1
|
Xiao G, Tang S, Zhang Y, Yuan Q, Sun D, Wang W. Downregulation of ferroptosis-related ATF3 alleviates lupus nephritis progression. Gene 2025; 934:149009. [PMID: 39427833 DOI: 10.1016/j.gene.2024.149009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 09/14/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The role of ferroptosis in lupus nephritis (LN) is unclear. This study aimed to explore the effects of ferroptosis-related genes in LN through bioinformatics prediction and experimental validation. METHODS Sample data were collected from the GEO dataset and divided into glomeruli and tubulointerstitium. We collected 382 ferroptosis-related genes. The intersection of ferroptosis-related genes with glomeruli and tubulointerstitium data, respectively, was performed. Machine learning methods (including unsupervised cluster typing and random forests) were operated to identify ferroptosis subtyping and ferroptosis important genes in LN. Immune infiltration and functional analysis were performed. The expression of ferroptosis important gene ATF3 was validated in vivo and in vitro. RESULTS 6 ferroptosis important genes common to glomeruli and tubulointerstitium were screened, including ATF3, CD44, CYBB, JUN, NCF2, and NNMT. ATF3 decreased in the LN group compared to the Control. Silencing ATF3 mitigated LPS/erastin-induced ferroptosis. Functional analysis showed that ATF3 was markedly enriched in the interferon-gamma-mediated signaling pathway, ECM-receptor interaction, and cell adhesion. In glomeruli, T cells regulatory (Tregs) infiltration decreased and Macrophages M1 levels increased with elevated ATF3 expression. Levels of immune cell infiltration were altered in different ferroptosis subtypes of LN glomeruli and tubulointerstitium. CONCLUSIONS Ferroptosis-related ATF3 levels decreased in LN. Inhibition of ATF3 might alleviate LN development by affecting the macrophage M1 and Treg cell infiltration. These implied that ATF3 might be a potential target for developing LN therapeutic strategies.
Collapse
Affiliation(s)
- Gong Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shumei Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Danni Sun
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
2
|
Liu H, Li M, Deng Y, Hou Y, Hou L, Zhang X, Zheng Z, Guo F, Sun K. The Roles of DMT1 in Inflammatory and Degenerative Diseases. Mol Neurobiol 2025:10.1007/s12035-025-04687-x. [PMID: 39775481 DOI: 10.1007/s12035-025-04687-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Iron homeostasis is critical for multiple physiological and pathological processes. DMT1, a core iron transporter, is expressed in almost all cells and organs and altered in response to various conditions, whereas, there is few reviews focusing on DMT1 in diseases associated with aberrant iron metabolism. Based on available knowledge, this review described a full view of DMT1 and summarized the roles of DMT1 and DMT1-mediated iron metabolism in the onset and development of inflammatory and degenerative diseases. This review also provided an overview of DMT1-related treatment in these disorders, highlighting its therapeutic potential in chronic inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Haigang Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Mi Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yi Deng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yanjun Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xiong Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zehang Zheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
3
|
Wu B. Genetically Predicted Iron Status Is a Causal Risk of Rheumatoid Arthritis: A Mendelian Randomization Study. Glob Med Genet 2024; 11:270-277. [PMID: 39211802 PMCID: PMC11361779 DOI: 10.1055/s-0044-1789259] [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] [Indexed: 09/04/2024] Open
Abstract
Background Current knowledge on iron's role in rheumatoid arthritis (RA) development is very limited, with studies yielding inconsistent findings. We conducted a two-sample Mendelian randomization study to assess the associations of iron status with the risk of RA. Methods This study leveraged genetic data from a large genome-wide association study (GWAS) of 257,953 individuals to identify single nucleotide polymorphisms (SNPs) associated with iron status. We then analyzed these data in conjunction with summary-level data on RA from the IEU open GWAS project, which included 5,427 RA cases and 479,171 controls. An inverse-variance weighted method with random effects was employed, along with sensitivity analyses, to assess the relationship between iron status and RA risk. Results Genetic predisposition to high ferritin and serum iron status was causally associated with lower odds of RA. Ferritin had an odds ratio (OR) of 0.997 (95% confidence interval [CI]: 0.995-0.997; p = 0.010), indicating that a one-unit increase in ferritin is associated with a 0.3% decrease in the odds of RA. Similarly, serum iron had an OR of 0.997 (95% CI: 0.995-0.999; p = 0.014). However, MR analyses found no significant causal associations between total iron-binding capacity (OR = 1.0, 95% CI: 0.999-1.002; p = 0.592) or transferrin saturation percentage (OR = 0.998, 95% CI: 0.996-1.000; p = 0.080) and risk of developing RA. Conclusions This study suggests that individuals with genes linked to higher iron levels may have a lower risk of developing RA. Our findings indicate that the total amount of iron in the body, rather than how it is distributed, might be more important for RA. This raises the intriguing possibility that iron supplementation could be a preventative strategy, but further research is necessary.
Collapse
Affiliation(s)
- Boyuan Wu
- School of Global Public Health New York University, New York, New York, United States
| |
Collapse
|
4
|
Osman EEA, Neamati N. Ironing Out the Mechanism of gp130 Signaling. Pharmacol Rev 2024; 76:1399-1443. [PMID: 39414364 DOI: 10.1124/pharmrev.124.001245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 10/18/2024] Open
Abstract
gp130 functions as a shared signal-transducing subunit not only for interleukin (IL)-6 but also for eight other human cytokine receptor complexes. The IL-6 signaling pathway mediated through gp130 encompasses classical, trans, or cluster signaling, intricately regulated by a diverse array of modulators affecting IL-6, its receptor, and gp130. Currently, only a limited number of small molecule antagonists and agonists for gp130 are known. This review aims to comprehensively examine the current knowledge of these modulators and provide insights into their pharmacological properties, particularly in the context of cancer and other diseases. Notably, the prominent gp130 modulators SC144, bazedoxifene, and raloxifene are discussed in detail, with a specific focus on the discovery of SC144's iron-chelating properties. This adds a new dimension to the understanding of its pharmacological effects and therapeutic potential in conditions where iron homeostasis is significant. Our bioinformatic analysis of gp130 and genes related to iron homeostasis reveals insightful correlations, implicating the role of iron in the gp130 signaling pathway. Overall, this review contributes to the evolving understanding of gp130 modulation and its potential therapeutic applications in various disease contexts. SIGNIFICANCE STATEMENT: This perspective provides a timely and comprehensive analysis of advancements in gp130 signaling research, emphasizing the therapeutic implications of the currently available modulators. Bioinformatic analysis demonstrates potential interplay between gp130 and genes that regulate iron homeostasis, suggesting new therapeutic avenues. By combining original research findings with a broader discussion of gp130's therapeutic potential, this perspective significantly contributes to the field.
Collapse
Affiliation(s)
- Essam Eldin A Osman
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan (E.E.A.O., N.N.) and Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt (E.E.A.O.)
| |
Collapse
|
5
|
Leineweber WD, Rowell MZ, Ranamukhaarachchi SK, Walker A, Li Y, Villazon J, Mestre-Farrera A, Hu Z, Yang J, Shi L, Fraley SI. Divergent iron regulatory states contribute to heterogeneity in breast cancer aggressiveness. iScience 2024; 27:110661. [PMID: 39262774 PMCID: PMC11387597 DOI: 10.1016/j.isci.2024.110661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/19/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Contact with dense collagen I (Col1) can induce collective invasion of triple negative breast cancer (TNBC) cells and transcriptional signatures linked to poor patient prognosis. However, this response is heterogeneous and not well understood. Using phenotype-guided sequencing analysis of invasive vs. noninvasive subpopulations, we show that these two phenotypes represent opposite sides of the iron response protein 1 (IRP1)-mediated response to cytoplasmic labile iron pool (cLIP) levels. Invasive cells upregulate iron uptake and utilization machinery characteristic of a low cLIP response, which includes contractility regulating genes that drive migration. Non-invasive cells upregulate iron sequestration machinery characteristic of a high cLIP response, which is accompanied by upregulation of actin sequestration genes. These divergent IRP1 responses result from Col1-induced transient expression of heme oxygenase I (HO-1), which cleaves heme and releases iron. These findings lend insight into the emerging theory that heme and iron fluxes regulate TNBC aggressiveness.
Collapse
Affiliation(s)
- William D. Leineweber
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maya Z. Rowell
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Alyssa Walker
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yajuan Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jorge Villazon
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Aida Mestre-Farrera
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Lingyan Shi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Harnik Y, Yakubovsky O, Hoefflin R, Novoselsky R, Bahar Halpern K, Barkai T, Korem Kohanim Y, Egozi A, Golani O, Addadi Y, Kedmi M, Keidar Haran T, Levin Y, Savidor A, Keren-Shaul H, Mayer C, Pencovich N, Pery R, Shouval DS, Tirosh I, Nachmany I, Itzkovitz S. A spatial expression atlas of the adult human proximal small intestine. Nature 2024; 632:1101-1109. [PMID: 39112711 DOI: 10.1038/s41586-024-07793-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 07/05/2024] [Indexed: 08/17/2024]
Abstract
The mouse small intestine shows profound variability in gene expression along the crypt-villus axis1,2. Whether similar spatial heterogeneity exists in the adult human gut remains unclear. Here we use spatial transcriptomics, spatial proteomics and single-molecule fluorescence in situ hybridization to reconstruct a comprehensive spatial expression atlas of the adult human proximal small intestine. We describe zonated expression and cell type representation for epithelial, mesenchymal and immune cell types. We find that migrating enterocytes switch from lipid droplet assembly and iron uptake at the villus bottom to chylomicron biosynthesis and iron release at the tip. Villus tip cells are pro-immunogenic, recruiting γδ T cells and macrophages to the tip, in contrast to their immunosuppressive roles in mouse. We also show that the human small intestine contains abundant serrated and branched villi that are enriched at the tops of circular folds. Our study presents a detailed resource for understanding the biology of the adult human small intestine.
Collapse
Affiliation(s)
- Yotam Harnik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oran Yakubovsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rouven Hoefflin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Roy Novoselsky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Keren Bahar Halpern
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Barkai
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Sheba Medical Center, Ramat Gan, Israel
| | - Yael Korem Kohanim
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Adi Egozi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Kedmi
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Keidar Haran
- Department of Pathology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Yishai Levin
- The De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- The De Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Hadas Keren-Shaul
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Chen Mayer
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Niv Pencovich
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ron Pery
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dror S Shouval
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute of Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Nachmany
- Department of General Surgery and Transplantation, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
7
|
Geroyska S, Mejia I, Chan AA, Navarrete M, Pandey V, Kharpatin S, Noguti J, Wang F, Srole D, Chou TF, Wohlschlegel J, Nemeth E, Damoiseaux R, Shackelford DB, Lee DJ, Díaz B. N-Myristoytransferase Inhibition Causes Mitochondrial Iron Overload and Parthanatos in TIM17A-Dependent Aggressive Lung Carcinoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1815-1833. [PMID: 38949950 PMCID: PMC11270646 DOI: 10.1158/2767-9764.crc-23-0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 05/09/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024]
Abstract
Myristoylation is a type of protein acylation by which the fatty acid myristate is added to the N-terminus of target proteins, a process mediated by N-myristoyltransferases (NMT). Myristoylation is emerging as a promising cancer therapeutic target; however, the molecular determinants of sensitivity to NMT inhibition or the mechanism by which it induces cancer cell death are not completely understood. We report that NMTs are a novel therapeutic target in lung carcinoma cells with LKB1 and/or KEAP1 mutations in a KRAS-mutant background. Inhibition of myristoylation decreases cell viability in vitro and tumor growth in vivo. Inhibition of myristoylation causes mitochondrial ferrous iron overload, oxidative stress, elevated protein poly (ADP)-ribosylation, and death by parthanatos. Furthermore, NMT inhibitors sensitized lung carcinoma cells to platinum-based chemotherapy. Unexpectedly, the mitochondrial transporter translocase of inner mitochondrial membrane 17 homolog A (TIM17A) is a critical target of myristoylation inhibitors in these cells. TIM17A silencing recapitulated the effects of NMT inhibition at inducing mitochondrial ferrous iron overload and parthanatos. Furthermore, sensitivity of lung carcinoma cells to myristoylation inhibition correlated with their dependency on TIM17A. This study reveals the unexpected connection between protein myristoylation, the mitochondrial import machinery, and iron homeostasis. It also uncovers myristoylation inhibitors as novel inducers of parthanatos in cancer, and the novel axis NMT-TIM17A as a potential therapeutic target in highly aggressive lung carcinomas. SIGNIFICANCE KRAS-mutant lung carcinomas with LKB1 and/or KEAP1 co-mutations have intrinsic therapeutic resistance. We show that these tumors are sensitive to NMT inhibitors, which slow tumor growth in vivo and sensitize cells to platinum-based chemotherapy in vitro. Inhibition of myristoylation causes death by parthanatos and thus has the potential to kill apoptosis and ferroptosis-resistant cancer cells. Our findings warrant investigation of NMT as a therapeutic target in highly aggressive lung carcinomas.
Collapse
Affiliation(s)
- Sofia Geroyska
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Hematology and Oncology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Isabel Mejia
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Hematology and Oncology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Alfred A. Chan
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Marian Navarrete
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Vijaya Pandey
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Samuel Kharpatin
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Juliana Noguti
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Feng Wang
- Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| | - Daniel Srole
- UCLA Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Tsui-Fen Chou
- Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| | - James Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Elizabeta Nemeth
- UCLA Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California.
- California NanoSystems Institute at UCLA, Los Angeles, California.
- Department for Bioengineering, Samueli School of Engineering, UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| | - David B. Shackelford
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| | - Delphine J. Lee
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Dermatology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| | - Begoña Díaz
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California.
- Division of Hematology and Oncology at Harbor-UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, California.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California.
| |
Collapse
|
8
|
Shete PA, Ghatpande NS, Varma ME, Joshi PV, Suryavanshi KR, Misar AV, Jadhav SH, Apte PP, Kulkarni PP. Chronic dietary iron overload affects hepatic iron metabolism and cognitive behavior in Wistar rats. J Trace Elem Med Biol 2024; 84:127422. [PMID: 38492476 DOI: 10.1016/j.jtemb.2024.127422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Iron accumulation in organs affects iron metabolism, leading to deleterious effects on the body. Previously, it was studied that high dietary iron in various forms and concentrations influences iron metabolism, resulting in iron accumulation in the liver and spleen and cognitive impairment. However, the actual mechanism and impact of long-term exposure to high dietary iron remain unknown. As a result, we postulated that iron overload caused by chronic exposure to excessive dietary iron supplementation would play a role in iron dyshomeostasis and inflammation in the liver and brain of Wistar rats. METHODS Animals were segregated into control, low iron (FAC-Ferric Ammonium Citrate 5000 ppm), and high iron dose group (FAC 20,000 ppm). The outcome of dietary iron overload on Wistar rats was evaluated in terms of body weight, biochemical markers, histological examination of liver and brain tissue, and cognitive-behavioral studies. Also, gene expression of rat brain tissue involving iron transporters Dmt1, TfR1, iron storage protein Fpn1, inflammatory markers Nf-kB, Tnf-α, Il-6, and hepcidin was performed. RESULTS Our data indicate that excess iron supplementation for 30 weeks leads to decreased body weight, increased serum iron levels, and decreased RBC levels in iron fed Wistar rats. Morris water maze (MWM) studies after 30 weeks showed increased escape latency in the high iron dose group compared with the control group. Histological studies of the high iron dose group showed an iron accumulation in the liver and brain loss of cellular architecture, and cellular degeneration was observed. Excess iron treatment showed upregulation of the Dmt1 gene in iron metabolism and a remarkable increase in the Nf-kB gene in rat brain tissue. CONCLUSION The results show chronic excess iron supplementation leads to iron accumulation in the liver, leading to inflammation in Wistar rats.
Collapse
Affiliation(s)
- Padmaja Anil Shete
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India; Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra 411007, India.
| | - Niraj Sudhir Ghatpande
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India.
| | - Mokshada Evameshwar Varma
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India.
| | - Pranav Vijay Joshi
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India.
| | - Komal Ravindra Suryavanshi
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India; Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra 411007, India.
| | - Ashwini Vivek Misar
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India.
| | - Sachin Hanumantrao Jadhav
- Nanobioscience Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India.
| | - Priti Parag Apte
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India.
| | - Prasad Padmakar Kulkarni
- Bioprospecting Group, Agharkar Research Institute, G. G. Agarkar Road, Pune, Maharashtra 411004, India; Savitribai Phule Pune University, Ganeshkhind, Pune, Maharashtra 411007, India.
| |
Collapse
|
9
|
Zhou W, Liang Y, Liao X, Tong L, Du W, Fu W, Tian S, Deng Y, Jiang X. ISRIB improves white matter injury following TBI by inhibiting NCOA4-mediated ferritinophagy. Neurochem Int 2024; 177:105744. [PMID: 38663454 DOI: 10.1016/j.neuint.2024.105744] [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: 02/25/2024] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024]
Abstract
Traumatic brain injury (TBI) often results in persistent neurological dysfunction, which is closely associated with white matter injury. The mechanisms underlying white matter injury after TBI remain unclear. Ferritinophagy is a selective autophagic process that degrades ferritin and releases free iron, which may cause ferroptosis. Although ferroptosis has been demonstrated to be involved in TBI, it is unclear whether ferritinophagy triggers ferroptosis in TBI. Integrated stress response inhibitor (ISRIB) has neuroprotective properties. However, the effect of ISRIB on white matter after TBI remains uncertain. We aimed to investigate whether ferritinophagy was involved in white matter injury following TBI and whether ISRIB can mitigate white matter injury after TBI by inhibiting ferritinophagy. In this study, controlled cortical impact (CCI) was performed on rats to establish the TBI model. Ferritinophagy was measured by assessing the levels of nuclear receptor coactivator 4 (NCOA4), which regulates ferritinophagy, ferritin heavy chain 1(FTH1), LC3, ATG5, and FTH1 colocalization with LC3 in the white matter. Increased NCOA4 and decreased FTH1 were detected in our study. FTH1 colocalization with LC3 enhanced in the white matter after TBI, indicating that ferritinophagy was activated. Immunofluorescence co-localization results also suggested that ferritinophagy occurred in neurons and oligodendrocytes after TBI. Furthermore, ferroptosis was assessed by determining free iron content, MDA content, GSH content, and Perl's staining. The results showed that ferroptosis was suppressed by NCOA4 knockdown via shNCOA4 lentivirus infection, indicating that ferroptosis in TBI is triggered by ferritinophagy. Besides, NCOA4 deletion notably improved white matter injury following TBI, implying that ferritinophagy contributed to white matter injury. ISRIB treatment reduced the occurrence of ferritinophagy in neurons and oligodendrocytes, attenuated ferritinophagy-induced ferroptosis, and alleviated white matter injury. These findings suggest that NCOA4-mediated ferritinophagy is a critical mechanism underlying white matter injury after TBI. ISRIB holds promise as a therapeutic agent for this condition.
Collapse
Affiliation(s)
- Wenzhu Zhou
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yidan Liang
- Department of Chongqing Emergency Medical Center, Chongqing University Center Hospital, School of Medicine, Chongqing University, Chongqing, 400016, China
| | - Xinyu Liao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Luyao Tong
- Department of Medical Technology, Anhui Medical College, Hefei, 230601, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Wenqiao Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - ShanShan Tian
- Department of Chongqing Emergency Medical Center, Chongqing University Center Hospital, School of Medicine, Chongqing University, Chongqing, 400016, China
| | - Yongbing Deng
- Department of Chongqing Emergency Medical Center, Chongqing University Center Hospital, School of Medicine, Chongqing University, Chongqing, 400016, China.
| | - Xue Jiang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, College of Basic Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
10
|
Lakhal-Littleton S, Cleland JGF. Iron deficiency and supplementation in heart failure. Nat Rev Cardiol 2024; 21:463-486. [PMID: 38326440 DOI: 10.1038/s41569-024-00988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Non-anaemic iron deficiency (NAID) is a strategic target in cardiovascular medicine because of its association with a range of adverse effects in various conditions. Endeavours to tackle NAID in heart failure have yielded mixed results, exposing knowledge gaps in how best to define 'iron deficiency' and the handling of iron therapies by the body. To address these gaps, we harness the latest understanding of the mechanisms of iron homeostasis outside the erythron and integrate clinical and preclinical lines of evidence. The emerging picture is that current definitions of iron deficiency do not assimilate the multiple influences at play in patients with heart failure and, consequently, fail to identify those with a truly unmet need for iron. Additionally, current iron supplementation therapies benefit only certain patients with heart failure, reflecting differences in the nature of the unmet need for iron and the modifying effects of anaemia and inflammation on the handling of iron therapies by the body. Building on these insights, we identify untapped opportunities in the management of NAID, including the refinement of current approaches and the development of novel strategies. Lessons learned from NAID in cardiovascular disease could ultimately translate into benefits for patients with other chronic conditions such as chronic kidney disease, chronic obstructive pulmonary disease and cancer.
Collapse
Affiliation(s)
| | - John G F Cleland
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
11
|
Shin HE, Pan C, Curran DM, Bateman TJ, Chong DHY, Ng D, Shah M, Moraes TF. Prevalence of Slam-dependent hemophilins in Gram-negative bacteria. J Bacteriol 2024; 206:e0002724. [PMID: 38814789 PMCID: PMC11332172 DOI: 10.1128/jb.00027-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024] Open
Abstract
Iron acquisition systems are crucial for pathogen growth and survival in iron-limiting host environments. To overcome nutritional immunity, bacterial pathogens evolved to use diverse mechanisms to acquire iron. Here, we examine a heme acquisition system that utilizes hemophores called hemophilins which are also referred to as HphAs in several Gram-negative bacteria. In this study, we report three new HphA structures from Stenotrophomonas maltophilia, Vibrio harveyi, and Haemophilus parainfluenzae. Structural determination of HphAs revealed an N-terminal clamp-like domain that binds heme and a C-terminal eight-stranded β-barrel domain that shares the same architecture as the Slam-dependent Neisserial surface lipoproteins. The genetic organization of HphAs consists of genes encoding a Slam homolog and a TonB-dependent receptor (TBDR). We investigated the Slam-HphA system in the native organism or the reconstituted system in Escherichia coli cells and found that the efficient secretion of HphA depends on Slam. The TBDR also played an important role in heme uptake and conferred specificity for its cognate HphA. Furthermore, bioinformatic analysis of HphA homologs revealed that HphAs are conserved in the alpha, beta, and gammaproteobacteria. Together, these results show that the Slam-dependent HphA-type hemophores are prevalent in Gram-negative bacteria and further expand the role of Slams in transporting soluble proteins. IMPORTANCE This paper describes the structure and function of a family of Slam (Type IX secretion System) secreted hemophores that bacteria use to uptake heme (iron) while establishing an infection. Using structure-based bioinformatics analysis to define the diversity and prevalence of this heme acquisition pathway, we discovered that a large portion of gammaproteobacterial harbors this system. As organisms, including Acinetobacter baumannii, utilize this system to facilitate survival during host invasion, the identification of this heme acquisition system in bacteria species is valuable information and may represent a target for antimicrobials.
Collapse
Affiliation(s)
- Hyejin Esther Shin
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Chuxi Pan
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - David M. Curran
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Thomas J. Bateman
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | | | - Dixon Ng
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Megha Shah
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Trevor F. Moraes
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Liu J, Deng L, Qu L, Li X, Wang T, Chen Y, Jiang M, Zou W. Herbal medicines provide regulation against iron overload in cardiovascular diseases: Informing future applications. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117941. [PMID: 38387684 DOI: 10.1016/j.jep.2024.117941] [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: 03/27/2023] [Revised: 02/04/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Iron is an essential micronutrient for maintaining physiological activities, especially for highly active cardiomyocytes. Inappropriate iron overload or deficiency has a significant impact on the incidence and severity of cardiovascular diseases (CVD). Iron overload exerts potentially deleterious effects on doxorubicin (DOX) cardiomyopathy, atherosclerosis, and myocardial ischemia-reperfusion injury (MI/RI) by participating in lipid peroxides production. Notably, iron overload-associated cell death has been defined as a possible mechanism for ferroptosis. At present, some traditional herbal medicines and extracts have been included in the study of regulating iron overload and the subsequent therapeutic effect on CVD. AIM OF THE STUDY To give an outline of iron metabolism and ferroptosis in cardiomyocytes and to focus on herbal medicines and extracts to prevent iron overload in CVD. MATERIALS AND METHODS Literature information was systematically collected from ScienceDirect, PubMed, Google Scholar, Web of Science, China National Knowledge Infrastructure, WanFang data, as well as classic books and clinical reports. RESULTS After understanding the mechanism of iron overload on CVD, this paper reviews the therapeutic function of various herbal medicines in eliminating iron overload in CVD. These include Chinese herbal compound prescriptions (Salvia miltiorrhiza injection, Gegen Qinlian decoction, Tongxinluo, Banxia-Houpu decoction), plant extracts, phenylpropanoids, flavonoids, terpenoids, and polyphenols. Among them, flavonoids are considered to be the most promising compounds because of their prominent iron chelation. Mechanically, these herbal medicines act on the Nrf2 signaling pathway, AMPK signaling pathway, and KAT5/GPX4 signaling pathway, thereby attenuating iron overload and lipid peroxidation in CVD. CONCLUSION Our review provides up-to-date information on herbal medicines that exert cardiovascular protective effects by modulating iron overload and ferroptosis. These herbal medicines hold promise as a template for preventing iron overload in CVD.
Collapse
Affiliation(s)
- Jia Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Liangyan Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Liping Qu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Xiaofen Li
- School of Basic Medicine Sciences, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Tao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yuanyuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Miao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Wenjun Zou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
13
|
Feng Z, Wang Y, Fu Z, Liao J, Liu H, Zhou M. Exploring the Causal Effects of Mineral Metabolism Disorders on Telomere and Mitochondrial DNA: A Bidirectional Two-Sample Mendelian Randomization Analysis. Nutrients 2024; 16:1417. [PMID: 38794655 PMCID: PMC11123946 DOI: 10.3390/nu16101417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The aim of this study was to assess the causal relationships between mineral metabolism disorders, representative of trace elements, and key aging biomarkers: telomere length (TL) and mitochondrial DNA copy number (mtDNA-CN). Utilizing bidirectional Mendelian randomization (MR) analysis in combination with the two-stage least squares (2SLS) method, we explored the causal relationships between mineral metabolism disorders and these aging indicators. Sensitivity analysis can be used to determine the reliability and robustness of the research results. The results confirmed that a positive causal relationship was observed between mineral metabolism disorders and TL (p < 0.05), while the causal relationship with mtDNA-CN was not significant (p > 0.05). Focusing on subgroup analyses of specific minerals, our findings indicated a distinct positive causal relationship between iron metabolism disorders and both TL and mtDNA-CN (p < 0.05). In contrast, disorders in magnesium and phosphorus metabolism did not exhibit significant causal effects on either aging biomarker (p > 0.05). Moreover, reverse MR analysis did not reveal any significant causal effects of TL and mtDNA-CN on mineral metabolism disorders (p > 0.05). The combination of 2SLS with MR analysis further reinforced the positive causal relationship between iron levels and both TL and mtDNA-CN (p < 0.05). Notably, the sensitivity analysis did not indicate significant pleiotropy or heterogeneity within these causal relationships (p > 0.05). These findings highlight the pivotal role of iron metabolism in cellular aging, particularly in regulating TL and sustaining mtDNA-CN, offering new insights into how mineral metabolism disorders influence aging biomarkers. Our research underscores the importance of trace element balance, especially regarding iron intake, in combating the aging process. This provides a potential strategy for slowing aging through the adjustment of trace element intake, laying the groundwork for future research into the relationship between trace elements and healthy aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China or (Z.F.); (Y.W.); (Z.F.); (J.L.); (H.L.)
| |
Collapse
|
14
|
Kumar A, Ye C, Nkansah A, Decoville T, Fogo GM, Sajjakulnukit P, Reynolds MB, Zhang L, Quaye O, Seo YA, Sanderson TH, Lyssiotis CA, Chang CH. Iron regulates the quiescence of naive CD4 T cells by controlling mitochondria and cellular metabolism. Proc Natl Acad Sci U S A 2024; 121:e2318420121. [PMID: 38621136 PMCID: PMC11047099 DOI: 10.1073/pnas.2318420121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/14/2024] [Indexed: 04/17/2024] Open
Abstract
In response to an immune challenge, naive T cells undergo a transition from a quiescent to an activated state acquiring the effector function. Concurrently, these T cells reprogram cellular metabolism, which is regulated by iron. We and others have shown that iron homeostasis controls proliferation and mitochondrial function, but the underlying mechanisms are poorly understood. Given that iron derived from heme makes up a large portion of the cellular iron pool, we investigated iron homeostasis in T cells using mice with a T cell-specific deletion of the heme exporter, FLVCR1 [referred to as knockout (KO)]. Our finding revealed that maintaining heme and iron homeostasis is essential to keep naive T cells in a quiescent state. KO naive CD4 T cells exhibited an iron-overloaded phenotype, with increased spontaneous proliferation and hyperactive mitochondria. This was evidenced by reduced IL-7R and IL-15R levels but increased CD5 and Nur77 expression. Upon activation, however, KO CD4 T cells have defects in proliferation, IL-2 production, and mitochondrial functions. Iron-overloaded CD4 T cells failed to induce mitochondrial iron and exhibited more fragmented mitochondria after activation, making them susceptible to ferroptosis. Iron overload also led to inefficient glycolysis and glutaminolysis but heightened activity in the hexosamine biosynthetic pathway. Overall, these findings highlight the essential role of iron in controlling mitochondrial function and cellular metabolism in naive CD4 T cells, critical for maintaining their quiescent state.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Chenxian Ye
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Afia Nkansah
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, AccraG4522, Ghana
| | - Thomas Decoville
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Garrett M. Fogo
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI48109
| | - Peter Sajjakulnukit
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI48109
| | - Mack B. Reynolds
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Li Zhang
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI48109
| | - Osbourne Quaye
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, AccraG4522, Ghana
| | - Young-Ah Seo
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI48109
| | - Thomas H. Sanderson
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Costas A. Lyssiotis
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, MI48109
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Cheong-Hee Chang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI48109
| |
Collapse
|
15
|
Ashoub MH, Razavi R, Heydaryan K, Salavati-Niasari M, Amiri M. Targeting ferroptosis for leukemia therapy: exploring novel strategies from its mechanisms and role in leukemia based on nanotechnology. Eur J Med Res 2024; 29:224. [PMID: 38594732 PMCID: PMC11003188 DOI: 10.1186/s40001-024-01822-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
The latest findings in iron metabolism and the newly uncovered process of ferroptosis have paved the way for new potential strategies in anti-leukemia treatments. In the current project, we reviewed and summarized the current role of nanomedicine in the treatment and diagnosis of leukemia through a comparison made between traditional approaches applied in the treatment and diagnosis of leukemia via the existing investigations about the ferroptosis molecular mechanisms involved in various anti-tumor treatments. The application of nanotechnology and other novel technologies may provide a new direction in ferroptosis-driven leukemia therapies. The article explores the potential of targeting ferroptosis, a new form of regulated cell death, as a new therapeutic strategy for leukemia. It discusses the mechanisms of ferroptosis and its role in leukemia and how nanotechnology can enhance the delivery and efficacy of ferroptosis-inducing agents. The article not only highlights the promise of ferroptosis-targeted therapies and nanotechnology in revolutionizing leukemia treatment, but also calls for further research to overcome challenges and fully realize the clinical potential of this innovative approach. Finally, it discusses the challenges and opportunities in clinical applications of ferroptosis.
Collapse
Affiliation(s)
- Muhammad Hossein Ashoub
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Kamran Heydaryan
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Masoud Salavati-Niasari
- Institute of Nano Science and Nano Technology, University of Kashan, P.O. Box 87317-51167, Kashan, Iran
| | - Mahnaz Amiri
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran.
| |
Collapse
|
16
|
Li SL, Hou HY, Chu X, Zhu YY, Zhang YJ, Duan MD, Liu J, Liu Y. Nanomaterials-Involved Tumor-Associated Macrophages' Reprogramming for Antitumor Therapy. ACS NANO 2024; 18:7769-7795. [PMID: 38420949 DOI: 10.1021/acsnano.3c12387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Tumor-associated macrophages (TAMs) play pivotal roles in tumor development. As primary contents of tumor environment (TME), TAMs secrete inflammation-related substances to regulate tumoral occurrence and development. There are two kinds of TAMs: the tumoricidal M1-like TAMs and protumoral M2-like TAMs. Reprogramming TAMs from immunosuppressive M2 to immunocompetent M1 phenotype is considered a feasible way to improve immunotherapeutic efficiency. Notably, nanomaterials show great potential for biomedical fields due to their controllable structures and properties. There are many types of nanomaterials that exhibit great regulatory activities for TAMs' reprogramming. In this review, the recent progress of nanomaterials-involved TAMs' reprogramming is comprehensively discussed. The various nanomaterials for TAMs' reprogramming and the reprogramming strategies are summarized and introduced. Additionally, the challenges and perspectives of TAMs' reprogramming for efficient therapy are discussed, aiming to provide inspiration for TAMs' regulator design and promote the development of TAMs-mediated immunotherapy.
Collapse
Affiliation(s)
- Shu-Lan Li
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Hua-Ying Hou
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Xu Chu
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Ying Zhu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
| | - Yu-Juan Zhang
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Meng-Die Duan
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
| | - Junyi Liu
- Albany Medical College, New York 12208, United States
| | - Yi Liu
- State Key Laboratory of Separation Membrane and Membrane Process, School of Chemistry & School of Electronic and Information Engineering, Tiangong University, Tianjin 300387, P. R. China
- School of Materials Science and Engineering & School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, P. R. China
- School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, P. R. China
| |
Collapse
|
17
|
Zhang EX, Hao WW, Wang ZH, Shi YR. Mechanism of prevention and treatment of ulcerative colitis from the perspective of iron death. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:109-115. [DOI: 10.11569/wcjd.v32.i2.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
|
18
|
Liu Y, Liu X, Duan L, Zhao Y, He Y, Li W, Cui J. Associations of micronutrient dietary patterns with sarcopenia among US adults: a population-based study. Front Nutr 2024; 11:1301831. [PMID: 38410638 PMCID: PMC10894935 DOI: 10.3389/fnut.2024.1301831] [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: 09/25/2023] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Background Current epidemiological evidence points to an association between micronutrient (MN) intake and sarcopenia, but studies have focused on single MN, and no combined effects on MNs have been reported. The aim of this study was to investigate the relationship between different MN intake patterns and sarcopenia and skeletal muscle mass. Methods We performed a population-based cross-sectional study, with a total of 5,256 U.S. adults aged 20-59 years, and we collected total daily MN intake and appendicular skeletal muscle mass measured by Dual-Energy X-ray Absorptiometry (DXA). Principal component analysis (PCA) was used to obtain nutrient patterns and principal component scores based on the intake of 14 MNs, and logistic regression analysis was used to assess the effects of single MN and MN intake patterns on sarcopenia and muscle mass. Results We defined three MN intake patterns by PCA: (1) adherence to VitB-mineral, high intake of vitamin B and minerals; (2) adherence to VitAD-Ca-VB12, high intake of vitamin A, vitamin D, calcium and vitamin B12; and (3) adherence to Antioxidant Vit, high intake of antioxidant vitamins A, C, E, and K. These three nutrient patterns explained 73.26% of the variance of the population. A negative association was observed between most single MN intakes and sarcopenia, and after adjusting for confounders, adherence to the highest tertile of the three nutrient patterns was associated with a lower risk of sarcopenia and relatively higher skeletal muscle mass compared to the lowest adherence. In subgroup analysis, MN intake patterns were significantly correlated with sarcopenia in middle-aged females. Conclusion Nutritional patterns based on MN intake were significantly related to sarcopenia, indicating that MNs interact with each other while exerting their individual functions, and that MN dietary patterns may provide promising strategies for preventing the loss of muscle mass, with further prospective studies warranted in the future.
Collapse
Affiliation(s)
- Yining Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xiangliang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Linnan Duan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yixin Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yuwei He
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Troike KM, Wang SZ, Silver DJ, Lee J, Mulkearns-Hubert EE, Hajdari N, Ghosh PK, Kay KE, Beilis JL, Mitchell SE, Bishop CW, Hong ES, Artomov M, Hubert CG, Rajappa P, Connor JR, Fox PL, Kristensen BW, Lathia JD. Homeostatic iron regulatory protein drives glioblastoma growth via tumor cell-intrinsic and sex-specific responses. Neurooncol Adv 2024; 6:vdad154. [PMID: 38239626 PMCID: PMC10794878 DOI: 10.1093/noajnl/vdad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2024] Open
Abstract
Background Glioblastoma (GBM) displays alterations in iron that drive proliferation and tumor growth. Iron regulation is complex and involves many regulatory mechanisms, including the homeostatic iron regulator (HFE) gene, which encodes the homeostatic iron regulatory protein. While HFE is upregulated in GBM and correlates with poor survival outcomes, the function of HFE in GBM remains unclear. Methods We interrogated the impact of cell-intrinsic Hfe expression on proliferation and survival of intracranially implanted animals through genetic gain- and loss-of-function approaches in syngeneic mouse glioma models, along with in vivo immune assessments. We also determined the expression of iron-associated genes and their relationship to survival in GBM using public data sets and used transcriptional profiling to identify differentially expressed pathways in control compared to Hfe-knockdown cells. Results Overexpression of Hfe accelerated GBM proliferation and reduced animal survival, whereas suppression of Hfe induced apoptotic cell death and extended survival, which was more pronounced in females and associated with attenuation of natural killer cells and CD8+ T cell activity. Analysis of iron gene signatures in Hfe-knockdown cells revealed alterations in the expression of several iron-associated genes, suggesting global disruption of intracellular iron homeostasis. Further analysis of differentially expressed pathways revealed oxidative stress as the top pathway upregulated following Hfe loss. Hfe knockdown indeed resulted in enhanced 55Fe uptake and generation of reactive oxygen species. Conclusions These findings reveal an essential function for HFE in GBM cell growth and survival, as well as a sex-specific interaction with the immune response.
Collapse
Affiliation(s)
- Katie M Troike
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sabrina Z Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Daniel J Silver
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Erin E Mulkearns-Hubert
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicole Hajdari
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Prabar K Ghosh
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kristen E Kay
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Julia L Beilis
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sofia E Mitchell
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christopher W Bishop
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ellen S Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Mykyta Artomov
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher G Hubert
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Prajwal Rajappa
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Neurological Surgery, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Paul L Fox
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Bjarne W Kristensen
- Department of Clinical Medicine, Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
20
|
Liang Z, Zheng Z, Guo Q, Tian M, Yang J, Liu X, Zhu X, Liu S. The role of HIF-1α/HO-1 pathway in hippocampal neuronal ferroptosis in epilepsy. iScience 2023; 26:108098. [PMID: 37876811 PMCID: PMC10590818 DOI: 10.1016/j.isci.2023.108098] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/12/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023] Open
Abstract
Epilepsy, a common central nervous system disorder, remains an enigma in pathogenesis. Emerging consensus designates hippocampal neuronal injury as a cornerstone for epileptogenic foci, pivotal in epileptic genesis and progression. Ferroptosis, a regulated cell death modality hinging on iron, catalyzes lipid reactive oxygen species formation through iron and membrane polyunsaturated fatty acid interplay, culminating in oxidative cell death. This research investigates the role of hypoxia-inducible factor (HIF)-1α/heme oxygenase (HO)-1 in hippocampal neuron ferroptosis during epilepsy. Untargeted metabolomics exposes metabolite discrepancies between epilepsy patients and healthy individuals, unveiling escalated oxidative stress, heightened bilirubin, and augmented iron metabolism in epileptic blood. Enrichment analyses unveil active HIF-1 pathway in epileptic pathogenesis, reinforced by HIF-1α signaling perturbations in DisGeNET database. PTZ-kindled mice model confirms increased ferroptotic markers, oxidative stress, HIF-1α, and HO-1 in epilepsy. Study implicates HIF-1α/HO-1 potentially regulates hippocampal neuronal ferroptosis, iron metabolism, and oxidative stress, thereby promoting the propagation of epilepsy.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Zhaoshi Zheng
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qi Guo
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Tian
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Jing Yang
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xiu Liu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Xiaojuan Zhu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Zhao H, Tang C, Wang M, Zhao H, Zhu Y. Ferroptosis as an emerging target in rheumatoid arthritis. Front Immunol 2023; 14:1260839. [PMID: 37928554 PMCID: PMC10620966 DOI: 10.3389/fimmu.2023.1260839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/22/2023] [Indexed: 11/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology. Due to the rise in the incidence rate of RA and the limitations of existing therapies, the search for new treatment strategies for RA has become a global focus. Ferroptosis is a novel programmed cell death characterized by iron-dependent lipid peroxidation, with distinct differences from apoptosis, autophagy, and necrosis. Under the conditions of iron accumulation and the glutathione peroxidase 4 (GPX4) activity loss, the lethal accumulation of lipid peroxide is the direct cause of ferroptosis. Ferroptosis mediates inflammation, oxidative stress, and lipid oxidative damage processes, and also participates in the occurrence and pathological progression of inflammatory joint diseases including RA. This review provides insight into the role and mechanism of ferroptosis in RA and discusses the potential and challenges of ferroptosis as a new therapeutic strategy for RA, with an effort to provide new targets for RA prevention and treatment.
Collapse
Affiliation(s)
- Hui Zhao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Cheng Tang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Miao Wang
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Hongfang Zhao
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Yan Zhu
- The Geriatrics, Graduate School of Anhui University of Chinese Medicine, Hefei, China
- The Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
22
|
Anne Marie U, Murererehe J, Rehman M, Chittilla M, Uwambaye P, Razzaque MS. Oral manifestations of iron imbalance. Front Nutr 2023; 10:1272902. [PMID: 37899821 PMCID: PMC10611504 DOI: 10.3389/fnut.2023.1272902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/11/2023] [Indexed: 10/31/2023] Open
Affiliation(s)
- Uwitonze Anne Marie
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
| | - Julienne Murererehe
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
| | - Mahum Rehman
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Mythri Chittilla
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Peace Uwambaye
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
| | - Mohammed S. Razzaque
- Department of Preventive and Community Dentistry, School of Dentistry, University of Rwanda College of Medicine and Health Sciences, Kigali, Rwanda
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| |
Collapse
|
23
|
El-Fakharany EM, El-Maradny YA, Ashry M, Abdel-Wahhab KG, Shabana ME, El-Gendi H. Green synthesis, characterization, anti-SARS-CoV-2 entry, and replication of lactoferrin-coated zinc nanoparticles with halting lung fibrosis induced in adult male albino rats. Sci Rep 2023; 13:15921. [DOI: https:/doi.org/10.1038/s41598-023-42702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/13/2023] [Indexed: 11/09/2023] Open
Abstract
AbstractThe ethanolic extract of Coleus forskohlii Briq leaves was employed in the green synthesis of zinc nanoparticles (Zn-NPs) by an immediate, one-step, and cost-effective method in the present study. Zn-NPs were coated with purified bovine lactoferrin (LF) and characterized through different instrumental analysis. The biosynthesized Zn-NPs were white in color revealing oval to spherical-shaped particles with an average size of 77 ± 5.50 nm, whereas LF-coated Zn-NPs (LF-Zn-NPs) revealed a larger particles size of up to 98 ± 6.40 nm. The biosynthesized Zn-NPs and LF-Zn-NPs revealed negatively charged surfaces with zeta-potentials of – 20.25 ± 0.35 and – 44.3 ± 3.25 mV, respectively. Interestingly, the LF-Zn-NPs showed potent in vitro retardation for SARS-CoV-2 entry to host cells by binding to the ACE2-receptor and spike protein receptor binding domain at IC50 values of 59.66 and μg/mL, respectively. Additionally, the results indicated the ability of LF-Zn-NPs to inhibit SARS-CoV-2 replication by interfering with RNA-dependent RNA polymerase “RdRp” activity at IC50 of 49.23 μg/mL. In vivo, the LF-Zn-NPs displayed a protective and therapeutic activity against induced pulmonary fibrosis in Bleomycin-treated male albino rats owing to its anti-inflammatory, antioxidant, and significant reduction in CRP, LDH, ferritin, and D-dimer levels. The obtained findings offer a promising route for biosynthesized Zn-NPs and LF-Zn-NPs as promising candidates against COVID-19.
Collapse
|
24
|
El-Fakharany EM, El-Maradny YA, Ashry M, Abdel-Wahhab KG, Shabana ME, El-Gendi H. Green synthesis, characterization, anti-SARS-CoV-2 entry, and replication of lactoferrin-coated zinc nanoparticles with halting lung fibrosis induced in adult male albino rats. Sci Rep 2023; 13:15921. [PMID: 37741872 PMCID: PMC10518009 DOI: 10.1038/s41598-023-42702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023] Open
Abstract
The ethanolic extract of Coleus forskohlii Briq leaves was employed in the green synthesis of zinc nanoparticles (Zn-NPs) by an immediate, one-step, and cost-effective method in the present study. Zn-NPs were coated with purified bovine lactoferrin (LF) and characterized through different instrumental analysis. The biosynthesized Zn-NPs were white in color revealing oval to spherical-shaped particles with an average size of 77 ± 5.50 nm, whereas LF-coated Zn-NPs (LF-Zn-NPs) revealed a larger particles size of up to 98 ± 6.40 nm. The biosynthesized Zn-NPs and LF-Zn-NPs revealed negatively charged surfaces with zeta-potentials of - 20.25 ± 0.35 and - 44.3 ± 3.25 mV, respectively. Interestingly, the LF-Zn-NPs showed potent in vitro retardation for SARS-CoV-2 entry to host cells by binding to the ACE2-receptor and spike protein receptor binding domain at IC50 values of 59.66 and μg/mL, respectively. Additionally, the results indicated the ability of LF-Zn-NPs to inhibit SARS-CoV-2 replication by interfering with RNA-dependent RNA polymerase "RdRp" activity at IC50 of 49.23 μg/mL. In vivo, the LF-Zn-NPs displayed a protective and therapeutic activity against induced pulmonary fibrosis in Bleomycin-treated male albino rats owing to its anti-inflammatory, antioxidant, and significant reduction in CRP, LDH, ferritin, and D-dimer levels. The obtained findings offer a promising route for biosynthesized Zn-NPs and LF-Zn-NPs as promising candidates against COVID-19.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Alexandria, 21934, Egypt.
| | - Yousra A El-Maradny
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Alexandria, 21934, Egypt
- Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Alamein, 51718, Egypt
| | - Mahmoud Ashry
- Zoology Department, Faculty of Science, Al-Azhar University, Assuit, Egypt
| | | | | | - Hamada El-Gendi
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Alexandria, 21934, Egypt
| |
Collapse
|
25
|
El-Fakharany EM, El-Gendi H, El-Maradny YA, Abu-Serie MM, Abdel-Wahhab KG, Shabana ME, Ashry M. Inhibitory effect of lactoferrin-coated zinc nanoparticles on SARS-CoV-2 replication and entry along with improvement of lung fibrosis induced in adult male albino rats. Int J Biol Macromol 2023; 245:125552. [PMID: 37356684 PMCID: PMC10290166 DOI: 10.1016/j.ijbiomac.2023.125552] [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: 04/11/2023] [Revised: 06/12/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Severe acute respiratory syndrome 2019-new coronavirus (SARS-CoV-2) is a major global challenge caused by a pandemic disease, named 'COVID-19' with no effective and selective therapy available so far. COVID-19-associated mortality is directly related to the inability to suppress the viral infection and the uncontrolled inflammatory response. So, we investigated the antiviral efficiency of the nanofabricated and well-characterized lactoferrin-coated zinc nanoparticles (Lf-Zn-NPs) on SARS-CoV-2 replication and entry into host cells. Lf-Zn-NPs showed potent inhibition of the entry of SARS-CoV-2 into the host cells by inhibition of ACE2, the SARS-CoV-2 receptor. This inhibitory activity of Lf-Zn-NPs to target the interaction between the SARS-CoV-2 spike protein and the ACE2 receptor offers potent protection against COVID-19 outbreaks. Moreover, the administration of Lf-Zn-NPs markedly improved lung fibrosis disorders, as supported by histopathological findings and monitored by the significant reduction in the values of CRP, LDH, ferritin, and D-dimer, with a remarkable rise in CD4+, lung SOD, GPx, GSH, and CAT levels. Lf-Zn-NPs revealed therapeutic efficiency against lung fibrosis owing to their anti-inflammatory, antioxidant, and ACE2-inhibiting activities. These findings suggest a promising nanomedicine agent against COVID-19 and its complications, with improved antiviral and immunomodulatory properties as well as a safer mode of action.
Collapse
Affiliation(s)
- Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA- City), New Borg El-Arab City 21934, Alexandria, Egypt.
| | - Hamada El-Gendi
- Bioprocess Development Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City 21934, Alexandria, Egypt.
| | - Yousra A El-Maradny
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA- City), New Borg El-Arab City 21934, Alexandria, Egypt; Microbiology and Immunology, Faculty of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Alamein 51718, Egypt
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab 21934, Alexandria, Egypt
| | | | | | - Mahmoud Ashry
- Zoology Department, Faculty of Science, Al-Azhar University, Assuit, Egypt
| |
Collapse
|
26
|
Sande R, Doshi G, Godad A. Deciphering the role of metal and non-metals in the treatment of epilepsy. Neurochem Int 2023; 167:105536. [PMID: 37178926 DOI: 10.1016/j.neuint.2023.105536] [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: 02/18/2023] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Metals and non-metals have known to play a significant role in various physiological roles in the body including the central nervous system (CNS). The alterations in their concentration in the CNS leads to abnormalities in the normal functions which may lead to various neurological conditions including epilepsy. Manganese is a cofactor required for antioxidant enzymes such as Superoxide dismutase, Glutamine synthetase, etc. The accumulation of iron leads to formation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) which have the potential to cause ferroptosis, one of the reasons for epileptogenesis. Zinc has biphasic response, both neurotoxic and neuroprotective, based on concentration levels in the CNS. Selenium is a main element for selenoproteins which is responsible for the regulation of oxidative state and antioxidant defence mechanism. The reduction in the phosphorous levels in the CNS is widely observed after generalised tonic clonic seizures (GTC), which can be a potential diagnostic biomarker. Copper acts in the CNS in an identical manner, i.e., by blocking both AMPA mediated and GABA mediated neuronal transmission. Magnesium blocks calcium channels in the NMDA receptor and prevents glutamatergic transmission, thus inhibiting excitotoxicity. Lithium acts as a proconvulsive agent and is used in combination with pilocarpine to induce seizures. The identified potential of metals and non-metals in epilepsy can be utilised in order to devise new adjuvant therapies for the management of epilepsy. The article summaries in depth the role of metals and non-metals in the treatment of epilepsy supported with special paragraph on author perspective on to the topic. Furthermore, an update of preclinical and clinical evidences are discussed in the review to give evidence on metal and non-metal based therapies in epilepsy.
Collapse
Affiliation(s)
- Ruksar Sande
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India
| | - Angel Godad
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai, 400056, India.
| |
Collapse
|
27
|
Qin Y, Bai D, Tang M, Zhang M, Zhao L, Li J, Yang R, Jiang G. Ketogenic Diet Alleviates Brain Iron Deposition and Cognitive Dysfunction via Nrf2-mediated Ferroptosis pathway in APP/PS1 Mouse. Brain Res 2023; 1812:148404. [PMID: 37164173 DOI: 10.1016/j.brainres.2023.148404] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/26/2023] [Accepted: 05/06/2023] [Indexed: 05/12/2023]
Abstract
Progressive cognitive decline and increased brain iron deposition with age are important features of Alzheimer's disease. Previous studies have found that the short-term ketogenic diet has neuroprotective effects in a variety of neurodegenerative diseases, but the effects of an early and long-term ketogenic diet on brain iron content and cognition of Alzheimer's disease have not been reported. In our study, 8-week-old APP/PS1 mice were given a 12-month ketogenic or standard diet, while C57BL/6 mice matched with the age and genetic background of APP/PS1 mice were used as normal controls to be given a standard diet for the same length of time. We found that 12 months of an early ketogenic diet improved the impaired learning and memory ability of APP/PS1 mice. The improvement of cognitive function may be related to the reduction of amyloid-beta deposition and neuronal ferroptosis. The mechanism was achieved by the regulation of ferroptosis-related pathways after activation of nuclear factor erythroid 2-related factor 2 by ketogenic diet-induced elevated β-hydroxybutyrate. In addition, blood biochemical results showed that compared with the standard diet group of the disease, although the early and long-term ketogenic diet increased blood lipids to some extent, it seemed to reduce liver, renal, and myocardial damage caused by genetic differences. This will provide a piece of positive evidence for the early and long-term use of ketogenic diets in people at risk of Alzheimer's disease.
Collapse
Affiliation(s)
- Yaya Qin
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Dazhang Bai
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Ming Tang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Ming Zhang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Li Zhao
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Jia Li
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Rui Yang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China
| | - Guohui Jiang
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Road, Nanchong 637000, Sichuan, China; Institute of Neurological Diseases, North Sichuan Medical College, 234 Fujiang Road, Nanchong, Sichuan, China.
| |
Collapse
|
28
|
Rafati Rahimzadeh M, Rafati Rahimzadeh M, Kazemi S, Moghadamnia AR, Ghaemi Amiri M, Moghadamnia AA. Iron; Benefits or threatens (with emphasis on mechanism and treatment of its poisoning). Hum Exp Toxicol 2023; 42:9603271231192361. [PMID: 37526177 DOI: 10.1177/09603271231192361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Iron is a necessary biological element and one of the richest in the human body, but it can cause changes in cell function and activity control. Iron is involved in a wide range of oxidation - reduction activities. Whenever iron exceeds the cellular metabolic needs, its excess causes changes in the products of cellular respiration, such as superoxide, hydrogen peroxide and hydroxyl. The formation of these compounds causes cellular toxicity. Lack of control over reactive oxygen species causes damages to DNA, proteins, and lipids. Conversely, superoxide, hydrogen peroxide and hydroxyl are reactive oxygen species, using antioxidants, restoring DNA function, and controlling iron stores lead to natural conditions. Iron poisoning causes clinical manifestations in the gastrointestinal tract, liver, heart, kidneys, and hematopoietic system. When serum iron is elevated, serum iron concentrations, total iron-binding capacity (TIBC) and ferritin will also increase. Supportive care is provided by whole bowel irrigation (WBI), esophagogastroduodenoscopy is required to evaluate mucosal injury and remove undissolved iron tablets. The use of chelator agents such as deferoxamine mesylate, deferasirox, deferiprone, deferitrin are very effective in removing excess iron. Of course, the combined treatment of these chelators plays an important role in increasing iron excretion, and reducing side effects.
Collapse
Affiliation(s)
| | | | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Maryam Ghaemi Amiri
- Faculty of Education Development Center (EDC), Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
29
|
Ghio C, Soukup JM, Dailey LA, Ghio AJ, Schreinemachers DM, Koppes RA, Koppes AN. Lactate Production can Function to Increase Human Epithelial Cell Iron Concentration. Cell Mol Bioeng 2022; 15:571-585. [PMID: 36531860 PMCID: PMC9751240 DOI: 10.1007/s12195-022-00741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 09/21/2022] [Indexed: 11/28/2022] Open
Abstract
Introduction Under conditions of limited iron availability, plants and microbes have evolved mechanisms to acquire iron. For example, metal deficiency stimulates reprogramming of carbon metabolism, increasing activity of enzymes involved in the Krebs cycle and the glycolytic pathway. Resultant carboxylates/hydroxycarboxylates then function as ligands to complex iron and facilitate solubilization and uptake, reversing the metal deficiency. Similarly, human intestinal epithelial cells may produce lactate, a hydroxycarboxylate, during absolute and functional iron deficiency to import metal to reverse limited availability. Methods Here we investigate (1) if lactate can increase cell metal import of epithelial cells in vitro, (2) if lactate dehydrogenase (LDH) activity in and lactate production by epithelial cells correspond to metal availability, and (3) if blood concentrations of LDH in a human cohort correlate with indices of iron homeostasis. Results Results show that exposures of human epithelial cells, Caco-2, to both sodium lactate and ferric ammonium citrate (FAC) increase metal import relative to FAC alone. Similarly, fumaric, isocitric, malic, and succinic acid coincubation with FAC increase iron import relative to FAC alone. Increased iron import following exposures to sodium lactate and FAC elevated both ferritin and metal associated with mitochondria. LDH did not change after exposure to deferoxamine but decreased with 24 h exposure to FAC. Lactate levels revealed decreased levels with FAC incubation. Review of the National Health and Nutrition Examination Survey demonstrated significant negative relationships between LDH concentrations and serum iron in human cohorts. Conclusions Therefore, we conclude that iron import in human epithelial cells can involve lactate, LDH activity can reflect the availability of this metal, and blood LDH concentrations can correlate with indices of iron homeostasis.
Collapse
Affiliation(s)
- Caroline Ghio
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| | | | - Lisa A. Dailey
- US Environmental Protection Agency, Chapel Hill, NC 27514 USA
| | - Andrew J. Ghio
- US Environmental Protection Agency, Chapel Hill, NC 27514 USA
| | | | - Ryan A. Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| | - Abigail N. Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
- Department of Biology, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
- Department of Bioengineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
- Northeastern University, 360 Huntington Ave., 332 Mugar Life Science Building, Boston, MA 02115 USA
| |
Collapse
|
30
|
Zhang X, Kong F, Wang T, Huang X, Li W, Zhang M, Wen T, Liu J, Zhang Y, Meng J, Xu H. Iron oxide nanoparticles cause surface coating- and core chemistry-dependent endothelial cell ferroptosis. Nanotoxicology 2022; 16:829-843. [PMID: 36660964 DOI: 10.1080/17435390.2022.2154176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Iron oxide nanoparticles (IONPs) are mostly intended to be administrated intravenously, understanding the interaction of IONPs with vascular endothelial cells is extremely crucial for developing safe application regimes of IONPs. In this work, interactions of three kinds of IONPs to endothelial cells were investigated both in human umbilical vein endothelial cells (HUVECs) and in healthy mice. Both meso-2,3-dimercaptosuccinic acid (DMSA) coated Fe3O4 NPs (DMSA-Fe3O4 NPs) and DMSA-Fe2O3 NPs induced cell growth inhibition, while polyglucose sorbitol carboxymethyether coated Fe2O3 NPs(PSC-Fe2O3 NPs) did not. The PSC coating inhibited the cellular uptake of the IONPs. Both DMSA-Fe3O4 and DMSA-Fe2O3 NPs induced ferroptosis of HUVEC through upregulating phospholipid peroxides, which could be inhibited by typical ferroptosis inhibitors ferrostatin-1, Trolox and deferoxamine. Moreover, transforming growth factor beta 1 (TGFβ1) was upregulated by DMSA-Fe3O4 NPs at protein and gene level. The inhibitor of TGFβ1 receptor LY210 could reduce the effect. When being intravenously injected in mice, DMSA-Fe3O4 NPs were observed locating in the liver, increased the levels of lipid peroxidation (4-hydroxynonenal), acyl-CoA synthetase long-chain family member 4(ACSL4) and TGFβ1, indicating ferroptosis occurrence in vivo. The ferroptosis of vascular endothelial cells in exposure with IONPs depended on the surface coating and core chemistry of the NPs. Both DMSA-Fe3O4 NPs and DMSA-Fe2O3 NPs could induce the ferroptosis of endothelial cells, while PSC-Fe2O3 NPs did not induce ferroptosis and apoptosis possibly due to the very low cellular uptake. DMSA-Fe3O4 NPs and TGFβ1 formed feedforward loop to induce ferroptosis.
Collapse
Affiliation(s)
- Xue Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Kong
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Tian Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Huang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meichen Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Wen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Jie Meng
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Zhang L, Liu J, Dai Z, Wang J, Wu M, Su R, Zhang D. Crosstalk between regulated necrosis and micronutrition, bridged by reactive oxygen species. Front Nutr 2022; 9:1003340. [PMID: 36211509 PMCID: PMC9543034 DOI: 10.3389/fnut.2022.1003340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/26/2022] [Indexed: 11/15/2022] Open
Abstract
The discovery of regulated necrosis revitalizes the understanding of necrosis from a passive and accidental cell death to a highly coordinated and genetically regulated cell death routine. Since the emergence of RIPK1 (receptor-interacting protein kinase 1)-RIPK3-MLKL (mixed lineage kinase domain-like) axis-mediated necroptosis, various other forms of regulated necrosis, including ferroptosis and pyroptosis, have been described, which enrich the understanding of pathophysiological nature of diseases and provide novel therapeutics. Micronutrients, vitamins, and minerals, position centrally in metabolism, which are required to maintain cellular homeostasis and functions. A steady supply of micronutrients benefits health, whereas either deficiency or excessive amounts of micronutrients are considered harmful and clinically associated with certain diseases, such as cardiovascular disease and neurodegenerative disease. Recent advance reveals that micronutrients are actively involved in the signaling pathways of regulated necrosis. For example, iron-mediated oxidative stress leads to lipid peroxidation, which triggers ferroptotic cell death in cancer cells. In this review, we illustrate the crosstalk between micronutrients and regulated necrosis, and unravel the important roles of micronutrients in the process of regulated necrosis. Meanwhile, we analyze the perspective mechanism of each micronutrient in regulated necrosis, with a particular focus on reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Lei Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Jinting Liu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Ziyan Dai
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Jia Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Mengyang Wu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Ruicong Su
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, China
- *Correspondence: Di Zhang,
| |
Collapse
|
32
|
Fagundes RR, Bourgonje AR, Hu S, Barbieri R, Jansen BH, Sinnema N, Blokzijl T, Taylor CT, Weersma RK, Faber KN, Dijkstra G. HIF1α-Dependent Induction of TFRC by a Combination of Intestinal Inflammation and Systemic Iron Deficiency in Inflammatory Bowel Disease. Front Physiol 2022; 13:889091. [PMID: 35755436 PMCID: PMC9214203 DOI: 10.3389/fphys.2022.889091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/20/2022] [Indexed: 01/04/2023] Open
Abstract
Background and Aims: Iron deficiency (ID) is a frequent extra-intestinal manifestation in patients with Inflammatory Bowel Disease (IBD), who often do not respond to iron supplementation. Iron is a cofactor for hydroxylases that suppress the hypoxia-inducible factor-1α (HIF1α), a transcription factor regulating iron homeostasis. We hypothesized that iron deficiency affects mucosal HIF1α activity in IBD. Methods: IBD patients (n = 101) were subdivided based on iron status (ferritin levels or transferrin saturation) and systemic inflammation (C-reactive protein levels). 154 corresponding ileal and colonic biopsies were analyzed for differential expression of 20 HIF1α pathway-associated genes and related to iron and inflammation status. In vitro expression of selected HIF1α pathway genes were analyzed in wild-type and HIF1A-null Caco-2 cells. Results: Gene expression of the mucosal HIF1α pathway was most affected by intestinal location and inflammatory status. Especially, ileal mucosal TFRC expression, encoding the transferrin receptor TFR1, was increased in inflamed tissue (p < 0.001), and further enhanced in ID. Accordingly, TFRC expression in inflamed tissue associated negatively with serum iron levels, which was not observed in the non-inflamed mucosa. The HIF1α pathway agonist DMOG increased TFRC expression in Caco-2 cells, which was blunted in HIF1A-null cells. Conclusion: We demonstrate that inflammation and anatomical location primarily determine HIF1α pathway activation and downstream TFRC expression in the intestinal mucosa. IBD patients with ID may benefit from treatment with HIF1α-agonists by 1) increasing TFRC-mediated iron absorption in non-inflamed tissue and 2) decreasing mucosal inflammation, thereby improving their responsiveness to oral iron supplementation.
Collapse
Affiliation(s)
- Raphael R Fagundes
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Ruggero Barbieri
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Bernadien H Jansen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Nienke Sinnema
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Tjasso Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Cormac T Taylor
- School of Medicine and Medical Science and the Conway Institute, University College Dublin, Dublin, Ireland
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
33
|
Elazab MFA, Elbaiomy AEA, Ahmed MS, Alsharif KF, Dahran N, Elmahallawy EK, Mokhbatly AA. Ameliorative Effects of Bovine Lactoferrin on Benzene-Induced Hematotoxicity in Albino Rats. Front Vet Sci 2022; 9:907580. [PMID: 35812844 PMCID: PMC9257330 DOI: 10.3389/fvets.2022.907580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Benzene (Bz) is one of the major products of the petrochemical industry globally, which induces aplastic anemia and leukemia in humans and animals. This study aimed to investigate the modulatory effects of bovine lactoferrin (bLf) on Bz-induced hematotoxicity in albino rats. Eighty male rats were randomly divided into eight groups: corn oil group [2 mL/kg body weight (BW)], bLf groups (100, 200, and 300 mg/kg BW), Bz group (Bz 2 mL/kg BW; corn oil 2 mL/kg BW), and Bz + bLf groups (Bz 2 mL/kg BW; corn oil 2 mL/kg BW; bLf 100, 200, and 300 mg/kg BW). Hematobiochemical results exhibited marked pancytopenia, a significant decrease in total protein, albumin, α2- and γ-globulin, ferritin, serum iron, and total iron-binding capacity (TIBC), and an increase in serum bioactivities of aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase (ALP), lactate dehydrogenase (LDH), and erythropoietin hormone levels in Bz-treated rats. Histopathological examination revealed a marked reduction in all hematopoietic cell lines in the bone marrow (BM), necrosis in the white pulp of the spleen and cytosolic hydrops, and apoptosis of hepatocytes in the Bz-treated group. Rats treated with bLf (300 mg/kg BW) revealed marked increases in total protein, albumin, α2- and γ-globulin, ferritin, serum iron, and TIBC levels and decreases both in ALP and LDH bioactivities and erythropoietin hormone levels compared with the Bz-treated group. Histopathological results were concomitant with hematobiochemical parameters in rats treated with bLf (300 mg/kg BW), almost showing restoration of the normal cellularity of BM, the architecture of red and white pulps of the spleen, and even the normal hypertrophy of hepatocytes compared with the control groups. To conclude, bLf (300 mg/kg BW) can be recommended to treat Bz-induced hematotoxicity.
Collapse
Affiliation(s)
- Mohamed F. Abou Elazab
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Asmaa E. A. Elbaiomy
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Mohamed S. Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Naief Dahran
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Ehab Kotb Elmahallawy
- Department of Zoonoses, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Abdallah A. Mokhbatly
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| |
Collapse
|
34
|
Lai B, Wu CH, Wu CY, Luo SF, Lai JH. Ferroptosis and Autoimmune Diseases. Front Immunol 2022; 13:916664. [PMID: 35720308 PMCID: PMC9203688 DOI: 10.3389/fimmu.2022.916664] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022] Open
Abstract
Adequate control of autoimmune diseases with an unclear etiology resulting from autoreactivation of the immune system remains a major challenge. One of the factors that trigger autoimmunity is the abnormal induction of cell death and the inadequate clearance of dead cells that leads to the exposure or release of intracellular contents that activate the immune system. Different from other cell death subtypes, such as apoptosis, necroptosis, autophagy, and pyroptosis, ferroptosis has a unique association with the cellular iron load (but not the loads of other metals) and preserves its distinguishable morphological, biological, and genetic features. This review addresses how ferroptosis is initiated and how it contributes to the pathogenesis of autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel diseases. The mechanisms responsible for ferroptosis-associated events are discussed. We also cover the perspective of targeting ferroptosis as a potential therapeutic for patients with autoimmune diseases. Collectively, this review provides up-to-date knowledge regarding how ferroptosis occurs and its significance in autoimmune diseases.
Collapse
Affiliation(s)
- Benjamin Lai
- Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chien-Hsiang Wu
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chao-Yi Wu
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shue-Fen Luo
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
- *Correspondence: Jenn-Haung Lai,
| |
Collapse
|
35
|
Effects of Iron Supplementation on Testicular Function and Spermatogenesis of Iron-Deficient Rats. Nutrients 2022; 14:nu14102063. [PMID: 35631204 PMCID: PMC9144601 DOI: 10.3390/nu14102063] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 01/27/2023] Open
Abstract
Iron deficiency is the most common micronutrient deficiency in the world. Previous studies have shown that iron deficiency increases oxidative stress and decreases antioxidant enzymes, and studies of male infertility indicated that oxidative stress may affect male reproductive functions. The aim of this study was to investigate the effects of iron supplementation on spermatogenesis and testicular functions in iron-deficient rats. Three-week-old male Sprague Dawley (SD) rats were randomly divided into two groups: an iron-adequate control (AI group, 35 ppm FeSO4) and an iron-deficient group (ID group, <5 ppm FeSO4). After three weeks, the iron-deficient group was divided into an original iron-deficient group and five iron-supplemented groups, the latter fed diets containing different doses of FeSO4 (6, 12, 18, 24, and 35 ppm). After five weeks, blood and testis tissue were analyzed. We presented as median (interquartile range, IQR) for continuous measurements and compared their differences using the Kruskal−Wallis test followed by the Mann−Whitney U test among groups. The results showed that as compared with the AI group, the ID group had significantly lower serum testosterone and poorer spermatogenesis (The medians (QR) were 187.4 (185.6−190.8) of AI group vs. 87.5 (85.7−90.4) of ID group in serum testosterone, p < 0.05; 9.3 (8.8−10.6) of AI group vs. 4.9 (3.4−5.4) of ID group in mean testicular biopsy score (MTBS], p < 0.05); iron supplementation reversed the impairment of testis tissue. In the testosterone biosynthesis pathway, iron supplementation improved the lowered protein expressions of hydroxysteroid dehydrogenases caused by iron deficiency. Additionally, decreased activities of glutathione peroxidase and catalase, and increased cleaved-caspase 8 and caspase 3 expression, were found in the iron-deficient rats. The iron-supplemented rats that received > 12 ppm FeSO4 exhibited improvements in antioxidant levels. In conclusion, iron supplementation can abrogate testis dysfunction due to iron deficiency through regulation of the testicular antioxidant capacity.
Collapse
|
36
|
Gao X, Song Y, Wu J, Lu S, Min X, Liu L, Hu L, Zheng M, Du P, Yu Y, Long H, Wu H, Jia S, Yu D, Lu Q, Zhao M. Iron-dependent epigenetic modulation promotes pathogenic T cell differentiation in lupus. J Clin Invest 2022; 132:152345. [PMID: 35499082 PMCID: PMC9057600 DOI: 10.1172/jci152345] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 03/01/2022] [Indexed: 12/20/2022] Open
Affiliation(s)
- Xiaofei Gao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Yang Song
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Jiali Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Shuang Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Xiaoli Min
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Limin Liu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Longyuan Hu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Meiling Zheng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Pei Du
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Yaqin Yu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Hai Long
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Haijing Wu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| | - Sujie Jia
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha, China
| | - Di Yu
- Diamantina Institute, The University of Queensland, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, China
- Research Unit of Key Technologies of Diagnosis and Treatment for Immune-related Skin Diseases, Chinese Academy of Medical Sciences, Changsha, China
- Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
- Clinical Immunology Research Center, Central South University, Changsha, China
| |
Collapse
|
37
|
Wang C, Zhang W, Xu W, Liu Z, Huang K. AMP-activated protein kinase α1 phosphorylates PHD2 to maintain systemic iron homeostasis. Clin Transl Med 2022; 12:e854. [PMID: 35538889 PMCID: PMC9091988 DOI: 10.1002/ctm2.854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Iron is essential for all mammalian life, and either a deficiency or excess of iron can cause diseases. AMP-activated protein kinase (AMPK) is a critical regulator of metabolic homeostasis; however, it has not been established whether AMPK regulates iron metabolism. METHODS Iron, hepcidin and ferroportin levels were examined in mice with global and hepatocyte-specific knockout of AMPKα1 and AMPKα2. Primary AMPKα1 or AMPKα2 deleted hepatocytes were isolated and cultured in hypoxia condition to explore PHD2, HIF and hydroxylated HIF1α levels. We performed immunoprecipitation, in vitro AMPK kinase assay and site-direct mutant assay to detect phosphorylation sites of PHD2. We also obtained liver tissues from patients with anaemia of chronic disease undergoing surgery, AMPKα1 and hydroxylated HIF1α levels were measured by immunohistochemical analysis. RESULTS We found that mice with global deficiency of AMPKα1, but not AMPKα2, exhibited hypoferraemia as well as iron sequestration in the spleen and liver. Hepatocyte-specific, but not myeloid-specific, ablation of AMPKα1 also reduced serum iron levels in association with increased hepcidin and decreased ferroportin protein levels. Mechanistically, AMPKα1 directly phosphorylated prolyl hydroxylase domain-containing (PHD)2 at serines 61 and 136, which suppressed PHD2-dependent hydroxylation of hypoxia-inducible factor (HIF)1α and subsequent regulation of hepatic hepcidin-related iron signalling. Inhibition of PHD2 hydroxylation ameliorated abnormal iron metabolism in hepatic AMPKα1-deficient mice. Furthermore, we found hepatic AMPKα/PHD2/HIFα/ hepcidin axes were highly clinically relevant to anaemia of chronic disease. CONCLUSION In conclusion, these observations suggest that hepatic AMPKα1 has an essential role in maintaining iron homeostasis by PHD2-dependent regulation of hepcidin, thus providing a potentially promising approach for the treatment of iron disturbances in chronic diseases.
Collapse
Affiliation(s)
- Cheng Wang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular AgingTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of RheumatologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Wencheng Zhang
- Department of CardiologyQilu HospitalCheeloo College of MedicineShandong UniversityJinanChina
| | - Wenjing Xu
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhaoyu Liu
- Department of CardiologySun Yat‐sen Memorial HospitalSun Yat‐sen University, GuangzhouChina
| | - Kai Huang
- Clinic Center of Human Gene ResearchUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular AgingTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
38
|
Valencia E, García M, Fernández-Vega B, Pereiro R, Lobo L, González-Iglesias H. Targeted Analysis of Tears Revealed Specific Altered Metal Homeostasis in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2022; 63:10. [PMID: 35426907 PMCID: PMC9034717 DOI: 10.1167/iovs.63.4.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Specific altered metal homeostasis has been investigated in the tear film of age-related macular degeneration (AMD) patients considering that metal dyshomeostasis contributes to the production of free radicals, inflammation, and apoptosis and results in conformational changes of proteins. Methods A multitargeted approach based on spectrophotometry and mass spectrometry techniques has been implemented to the multiplexed quantitation of lactoferrin (LF), S100 calcium binding protein A6 (S100A6), metallothionein 1A (MT1A), complement factor H (CFH), clusterin (CLU), amyloid precursor protein (APP), Mg, P, Na, Fe, Cu, Zn, and Ca, in the tear film from 60 subjects, 31 patients diagnosed with the dry form of AMD, and 29 healthy individuals Results Significant up-regulations of MT1A (1.9-fold) and S100A6 (1.4-fold) and down-regulations of LF (0.7-fold), Fe (0.6-fold), Mg (0.7-fold), and Cu (0.7-fold) were observed in AMD patients, when compared to control subjects. Of all the studied variables, only APP showed negative correlation with age in the AMD group. Also, positive correlations were observed for the variables Mg and Na, Cu and Mg, and P and Mg in both the AMD and control groups, whereas positive correlations were exclusively determined in the AMD group for Cu and LF, Na and Ca, and Mg and Ca. The panel constituted of MT1A, Na, and Mg predicts AMD disease in 73% of cases. Conclusions The different levels of target metals and (metallo-)proteins in the tear film suggest altered metal homeostasis in AMD patients. These observed pathophysiological changes may be related with the anomalous protein aggregation in the macula.
Collapse
Affiliation(s)
- Eva Valencia
- Ophtalmological Research Foundation, University Institute Fernández-Vega, University of Oviedo, Oviedo, Spain.,Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Oviedo, Spain
| | - Montserrat García
- Ophtalmological Research Foundation, University Institute Fernández-Vega, University of Oviedo, Oviedo, Spain.,Ophthalmological Institute Fernández-Vega, Oviedo, Spain
| | - Beatriz Fernández-Vega
- Ophtalmological Research Foundation, University Institute Fernández-Vega, University of Oviedo, Oviedo, Spain.,Ophthalmological Institute Fernández-Vega, Oviedo, Spain
| | - Rosario Pereiro
- Ophtalmological Research Foundation, University Institute Fernández-Vega, University of Oviedo, Oviedo, Spain.,Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Oviedo, Spain
| | - Lara Lobo
- Department of Physical and Analytical Chemistry, Faculty of Chemistry, University of Oviedo, Oviedo, Spain
| | - Héctor González-Iglesias
- Ophtalmological Research Foundation, University Institute Fernández-Vega, University of Oviedo, Oviedo, Spain.,Ophthalmological Institute Fernández-Vega, Oviedo, Spain
| |
Collapse
|
39
|
Grove A. Extracytoplasmic Function Sigma Factors Governing Production of the Primary Siderophores in Pathogenic Burkholderia Species. Front Microbiol 2022; 13:851011. [PMID: 35283809 PMCID: PMC8908255 DOI: 10.3389/fmicb.2022.851011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Bacteria respond to changing environments by modulating their gene expression programs. One of the mechanisms by which this may be accomplished is by substituting the primary σ factor with an alternative σ factor belonging to the family of extracytoplasmic function (ECF) σ factors. ECF σ factors are activated only in presence of specific signals, and they direct the RNA polymerase (RNAP) to transcribe a defined subset of genes. One condition, which may trigger the activation of an ECF σ factor, is iron limitation. To overcome iron starvation, bacteria produce and secrete siderophores, which chelate iron and facilitate its cellular uptake. In the genus Burkholderia, which includes several serious human pathogens, uptake of iron is critical for virulence, and expression of biosynthetic gene clusters encoding proteins involved in synthesis and transport of the primary siderophores are under control of an ECF σ factor. This review summarizes mechanisms involved in regulation of these gene clusters, including the role of global transcriptional regulators. Since siderophore-mediated iron acquisition is important for virulence, interference with this process constitutes a viable approach to the treatment of bacterial infections.
Collapse
Affiliation(s)
- Anne Grove
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
40
|
Arif HM, Qian Z, Wang R. Signaling Integration of Hydrogen Sulfide and Iron on Cellular Functions. Antioxid Redox Signal 2022; 36:275-293. [PMID: 34498949 DOI: 10.1089/ars.2021.0203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Hydrogen sulfide (H2S) is an endogenous signaling molecule, regulating numerous physiological functions from vasorelaxation to neuromodulation. Iron is a well-known bioactive metal ion, being the central component of hemoglobin for oxygen transportation and participating in biomolecule degradation, redox balance, and enzymatic actions. The interplay between H2S and iron metabolisms and functions impacts significantly on the fate and wellness of different types of cells. Recent Advances: Iron level in vivo affects the production of H2S via nonenzymatic reactions. On the contrary, H2S quenches excessive iron inside the cells and regulates the redox status of iron. Critical Issues: Abnormal metabolisms of both iron and H2S are associated with various conditions and diseases such as iron overload, anemia, oxidative stress, and cardiovascular and neurodegenerative diseases. The molecular mechanisms for the interactions between H2S and iron are unsettled yet. Here we review signaling links of the production, metabolism, and their respective and integrative functions of H2S and iron in normalcy and diseases. Future Directions: Physiological and pathophysiological importance of H2S and iron as well as their therapeutic applications should be evaluated jointly, not separately. Future investigation should expand from iron-rich cells and tissues to the others, in which H2S and iron interaction has not received due attention. Antioxid. Redox Signal. 36, 275-293.
Collapse
Affiliation(s)
| | - Zhongming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, China
| | - Rui Wang
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
41
|
Jayakumar D, S Narasimhan KK, Periandavan K. Triad role of hepcidin, ferroportin, and Nrf2 in cardiac iron metabolism: From health to disease. J Trace Elem Med Biol 2022; 69:126882. [PMID: 34710708 DOI: 10.1016/j.jtemb.2021.126882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 11/28/2022]
Abstract
Iron is an essential trace element required for several vital physiological and developmental processes, including erythropoiesis, bone, and neuronal development. Iron metabolism and oxygen homeostasis are interlinked to perform a vital role in the functionality of the heart. The metabolic machinery of the heart utilizes almost 90 % of oxygen through the electron transport chain. To handle this tremendous level of oxygen, the iron metabolism in the heart is utmost crucial. Iron availability to the heart is therefore tightly regulated by (i) the hepcidin/ferroportin axis, which controls dietary iron absorption, storage, and recycling, and (ii) iron regulatory proteins 1 and 2 (IRP1/2) via hypoxia inducible factor 1 (HIF1) pathway. Despite iron being vital to the heart, recent investigations have demonstrated that iron imbalance is a common manifestation in conditions of heart failure (HF), since free iron readily transforms between Fe2+ and Fe3+via the Fenton reaction, leading to reactive oxygen species (ROS) production and oxidative damage. Therefore, to combat iron-mediated oxidative stress, targeting Nrf2/ARE antioxidant signaling is rational. The involvement of Nrf2 in regulating several genes engaged in heme synthesis, iron storage, and iron export is beginning to be uncovered. Consequently, it is possible that Nrf2/hepcidin/ferroportin might act as an epicenter connecting iron metabolism to redox alterations. However, the mechanism bridging the two remains obscure. In this review, we tried to summarize the contemporary insight of how cardiomyocytes regulate intracellular iron levels and discussed the mechanisms linking cardiac dysfunction with iron imbalance. Further, we emphasized the impact of Nrf2 on the interplay between systemic/cardiac iron control in the context of heart disease, particularly in myocardial ischemia and HF.
Collapse
Affiliation(s)
- Deepthy Jayakumar
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India
| | - Kishore Kumar S Narasimhan
- Department of Pharmacology and Neurosciences, Creighton University, 2500 California Plaza, Omaha, NE, USA
| | - Kalaiselvi Periandavan
- Department of Medical Biochemistry, Dr. ALM Post Graduate Institute for Basic Medical Sciences, University of Madras, Chennai, 600113, Tamil Nadu, India.
| |
Collapse
|
42
|
Pecoraro L, Wang X, Shah D, Song X, Kumar V, Shakoor A, Tripathi K, Ramteke PW, Rani R. Biosynthesis Pathways, Transport Mechanisms and Biotechnological Applications of Fungal Siderophores. J Fungi (Basel) 2021; 8:21. [PMID: 35049961 PMCID: PMC8781417 DOI: 10.3390/jof8010021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Iron (Fe) is the fourth most abundant element on earth and represents an essential nutrient for life. As a fundamental mineral element for cell growth and development, iron is available for uptake as ferric ions, which are usually oxidized into complex oxyhydroxide polymers, insoluble under aerobic conditions. In these conditions, the bioavailability of iron is dramatically reduced. As a result, microorganisms face problems of iron acquisition, especially under low concentrations of this element. However, some microbes have evolved mechanisms for obtaining ferric irons from the extracellular medium or environment by forming small molecules often regarded as siderophores. Siderophores are high affinity iron-binding molecules produced by a repertoire of proteins found in the cytoplasm of cyanobacteria, bacteria, fungi, and plants. Common groups of siderophores include hydroxamates, catecholates, carboxylates, and hydroximates. The hydroxamate siderophores are commonly synthesized by fungi. L-ornithine is a biosynthetic precursor of siderophores, which is synthesized from multimodular large enzyme complexes through non-ribosomal peptide synthetases (NRPSs), while siderophore-Fe chelators cell wall mannoproteins (FIT1, FIT2, and FIT3) help the retention of siderophores. S. cerevisiae, for example, can express these proteins in two genetically separate systems (reductive and nonreductive) in the plasma membrane. These proteins can convert Fe (III) into Fe (II) by a ferrous-specific metalloreductase enzyme complex and flavin reductases (FREs). However, regulation of the siderophore through Fur Box protein on the DNA promoter region and its activation or repression depend primarily on the Fe availability in the external medium. Siderophores are essential due to their wide range of applications in biotechnology, medicine, bioremediation of heavy metal polluted environments, biocontrol of plant pathogens, and plant growth enhancement.
Collapse
Affiliation(s)
- Lorenzo Pecoraro
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, China; (X.W.); (D.S.); (X.S.); (A.S.); (R.R.)
| | - Xiao Wang
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, China; (X.W.); (D.S.); (X.S.); (A.S.); (R.R.)
| | - Dawood Shah
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, China; (X.W.); (D.S.); (X.S.); (A.S.); (R.R.)
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture Peshawar, Peshawar 25000, Pakistan
| | - Xiaoxuan Song
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, China; (X.W.); (D.S.); (X.S.); (A.S.); (R.R.)
| | - Vishal Kumar
- Department of Food Science and Technology, Yeungnam University, Gyongsan 38541, Korea;
| | - Abdul Shakoor
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, China; (X.W.); (D.S.); (X.S.); (A.S.); (R.R.)
- College of Geography and Environmental Science, Henan University, Kaifeng 475004, China
| | - Keshawanand Tripathi
- Center for Conservation and Utilization of Blue-Green Algae, ICAR-Indian Agricultural Research Institute, New Delhi 110012, India;
| | - Pramod W. Ramteke
- Faculty of Life Sciences, Mandsaur University, Mandsaur 458001, India;
| | - Rupa Rani
- School of Pharmaceutical Science and Technology, Tianjin University, 92 Weijin Road, Tianjin 300072, China; (X.W.); (D.S.); (X.S.); (A.S.); (R.R.)
- Department of Environmental Science and Engineering, Indian Institute of Technology (Indian School of Mines), Dhanbad 826004, India
| |
Collapse
|
43
|
Alnuwaysir RIS, Hoes MF, van Veldhuisen DJ, van der Meer P, Beverborg NG. Iron Deficiency in Heart Failure: Mechanisms and Pathophysiology. J Clin Med 2021; 11:125. [PMID: 35011874 PMCID: PMC8745653 DOI: 10.3390/jcm11010125] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Iron is an essential micronutrient for a myriad of physiological processes in the body beyond erythropoiesis. Iron deficiency (ID) is a common comorbidity in patients with heart failure (HF), with a prevalence reaching up to 59% even in non-anaemic patients. ID impairs exercise capacity, reduces the quality of life, increases hospitalisation rate and mortality risk regardless of anaemia. Intravenously correcting ID has emerged as a promising treatment in HF as it has been shown to alleviate symptoms, improve quality of life and exercise capacity and reduce hospitalisations. However, the pathophysiology of ID in HF remains poorly characterised. Recognition of ID in HF triggered more research with the aim to explain how correcting ID improves HF status as well as the underlying causes of ID in the first place. In the past few years, significant progress has been made in understanding iron homeostasis by characterising the role of the iron-regulating hormone hepcidin, the effects of ID on skeletal and cardiac myocytes, kidneys and the immune system. In this review, we summarise the current knowledge and recent advances in the pathophysiology of ID in heart failure, the deleterious systemic and cellular consequences of ID.
Collapse
Affiliation(s)
| | | | | | | | - Niels Grote Beverborg
- Department of Cardiology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (R.I.S.A.); (M.F.H.); (D.J.v.V.); (P.v.d.M.)
| |
Collapse
|
44
|
Douwes RM, Vinke JSJ, Gomes-Neto AW, Ayerdem G, van Hassel G, Berger SP, Touw DJ, Blokzijl H, Bakker SJL, de Borst MH, Eisenga MF. Type of proton-pump inhibitor and risk of iron deficiency in kidney transplant recipients - results from the TransplantLines Biobank and Cohort Study. Transpl Int 2021; 34:2305-2316. [PMID: 34519109 PMCID: PMC9293430 DOI: 10.1111/tri.14110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 08/16/2021] [Accepted: 09/03/2021] [Indexed: 12/16/2022]
Abstract
Proton‐pump inhibitors (PPIs) have been associated with iron deficiency (ID) in kidney transplant recipients (KTRs). Gastric acid plays a pivotal role in the intestinal absorption of non‐heme iron, but the pharmacodynamics of PPIs differs in potency of acid suppression. We hypothesized that the risk of ID might be lower in KTRs using a less potent PPI. In a cohort of 724 KTRs from the TransplantLines Biobank and Cohort Study (NCT03272841), PPI use was associated with ID [odds ratio (OR) 2.02; 95% CI 1.36–2.98]. Compared with no PPI use, the point estimate of the odds ratio for risk of ID for pantoprazole (OR 1.55; 95%CI 0.78–3.10) was lower than for esomeprazole and omeprazole (3.58; 95%CI 1.73–7.40 and 1.96; 95%CI 1.31–2.94, respectively). When comparing pantoprazole users with omeprazole users on an equipotent dose (≤20 omeprazole equivalents (OE)/day) omeprazole, but not pantoprazole was associated with ID, although the lack of a significant effect of pantoprazole on the risk of ID could be caused by a lack of power. Furthermore, risk of ID was higher among users of a high PPI dose (≥ 20 OE/day) and OE as continuous variable was also independently associated with ID, indicating that risk of ID is higher while using a more potent PPI. Further investigation seems warranted to confirm whether pantoprazole leads to less ID in KTRs.
Collapse
Affiliation(s)
- Rianne M Douwes
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Joanna Sophia J Vinke
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - António W Gomes-Neto
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gizem Ayerdem
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gaston van Hassel
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Condeles AL, Toledo Junior JC. The Labile Iron Pool Reacts Rapidly and Catalytically with Peroxynitrite. Biomolecules 2021; 11:1331. [PMID: 34572543 PMCID: PMC8466499 DOI: 10.3390/biom11091331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/23/2022] Open
Abstract
While investigating peroxynitrite-dependent oxidation in murine RAW 264.7 macrophage cells, we observed that removal of the Labile Iron Pool (LIP) by chelation increases the intracellular oxidation of the fluorescent indicator H2DCF, so we concluded that the LIP reacts with peroxynitrite and decreases the yield of peroxynitrite-derived oxidants. This was a paradigm-shifting finding in LIP biochemistry and raised many questions. In this follow-up study, we address fundamental properties of the interaction between the LIP and peroxynitrite by using the same cellular model and fluorescence methodology. We have identified that the reaction between the LIP and peroxynitrite has catalytic characteristics, and we have estimated that the rate constant of the reaction is in the range of 106 to 107 M-1s-1. Together, these observations suggest that the LIP represents a constitutive peroxynitrite reductase system in RAW 264.7 cells.
Collapse
Affiliation(s)
| | - José Carlos Toledo Junior
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-901, Brazil;
| |
Collapse
|
46
|
Dietz JV, Fox JL, Khalimonchuk O. Down the Iron Path: Mitochondrial Iron Homeostasis and Beyond. Cells 2021; 10:cells10092198. [PMID: 34571846 PMCID: PMC8468894 DOI: 10.3390/cells10092198] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022] Open
Abstract
Cellular iron homeostasis and mitochondrial iron homeostasis are interdependent. Mitochondria must import iron to form iron–sulfur clusters and heme, and to incorporate these cofactors along with iron ions into mitochondrial proteins that support essential functions, including cellular respiration. In turn, mitochondria supply the cell with heme and enable the biogenesis of cytosolic and nuclear proteins containing iron–sulfur clusters. Impairment in cellular or mitochondrial iron homeostasis is deleterious and can result in numerous human diseases. Due to its reactivity, iron is stored and trafficked through the body, intracellularly, and within mitochondria via carefully orchestrated processes. Here, we focus on describing the processes of and components involved in mitochondrial iron trafficking and storage, as well as mitochondrial iron–sulfur cluster biogenesis and heme biosynthesis. Recent findings and the most pressing topics for future research are highlighted.
Collapse
Affiliation(s)
- Jonathan V. Dietz
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA;
| | - Jennifer L. Fox
- Department of Chemistry and Biochemistry, College of Charleston, Charleston, SC 29424, USA;
| | - Oleh Khalimonchuk
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA;
- Nebraska Redox Biology Center, University of Nebraska, Lincoln, NE 68588, USA
- Fred and Pamela Buffett Cancer Center, Omaha, NE 68198, USA
- Correspondence:
| |
Collapse
|
47
|
Bhukta S, Gopinath P, Dandela R. Target identification of anticancer natural products using a chemical proteomics approach. RSC Adv 2021; 11:27950-27964. [PMID: 35480761 PMCID: PMC9038044 DOI: 10.1039/d1ra04283a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
In recent years, there has been a strong demand worldwide for the identification and development of potential anticancer drugs based on natural products. Natural products have been explored for their diverse biological and therapeutic applications from ancient time. In order to enhance the efficacy and selectivity and to minimize the undesired side effects of anti cancer natural products (ANPs), it is essential to understand their target proteins and their mechanistic pathway. Chemical proteomics is one of the most powerful tools to connect ANP target identification and quantification where labeling and non-labeling based approaches have been used. Herein, we have discussed the various strategies to systemically develop selective ANP based chemical probes to characterise their specific and non-specific target proteins using a chemical proteomic approach in various cancer cell lysates.
Collapse
Affiliation(s)
- Swadhapriya Bhukta
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology Indianoil Odisha Campus, Samantpuri Bhubaneswar 751013 India
| | - Pushparathinam Gopinath
- Department of Chemistry, SRM-Institute of Science and Technology Kattankulathur 603203 Chennai Tamilnadu India
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology Indianoil Odisha Campus, Samantpuri Bhubaneswar 751013 India
| |
Collapse
|
48
|
Gu Z, Liu T, Liu C, Yang Y, Tang J, Song H, Wang Y, Yang Y, Yu C. Ferroptosis-Strengthened Metabolic and Inflammatory Regulation of Tumor-Associated Macrophages Provokes Potent Tumoricidal Activities. NANO LETTERS 2021; 21:6471-6479. [PMID: 34292757 DOI: 10.1021/acs.nanolett.1c01401] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Modulation of tumor-associated macrophages (TAMs) holds promise for cancer treatment, mainly relying on M1 signaling activation and pro-inflammatory promotion. Nevertheless, the antitumor activity is often limited by the anti-inflammatory factors in the tumor microenvironment. Moreover, the metabolic function of TAMs is also critical to tumor progression. However, there are a few strategies that can simultaneously regulate both inflammatory and metabolic functions to achieve safe and potent antitumor activation of TAMs. Herein, we demonstrate that an iron-based metal organic framework nanoparticle and a ferroptosis-inducing agent synergistically induce mitochondrial alternation in TAMs, resulting in a radical metabolic switch from mitochondrial oxidative phosphorylation to glycolysis, which is resistant to anti-inflammatory stimuli challenge. The ferroptosis stress strengthened by the nanoformulation also drives multiple pro-inflammatory signaling pathways, enabling macrophage activation with potent tumoricidal activities. The ferroptosis-strengthened macrophage regulation strategy present in this study paves the way for TAM-centered antitumoral treatment to overcome the limitations of conventional methods.
Collapse
Affiliation(s)
- Zhengying Gu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales 2145, Australia
| | - Chao Liu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Chengzhong Yu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, People's Republic of China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
49
|
Li YQ, Guo C. A Review on Lactoferrin and Central Nervous System Diseases. Cells 2021; 10:cells10071810. [PMID: 34359979 PMCID: PMC8307123 DOI: 10.3390/cells10071810] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Central nervous system (CNS) diseases are currently one of the major health issues around the world. Most CNS disorders are characterized by high oxidative stress levels and intense inflammatory responses in affected tissues. Lactoferrin (Lf), a multifunctional iron-binding glycoprotein, plays a significant role in anti-inflammatory, antibacterial, antiviral, reactive oxygen species (ROS) modulator, antitumor immunity, and anti-apoptotic processes. Previous studies have shown that Lf is abnormally expressed in a variety of neurological diseases, especially neurodegenerative diseases. Recently, the promotion of neurodevelopment and neuroprotection by Lf has attracted widespread attention, and Lf could be exploited both as an active therapeutic agent and drug nanocarrier. However, our understanding of the roles of Lf proteins in the initiation or progression of CNS diseases is limited, especially the roles of Lf in regulating neurogenesis. This review highlights recent advances in the understanding of the major pharmacological effects of Lf in CNS diseases, including neurodegenerative diseases, cerebrovascular disease, developmental delays in children, and brain tumors.
Collapse
Affiliation(s)
| | - Chuang Guo
- Correspondence: ; Tel.: +86-24-8365-6109
| |
Collapse
|
50
|
Jiang S, Guo T, Guo S, Gao J, Ni Y, Ma W, Zhao R. Chronic Variable Stress Induces Hepatic Fe(II) Deposition by Up-Regulating ZIP14 Expression via miR-181 Family Pathway in Rats. BIOLOGY 2021; 10:biology10070653. [PMID: 34356508 PMCID: PMC8301360 DOI: 10.3390/biology10070653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/25/2021] [Accepted: 07/09/2021] [Indexed: 12/17/2022]
Abstract
Simple Summary Modern intensive production methods attract accusations of poor animal welfare due to long-term exposure to stressors including high temperature, persistent humidity and overcrowding. Stress can be defined as any condition that threatens the physiological homoeostasis and hypothalamic-pituitary-adrenal (HPA) axis responses that tend to restore the prior stable status of the organism. Uncontrollable and unpredictable sources of stress can cause various forms of damage to the liver, which is the central mediator of systemic iron balance. Iron, notably, is an essential element for maintaining health in virtually all organisms. We found that chronic variable stress can cause weight loss and disorders of the liver iron metabolism in rats, thereby triggering liver oxidative damage. Our results also suggest that the miR-181 family is a potential target for treating iron overload-associated diseases. Abstract It is well-known that hepatic iron dysregulation, which is harmful to health, can be caused by stress. The aim of the study was to evaluate chronic variable stress (CVS) on liver damage, hepatic ferrous iron deposition and its molecular regulatory mechanism in rats. Sprague Dawley rats at seven weeks of age were randomly divided into two groups: a control group (Con) and a CVS group. CVS reduces body weight, but increases the liver-to-body weight ratio. The exposure of rats to CVS increased plasma aspartate aminotransferase (AST), alkaline phosphatase (ALP) and hepatic malondialdehyde (MDA) levels, but decreased glutathione peroxidase (GSH-Px) activity, resulting in liver damage. CVS lowered the total amount of hepatic iron content, but induced hepatic Fe(II) accumulation. CVS up-regulated the expression of transferrin receptor 1 (TFR1) and ZRT/IRT-like protein 14 (ZIP14), but down-regulated ferritin and miR-181 family members. In addition, miR-181 family expression was found to regulate ZIP14 expression in HEK-293T cells by the dual-luciferase reporter system. These results indicate that CVS results in liver damage and induces hepatic Fe(II) accumulation, which is closely associated with the up-regulation of ZIP14 expression via the miR-181 family pathway.
Collapse
Affiliation(s)
- Shuxia Jiang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Taining Guo
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Shihui Guo
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiang Gao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| | - Wenqiang Ma
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: ; Tel.: +86-25-8439-6413; Fax: +86-25-8439-8669
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (S.J.); (T.G.); (S.G.); (J.G.); (Y.N.); (R.Z.)
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|