen species, causing tissue damage. The amount and distribution of iron thus need to be tightly regulated by the liver-expressed hormone hepcidin. This review analyzes the roles of key oxygen-sensing pathways in cellular and systemic regulation of iron homeostasis; specifically, the prolyl hydroxylase domain (PHD)/hypoxia-inducible factor (HIF) and the Kelch-like ECH-associated protein 1/NF-E2 p45-related factor 2 (KEAP1/NRF2) pathways, which mediate tissue adaptation to low and high oxygen, respectively. Recent Advances: In macrophages, NRF2 regulates genes involved in hemoglobin catabolism, iron storage, and iron export. NRF2 was recently identified as the molecular sensor of iron-induced oxidative stress and is responsible for BMP6 expression by liver sinusoidal endothelial cells, which in turn activates hepcidin synthesis by hepatocytes to restore systemic iron levels. Moreover, NRF2 orchestrates the activation of antioxidant defenses that are crucial to protect against iron toxicity. On the contrary, low iron/hypoxia stabilizes renal HIF2a via inactivation of iron-dependent PHD dioxygenases, causing an erythropoietic stimulus that represses hepcidin via an inhibitory effect of erythroferrone on bone morphogenetic proteins. Intestinal HIF2a is also stabilized, increasing the expression of genes involved in dietary iron absorption. Critical Issues: An intimate crosstalk between oxygen-sensing pathways and iron regulatory mechanisms ensures that fluctuations in systemic iron levels are promptly detected and restored. Future Directions: The realization that redox-sensitive transcription factors regulate systemic iron levels suggests novel therapeutic approaches. Antioxid. Redox Signal. 35, 433-452.
Collapse
Affiliation(s)
- Tiago L Duarte
- Instituto de Biologia Molecular e Celular, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Nick P Talbot
- Department of Physiology, Anatomy & Genetics, University of Oxford, Oxford, United Kingdom
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Haematology Theme, Oxford Biomedical Research Centre, Oxford, United Kingdom
| |
Collapse
|
19
|
Schmidlin CJ, Shakya A, Dodson M, Chapman E, Zhang DD. The intricacies of NRF2 regulation in cancer. Semin Cancer Biol 2021; 76:110-119. [PMID: 34020028 DOI: 10.1016/j.semcancer.2021.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023]
Abstract
The complex role of NRF2 in the context of cancer continues to evolve. As a transcription factor, NRF2 regulates various genes involved in redox homeostasis, protein degradation, DNA repair, and xenobiotic metabolism. As such, NRF2 is critical in preserving cell function and viability, particularly during stress. Importantly, NRF2 itself is regulated via a variety of mechanisms, and the mode of NRF2 activation often dictates the duration of NRF2 signaling and its role in either preventing cancer initiation or promoting cancer progression. Herein, different modes of NRF2 regulation, including oxidative stress, autophagy dysfunction, protein-protein interactions, and epigenetics, as well as pharmacological modulators targeting this cascade in cancer, are explored. Specifically, how the timing and duration of these different mechanisms of NRF2 induction affect tumor initiation, progression, and metastasis are discussed. Additionally, progress in the discovery and development of NRF2 inhibitors for the treatment of NRF2-addicted cancers is highlighted, including modulators that inhibit specific NRF2 downstream targets. Overall, a better understanding of the intricate nature of NRF2 regulation in specific cancer contexts should facilitate the generation of novel therapeutics designed to not only prevent tumor initiation, but also halt progression and ultimately improve patient wellbeing and survival.
Collapse
Affiliation(s)
- Cody J Schmidlin
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Aryatara Shakya
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Matthew Dodson
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Eli Chapman
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Donna D Zhang
- Deparment of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
20
|
Si SJ, Tasian SK, Bassiri H, Fisher BT, Atalla J, Patel R, Romberg N, Lambert MP, Paessler M, Behrens EJ, Teachey DT, Sullivan KE. Diagnostic Challenges in Pediatric Hemophagocytic Lymphohistiocytosis. J Clin Immunol 2021; 41:1213-1218. [PMID: 33761058 PMCID: PMC7988244 DOI: 10.1007/s10875-021-01025-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/15/2021] [Indexed: 11/20/2022]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a syndrome of severe immune dysregulation that encompasses a broad range of underlying genetic diseases and infectious triggers. Monogenic conditions, autoimmune diseases, and infections can all drive the phenotype of HLH and associated immune hyperactivation with hypercytokinemia. A diagnosis of HLH usually requires a combination of clinical and laboratory findings; there is no single sensitive and specific diagnostic test, which often leads to “diagnostic dilemmas” and delays in treatment initiation. Ferritin levels, one of the most commonly used screening tests, were collected across a large tertiary care pediatric hospital to identify the positive predictive value for HLH. Herein, we present several cases that illustrate the clinical challenges of confirming an HLH diagnosis. Additionally, we report on the utility of establishing a formal multi-disciplinary group to aid the prompt diagnosis and treatment of patients presenting with HLH-like pathophysiologies.
Collapse
Affiliation(s)
- Stephanie J Si
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Division of Pediatric Oncology-Hematology, University of Hawai'i Cancer Center, Kapi'olani Medical Center for Women and Children, Honolulu, HI, USA. .,University of Hawai'i School of Medicine, 701 Ilalo St, Honolulu, HI, 96813, USA.
| | - Sarah K Tasian
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hamid Bassiri
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brian T Fisher
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jasmyn Atalla
- Department of Internal Medicine and Department of Pediatrics, Vidant Medical Center, East Carolina University, Greenville, NC, USA
| | - Reema Patel
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Neil Romberg
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michele P Lambert
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michele Paessler
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Edward J Behrens
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David T Teachey
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen E Sullivan
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
21
|
Kianpour S, Ebrahiminezhad A, Heidari R, Khalvati B, Shahbazi MA, Negahdaripour M, Mohkam M, Aghaei R, Berenjian A, Niknezhad SV, Ghasemi Y. Enterobacter sp. Mediated Synthesis of Biocompatible Nanostructured Iron-Polysaccharide Complexes: a Nutritional Supplement for Iron-Deficiency Anemia. Biol Trace Elem Res 2020; 198:744-755. [PMID: 32157632 DOI: 10.1007/s12011-020-02101-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/27/2020] [Indexed: 01/01/2023]
Abstract
FDA has approved iron oxide nanoparticles (IONs) coated with organic compounds as a safe material with less toxic effects compared with the naked metal ions and nanoparticles. In this study, the biological and physicochemical characteristics of a nanostructured iron-polysaccharide complexes (Nano-IPC) biosynthesized by Enterobacter sp. were evaluated. Furthermore, the serum biochemical parameters, tissue iron level, red blood cell parameters, and organ ferritin of rats were measured for investigating the effect of the Nano-IPCs in comparison with FeSO4 as a supplement for iron deficiency. The biosafety data demonstrated 35% increment of viability in Hep-G2 hepatocarcinoma cell lines when treated with nanoparticles (500 μg/mL) for 24 h. Besides, iron concentration in serum and tissue as well as the expression of ferritin L subunit in animals treated with the Nano-IPCs supplement were meaningfully higher than the FeSO4-supplemented and negative control animals. Moreover, the expression level of ferritin H subunit and biochemical factors remained similar to the negative control animals in the Nano-IPC-supplemented group. These results indicated that Nano-IPCs can be considered as a nontoxic supplement for patients carrying iron-deficiency anemia (IDA).
Collapse
Affiliation(s)
- Sedigheh Kianpour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 1583, 71345 Karafarin Street, Shiraz, Iran
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Ebrahiminezhad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Khalvati
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, 4513956184, Iran
| | - Manica Negahdaripour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 1583, 71345 Karafarin Street, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Mohkam
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 1583, 71345 Karafarin Street, Shiraz, Iran
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayyeh Aghaei
- Department of Marine Chemistry, Faculty of Marine Science, Chabahar Maritime University, Chabahar, Iran
| | - Aydin Berenjian
- School of Engineering, Faculty of Science and Engineering, University of Waikato, Hamilton, New Zealand
| | - Seyyed Vahid Niknezhad
- Biotechnology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 1583, 71345 Karafarin Street, Shiraz, Iran.
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
22
|
Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron. Cell Death Dis 2020; 11:787. [PMID: 32968051 PMCID: PMC7511955 DOI: 10.1038/s41419-020-02950-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/20/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
The consumption of red meat is probably carcinogenic to humans and is associated with an increased risk to develop colorectal cancer (CRC). Red meat contains high amounts of heme iron, which is thought to play a causal role in tumor formation. In this study, we investigated the genotoxic and cytotoxic effects of heme iron (i.e., hemin) versus inorganic iron in human colonic epithelial cells (HCEC), human CRC cell lines and murine intestinal organoids. Hemin catalyzed the formation of reactive oxygen species (ROS) and induced oxidative DNA damage as well as DNA strand breaks in both HCEC and CRC cells. In contrast, inorganic iron hardly affected ROS levels and only slightly increased DNA damage. Hemin, but not inorganic iron, caused cell death and reduced cell viability. This occurred preferentially in non-malignant HCEC, which was corroborated in intestinal organoids. Both hemin and inorganic iron were taken up into HCEC and CRC cells, however with differential kinetics and efficiency. Hemin caused stabilization and nuclear translocation of Nrf2, which induced heme oxygenase-1 (HO-1) and ferritin heavy chain (FtH). This was not observed after inorganic iron treatment. Chemical inhibition or genetic knockdown of HO-1 potentiated hemin-triggered ROS generation and oxidative DNA damage preferentially in HCEC. Furthermore, HO-1 abrogation strongly augmented the cytotoxic effects of hemin in HCEC, revealing its pivotal function in colonocytes and highlighting the toxicity of free intracellular heme iron. Taken together, this study demonstrated that hemin, but not inorganic iron, induces ROS and DNA damage, resulting in a preferential cytotoxicity in non-malignant intestinal epithelial cells. Importantly, HO-1 conferred protection against the detrimental effects of hemin.
Collapse
|
23
|
Torti SV, Torti FM. Iron and Cancer: 2020 Vision. Cancer Res 2020; 80:5435-5448. [PMID: 32928919 DOI: 10.1158/0008-5472.can-20-2017] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/06/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022]
Abstract
New and provocative insights into the relationships between iron and cancer have been uncovered in recent years. These include delineation of connections that link cellular iron to DNA repair, genomic integrity, and oncogenic signaling as well as the discovery of ferroptosis, a novel iron-dependent form of cell death. In parallel, new molecules and pathways that regulate iron influx, intracellular iron trafficking, and egress in normal cells, and their perturbations in cancer have been discovered. In addition, insights into the unique properties of iron handling in tumor-initiating cells (cancer stem cells), novel contributions of the tumor microenvironment to the uptake and regulation of iron in cancer cells, and new therapeutic modalities that leverage the iron dependence of cancer have emerged.
Collapse
Affiliation(s)
- Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut.
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
24
|
Żychowska M, Grzybkowska A, Wiech M, Urbański R, Pilch W, Piotrowska A, Czerwińska-Ledwig O, Antosiewicz J. Exercise Training and Vitamin C Supplementation Affects Ferritin mRNA in Leukocytes without Affecting Prooxidative/Antioxidative Balance in Elderly Women. Int J Mol Sci 2020; 21:ijms21186469. [PMID: 32899447 PMCID: PMC7554744 DOI: 10.3390/ijms21186469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/24/2022] Open
Abstract
Physical training and antioxidant supplementation may influence iron metabolism through reduced oxidative stress and subsequent lowering of mRNA levels of genes that are easily induced by this stress, including those responsible for iron homeostasis. Fifteen elderly women participated in our 12-week experiment, involving six weeks of training without supplementation and six weeks of training supported by oral supplementation of 1000 mg of vitamin C daily. The participants were divided into two groups (n = 7 in group 1 and n = 8 in group 2). In group 1, we applied vitamin C supplementation in the first six weeks of training, while in group 2 during the remaining six weeks of training. In both phases, the health-related training occurred three times per week. Training accompanied by vitamin C supplementation did not affect prooxidative/antioxidative balance but significantly decreased ferritin heavy chain (FTH) and ferritin light chain (FTL) mRNA in leukocytes (for FTH mRNA from 2^64.24 to 2^11.06, p = 0.03 in group 1 and from 2^60.54 to 2^16.03, p = 0.01 in group 2, for FTL mRNA from 2^20.22 to 2^4.53, p = 0.01 in group 2). We concluded that vitamin C supplementation might have caused a decrease in gene expression of two important antioxidative genes (FTH, FTL) and had no effect on plasma prooxidative/antioxidative balance.
Collapse
Affiliation(s)
- Małgorzata Żychowska
- Department of Sport, Faculty of Physical Education, Kazimierz Wielki University in Bydgoszcz, 85-064 Bydgoszcz, Poland
- Department of Biochemistry, Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
- Correspondence: (M.Ż.); (J.A.); Tel.: +48-881-555-337 (M.Ż. & J.A.)
| | - Agata Grzybkowska
- Department of Biochemistry, Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
| | - Monika Wiech
- Department of Health Promotion, Faculty of Tourism and Recreation, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
| | - Robert Urbański
- Department of Biomechanics and Sports Engineering, Faculty of Physical Education, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland;
| | - Wanda Pilch
- Department of Cosmetology, Faculty of Physiotherapy, University of Physical Education in Krakow, 31-571 Krakow, Poland; (W.P.); (A.P.); (O.C.-L.)
| | - Anna Piotrowska
- Department of Cosmetology, Faculty of Physiotherapy, University of Physical Education in Krakow, 31-571 Krakow, Poland; (W.P.); (A.P.); (O.C.-L.)
| | - Olga Czerwińska-Ledwig
- Department of Cosmetology, Faculty of Physiotherapy, University of Physical Education in Krakow, 31-571 Krakow, Poland; (W.P.); (A.P.); (O.C.-L.)
| | - Jędrzej Antosiewicz
- Department of Bioenergetics and Exercise Physiology, Faculty of Health, Medical University of Gdansk, 80-210 Gdansk, Poland
- Correspondence: (M.Ż.); (J.A.); Tel.: +48-881-555-337 (M.Ż. & J.A.)
| |
Collapse
|
25
|
Chen MH, Lin YT, Lai CC, Chou LS, Chen CY. Tissue concentrations of Fe, Zn, Cu and Mn of four Taiwanese toothed cetaceans. MARINE POLLUTION BULLETIN 2020; 158:111094. [PMID: 32753165 DOI: 10.1016/j.marpolbul.2020.111094] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 06/11/2023]
Abstract
Concentrations of Fe, Mn, Zn, and Cu in the muscle, lung, liver, and kidney tissues of 49 cetaceans, including 11 Kogia sima (Ks), 10 Lagenodelphis hosei (Lh), 14 Grampus griseus (Gg), and 14 Stenella attenuata (Sa) from 1994 to 2012 in Taiwan were measured. Ks exhibited the highest Fe, and the lowest Zn, Cu and Mn tissue concentrations. The Kogiid and Delphinid groups were significantly categorized by nMDS analysis. Fe muscle concentrations were found to be positively correlated with Ks, Lh, and Gg but not Sa body length. The different levels of Zn-, Cu-, and Mn- tissue concentrations of the two families were due to their different SOD systems. Their calves contained higher Cu liver concentrations. The four essential elements in cetaceans regulate homeostasis to meet their eco-physiological demand. The baseline levels for these four elements in these four tissues in the Delphinid group are defined.
Collapse
Affiliation(s)
- Meng-Hsien Chen
- Department of Oceanography (Marine Biology group), National Sun Yat-sen University, 804 Kaohsiung, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 807 Kaohsiung, Taiwan.
| | - Yu-Ting Lin
- Department of Oceanography (Marine Biology group), National Sun Yat-sen University, 804 Kaohsiung, Taiwan
| | - Chien-Cheng Lai
- Department of Oceanography (Marine Biology group), National Sun Yat-sen University, 804 Kaohsiung, Taiwan
| | - Lien-Siang Chou
- Institute of Ecology and Evolutionary Biology, National Taiwan University, 106 Taipei, Taiwan
| | - Chiee-Young Chen
- Department of Marine Environmental Engineering, National Kaohsiung University, 811 Kaohsiung, Taiwan
| |
Collapse
|
26
|
Ikeda Y, Watanabe H, Shiuchi T, Hamano H, Horinouchi Y, Imanishi M, Goda M, Zamami Y, Takechi K, Izawa-Ishizawa Y, Miyamoto L, Ishizawa K, Aihara KI, Tsuchiya K, Tamaki T. Deletion of H-ferritin in macrophages alleviates obesity and diabetes induced by high-fat diet in mice. Diabetologia 2020; 63:1588-1602. [PMID: 32430665 DOI: 10.1007/s00125-020-05153-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/09/2020] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Iron accumulation affects obesity and diabetes, both of which are ameliorated by iron reduction. Ferritin, an iron-storage protein, plays a crucial role in iron metabolism. H-ferritin exerts its cytoprotective action by reducing toxicity via its ferroxidase activity. We investigated the role of macrophage H-ferritin in obesity and diabetes. METHODS Conditional macrophage-specific H-ferritin (Fth, also known as Fth1) knockout (LysM-Cre Fth KO) mice were used and divided into four groups: wild-type (WT) and LysM-Cre Fth KO mice with normal diet (ND), and WT and LysM-Cre Fth KO mice with high-fat diet (HFD). These mice were analysed for characteristics of obesity and diabetes, tissue iron content, inflammation, oxidative stress, insulin sensitivity and metabolic measurements. RAW264.7 macrophage cells were used for in vitro experiments. RESULTS Iron concentration reduced, and mRNA expression of ferroportin increased, in macrophages from LysM-Cre Fth KO mice. HFD-induced obesity was lower in LysM-Cre Fth KO mice than in WT mice at 12 weeks (body weight: KO 34.6 ± 5.6 g vs WT 40.1 ± 5.2 g). mRNA expression of inflammatory cytokines and infiltrated macrophages and oxidative stress increased in the adipose tissue of HFD-fed WT mice, but was not elevated in HFD-fed LysM-Cre Fth KO mice. However, WT mice fed an HFD had elevated iron concentration in adipose tissue and spleen, which was not observed in LysM-Cre Fth KO mice fed an HFD (adipose tissue [μmol Fe/g protein]: KO 1496 ± 479 vs WT 2316 ± 866; spleen [μmol Fe/g protein]: KO 218 ± 54 vs WT 334 ± 83). Moreover, HFD administration impaired both glucose tolerance and insulin sensitivity in WT mice, which was ameliorated in LysM-Cre Fth KO mice. In addition, energy expenditure, mRNA expression of thermogenic genes, and body temperature were higher in KO mice with HFD than WT mice with HFD. In vitro experiments showed that iron content was reduced, and lipopolysaccharide-induced Tnf-α (also known as Tnf) mRNA upregulation was inhibited in a macrophage cell line transfected with Fth siRNA. CONCLUSIONS/INTERPRETATION Deletion of macrophage H-ferritin suppresses the inflammatory response by reducing intracellular iron levels, resulting in the prevention of HFD-induced obesity and diabetes. The findings from this study highlight macrophage iron levels as a potential therapeutic target for obesity and diabetes.
Collapse
Affiliation(s)
- Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan.
| | - Hiroaki Watanabe
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
- Department of Clinical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Hirofumi Hamano
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yuya Horinouchi
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Masaki Imanishi
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Mitsuhiro Goda
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yoshito Zamami
- Department of Clinical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Kenshi Takechi
- Clinical Trial Center for Developmental Therapeutics, Tokushima University Hospital, Tokushima, Japan
| | | | - Licht Miyamoto
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Keisuke Ishizawa
- Department of Clinical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Ken-Ichi Aihara
- Department of Community Medicine for Diabetes and Metabolic Disorders, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Toshiaki Tamaki
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
- Anan Medical Center, Tokushima, Japan
| |
Collapse
|
27
|
Yokel RA, Tseng MT, Butterfield DA, Hancock ML, Grulke EA, Unrine JM, Stromberg AJ, Dozier AK, Graham UM. Nanoceria distribution and effects are mouse-strain dependent. Nanotoxicology 2020; 14:827-846. [DOI: 10.1080/17435390.2020.1770887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Robert A. Yokel
- Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Michael T. Tseng
- Anatomical Sciences & Neurobiology, University of Louisville, Louisville, KY, USA
| | | | - Matthew L. Hancock
- Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA
| | - Eric A. Grulke
- Chemical and Materials Engineering, University of Kentucky, Lexington, KY, USA
| | - Jason M. Unrine
- Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA
| | | | | | - Uschi M. Graham
- Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
- CDC, NIOSH, Cincinnati, OH, USA
| |
Collapse
|
28
|
Abstract
Ferritins are evolutionarily conserved proteins that regulate cellular iron metabolism. It is the only intracellular protein that is capable of storing large quantities of iron. Although the ratio of different subunits determines the iron content of each ferritin molecule, the exact mechanism that dictates organization of these subunits still is unclear. In this review, we address renal ferritin expression and its implication in kidney disease. Specifically, we address the role of ferritin subunits in preventing kidney injury and also promoting tolerance against infection-associated kidney injury. We describe functions for ferritin that are independent of its ability to ferroxidize and store iron. We further discuss the implications of ferritin in body fluids, including blood and urine, during inflammation and kidney disease. Although there are several in-depth review articles on ferritin in the context of iron metabolism, we chose to focus on the role of ferritin particularly in kidney health and disease and highlight unanswered questions in the field.
Collapse
Affiliation(s)
- Kayla McCullough
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Subhashini Bolisetty
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL.
| |
Collapse
|
29
|
Wu YH, Wang SY, Li MX, He H, Yin WJ, Guo YH, Zhang HQ, Sun ZM, Zhang D, Wang X, Sun SY, Tang SX, Du R, Zhang CH. Serum Ferritin Independently Predicts the Incidence of Chronic Kidney Disease in Patients with Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2020; 13:99-105. [PMID: 32021356 PMCID: PMC6970239 DOI: 10.2147/dmso.s228335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/29/2019] [Indexed: 01/20/2023] Open
Abstract
AIM This study aimed to determine whether serum ferritin (SF) is an independent risk factor of the incidence of chronic kidney disease (CKD) and rapid renal function decline (RFD) in male Tibetan patients with type 2 diabetes mellitus (T2DM). METHODS We performed a retrospective cohort study that included 191 male Tibetan patients with T2DM without CKD. Patients were divided into three groups according to the level of SF. The following outcomes were measured: cumulative incidence of chronic kidney disease [i.e. estimated glomerular filtration rate (eGFR) <60 mL/min per 1.73 m2 and/or urinary albumin/creatine ratio (ACR) ≥30 mg/g] and RFD (i.e. decrease in eGFR of ≥25% from baseline or a decline rate of ≥3 mL/min per 1.73 m2 annually). RESULTS In total, over a median follow-up period of 23 months, 30 (15.7%) and 89 patients (46.6%) developed CKD and RFD. In multivariable Cox models, a 100 ng/mL increment in SF was associated with a 1.12-fold (95% CI: 1.02-1.24) higher adjusted risk for incidence of CKD. The adjusted-HR of CKD was 1.31 (95% CI: 0.38-4.53) and 2.92 (95% CI: 0.87-9.77) for those in tertile 2 and tertile 3, respectively, compared with the patients in tertile 1. However, SF was not significantly associated with RFD (adjusted-HR: 1.06, 95% CI: 0.99-1.14). CONCLUSION Serum ferritin independently predicts the incidence of CKD in male Tibetan patients with T2DM. High levels of serum ferritin may play a role in the pathogenesis leading to the development of CKD in T2DM.
Collapse
Affiliation(s)
- Yun Hong Wu
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Su Yuan Wang
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Ming Xia Li
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Hua He
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Wei Jin Yin
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Yan Hong Guo
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Hui Qin Zhang
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Zeng Mei Sun
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Dan Zhang
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Xi Wang
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Shu Yao Sun
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Shu Xi Tang
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Rong Du
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
| | - Cheng Hui Zhang
- Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, Chengdu, People’s Republic of China
- Correspondence: Cheng Hui Zhang Department of Endocrinology and Metabolism, Hospital of Chengdu Office of People’s Government of Tibetan Autonomous Region, No. 20 Ximianqiao Street, Chengdu, Sichuan610041, People’s Republic of China Email
| |
Collapse
|
30
|
Hernandez EP, Talactac MR, Fujisaki K, Tanaka T. The case for oxidative stress molecule involvement in the tick-pathogen interactions -an omics approach. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 100:103409. [PMID: 31200008 DOI: 10.1016/j.dci.2019.103409] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 06/09/2023]
Abstract
The blood-feeding behavior of ticks has resulted in them becoming one of the most important vectors of disease-causing pathogens. Ticks possess a well-developed innate immune system to counter invading pathogens. However, the coevolution of ticks with tick-borne pathogens has adapted these pathogens to the tick's physiology and immune response through several mechanisms including transcriptional regulation. The recent development in tick and tick-borne disease research greatly involved the "omics" approach. The omics approach takes a look en masse at the different genes, proteins, metabolomes, and the microbiome of the ticks that could be differentiated during pathogen infection. Data from this approach revealed that oxidative stress-related molecules in ticks are differentiated and possibly being exploited by the pathogens to evade the tick's immune response. In this study, we review and discuss transcriptomic and proteomic data for some oxidative stress molecules differentially expressed during pathogen infection. We also discuss metabolomics and microbiome data as well as functional genomics in order to provide insight into the tick-pathogen interaction.
Collapse
Affiliation(s)
- Emmanuel Pacia Hernandez
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan
| | - Melbourne Rio Talactac
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan; Department of Clinical and Population Health, College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Cavite, 4122, Philippines
| | - Kozo Fujisaki
- National Agricultural and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki, 305-0856, Japan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan; Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan.
| |
Collapse
|
31
|
Agarwal HS, Kuttesch JF. Early Testing of Serum Ferritin Facilitates Hemophagocytic Lymphohistiocytosis Diagnosis in Children. J Pediatr Hematol Oncol 2019; 41:e478-e480. [PMID: 30222642 DOI: 10.1097/mph.0000000000001312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The clinical and laboratory features of hemophagocytic lymphohistiocytosis (HLH) are nonspecific that makes the definitive diagnosis of HLH very challenging. The disease is almost universally fatal in the absence of early recognition and appropriate therapy. Elevated serum ferritin level is one of the diagnostic markers of HLH disease. We report the value of testing serum ferritin level early in the disease process in 3 pediatric patients who presented with persistent fever and sepsis-like features. Detection of elevated serum ferritin levels facilitated further testing to confirm the diagnosis of HLH and initiate early therapy with good outcomes.
Collapse
Affiliation(s)
- Hemant S Agarwal
- Department of Pediatrics, University of New Mexico Health Sciences Center, 1 University of New Mexico, Albuquerque, NM
| | | |
Collapse
|
32
|
Rubio-Navarro A, Vázquez-Carballo C, Guerrero-Hue M, García-Caballero C, Herencia C, Gutiérrez E, Yuste C, Sevillano Á, Praga M, Egea J, Cannata P, Cortegano I, de Andrés B, Gaspar ML, Cadenas S, Michalska P, León R, Ortiz A, Egido J, Moreno JA. Nrf2 Plays a Protective Role Against Intravascular Hemolysis-Mediated Acute Kidney Injury. Front Pharmacol 2019; 10:740. [PMID: 31333462 PMCID: PMC6619398 DOI: 10.3389/fphar.2019.00740] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Massive intravascular hemolysis is associated with acute kidney injury (AKI). Nuclear factor erythroid-2-related factor 2 (Nrf2) plays a central role in the defense against oxidative stress by activating the expression of antioxidant proteins. We investigated the role of Nrf2 in intravascular hemolysis and whether Nrf2 activation protected against hemoglobin (Hb)/heme-mediated renal damage in vivo and in vitro. We observed renal Nrf2 activation in human hemolysis and in an experimental model of intravascular hemolysis promoted by phenylhydrazine intraperitoneal injection. In wild-type mice, Hb/heme released from intravascular hemolysis promoted AKI, resulting in decreased renal function, enhanced expression of tubular injury markers (KIM-1 and NGAL), oxidative and endoplasmic reticulum stress (ER), and cell death. These features were more severe in Nrf2-deficient mice, which showed decreased expression of Nrf2-related antioxidant enzymes, including heme oxygenase 1 (HO-1) and ferritin. Nrf2 activation with sulforaphane protected against Hb toxicity in mice and cultured tubular epithelial cells, ameliorating renal function and kidney injury and reducing cell stress and death. Nrf2 genotype or sulforaphane treatment did not influence the severity of hemolysis. In conclusion, our study identifies Nrf2 as a key molecule involved in protection against renal damage associated with hemolysis and opens novel therapeutic approaches to prevent renal damage in patients with severe hemolytic crisis. These findings provide new insights into novel aspects of Hb-mediated renal toxicity and may have important therapeutic implications for intravascular hemolysis-related diseases.
Collapse
Affiliation(s)
- Alfonso Rubio-Navarro
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Cristina Vázquez-Carballo
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Melania Guerrero-Hue
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Cristina García-Caballero
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Carmen Herencia
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | | | - Claudia Yuste
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | - Ángel Sevillano
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital 12 de Octubre, Madrid, Spain
| | - Javier Egea
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain.,Hospital Santa Cristina, Madrid, Spain
| | - Pablo Cannata
- Pathology Department, Fundación Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Isabel Cortegano
- Immunology Department, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Belén de Andrés
- Immunology Department, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María Luisa Gaspar
- Immunology Department, Centro Nacional de Microbiologìa, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Susana Cadenas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Patrycja Michalska
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Rafael León
- Instituto de Investigación Sanitaria-Hospital Universitario de la Princesa, Madrid, Spain.,Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, UAM, Madrid, Spain
| | - Alberto Ortiz
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain
| | - Juan Antonio Moreno
- Renal, Vascular and Diabetes Research Lab, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma University, Madrid, Spain.,Department of Cell Biology, Physiology and Immunology, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, Cordoba, Spain
| |
Collapse
|
33
|
Gkamprela E, Mani I, Alexopoulou A, Vasilieva L, Pouriki S, Dourakis SP. Letter: high ferritin to haemoglobin ratio is related to early mortality in patients with alcoholic hepatitis. Aliment Pharmacol Ther 2019; 49:1456-1457. [PMID: 31074902 DOI: 10.1111/apt.15266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Eleana Gkamprela
- 2nd Department of Medicine, Medical School, National &, Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Iliana Mani
- 2nd Department of Medicine, Medical School, National &, Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Alexandra Alexopoulou
- 2nd Department of Medicine, Medical School, National &, Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Larisa Vasilieva
- 2nd Department of Medicine, Medical School, National &, Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Sophia Pouriki
- 2nd Department of Medicine, Medical School, National &, Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| | - Spyros P Dourakis
- 2nd Department of Medicine, Medical School, National &, Kapodistrian University of Athens, Hippokration General Hospital, Athens, Greece
| |
Collapse
|
34
|
Czaja AJ. Review article: iron disturbances in chronic liver diseases other than haemochromatosis - pathogenic, prognostic, and therapeutic implications. Aliment Pharmacol Ther 2019; 49:681-701. [PMID: 30761559 DOI: 10.1111/apt.15173] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Disturbances in iron regulation have been described in diverse chronic liver diseases other than hereditary haemochromatosis, and iron toxicity may worsen liver injury and outcome. AIMS To describe manifestations and consequences of iron dysregulation in chronic liver diseases apart from hereditary haemochromatosis and to encourage investigations that clarify pathogenic mechanisms, define risk thresholds for iron toxicity, and direct management METHODS: English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. RESULTS Hyperferritinemia is present in 4%-65% of patients with non-alcoholic fatty liver disease, autoimmune hepatitis, chronic viral hepatitis, or alcoholic liver disease, and hepatic iron content is increased in 11%-52%. Heterozygosity for the C282Y mutation is present in 17%-48%, but this has not uniformly distinguished patients with adverse outcomes. An inappropriately low serum hepcidin level has characterised most chronic liver diseases with the exception of non-alcoholic fatty liver disease, and the finding has been associated mainly with suppression of transcriptional activity of the hepcidin gene. Iron overload has been associated with oxidative stress, advanced fibrosis and decreased survival, and promising therapies beyond phlebotomy and oral iron chelation have included hepcidin agonists. CONCLUSIONS Iron dysregulation is common in chronic liver diseases other than hereditary haemochromatosis, and has been associated with liver toxicity and poor prognosis. Further evaluation of iron overload as a co-morbid factor should identify the key pathogenic disturbances, establish the risk threshold for iron toxicity, and promote molecular interventions.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
35
|
Up-regulated ferritin in periodontitis promotes inflammatory cytokine expression in human periodontal ligament cells through transferrin receptor via ERK/P38 MAPK pathways. Clin Sci (Lond) 2019; 133:135-148. [PMID: 30552136 DOI: 10.1042/cs20180679] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/29/2018] [Accepted: 12/14/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Ferritin, an iron-binding protein, is ubiquitous and highly conserved; it plays a crucial role in inflammation, which is the main symptom of periodontitis. Full-length cDNA library analyses have demonstrated abundant expression of ferritin in human periodontal ligament. The aims of the present study were to explore how ferritin is regulated by local inflammation, and to investigate its functions and mechanisms of action in the process of periodontitis. METHODS Human gingival tissues were collected from periodontitis patients and healthy individuals. Experimental periodontitis was induced by ligature of second molars in mice. The expression of ferritin light polypeptide (FTL) and ferritin heavy polypeptide (FTH) were assessed by immunohistochemistry. Meanwhile, after stimulating human periodontal ligament cells (HPDLCs) with P. gingivalis-lipopolysaccharide (LPS), interleukin (IL)-6, and tumor necrosis factor-α (TNF-α), the expression of FTH and FTL were measured. Then, IL-6 and IL-8 were measured after incubation with different concentrations of apoferritin (iron-free ferritin) and several intracellular signaling pathway inhibitors, or after knockdown of the transferrin receptor. RESULTS Both FTH and FTL were substantially higher in inflamed periodontal tissues than in healthy tissues. The location of the elevated expression correlated well with the extent of inflammatory infiltration. Moreover, expression of FTH and FTL were enhanced after stimulation with P. gingivalis-LPS, IL-6, TNF-α. Apoferritin induced the production of IL-6 and IL-8 in a dose-dependent manner partly through binding to the transferrin receptor and activating ERK/P38 signaling pathways in HPDLCs. CONCLUSIONS Ferritin is up-regulated by inflammation and exhibits cytokine-like activity in HPDLCs inducing a signaling cascade that promotes expression of pro-inflammatory cytokines associated with periodontitis.
Collapse
|
36
|
Abstract
SIGNIFICANCE Iron and oxygen are intimately linked: iron is an essential nutrient utilized as a cofactor in enzymes for oxygen transport, oxidative phosphorylation, and metabolite oxidation. However, excess labile iron facilitates the formation of oxygen-derived free radicals capable of damaging biomolecules. Therefore, biological utilization of iron is a tightly regulated process. The nuclear factor (erythroid-derived 2)-like 2 (NRF2) transcription factor, which can respond to oxidative and electrophilic stress, regulates several genes involved in iron metabolism. Recent Advances: The bulk of NRF2 transcription factor research has focused on its roles in detoxification and cancer prevention. Recent works have identified that several genes involved in heme synthesis, hemoglobin catabolism, iron storage, and iron export are under the control of NRF2. Constitutive NRF2 activation and subsequent deregulation of iron metabolism have been implicated in cancer development: NRF2-mediated upregulation of the iron storage protein ferritin or heme oxygenase 1 can lead to enhanced proliferation and therapy resistance. Of note, NRF2 activation and alterations to iron signaling in cancers may hinder efforts to induce the iron-dependent cell death process known as ferroptosis. CRITICAL ISSUES Despite growing recognition of NRF2 as a modulator of iron signaling, exactly how iron metabolism is altered due to NRF2 activation in normal physiology and in pathologic conditions remains imprecise; moreover, the roles of NRF2-mediated iron signaling changes in disease progression are only beginning to be uncovered. FUTURE DIRECTIONS Further studies are necessary to connect NRF2 activation with physiological and pathological changes to iron signaling and oxidative stress. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Michael John Kerins
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona , Tucson, Arizona
| | - Aikseng Ooi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona , Tucson, Arizona
| |
Collapse
|
37
|
Manjunathachar HV, Kumar B, Saravanan BC, Choudhary S, Mohanty AK, Nagar G, Chigure G, Ravi Kumar GVPPS, de la Fuente J, Ghosh S. Identification and characterization of vaccine candidates against Hyalomma anatolicum-Vector of Crimean-Congo haemorrhagic fever virus. Transbound Emerg Dis 2018; 66:422-434. [PMID: 30300470 DOI: 10.1111/tbed.13038] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/27/2018] [Accepted: 09/30/2018] [Indexed: 12/24/2022]
Abstract
Crimean-Congo haemorrhagic fever (CCHF) is a tick borne viral disease reported from different parts of the world. The distribution of the CCHF cases are linked with the distribution of the principal vector, Hyalomma anatolicum in the ecosystem. Presently, vector control is mainly dependent on repeated application of acaricides, results in partial efficacy and generated acaricide resistant tick strains. Amongst the different components of integrated management programme, immunization of hosts is considered as one of the sustainable component. To restrict CCHF virus spreading, use of anti-Hyalomma vaccines appears as a viable solution. Accordingly, present study was under taken to characterize and evaluate vaccine potential of two conserved molecules, ferritin2 (FER2) and tropomyosin (TPM). Silencing of the genes conferred a cumulative reduction (rejection + unable to engorge) of 61.3% in FER2 and 70.2% in TPM respectively. Furthermore, 44.2% and 72.7% reduction in engorgement weight, 63.6% and 94.9% reduction in egg masses in FER2 and TPM silenced ticks in comparison to LUC-control group was recorded. The recombinant protein, rHaFER2 was characterized as 35 kDa protein with pI of 5.84 and possesses iron binding domains. While rHaTPM is a 51kDa protein with pI of 4.94 having calcium binding domains. Immunization of cross-bred calves by rHaFER2 conferred 51.7% and 51.2% protection against larvae and adults of H. anatolicum challenge infestations. While rHaTPM conferred 63.7% and 66.4% protection against larvae and adults infestations, respectively. The results were comparable with the data generated by RNAi and it clearly showed the possibility for the development of anti-hyalomma vaccine to manage CCHF virus and Theileria annulata infection in human and animals.
Collapse
Affiliation(s)
| | - Binod Kumar
- Entomology Laboratory, Division of Parasitology, Indian Veterinary Research Institute (IVRI)-Izatnagar, Bareilly, India
| | | | - Suman Choudhary
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Ashok K Mohanty
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal, Haryana, India
| | - Gaurav Nagar
- Entomology Laboratory, Division of Parasitology, Indian Veterinary Research Institute (IVRI)-Izatnagar, Bareilly, India
| | - Gajanan Chigure
- Entomology Laboratory, Division of Parasitology, Indian Veterinary Research Institute (IVRI)-Izatnagar, Bareilly, India
| | | | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC (CSIC-UCLM-JCCM), Ciudad Real, Spain.,Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma
| | - Srikant Ghosh
- Entomology Laboratory, Division of Parasitology, Indian Veterinary Research Institute (IVRI)-Izatnagar, Bareilly, India
| |
Collapse
|
38
|
Hernandez EP, Kusakisako K, Talactac MR, Galay RL, Yoshii K, Tanaka T. Induction of intracellular ferritin expression in embryo-derived Ixodes scapularis cell line (ISE6). Sci Rep 2018; 8:16566. [PMID: 30410072 PMCID: PMC6224502 DOI: 10.1038/s41598-018-34860-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/25/2018] [Indexed: 11/26/2022] Open
Abstract
Iron is a very important nutrient for cells; however, it could also cause fatal effects because of its capability to trigger oxidative stress. Due to high exposure to iron from their blood diet, ticks make use of several mechanisms to cope up with oxidative stress. One mechanism is iron sequestration by ferritin and its control protein (IRP). Since the IRP activity is dependent on the ferrous iron concentration, we tried to induce intracellular ferritin (FER1) protein expression by exposing Ixodes scapularis embryo-derived cell line (ISE6) to different concentrations of ferrous sulphate at different time points. We were able to induce FER1 protein after exposure to 2 mM of ferrous sulphate for 48 h, as observed in both Western blotting and indirect immunofluorescent antibody tests. This could indicate that the FER1 produced could be a product of the release of IRPs from the FER1 mRNA leading to its translation. The RNA interference of FER1, through the transfection of dsRNA, led to an increase in mortality and decrease in the cellular proliferation of ISE6 cells. Overall, ISE6 cells could be a good tool in further understanding the mechanism of FER1 action, not just in Ixodes ticks but in other tick species as well.
Collapse
Affiliation(s)
- Emmanuel Pacia Hernandez
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan.,Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan
| | - Kodai Kusakisako
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan.,Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan
| | - Melbourne Rio Talactac
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan.,Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan.,Department of Clinical and Population Health, College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Cavite, 4122, Philippines
| | - Remil Linggatong Galay
- Department of Veterinary Paraclinical Sciences, University of the Philippines Los Baños, College, Laguna, 3004, Philippines
| | - Kentaro Yoshii
- Laboratory of Public Health, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Tetsuya Tanaka
- Laboratory of Infectious Diseases, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima, 890-0056, Japan. .,Department of Pathological and Preventive Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yoshida, Yamaguchi, 753-8515, Japan.
| |
Collapse
|
39
|
Raghunath A, Sundarraj K, Nagarajan R, Arfuso F, Bian J, Kumar AP, Sethi G, Perumal E. Antioxidant response elements: Discovery, classes, regulation and potential applications. Redox Biol 2018; 17:297-314. [PMID: 29775961 PMCID: PMC6007815 DOI: 10.1016/j.redox.2018.05.002] [Citation(s) in RCA: 332] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/25/2018] [Accepted: 05/05/2018] [Indexed: 12/20/2022] Open
Abstract
Exposure to antioxidants and xenobiotics triggers the expression of a myriad of genes encoding antioxidant proteins, detoxifying enzymes, and xenobiotic transporters to offer protection against oxidative stress. This articulated universal mechanism is regulated through the cis-acting elements in an array of Nrf2 target genes called antioxidant response elements (AREs), which play a critical role in redox homeostasis. Though the Keap1/Nrf2/ARE system involves many players, AREs hold the key in transcriptional regulation of cytoprotective genes. ARE-mediated reporter constructs have been widely used, including xenobiotics profiling and Nrf2 activator screening. The complexity of AREs is brought by the presence of other regulatory elements within the AREs. The diversity in the ARE sequences not only bring regulatory selectivity of diverse transcription factors, but also confer functional complexity in the Keap1/Nrf2/ARE pathway. The different transcription factors either homodimerize or heterodimerize to bind the AREs. Depending on the nature of partners, they may activate or suppress the transcription. Attention is required for deeper mechanistic understanding of ARE-mediated gene regulation. The computational methods of identification and analysis of AREs are still in their infancy. Investigations are required to know whether epigenetics mechanism plays a role in the regulation of genes mediated through AREs. The polymorphisms in the AREs leading to oxidative stress related diseases are warranted. A thorough understanding of AREs will pave the way for the development of therapeutic agents against cancer, neurodegenerative, cardiovascular, metabolic and other diseases with oxidative stress.
Collapse
Affiliation(s)
- Azhwar Raghunath
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, Tamilnadu, India
| | - Kiruthika Sundarraj
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, Tamilnadu, India
| | - Raju Nagarajan
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai 600036, Tamilnadu, India
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia
| | - Jinsong Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore
| | - Alan P Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, Australia.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore, Singapore.
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641046, Tamilnadu, India.
| |
Collapse
|
40
|
Anti-Inflammatory Effect of Gamisoyo-San in an Animal Model of Amyotrophic Lateral Sclerosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8580152. [PMID: 30034501 PMCID: PMC6032819 DOI: 10.1155/2018/8580152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/11/2022]
Abstract
Inflammation is considered a critical factor in the pathogenesis of amyotrophic lateral sclerosis (ALS). We aimed to evaluate the effect of the herbal formula Gamisoyo-San (GSS) on the muscles of hSOD1G93A transgenic mice, a mouse model of ALS, by examining the tissue expression of inflammation- and oxidative stress-related proteins. The mice were randomly divided into three groups: nontransgenic mice (non-Tg, n = 4), hSOD1G93A transgenic mice (Tg, n = 4), and GSS-treated hSOD1G93A transgenic mice (Tg+GSS, n = 4). Eight-week-old female hSOD1G93A transgenic mice were fed GSS (1 mg/g body weight) for 6 weeks. Gastrocnemius (GA) tissues were analyzed for inflammatory proteins [CD11b and toll-like receptor 4 (TLR4)] and oxidative stress-related proteins [heme oxygenase 1 (HO1) and ferritin] by western blot analysis. Administration of GSS significantly reduced the level of inflammation- and oxidative stress-related proteins in hSOD1G93A transgenic mice. GSS ameliorated inflammation by downregulating TLR4 and CD11b expression and regulated iron homeostasis in the GA muscle of hSOD1G93A mice. GSS could help reduce inflammation by regulating immune reactions in patients with ALS. To the best of our knowledge, this is the first study to demonstrate the effect of GSS on muscle inflammation in an ALS animal model.
Collapse
|
41
|
Agoro R, Taleb M, Quesniaux VFJ, Mura C. Cell iron status influences macrophage polarization. PLoS One 2018; 13:e0196921. [PMID: 29771935 PMCID: PMC5957380 DOI: 10.1371/journal.pone.0196921] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/23/2018] [Indexed: 12/21/2022] Open
Abstract
Macrophages play crucial roles in innate immune response and in the priming of adaptive immunity, and are characterized by their phenotypic heterogeneity and plasticity. Reprogramming intracellular metabolism in response to microenvironmental signals is required for M1/M2 macrophage polarization and function. Here we assessed the influence of iron on the polarization of the immune response in vivo and in vitro. Iron-enriched diet increased M2 marker Arg1 and Ym1 expression in liver and peritoneal macrophages, while iron deficiency decreased Arg1 expression. Under LPS-induced inflammatory conditions, low iron diet exacerbated the proinflammatory response, while the IL-12/IL-10 balance decreased with iron-rich diet, thus polarizing toward type 2 response. Indeed, in vitro macrophage iron loading reduced the basal percentage of cells expressing M1 co-stimulatory CD86 and MHC-II molecules. Further, iron loading of macrophages prevented the pro-inflammatory response induced by LPS through reduction of NF-κB p65 nuclear translocation with decreased iNOS, IL-1β, IL-6, IL-12 and TNFα expression. The increase of intracellular iron also reduced LPS-induced hepcidin gene expression and abolished ferroportin down-regulation in macrophages, in line with macrophage polarization. Thus, iron modulates the inflammatory response outcome, as elevated iron levels increased M2 phenotype and negatively regulated M1 proinflammatory LPS-induced response.
Collapse
Affiliation(s)
- Rafiou Agoro
- Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS, Orléans, France
- University of Orléans, Orléans, France
| | - Meriem Taleb
- Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS, Orléans, France
- University of Orléans, Orléans, France
| | - Valerie F. J. Quesniaux
- Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS, Orléans, France
- University of Orléans, Orléans, France
| | - Catherine Mura
- Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS, Orléans, France
- University of Orléans, Orléans, France
- * E-mail:
| |
Collapse
|
42
|
Lewerenz J, Ates G, Methner A, Conrad M, Maher P. Oxytosis/Ferroptosis-(Re-) Emerging Roles for Oxidative Stress-Dependent Non-apoptotic Cell Death in Diseases of the Central Nervous System. Front Neurosci 2018; 12:214. [PMID: 29731704 PMCID: PMC5920049 DOI: 10.3389/fnins.2018.00214] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022] Open
Abstract
Although nerve cell death is the hallmark of many neurological diseases, the processes underlying this death are still poorly defined. However, there is a general consensus that neuronal cell death predominantly proceeds by regulated processes. Almost 30 years ago, a cell death pathway eventually named oxytosis was described in neuronal cells that involved glutathione depletion, reactive oxygen species production, lipoxygenase activation, and calcium influx. More recently, a cell death pathway that involved many of the same steps was described in tumor cells and termed ferroptosis due to a dependence on iron. Since then there has been a great deal of discussion in the literature about whether these are two distinct pathways or cell type- and insult-dependent variations on the same pathway. In this review, we compare and contrast in detail the commonalities and distinctions between the two pathways concluding that the molecular pathways involved in the regulation of ferroptosis and oxytosis are highly similar if not identical. Thus, we suggest that oxytosis and ferroptosis should be regarded as two names for the same cell death pathway. In addition, we describe the potential physiological relevance of oxytosis/ferroptosis in multiple neurological diseases.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, Ulm University, Ulm, Germany
| | - Gamze Ates
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Axel Methner
- Department of Neurology, University Medical Center and Focus Program Translational Neuroscience of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
43
|
Urrutia PJ, Aguirre P, Tapia V, Carrasco CM, Mena NP, Núñez MT. Cell death induced by mitochondrial complex I inhibition is mediated by Iron Regulatory Protein 1. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2202-2209. [DOI: 10.1016/j.bbadis.2017.05.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/18/2017] [Accepted: 05/10/2017] [Indexed: 01/14/2023]
|
44
|
Gonzalez-Moragas L, Yu SM, Benseny-Cases N, Stürzenbaum S, Roig A, Laromaine A. Toxicogenomics of iron oxide nanoparticles in the nematode C. elegans. Nanotoxicology 2017; 11:647-657. [PMID: 28673184 DOI: 10.1080/17435390.2017.1342011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We present a mechanistic study of the effect of iron oxide nanoparticles (SPIONs) in Caenorhabditis elegans combining a genome-wide analysis with the investigation of specific molecular markers frequently linked to nanotoxicity. The effects of two different coatings were explored: citrate, an anionic stabilizer, and bovine serum albumin, as a pre-formed protein corona. The transcriptomic study identified differentially expressed genes following an exposure to SPIONs. The expression of genes involved in oxidative stress, metal detoxification response, endocytosis, intestinal integrity and iron homeostasis was quantitatively evaluated. The role of oxidative stress was confirmed by gene expression analysis and by synchrotron Fourier Transform infrared microscopy based on the higher tissue oxidation of NP-treated animals. The observed transcriptional modulation of key signaling pathways such as MAPK and Wnt suggests that SPIONs might be endocytosed by clathrin-mediated processes, a putative mechanism of nanotoxicity which deserves further mechanistic investigations.
Collapse
Affiliation(s)
- Laura Gonzalez-Moragas
- a Group of Nanoparticles and Nanocomposites, Crystallography Department , Institut de Ciència de Materials de Barcelona, ICMAB-CSIC , Barcelona , Campus UAB , Spain
| | - Si-Ming Yu
- a Group of Nanoparticles and Nanocomposites, Crystallography Department , Institut de Ciència de Materials de Barcelona, ICMAB-CSIC , Barcelona , Campus UAB , Spain.,b Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering , Jinan University , Guangzhou , China
| | | | - Stephen Stürzenbaum
- d Faculty of Life Sciences & Medicine, Analytical and Environmental Sciences Division , King's College London , London , UK
| | - Anna Roig
- a Group of Nanoparticles and Nanocomposites, Crystallography Department , Institut de Ciència de Materials de Barcelona, ICMAB-CSIC , Barcelona , Campus UAB , Spain
| | - Anna Laromaine
- a Group of Nanoparticles and Nanocomposites, Crystallography Department , Institut de Ciència de Materials de Barcelona, ICMAB-CSIC , Barcelona , Campus UAB , Spain
| |
Collapse
|
45
|
Belcher JD, Chen C, Nguyen J, Zhang P, Abdulla F, Nguyen P, Killeen T, Xu P, O'Sullivan G, Nath KA, Vercellotti GM. Control of Oxidative Stress and Inflammation in Sickle Cell Disease with the Nrf2 Activator Dimethyl Fumarate. Antioxid Redox Signal 2017; 26:748-762. [PMID: 26914345 PMCID: PMC5421647 DOI: 10.1089/ars.2015.6571] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS Heme derived from hemolysis is pro-oxidative and proinflammatory and promotes vaso-occlusion in murine models of sickle cell disease (SCD), suggesting that enhanced detoxification of heme may be beneficial. Nuclear factor erythroid-2-related factor-2 (Nrf2) transcription pathway is the principal cellular defense system responding to pro-oxidative and proinflammatory stress. Dimethyl fumarate (DMF), a drug approved for treatment of multiple sclerosis, provides neuroprotection by activating Nrf2-responsive genes. We hypothesized that induction of Nrf2 with DMF would be beneficial in murine SCD models. RESULTS DMF (30 mg/kg/day) or vehicle (0.08% methyl cellulose) was administered for 3-7 days to NY1DD and HbSS-Townes SCD mice. Vaso-occlusion, a hallmark of SCD, measured in sickle mice with dorsal skinfold chambers, was inhibited by DMF. The inhibitory effect of DMF was abrogated by the heme oxygenase-1 (HO-1) inhibitor tin protoporphyrin. DMF increased nuclear Nrf2 and cellular mRNA of Nrf2-responsive genes in livers and kidneys. DMF increased heme defenses, including HO-1, haptoglobin, hemopexin, and ferritin heavy chain, although plasma hemoglobin and heme levels were unchanged. DMF decreased markers of inflammation, including nuclear factor-kappa B phospho-p65, adhesion molecules, and toll-like receptor 4. DMF administered for 24 weeks to HbSS-Townes mice decreased hepatic necrosis, inflammatory cytokines, and irregularly shaped erythrocytes and increased hemoglobin F, but did not alter hematocrits, reticulocyte counts, lactate dehydrogenase, plasma heme, or spleen weights, indicating that the beneficial effects of DMF were not attributable to decreased hemolysis. INNOVATION These studies identify Nrf2 activation as a new therapeutic target for the treatment of SCD. CONCLUSION DMF activates Nrf2, enhances antioxidant defenses, and inhibits inflammation and vaso-occlusion in SCD mice. Antioxid. Redox Signal. 26, 748-762.
Collapse
Affiliation(s)
- John D Belcher
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Chunsheng Chen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Julia Nguyen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Ping Zhang
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Fuad Abdulla
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Phong Nguyen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Trevor Killeen
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Pauline Xu
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| | - Gerry O'Sullivan
- 2 Veterinary Population Medicine, University of Minnesota , St. Paul, Minnesota
| | - Karl A Nath
- 3 Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic/Foundation , Rochester, Minnesota
| | - Gregory M Vercellotti
- 1 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
46
|
Sternberg Z, Hu Z, Sternberg D, Waseh S, Quinn JF, Wild K, Jeffrey K, Zhao L, Garrick M. Serum Hepcidin Levels, Iron Dyshomeostasis and Cognitive Loss in Alzheimer's Disease. Aging Dis 2017; 8:215-227. [PMID: 28400987 PMCID: PMC5362180 DOI: 10.14336/ad.2016.0811] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 08/11/2016] [Indexed: 01/06/2023] Open
Abstract
This pilot study examined the status of the master iron regulatory peptide, hepcidin, and peripheral related iron parameters in Alzheimer's disease (AD) and mild cognitive impairment patients, and evaluated the relationship between iron dyshomeostasis and amyloid-beta (Aβ), cognitive assessment tests, neuroimaging and clinical data. Frozen serum samples from the Oregon Tissue Bank were used to measure serum levels of hepcidin, ferritin, Aβ40, Aβ42 using enzyme-linked immunosorbent assay. Serum transferrin levels were determined indirectly as total iron binding capacity, serum iron was measured and the percent saturation of transferrin calculated. The study variables were correlated with the patients' existing cognitive assessment tests, neuroimaging, and clinical data. Hepcidin, and iron-related proteins tended to be higher in AD patients than controls, reaching statistical significance for ferritin, whereas Aβ40, Aβ42 serum levels tended to be lower. Patients with pure AD had three times higher serum hepcidin levels than controls; gender differences in hepcidin and iron-related proteins were observed. Patient stratification based on clinical dementia rating-sum of boxes revealed significantly higher levels of iron and iron-related proteins in AD patients in the upper 50% as compared to controls, suggesting that iron dyshomeostasis worsens as cognitive impairment increases. Unlike Aβ peptides, iron and iron-related proteins showed significant association with cognitive assessment tests, neuroimaging, and clinical data. Hepcidin and iron-related proteins comprise a group of serum biomarkers that relate to AD diagnosis and AD disease progression. Future studies should determine whether strategies targeted to diminishing hepcidin synthesis/secretion and improving iron homeostasis could have a beneficial impact on AD progression.
Collapse
Affiliation(s)
- Zohara Sternberg
- Department of Neurology, Stroke Center, Buffalo Medical Center, Buffalo, NY, USA.
| | - Zihua Hu
- Center for Computational Research, New York State Center for Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Daniel Sternberg
- Department of Neurology, Stroke Center, Buffalo Medical Center, Buffalo, NY, USA.
| | - Shayan Waseh
- Department of Biology, State University of New York at Buffalo, Buffalo, NY 14260 USA.
| | - Joseph F. Quinn
- Layton Aging & Alzheimer's Research Center, Oregon Health and Science University, Portland, Oregon, USA.
| | - Katharine Wild
- Layton Aging & Alzheimer's Research Center, Oregon Health and Science University, Portland, Oregon, USA.
| | - Kaye Jeffrey
- Layton Aging & Alzheimer's Research Center, Oregon Health and Science University, Portland, Oregon, USA.
| | - Lin Zhao
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14214 USA.
| | - Michael Garrick
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY 14214 USA.
- Department of Pediatrics, State University of New York at Buffalo, Buffalo, NY 14214 USA.
| |
Collapse
|
47
|
Nanno S, Koh H, Nakashima Y, Katayama T, Okamura H, Koh S, Yoshimura T, Nishimoto M, Hayashi Y, Nakamae M, Hirose A, Nakane T, Hino M, Nakamae H. Diagnostic value of serum ferritin and the risk factors and cytokine profiles of hemophagocytic syndrome following allogeneic hematopoietic cell transplantation. Leuk Lymphoma 2016; 58:1664-1672. [DOI: 10.1080/10428194.2016.1262034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Satoru Nanno
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hideo Koh
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Yasuhiro Nakashima
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Takako Katayama
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hiroshi Okamura
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Shiro Koh
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Takuro Yoshimura
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Mitsutaka Nishimoto
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Yoshiki Hayashi
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Mika Nakamae
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Asao Hirose
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Takahiko Nakane
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Masayuki Hino
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hirohisa Nakamae
- Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
48
|
Banerjee P, Sahoo A, Anand S, Bir A, Chakrabarti S. The Oral Iron Chelator, Deferasirox, Reverses the Age-Dependent Alterations in Iron and Amyloid-β Homeostasis in Rat Brain: Implications in the Therapy of Alzheimer's Disease. J Alzheimers Dis 2016; 49:681-93. [PMID: 26484920 DOI: 10.3233/jad-150514] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The altered metabolism of iron impacts the brain function in multiple deleterious ways during normal aging as well as in Alzheimer's disease. We have shown in this study that chelatable iron accumulates in the aged rat brain along with overexpression of transferrin receptor 1 (TfR1) and ferritin, accompanied by significant alterations in amyloid-β (Aβ) peptide homeostasis in the aging brain, such as an increased production of the amyloid-β protein precursor, a decreased level of neprilysin, and increased accumulation of Aβ42. When aged rats are given daily the iron chelator, deferasirox, over a period of more than 4 months starting from the 18th month, the age-related accumulation of iron and overexpression of TfR1 and ferritin in the brain are significantly prevented. More interestingly, the chelator treatment also considerably reverses the altered Aβ peptide metabolism in the aging brain implying a significant role of iron in the latter phenomenon. Further, other results indicate that iron accumulation results in oxidative stress and the activation of NF-κB in the aged rat brain, which are also reversed by the deferasirox treatment. The analysis of the results together suggests that iron accumulation and oxidative stress interact at multiple levels that include transcriptional and post-transcriptional mechanisms to bring about changes in the expression levels of TfR1 and ferritin and also alterations in Aβ peptide metabolism in the aging rat brain. The efficacy of deferasirox in preventing age-related changes in iron and Aβ peptide metabolism in the aging brain, as shown here, has obvious therapeutic implications for Alzheimer's disease.
Collapse
Affiliation(s)
- Priyanjalee Banerjee
- Department of Biochemistry, Institute of Post Graduate Medical and Educational Research, Kolkata, India
| | - Arghyadip Sahoo
- Department of Biochemistry, Institute of Post Graduate Medical and Educational Research, Kolkata, India
| | - Shruti Anand
- Department of Biochemistry, Institute of Post Graduate Medical and Educational Research, Kolkata, India
| | - Aritri Bir
- Department of Biochemistry, ICARE Institute of Medical Sciences and Research, Haldia, India
| | - Sasanka Chakrabarti
- Department of Biochemistry, ICARE Institute of Medical Sciences and Research, Haldia, India
| |
Collapse
|
49
|
Vercellotti GM, Zhang P, Nguyen J, Abdulla F, Chen C, Nguyen P, Nowotny C, Steer CJ, Smith A, Belcher JD. Hepatic Overexpression of Hemopexin Inhibits Inflammation and Vascular Stasis in Murine Models of Sickle Cell Disease. Mol Med 2016; 22:437-451. [PMID: 27451971 DOI: 10.2119/molmed.2016.00063] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/11/2016] [Indexed: 02/02/2023] Open
Abstract
Sickle cell disease (SCD) patients have low serum hemopexin (Hpx) levels due to chronic hemolysis. We hypothesize that in SCD mice, hepatic overexpression of hemopexin will scavenge the proximal mediator of vascular activation, heme, and will inhibit inflammation and microvascular stasis. To examine the protective role of Hpx in SCD, we transplanted bone marrow from NY1DD SCD mice into Hpx™/™ or Hpx+/+ C57BL/6 mice. Dorsal skin fold chambers were implanted in week 13 post-transplant and microvascular stasis (% non-flowing venules) evaluated in response to heme infusion. Hpx™/™ sickle mice had significantly greater microvascular stasis in response to heme infusion than Hpx+/+ sickle mice (p<0.05), demonstrating the protective effect of Hpx in SCD. We utilized Sleeping Beauty (SB) transposon-mediated gene transfer to overexpress wild-type rat Hpx (wt-Hpx) in NY1DD and Townes-SS SCD mice. Control SCD mice were treated with lactated Ringer's solution (LRS) or a luciferase (Luc) plasmid. Plasma and hepatic Hpx were significantly increased compared to LRS and Luc controls. Microvascular stasis in response to heme infusion in NY1DD and Townes-SS mice overexpressing wt-Hpx had significantly less stasis than controls (p<0.05). Wt-Hpx overexpression markedly increased hepatic nuclear Nrf2 expression, HO-1 activity and protein, the heme-Hpx binding protein and scavenger receptor, CD91/LRP1 and decreased NF-κB activation. Two missense (ms)-Hpx SB-constructs that bound neither heme nor the Hpx receptor, CD91/LRP1, did not prevent heme-induced stasis. In conclusion, increasing Hpx levels in transgenic sickle mice via gene transfer activates the Nrf2/HO-1 anti-oxidant axis and ameliorates inflammation and vaso-occlusion.
Collapse
Affiliation(s)
- Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ping Zhang
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia Nguyen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Fuad Abdulla
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chunsheng Chen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Phong Nguyen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Carlos Nowotny
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clifford J Steer
- Division of Gastroenterology, Department of Medicine, and Department of Genetics, Cell Biology and Development, 420 Delaware St SE, MMC 36, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ann Smith
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - John D Belcher
- Division of Hematology, Oncology and Transplantation, Department of Medicine, 420 Delaware St SE, MMC 480, University of Minnesota, Minneapolis, Minnesota, USA.,Vascular Biology Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
50
|
Bayele HK, Balesaria S, Srai SKS. Phytoestrogens modulate hepcidin expression by Nrf2: Implications for dietary control of iron absorption. Free Radic Biol Med 2015; 89:1192-202. [PMID: 26546695 PMCID: PMC4698393 DOI: 10.1016/j.freeradbiomed.2015.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 10/27/2015] [Accepted: 11/02/2015] [Indexed: 12/30/2022]
Abstract
Hepcidin is a liver-derived antimicrobial peptide that regulates iron absorption and is also an integral part of the acute phase response. In a previous report, we found evidence that this peptide could also be induced by toxic heavy metals and xenobiotics, thus broadening its teleological role as a defensin. However it remained unclear how its sensing of disparate biotic and abiotic stressors might be integrated at the transcriptional level. We hypothesized that its function in cytoprotection may be regulated by NFE2-related factor 2 (Nrf2), the master transcriptional controller of cellular stress defenses. In this report, we show that hepcidin regulation is inextricably linked to the acute stress response through Nrf2 signaling. Nrf2 regulates hepcidin expression from a prototypical antioxidant response element in its promoter, and by synergizing with other basic leucine-zipper transcription factors. We also show that polyphenolic small molecules or phytoestrogens commonly found in fruits and vegetables including the red wine constituent resveratrol can induce hepcidin expression in vitro and post-prandially, with concomitant reductions in circulating iron levels and transferrin saturation by one such polyphenol quercetin. Furthermore, these molecules derepress hepcidin promoter activity when its transcription by Nrf2 is repressed by Keap1. Taken together, the data show that hepcidin is a prototypical antioxidant response or cytoprotective gene within the Nrf2 transcriptional circuitry. The ability of phytoestrogens to modulate hepcidin expression in vivo suggests a novel mechanism by which diet may impact iron homeostasis.
Collapse
Affiliation(s)
- Henry K Bayele
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT United Kingdom.
| | - Sara Balesaria
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT United Kingdom
| | - Surjit K S Srai
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT United Kingdom.
| |
Collapse
|