ijms-logo

Journal Browser

Journal Browser

Iron Metabolism in Health and Disease

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Biochemistry".

Deadline for manuscript submissions: closed (15 April 2023) | Viewed by 20754

Special Issue Editors


E-Mail Website
Guest Editor
1. Department of Molecular Medicine and Medical Biotechnologies, University Federico II of Naples, 80136 Napoli, Italy
2. CEINGE, Biotecnologie Avanzate, 80145 Napoli, Italy
Interests: red blood cell membrane defects; iron metabolism; molecular genetics; hereditary anemias; cell signaling; pathophysiologic mechanisms of hereditary anemias; dehydrated hereditary stomatocytosis; PIEZO1

E-Mail Website
Guest Editor
1. Department of Molecular Medicine and Medical Biotechnologies, University Federico II of Naples, 80136 Napoli, Italy
2. CEINGE, Biotecnologie Avanzate, 80145 Napoli, Italy
Interests: congenital dyserythropoietic anemias; erythropoiesis; abnormal erythropoiesis; hereditary anemias; molecular genetics; genomics of hereditary anemias
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Iron is biologically essential, but also potentially toxic; as such, it is tightly controlled at cell and systemic levels to prevent both deficiency and overload. Iron regulatory proteins post-transcriptionally control genes encoding proteins that modulate iron uptake, recycling, and storage and are themselves regulated by iron. The master regulator of systemic iron homeostasis is the liver peptide hepcidin, which controls serum iron through the degradation of ferroportin in iron-absorptive enterocytes and iron-recycling macrophages. Research in this field has made great strides in recent years and clarified the role of iron in physiology and diseases. The disorders associated with alterations in iron metabolism span from iron overload to iron deficiency. The studies of genetic and acquired iron disorders have identified novel iron genes, proteins, and pathways and revealed the essential role of the hepcidin–ferroportin axis in systemic iron homeostasis.

The aim of this issue is to obtain an overview of the regulation of iron metabolism at physiological levels and in the diseases associated with iron metabolism alterations.

Dr. Immacolata Andolfo
Dr. Roberta Russo
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • physiology of iron metabolism
  • iron overload
  • iron deficiency
  • mechanisms of alteration of iron metabolism

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

20 pages, 3194 KiB  
Article
Pro-Inflammatory Priming of the Brain: The Underlying Cause of Parkinson’s Disease
by Ana Catarina Martins, Illyane Sofia Lima, Ana Catarina Pêgo, Inês Sá Pereira, Gracelino Martins, Antonino Kapitão and Raffaella Gozzelino
Int. J. Mol. Sci. 2023, 24(9), 7949; https://doi.org/10.3390/ijms24097949 - 27 Apr 2023
Cited by 2 | Viewed by 1702
Abstract
Parkinson’s disease (PD) is a multifactorial neurodegenerative pathology characterized by the progressive loss of dopaminergic neurons in the substantia nigra of the brain. Aging is considered the main risk factor for the development of idiopathic PD. However, immunity and inflammation play a crucial [...] Read more.
Parkinson’s disease (PD) is a multifactorial neurodegenerative pathology characterized by the progressive loss of dopaminergic neurons in the substantia nigra of the brain. Aging is considered the main risk factor for the development of idiopathic PD. However, immunity and inflammation play a crucial role in the pathogenesis of this disorder. In mice, we showed that pro-inflammatory priming of the brain sensitizes to severe PD development, regardless of animal age. Age-related sub-acute inflammation, as well as the activation of the immune response upon exposure to harmful stimuli, enhances PD manifestations. The severity of PD is influenced by the engagement of host resistance mechanisms against infection based on the removal of iron (Fe) from the circulation. The sequestration of Fe by immune cells prevents pathogens from proliferating. However, it leads to the formation of a Fe-loaded circulating compartment. When entering the brain through a compromised blood-brain barrier, Fe-loaded immune cells contribute to enhancing neuroinflammation and brain Fe overload. Thus, pro-inflammatory priming of the brain exacerbates neuronal damage and represents a risk factor for the development of severe PD symptoms. Further investigations are now required to better understand whether therapeutic interventions inhibiting this phenomenon might protect against PD. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

12 pages, 886 KiB  
Article
Differential Effects of Iron Chelates vs. Iron Salts on Induction of Pro-Oncogenic Amphiregulin and Pro-Inflammatory COX-2 in Human Intestinal Adenocarcinoma Cell Lines
by Agata Tarczykowska, Niklas Engström, Darja Dobermann, Jonathan Powell and Nathalie Scheers
Int. J. Mol. Sci. 2023, 24(6), 5507; https://doi.org/10.3390/ijms24065507 - 14 Mar 2023
Cited by 1 | Viewed by 1744
Abstract
We previously showed that two iron compounds that are orally ingested by humans, namely ferric EDTA and ferric citrate, can induce an oncogenic growth factor (amphiregulin) in human intestinal epithelial adenocarcinoma cell lines. Here, we further screened these iron compounds, plus four other [...] Read more.
We previously showed that two iron compounds that are orally ingested by humans, namely ferric EDTA and ferric citrate, can induce an oncogenic growth factor (amphiregulin) in human intestinal epithelial adenocarcinoma cell lines. Here, we further screened these iron compounds, plus four other iron chelates and six iron salts (i.e., 12 oral iron compounds in total), for their effects on biomarkers of cancer and inflammation. Ferric pyrophosphate and ferric EDTA were the main inducers of amphiregulin and its receptor monomer, IGFr1. Moreover, at the maximum iron concentrations investigated (500 µM), the highest levels of amphiregulin were induced by the six iron chelates, while four of these also increased IGfr1. In addition, we observed that ferric pyrophosphate promoted signaling via the JAK/STAT pathway by up-regulating the cytokine receptor subunit IFN-γr1 and IL-6. For pro-inflammatory cyclooxygenase-2 (COX-2), ferric pyrophosphate but not ferric EDTA elevated intracellular levels. This, however, did not drive the other biomarkers based on COX-2 inhibition studies and was probably downstream of IL-6. We conclude that of all oral iron compounds, iron chelates may particularly elevate intracellular amphiregulin. Ferric pyrophosphate additionally induced COX-2, probably because of the high IL-6 induction that was observed with this compound. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

14 pages, 1742 KiB  
Article
Dysregulation of Iron Metabolism-Linked Genes at Myocardial Tissue and Cell Levels in Dilated Cardiomyopathy
by Ilaria Massaiu, Jeness Campodonico, Massimo Mapelli, Elisabetta Salvioni, Vincenza Valerio, Donato Moschetta, Veronika A. Myasoedova, Maria Domenica Cappellini, Giulio Pompilio, Paolo Poggio and Piergiuseppe Agostoni
Int. J. Mol. Sci. 2023, 24(3), 2887; https://doi.org/10.3390/ijms24032887 - 2 Feb 2023
Cited by 5 | Viewed by 2571
Abstract
In heart failure, the biological and clinical connection between abnormal iron homeostasis, myocardial function, and prognosis is known; however, the expression profiles of iron-linked genes both at myocardial tissue and single-cell level are not well defined. Through publicly available bulk and single-nucleus RNA [...] Read more.
In heart failure, the biological and clinical connection between abnormal iron homeostasis, myocardial function, and prognosis is known; however, the expression profiles of iron-linked genes both at myocardial tissue and single-cell level are not well defined. Through publicly available bulk and single-nucleus RNA sequencing (RNA-seq) datasets of left ventricle samples from adult non-failed (NF) and dilated cardiomyopathy (DCM) subjects, we aim to evaluate the altered iron metabolism in a diseased condition, at the whole cardiac tissue and single-cell level. From the bulk RNA-seq data, we found 223 iron-linked genes expressed at the myocardial tissue level and 44 differentially expressed between DCM and NF subjects. At the single-cell level, at least 18 iron-linked expressed genes were significantly regulated in DCM when compared to NF subjects. Specifically, the iron metabolism in DCM cardiomyocytes is altered at several levels, including: (1) imbalance of Fe3+ internalization (SCARA5 down-regulation) and reduction of internal conversion from Fe3+ to Fe2+ (STEAP3 down-regulation), (2) increase of iron consumption to produce hemoglobin (HBA1/2 up-regulation), (3) higher heme synthesis and externalization (ALAS2 and ABCG2 up-regulation), (4) lower cleavage of heme to Fe2+, biliverdin and carbon monoxide (HMOX2 down-regulation), and (5) positive regulation of hepcidin (BMP6 up-regulation). Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

12 pages, 2287 KiB  
Article
Receptor-Independent Anti-Ferroptotic Activity of TrkB Modulators
by Md. Jakaria, Abdel A. Belaidi, Adam Southon, Krista A. Dent, Darius J. R. Lane, Ashley I. Bush and Scott Ayton
Int. J. Mol. Sci. 2022, 23(24), 16205; https://doi.org/10.3390/ijms232416205 - 19 Dec 2022
Cited by 2 | Viewed by 1881
Abstract
Dysregulated brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signalling is implicated in several neurodegenerative diseases, including Alzheimer’s disease. A failure of neurotrophic support may participate in neurodegenerative mechanisms, such as ferroptosis, which has likewise been implicated in this disease class. The current [...] Read more.
Dysregulated brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signalling is implicated in several neurodegenerative diseases, including Alzheimer’s disease. A failure of neurotrophic support may participate in neurodegenerative mechanisms, such as ferroptosis, which has likewise been implicated in this disease class. The current study investigated whether modulators of TrkB signalling affect ferroptosis. Cell viability, C11 BODIPY, and cell-free oxidation assays were used to observe the impact of TrkB modulators, and an immunoblot assay was used to detect TrkB expression. TrkB modulators such as agonist BDNF, antagonist ANA-12, and inhibitor K252a did not affect RSL3-induced ferroptosis sensitivity in primary cortical neurons expressing detectable TrkB receptors. Several other modulators of the TrkB receptor, including agonist 7,8-DHF, activator phenelzine sulphate, and inhibitor GNF-5837, conferred protection against a range of ferroptosis inducers in several immortalised neuronal and non-neuronal cell lines, such as N27 and HT-1080 cells. We found these immortalised cell lines lack detectable TrkB receptor expression, so the anti-ferroptotic activity of these TrkB modulators was most likely due to their inherent radical-trapping antioxidant properties, which should be considered when interpreting their experimental findings. These modulators or their variants could be potential anti-ferroptotic therapeutics for various diseases. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

18 pages, 2866 KiB  
Article
Neuroprotective Effects of Transferrin in Experimental Glaucoma Models
by Jenny Youale, Karine Bigot, Bindu Kodati, Thara Jaworski, Yan Fan, Nana Yaa Nsiah, Nathaniel Pappenhagen, Denise M. Inman, Francine Behar-Cohen, Thierry Bordet and Emilie Picard
Int. J. Mol. Sci. 2022, 23(21), 12753; https://doi.org/10.3390/ijms232112753 - 22 Oct 2022
Cited by 8 | Viewed by 2630
Abstract
Iron is essential for retinal metabolism, but an excess of ferrous iron causes oxidative stress. In glaucomatous eyes, retinal ganglion cell (RGC) death has been associated with dysregulation of iron homeostasis. Transferrin (TF) is an endogenous iron transporter that controls ocular iron levels. [...] Read more.
Iron is essential for retinal metabolism, but an excess of ferrous iron causes oxidative stress. In glaucomatous eyes, retinal ganglion cell (RGC) death has been associated with dysregulation of iron homeostasis. Transferrin (TF) is an endogenous iron transporter that controls ocular iron levels. Intraocular administration of TF is neuroprotective in various models of retinal degeneration, preventing iron overload and reducing iron-induced oxidative stress. Herein, we assessed the protective effects of TF on RGC survival, using ex vivo rat retinal explants exposed to iron, NMDA-induced excitotoxicity, or CoCl2-induced hypoxia, and an in vivo rat model of ocular hypertension (OHT). TF significantly preserved RGCs against FeSO4-induced toxicity, NMDA-induced excitotoxicity, and CoCl2-induced hypoxia. TF protected RGCs from apoptosis, ferroptosis, and necrosis. In OHT rats, TF reduced RGC loss by about 70% compared to vehicle-treated animals and preserved about 47% of the axons. Finally, increased iron staining was shown in the retina of a glaucoma patient’s eye as compared to non-glaucomatous eyes. These results indicate that TF can interfere with different cell-death mechanisms involved in glaucoma pathogenesis and demonstrate the ability of TF to protect RGCs exposed to elevated IOP. Altogether, these results suggest that TF is a promising treatment against glaucoma neuropathy. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

7 pages, 1529 KiB  
Communication
Human IRP1 Translocates to the Nucleus in a Cell-Specific and Iron-Dependent Manner
by Wen Gu, Carine Fillebeen and Kostas Pantopoulos
Int. J. Mol. Sci. 2022, 23(18), 10740; https://doi.org/10.3390/ijms231810740 - 15 Sep 2022
Cited by 1 | Viewed by 1799
Abstract
Iron regulatory protein 1 (IRP1) is a bifunctional protein with mutually exclusive RNA-binding or enzymatic activities that depend on the presence of a 4Fe-4S cluster. While IRP1 is a well-established cytosolic protein, work in a Drosophila model suggested that it may also exhibit [...] Read more.
Iron regulatory protein 1 (IRP1) is a bifunctional protein with mutually exclusive RNA-binding or enzymatic activities that depend on the presence of a 4Fe-4S cluster. While IRP1 is a well-established cytosolic protein, work in a Drosophila model suggested that it may also exhibit nuclear localization. Herein, we addressed whether mammalian IRP1 can likewise translocate to the nucleus. We utilized primary cells and tissues from wild type and Irp1−/− mice, as well as human cell lines and tissue biopsy sections. IRP1 subcellular localization was analyzed by Western blotting, immunofluorescence and immunohistochemistry. We did not detect presence of nuclear IRP1 in wild type mouse embryonic fibroblasts (MEFs), primary hepatocytes or whole mouse liver. However, we observed IRP1-positive nuclei in human liver but not ovary sections. Biochemical fractionation studies revealed presence of IRP1 in the nucleus of human Huh7 and HepG2 hepatoma cells, but not HeLa cervical cancer cells. Importantly, nuclear IRP1 was only evident in iron-replete cells and disappeared following pharmacological iron chelation. These data provide the first experimental evidence for nuclear IRP1 expression in mammals, which appears to be species- and cell-specific. Furthermore, they suggest that the nuclear translocation of IRP1 is mediated by an iron-dependent mechanism. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

16 pages, 1932 KiB  
Article
Mutant WDR45 Leads to Altered Ferritinophagy and Ferroptosis in β-Propeller Protein-Associated Neurodegeneration
by Sokhna Haissatou Diaw, Christos Ganos, Simone Zittel, Kirstin Plötze-Martin, Leonora Kulikovskaja, Melissa Vos, Ana Westenberger, Aleksandar Rakovic, Katja Lohmann and Marija Dulovic-Mahlow
Int. J. Mol. Sci. 2022, 23(17), 9524; https://doi.org/10.3390/ijms23179524 - 23 Aug 2022
Cited by 12 | Viewed by 2788
Abstract
Beta-propeller protein-associated neurodegeneration (BPAN) is a subtype of neurodegeneration with brain iron accumulation (NBIA) caused by loss-of-function variants in WDR45. The underlying mechanism of iron accumulation in WDR45 deficiency remains elusive. We established a primary skin fibroblast culture of a new BPAN patient [...] Read more.
Beta-propeller protein-associated neurodegeneration (BPAN) is a subtype of neurodegeneration with brain iron accumulation (NBIA) caused by loss-of-function variants in WDR45. The underlying mechanism of iron accumulation in WDR45 deficiency remains elusive. We established a primary skin fibroblast culture of a new BPAN patient with a missense variant p.(Asn61Lys) in WDR45 (NM_007075.3: c.183C>A). The female patient has generalized dystonia, anarthria, parkinsonism, spasticity, stereotypies, and a distinctive cranial MRI with generalized brain atrophy, predominantly of the cerebellum. For the functional characterization of this variant and to provide a molecular link of WDR45 and iron accumulation, we looked for disease- and variant-related changes in the patient’s fibroblasts by qPCR, immunoblotting and immunofluorescence comparing to three controls and a previously reported WDR45 patient. We demonstrated molecular changes in mutant cells comprising an impaired mitochondrial network, decreased levels of lysosomal proteins and enzymes, and altered autophagy, confirming the pathogenicity of the variant. Compared to increased levels of the ferritinophagy marker Nuclear Coactivator 4 (NCOA4) in control cells upon iron treatment, patients’ cells revealed unchanged NCOA4 protein levels, indicating disturbed ferritinophagy. Additionally, we observed abnormal protein levels of markers of the iron-dependent cell death ferroptosis in patients’ cells. Altogether, our data suggests that WDR45 deficiency affects ferritinophagy and ferroptosis, consequentially disturbing iron recycling. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

Review

Jump to: Research

18 pages, 9065 KiB  
Review
Hypoxia-Inducible Factor Prolyl Hydroxylase Inhibitors and Iron Metabolism
by Chie Ogawa, Ken Tsuchiya and Kunimi Maeda
Int. J. Mol. Sci. 2023, 24(3), 3037; https://doi.org/10.3390/ijms24033037 - 3 Feb 2023
Cited by 11 | Viewed by 4334
Abstract
The production of erythropoietin (EPO), the main regulator of erythroid differentiation, is regulated by hypoxia-inducible factor (HIF). HIF2α seems to be the principal regulator of EPO transcription, but HIF1α and 3α also may have additional influences on erythroid maturation. HIF is also involved [...] Read more.
The production of erythropoietin (EPO), the main regulator of erythroid differentiation, is regulated by hypoxia-inducible factor (HIF). HIF2α seems to be the principal regulator of EPO transcription, but HIF1α and 3α also may have additional influences on erythroid maturation. HIF is also involved in the regulation of iron, an essential component in erythropoiesis. Iron is essential for the organism but is also highly toxic, so its absorption and retention are strictly controlled. HIF also induces the synthesis of proteins involved in iron regulation, thereby ensuring the availability of iron necessary for hematopoiesis. Iron is a major component of hemoglobin and is also involved in erythrocyte differentiation and proliferation and in the regulation of HIF. Renal anemia is a condition in which there is a lack of stimulation of EPO synthesis due to decreased HIF expression. HIF prolyl hydroxylase inhibitors (HIF-PHIs) stabilize HIF and thereby allow it to be potent under normoxic conditions. Therefore, unlike erythropoiesis-stimulating agents, HIF-PHI may enhance iron absorption from the intestinal tract and iron supply from reticuloendothelial macrophages and hepatocytes into the plasma, thus facilitating the availability of iron for hematopoiesis. The only HIF-PHI currently on the market worldwide is roxadustat, but in Japan, five products are available. Clinical studies to date in Japan have also shown that HIF-PHIs not only promote hematopoiesis, but also decrease hepcidin, the main regulator of iron metabolism, and increase the total iron-binding capacity (TIBC), which indicates the iron transport capacity. However, concerns about the systemic effects of HIF-PHIs have not been completely dispelled, warranting further careful monitoring. Full article
(This article belongs to the Special Issue Iron Metabolism in Health and Disease)
Show Figures

Figure 1

Back to TopTop