1
|
Molenaars M, Mir H, Alvarez SW, Arivazhagan L, Rosselot C, Zhan D, Park CY, Garcia-Ocana A, Schmidt AM, Possemato R. Acute inhibition of iron-sulfur cluster biosynthesis disrupts metabolic flexibility in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608291. [PMID: 39229169 PMCID: PMC11370322 DOI: 10.1101/2024.08.19.608291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Iron-sulfur clusters (ISCs) are cell-essential cofactors present in ∼60 proteins including subunits of OXPHOS complexes I-III, DNA polymerases, and iron-sensing proteins. Dysfunctions in ISC biosynthesis are associated with anemias, neurodegenerative disorders, and metabolic diseases. To assess consequences of acute ISC inhibition in a whole body setting, we developed a mouse model in which key ISC biosynthetic enzyme NFS1 can be acutely and reversibly suppressed. Contrary to in vitro ISC inhibition and pharmacological OXPHOS suppression, global NFS1 inhibition rapidly enhances lipid utilization and decreases adiposity without affecting caloric intake and physical activity. ISC proteins decrease, including key proteins involved in OXPHOS (SDHB), lipoic acid synthesis (LIAS), and insulin mRNA processing (CDKAL1), causing acute metabolic inflexibility. Age-related metabolic changes decelerate loss of adiposity substantially prolonged survival of mice with NFS1 inhibition. Thus, the observation that ISC metabolism impacts organismal fuel choice will aid in understanding the mechanisms underlying ISC diseases with increased risk for diabetes. Graphical abstract Highlights Acute ISC inhibition leads to rapid loss of adiposity in miceMulti-metabolic pathway disruption upon ISC deficiency blocks energy storageNfs1 inhibition induces glucose dyshomeostasis due to ISC deficiency in β-cellsEnergy distress caused by inhibition of ISC synthesis is attenuated in aged mice.
Collapse
|
2
|
Terzi EM, Possemato R. Iron, Copper, and Selenium: Cancer's Thing for Redox Bling. Cold Spring Harb Perspect Med 2024; 14:a041545. [PMID: 37932129 PMCID: PMC10982729 DOI: 10.1101/cshperspect.a041545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Cells require micronutrients for numerous basic functions. Among these, iron, copper, and selenium are particularly critical for redox metabolism, and their importance is heightened during oncogene-driven perturbations in cancer. In this review, which particularly focuses on iron, we describe how these micronutrients are carefully chaperoned about the body and made available to tissues, a process that is designed to limit the toxicity of free iron and copper or by-products of selenium metabolism. We delineate perturbations in iron metabolism and iron-dependent proteins that are observed in cancer, and describe the current approaches being used to target iron metabolism and iron-dependent processes.
Collapse
Affiliation(s)
- Erdem M Terzi
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York, New York 10016, USA
| | - Richard Possemato
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York, New York 10016, USA
| |
Collapse
|
3
|
Cao C, Cai Y, Li Y, Li T, Zhang J, Hu Z, Zhang J. Characterization and comparative transcriptomic analysis of skeletal muscle in female Pekin duck and Hanzhong Ma duck during different growth stages using RNA-seq. Poult Sci 2023; 102:103122. [PMID: 37832186 PMCID: PMC10568565 DOI: 10.1016/j.psj.2023.103122] [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: 06/27/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023] Open
Abstract
Duck is an economically important poultry, and there is currently a major focus on improving its meat quality through breeding. There are wide variations in the growth regulation mechanisms of different duck breeds, that fundamental research on skeletal muscle growth is essential for understanding the regulation of unknown genes. The study aimed to broaden the understanding the duck skeletal muscle development and thereby to improve the performance of domestic ducks. In this study, RNA-seq data from skeletal muscles (breast muscle and leg muscle) of Pekin duck and Hanzhong Ma duck sampled at d 17, 21, and 27 of embryo (E17d, E21d, and E27d), as well as at 6-mo-old following birth (M6), to investigate and compare the mRNA temporal expression profiles and associated pathways that regulate skeletal myogenesis of different duck breeds. There were 331 to 1,440 annotated differentially expressed genes (DEGs) in breast muscle and 380 to 1,790 annotated DEGs in leg muscle from different databases between 2 duck breeds. Gene ontology (GO) enrichment in skeletal muscles indicated that these DEGs were mainly involved in biosynthetic process, developmental process, regulation of protein metabolic process and regulation of gene expression. KEGG analysis in skeletal muscles showed that a total of 41 DEGs were mapped to 7 KEGG pathways, including ECM-receptor interaction, focal adhesion, carbon metabolism, regulation of actin cytoskeleton, calcium signaling pathway, biosynthesis of amino acids and PPAR signaling pathway. The differential expression of 8 selected DEGs was verified by qRT-PCR, and the results were consistent with RNA-seq data. The identified DEGs, such as SDC, SPP1, PAK1, MYL9, PGK1, NOS1, PHGDH, TNNT2, FN1, and AQP4, were specially highlighted, indicating their associations with muscle development in the Pekin duck and Hanzhong Ma duck. This study provides a basis for revealing the differences in skeletal muscle development between Pekin duck and Hanzhong Ma duck.
Collapse
Affiliation(s)
- Chang Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Yingjie Cai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Yuxiao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Tao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Jiqiao Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Zhigang Hu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China
| | - Jianqin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, PR China.
| |
Collapse
|
4
|
Asgari A, Jurasz P. Role of Nitric Oxide in Megakaryocyte Function. Int J Mol Sci 2023; 24:ijms24098145. [PMID: 37175857 PMCID: PMC10179655 DOI: 10.3390/ijms24098145] [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: 02/07/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Megakaryocytes are the main members of the hematopoietic system responsible for regulating vascular homeostasis through their progeny platelets, which are generally known for maintaining hemostasis. Megakaryocytes are characterized as large polyploid cells that reside in the bone marrow but may also circulate in the vasculature. They are generated directly or through a multi-lineage commitment step from the most primitive progenitor or Hematopoietic Stem Cells (HSCs) in a process called "megakaryopoiesis". Immature megakaryocytes enter a complicated development process defined as "thrombopoiesis" that ultimately results in the release of extended protrusions called proplatelets into bone marrow sinusoidal or lung microvessels. One of the main mediators that play an important modulatory role in hematopoiesis and hemostasis is nitric oxide (NO), a free radical gas produced by three isoforms of nitric oxide synthase within the mammalian cells. In this review, we summarize the effect of NO and its signaling on megakaryopoiesis and thrombopoiesis under both physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Amir Asgari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G-2E1, Canada
- Department of Pharmacology, University of Alberta, Edmonton, AB T6G-2H7, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB T6G-2S2, Canada
- Mazankowski Alberta Heart Institute, Edmonton, AB T6G-2R7, Canada
| |
Collapse
|
5
|
Yakout SM, Abdi S, Alaskar AH, Khattak MNK, Al-Masri AA, Al-Daghri NM. Impact of Vitamin D Status Correction on Serum Lipid Profile, Carboxypeptidase N and Nitric Oxide Levels in Saudi Adults. Int J Mol Sci 2023; 24:ijms24097711. [PMID: 37175418 PMCID: PMC10177893 DOI: 10.3390/ijms24097711] [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: 03/08/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
This study aimed to determine the impact on the lipid profile, carboxypeptidase N (CPN) and nitric oxide (NOx) associated with vitamin D (VD) status correction among Saudi adults with VD deficiency. A total 111 VD deficient (25(OH)D < 50 nmol/L)) adult Saudis aged 18-50 years old (57 females and 54 males) were enrolled in this 6-month interventional study. They were given 50,000 IU VD weekly for the first 2 months and then twice a month for the next 2 months, followed by 1000 IU daily for the last 2 months. The fasting lipid profile and the blood glucose, VD, NOx and CPN concentrations were measured at baseline and after intervention. Post-supplementation, the median VD was significantly higher (p < 0.001) in females [58.3 (50.6-71.2)] and males [57.8 (51.0-71.8)]. HDL cholesterol significantly increased (p = 0.05) and NOx significantly decreased (p = 0.02) in males post-supplementation. Triglycerides were positively associated with NOx in all subjects before (r = 0.44, p = 0.01) and after (r = 0.37, p = 0.01) VD status correction. There was a significant increase in serum levels of CPN2 (p = 0.02) in all subjects. Furthermore, CPN was inversely correlated with NOx (r = -0.35, p = 0.05) in males post-supplementation. In conclusion, VD status correction reduced serum NOx, particularly in males. The inhibition of NOx synthesis may be responsible for the anti-inflammatory effects of VD supplementation. An inverse association was found between NOx and CPN2.
Collapse
Affiliation(s)
- Sobhy M Yakout
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saba Abdi
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alhanouf H Alaskar
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Malak Nawaz Khan Khattak
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abeer A Al-Masri
- Department of Physiology, College Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nasser M Al-Daghri
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
6
|
Amer OE, Sabico S, Khattak MNK, Al-Daghri NM. Circulating Nitric Oxide and Metabolic Syndrome in Arab Children and Adolescents: A Case-Control Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020210. [PMID: 36832339 PMCID: PMC9954847 DOI: 10.3390/children10020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/12/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Metabolic syndrome (MetS) represents a cluster of known cardiometabolic risk factors, which elevates the risk of type 2 diabetes mellitus (T2DM), atherosclerotic cardiovascular disease (CVD) and chronic kidney disease (CKD) in adults and, only recently, even in children and adolescents. Circulating nitric oxide (NOx) has been observed to influence MetS risk factors in adults, but this has been scarcely investigated in children. The aim of the present study was to determine whether circulating NOx levels correlate with known components of MetS in Arab children and adolescents. METHODS Anthropometrics, serum NOx, lipid profile and fasting glucose levels were measured in 740 Saudi Arabs aged 10-17 years (68.8% girls). The presence of MetS was screened using the criteria of de Ferranti et al. Results: Overall, serum NOx levels were significantly higher in MetS participants compared to non-MetS (25.7 µmol/L (10.1-46.7) versus 11.9 µmol/L (5.5-22.9), p < 0.001) even after adjustments for age, BMI and sex. With the exception of elevated blood pressure, higher circulating NOx significantly increased the odds for MetS and its components. Lastly, receiver operating characteristics (ROC) showed that NOx, as a diagnostic marker for MetS, had good sensitivity and was higher in boys than girls (all MetS participants: area under the curve (AUC) = 0.68, p < 0.001), (girls with MetS: AUC = 0.62, p = 0.002), (boys with MetS: AUC = 0.83, p < 0.001)). CONCLUSIONS MetS and most of its components were significantly associated with circulating NOx levels in Arab adolescents and may be a promising diagnostic biomarker for MetS.
Collapse
|
7
|
Ranjana M, Sunil D. Naphthalimide derivatives as fluorescent probes for imaging endogenous gasotransmitters. Chem Biol Interact 2022; 363:110022. [PMID: 35753358 DOI: 10.1016/j.cbi.2022.110022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/07/2022] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Gasotransmitters have gained significant recognition attributed to their evident biological impacts, and is accepted as a promising and less-explored area with immense research scope. The three-member family comprising of nitric oxide, carbon monoxide and hydrogen sulphide as endogenous gaseous signaling molecules have been found to elicit a plethora of crucial biological functions, spawning a new research area. The sensing of these small molecules is vital to gain deeper insights into their functions, as they can act both as a friend or a foe in mammalian systems. The initial sections of the review present the physiological and pathophysiological roles of these endogenous gas transmitters and their synergistic interactions. Further, various detection approaches, especially the usage of fascinating features of 1,8-naphthalimide as fluorescent probe in the detection and monitoring of these small signaling molecules are highlighted. The current limitations and the future scope of improving the sensing of the three gasotransmitters are also discussed.
Collapse
Affiliation(s)
- M Ranjana
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India.
| |
Collapse
|
8
|
Zeidan RS, Han SM, Leeuwenburgh C, Xiao R. Iron homeostasis and organismal aging. Ageing Res Rev 2021; 72:101510. [PMID: 34767974 DOI: 10.1016/j.arr.2021.101510] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
Iron is indispensable for normal body functions across species because of its critical roles in red blood cell function and many essential proteins and enzymes required for numerous physiological processes. Regulation of iron homeostasis is an intricate process involving multiple modulators at the systemic, cellular, and molecular levels. Interestingly, emerging evidence has demonstrated that many modulators of iron homeostasis contribute to organismal aging and longevity. On the other hand, the age-related dysregulation of iron homeostasis is often associated with multiple age-related pathologies including bone resorption and neurodegenerative diseases such as Alzheimer's disease. Thus, a thorough understanding on the interconnections between systemic and cellular iron balance and organismal aging may help decipher the etiologies of multiple age-related diseases, which could ultimately lead to developing therapeutic strategies to delay aging and treat various age-related diseases. Here we present the current understanding on the mechanisms of iron homeostasis. We also discuss the impacts of aging on iron homeostatic processes and how dysregulated iron metabolism may affect aging and organismal longevity.
Collapse
|
9
|
Urrutia PJ, Bórquez DA, Núñez MT. Inflaming the Brain with Iron. Antioxidants (Basel) 2021; 10:antiox10010061. [PMID: 33419006 PMCID: PMC7825317 DOI: 10.3390/antiox10010061] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Iron accumulation and neuroinflammation are pathological conditions found in several neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Iron and inflammation are intertwined in a bidirectional relationship, where iron modifies the inflammatory phenotype of microglia and infiltrating macrophages, and in turn, these cells secrete diffusible mediators that reshape neuronal iron homeostasis and regulate iron entry into the brain. Secreted inflammatory mediators include cytokines and reactive oxygen/nitrogen species (ROS/RNS), notably hepcidin and nitric oxide (·NO). Hepcidin is a small cationic peptide with a central role in regulating systemic iron homeostasis. Also present in the cerebrospinal fluid (CSF), hepcidin can reduce iron export from neurons and decreases iron entry through the blood-brain barrier (BBB) by binding to the iron exporter ferroportin 1 (Fpn1). Likewise, ·NO selectively converts cytosolic aconitase (c-aconitase) into the iron regulatory protein 1 (IRP1), which regulates cellular iron homeostasis through its binding to iron response elements (IRE) located in the mRNAs of iron-related proteins. Nitric oxide-activated IRP1 can impair cellular iron homeostasis during neuroinflammation, triggering iron accumulation, especially in the mitochondria, leading to neuronal death. In this review, we will summarize findings that connect neuroinflammation and iron accumulation, which support their causal association in the neurodegenerative processes observed in AD and PD.
Collapse
Affiliation(s)
- Pamela J. Urrutia
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
| | - Daniel A. Bórquez
- Center for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, 8370007 Santiago, Chile;
| | - Marco Tulio Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
- Correspondence: ; Tel.: +56-2-29787360
| |
Collapse
|
10
|
The role of cellular iron deficiency in controlling iron export. Biochim Biophys Acta Gen Subj 2020; 1865:129829. [PMID: 33340587 DOI: 10.1016/j.bbagen.2020.129829] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/25/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Iron export via the transport protein ferroportin (Fpn) plays a critical role in the regulation of dietary iron absorption and iron recycling in macrophages. Fpn plasma membrane expression is controlled by the hepatic iron-regulated hormone hepcidin in response to high iron availability and inflammation. Hepcidin binds to the central cavity of the Fpn transporter to block iron export either directly or by inducing Fpn internalization and lysosomal degradation. Here, we investigated whether iron deficiency affects Fpn protein turnover. METHODS We ectopically expressed Fpn in HeLa cells and used cycloheximide chase experiments to study basal and hepcidin-induced Fpn degradation under extracellular and intracellular iron deficiency. CONCLUSIONS/GENERAL SIGNIFICANCE We show that iron deficiency does not affect basal Fpn turnover but causes a significant delay in hepcidin-induced degradation when cytosolic iron levels are low. These data have important mechanistic implications supporting the hypothesis that iron export is required for efficient targeting of Fpn by hepcidin. Additionally, we show that Fpn degradation is not involved in protecting cells from intracellular iron deficiency.
Collapse
|
11
|
Iron Dysregulation in Human Cancer: Altered Metabolism, Biomarkers for Diagnosis, Prognosis, Monitoring and Rationale for Therapy. Cancers (Basel) 2020; 12:cancers12123524. [PMID: 33255972 PMCID: PMC7761132 DOI: 10.3390/cancers12123524] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Iron is the more abundant metal ion in humans. It is essential for life as it has a role in various cellular processes involved, for instance, in cell metabolism and DNA synthesis. These functions are crucial for cell proliferation, and it is therefore not surprising that iron is accumulated in tumors. In this review, we describe normal and altered iron homeostasis mechanisms. We also provide a vision of iron-related proteins with altered expression in cancers and discuss their potential as diagnostic and/or prognostic biomarkers. Finally, we give an overview of therapeutic strategies acting on iron metabolism to fight against cancers. Abstract Iron (Fe) is a trace element that plays essential roles in various biological processes such as DNA synthesis and repair, as well as cellular energy production and oxygen transport, and it is currently widely recognized that iron homeostasis is dysregulated in many cancers. Indeed, several iron homeostasis proteins may be responsible for malignant tumor initiation, proliferation, and for the metastatic spread of tumors. A large number of studies demonstrated the potential clinical value of utilizing these deregulated proteins as prognostic and/or predictive biomarkers of malignancy and/or response to anticancer treatments. Additionally, the iron present in cancer cells and the importance of iron in ferroptosis cell death signaling pathways prompted the development of therapeutic strategies against advanced stage or resistant cancers. In this review, we select relevant and promising studies in the field of iron metabolism in cancer research and clinical oncology. Besides this, we discuss some co-existing discrepant findings. We also present and discuss the latest lines of research related to targeting iron, or its regulatory pathways, as potential promising anticancer strategies for human therapy. Iron chelators, such as deferoxamine or iron-oxide-based nanoparticles, which are already tested in clinical trials, alone or in combination with chemotherapy, are also reported.
Collapse
|
12
|
Berry T, Abohamza E, Moustafa AA. A disease-modifying treatment for Alzheimer's disease: focus on the trans-sulfuration pathway. Rev Neurosci 2020; 31:319-334. [PMID: 31751299 DOI: 10.1515/revneuro-2019-0076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/31/2019] [Indexed: 12/16/2022]
Abstract
High homocysteine levels in Alzheimer's disease (AD) result from low activity of the trans-sulfuration pathway. Glutathione levels are also low in AD. L-cysteine is required for the synthesis of glutathione. The synthesis of coenzyme A (CoA) requires L-cysteine, which is synthesized via the trans-sulfuration pathway. CoA is required for the synthesis of acetylcholine and appropriate cholinergic neurotransmission. L-cysteine is required for the synthesis of molybdenum-containing proteins. Sulfite oxidase (SUOX), which is a molybdenum-containing protein, could be dysregulated in AD. SUOX detoxifies the sulfites. Glutaminergic neurotransmission could be dysregulated in AD due to low levels of SUOX and high levels of sulfites. L-cysteine provides sulfur for iron-sulfur clusters. Oxidative phosphorylation (OXPHOS) is heavily dependent on iron-sulfur proteins. The decrease in OXPHOS seen in AD could be due to dysregulations of the trans-sulfuration pathway. There is a decrease in aconitase 1 (ACO1) in AD. ACO1 is an iron-sulfur enzyme in the citric acid cycle that upon loss of an iron-sulfur cluster converts to iron regulatory protein 1 (IRP1). With the dysregulation of iron-sulfur cluster formation ACO1 will convert to IRP1 which will decrease the 2-oxglutarate synthesis dysregulating the citric acid cycle and also dysregulating iron metabolism. Selenomethionine is also metabolized by the trans-sulfuration pathway. With the low activity of the trans-sulfuration pathway in AD selenoproteins will be dysregulated in AD. Dysregulation of selenoproteins could lead to oxidant stress in AD. In this article, we propose a novel treatment for AD that addresses dysregulations resulting from low activity of the trans-sulfuration pathway and low L-cysteine.
Collapse
Affiliation(s)
- Thomas Berry
- School of Social Sciences and Psychology, Western Sydney University, 2 Bullecourt Ave, Milperra, 2214 Sydney, New South Wales, Australia
| | - Eid Abohamza
- Department of Social Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Ahmed A Moustafa
- School of Social Sciences and Psychology, Western Sydney University, 2 Bullecourt Ave, Milperra, 2214 Sydney, New South Wales, Australia
| |
Collapse
|
13
|
A comprehensive mechanistic review insight into the effects of micronutrients on toll-like receptors functions. Pharmacol Res 2019; 152:104619. [PMID: 31887355 DOI: 10.1016/j.phrs.2019.104619] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/23/2019] [Accepted: 12/26/2019] [Indexed: 12/29/2022]
Abstract
Toll-like receptors (TLRs) are the special proteins receptors for recognition of molecules related to the pathogens. In this way, TLRs and secreted cytokines as a result of TLRs activation are involved in the inflammation pathways. So far, in vivo and in vitro studies have demonstrated that micronutrients (vitamins & minerals) with a broad range of effects on body health, can regulate TLRs signaling pathways. Current review aimed at determining the possible mechanisms of micronutrient effects on TLRs functions. In the aspect of gene expression, micronutrients have inconsistent effects on mRNA level of TLRs which are dependent on time, dose and type of studied TLR. Also, some micronutrients affect gene expression of TLRs signaling mediators namely TLRs adaptors like Myeloid differentiation primary response 88 (MyD88). In the aspect of TLRs signaling pathways, nuclear factor-κB (NF-κB) is an important mediator which is regulated by micronutrients. Also, the regulatory effects of micronutrients on phosphorylation reactions may be effective in the activation/inactivation of TLRs signaling mediators. In addition, zinc can regulate TLRs signaling indirectly via the zinc finger proteins which have contradictory effects on TLRs cascade. In conclusion, the relationship between micronutrients and TLRs signaling is complicated and depends on some known internal, external and genetic factors like form of studied micronutrient, cell type, TLR agonist, dose and time of exposure, inflammation, apoptosis, cell cycle, and environmental factors. Some unknown factors may be effective in TLRs response and as a result additional mechanistic studies are needed to elucidate exact effect of micronutrients on TLRs signaling.
Collapse
|
14
|
Chiou B, Neal EH, Bowman AB, Lippmann ES, Simpson IA, Connor JR. Endothelial cells are critical regulators of iron transport in a model of the human blood-brain barrier. J Cereb Blood Flow Metab 2019; 39:2117-2131. [PMID: 29911470 PMCID: PMC6827128 DOI: 10.1177/0271678x18783372] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Iron delivery to the brain is essential for multiple neurological processes such as myelination, neurotransmitter synthesis, and energy production. Loss of brain iron homeostasis is a significant factor in multiple neurological disorders. Understanding the mechanism by which the transport of iron across the blood-brain barrier (BBB) is regulated is crucial to address the impact of iron deficiency on brain development and excessive accumulation of iron in neurodegenerative diseases. Using induced pluripotent stem cell (iPSC)-derived brain endothelial cells (huECs) as a human BBB model, we demonstrate the ability of transferrin, hepcidin, and DMT1 to impact iron transport and release. Our model reveals a new function for H-ferritin to transport iron across the BBB by binding to the T-cell immunoglobulin and mucin receptor 1. We show that huECs secrete both transferrin and H-ferritin, which can serve as iron sources for the brain. Based on our data, brain iron status can exert control of iron transport across the endothelial cells that constitute the BBB. These data address a number of pertinent questions such as how brain iron uptake is regulated at the regional level, the source of iron delivery to the brain, and the clinical strategies for attempting to treat brain iron deficiency.
Collapse
Affiliation(s)
- Brian Chiou
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - Emma H Neal
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Aaron B Bowman
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ian A Simpson
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - James R Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
15
|
Dutta A, Islam ASM, Maiti D, Sasmal M, Pradhan C, Ali M. A smart molecular probe for selective recognition of nitric oxide in 100% aqueous solution with cell imaging application and DFT studies. Org Biomol Chem 2019; 17:2492-2501. [DOI: 10.1039/c9ob00177h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A simple, least-cytotoxic as well as an efficient fluorescent sensor HqEN480 recognizes NO in 100% aqueous solution with cell imaging application.
Collapse
Affiliation(s)
- Ananya Dutta
- Department of Chemistry Jadavpur University
- Kolkata 700 032
- India
| | | | - Debjani Maiti
- Department of Chemistry Jadavpur University
- Kolkata 700 032
- India
| | - Mihir Sasmal
- Department of Chemistry Jadavpur University
- Kolkata 700 032
- India
| | - Chandradoy Pradhan
- Molecular & Human Genetics Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Mahammad Ali
- Department of Chemistry Jadavpur University
- Kolkata 700 032
- India
- Vice-Chancellor
- Aliah University
| |
Collapse
|
16
|
Associations of Serum Nitric Oxide with Vitamin D and Other Metabolic Factors in Apparently Healthy Adolescents. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1489132. [PMID: 30175114 PMCID: PMC6098934 DOI: 10.1155/2018/1489132] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/26/2018] [Indexed: 12/13/2022]
Abstract
Introduction Nitric oxide (NOx) is an important biomolecule which interacts with other molecules including 25(OH)D to mediate various metabolic pathways. Interactions and associations of NOx with 25(OH)D have been well studied both in vitro and in vivo, yet associations in apparently healthy adolescents have never been studied. Methods A total of 740 (245 boys and 495 girls) apparently healthy Saudi adolescents aged 10-17 years were included in this cross-sectional study, to determine the associations of NOx with 25(OH)D and other biomarkers in Saudi adolescents. Serum NOx, 25(OH)D, and other biochemical and anthropometric parameters were measured following standard protocols and manufacturers' guidelines. Results NOx level was significantly higher in boys than girls (p<0.001). In all subjects, NOx showed a significant inverse correlation with 25(OH)D. After stratification according to sex however this significant association was observed only in boys and not in girls. NOx was also significantly associated with BMI, serum triglycerides, and systolic blood pressure in all subjects. Conclusion The significantly inverse association of NOx and 25(OH)D among apparently healthy adolescents is influenced by sex and further strengthens the extraskeletal role of 25(OH)D in maintaining endothelial homeostasis in this age group, particularly in boys. Whether vitamin D correction can influence NOx production over time among adolescents remains to be proven.
Collapse
|
17
|
Lim D, Kim KS, Jeong JH, Marques O, Kim HJ, Song M, Lee TH, Kim JI, Choi HS, Min JJ, Bumann D, Muckenthaler MU, Choy HE. The hepcidin-ferroportin axis controls the iron content of Salmonella-containing vacuoles in macrophages. Nat Commun 2018; 9:2091. [PMID: 29844422 PMCID: PMC5974375 DOI: 10.1038/s41467-018-04446-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/27/2018] [Indexed: 02/06/2023] Open
Abstract
Macrophages release iron into the bloodstream via a membrane-bound iron export protein, ferroportin (FPN). The hepatic iron-regulatory hormone hepcidin controls FPN internalization and degradation in response to bacterial infection. Salmonella typhimurium can invade macrophages and proliferate in the Salmonella-containing vacuole (SCV). Hepcidin is reported to increase the mortality of Salmonella-infected animals by increasing the bacterial load in macrophages. Here we assess the iron levels and find that hepcidin increases iron content in the cytosol but decreases it in the SCV through FPN on the SCV membrane. Loss-of-FPN from the SCV via the action of hepcidin impairs the generation of bactericidal reactive oxygen species (ROS) as the iron content decreases. We conclude that FPN is required to provide sufficient iron to the SCV, where iron serves as a cofactor for the generation of antimicrobial ROS rather than as a nutrient for Salmonella. The effects of iron on vacuole-resident Salmonella in macrophages are unclear. Here the authors show that the bacteria are not subject to nutritional inhibition by iron deprivation, but that iron depletion in the vacuole, via the hepcidin-ferroportin axis, inhibits the bactericidal effect of oxidative burst.
Collapse
Affiliation(s)
- Daejin Lim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea.,Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea
| | - Kwang Soo Kim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea.,Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea.,Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea
| | - Oriana Marques
- Department of Pediatric Hematology, Oncology and Immunology - University of Heidelberg, Im Neuenheimer Feld 350, Heidelberg, D-69120, Germany.,Molecular Medicine Partnership Unit, Heidelberg, D-69120, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, D-69120, Germany
| | - Hyun-Ju Kim
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea.,Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea
| | - Miryoung Song
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea.,Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea
| | - Tae-Hoon Lee
- Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea.,Department of Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University and Korea mouse phenotype center (KMPC), Gwangju, 61186, Republic of Korea
| | - Jae Il Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.,AnyGen, Gwangju Technopark, Gwangju, 61008, Republic of Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jung-Joon Min
- Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea.,Department of Nuclear Medicine, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Dirk Bumann
- Focal Area Infection Biology, University of Basel, Basel, CH-4056, Switzerland
| | - Martina U Muckenthaler
- Department of Pediatric Hematology, Oncology and Immunology - University of Heidelberg, Im Neuenheimer Feld 350, Heidelberg, D-69120, Germany.,Molecular Medicine Partnership Unit, Heidelberg, D-69120, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, D-69120, Germany
| | - Hyon E Choy
- Department of Microbiology, Chonnam National University Medical School, Gwangju, 61468, Republic of Korea. .,Department of Molecular Medicine (BK21plus), Chonnam National University Graduate School, Gwangju, 61468, Republic of Korea.
| |
Collapse
|
18
|
Tharmalingam S, Alhasawi A, Appanna VP, Lemire J, Appanna VD. Reactive nitrogen species (RNS)-resistant microbes: adaptation and medical implications. Biol Chem 2017. [PMID: 28622140 DOI: 10.1515/hsz-2017-0152] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nitrosative stress results from an increase in reactive nitrogen species (RNS) within the cell. Though the RNS - nitric oxide (·NO) and peroxynitrite (ONOO-) - play pivotal physiological roles, at elevated concentrations, these moieties can be poisonous to both prokaryotic and eukaryotic cells alike due to their capacity to disrupt a variety of essential biological processes. Numerous microbes are known to adapt to nitrosative stress by elaborating intricate strategies aimed at neutralizing RNS. In this review, we will discuss both the enzymatic systems dedicated to the elimination of RNS as well as the metabolic networks that are tailored to generate RNS-detoxifying metabolites - α-keto-acids. The latter has been demonstrated to nullify RNS via non-enzymatic decarboxylation resulting in the production of a carboxylic acid, many of which are potent signaling molecules. Furthermore, as aerobic energy production is severely impeded during nitrosative stress, alternative ATP-generating modules will be explored. To that end, a holistic understanding of the molecular adaptation to nitrosative stress, reinforces the notion that neutralization of toxicants necessitates significant metabolic reconfiguration to facilitate cell survival. As the alarming rise in antimicrobial resistant pathogens continues unabated, this review will also discuss the potential for developing therapies that target the alternative ATP-generating machinery of bacteria.
Collapse
|
19
|
Restini CBA, Gonçalves L. Nitric Oxide and Related Aspects Underlying Angina. Open Cardiovasc Med J 2017; 11:33-46. [PMID: 28567132 PMCID: PMC5418930 DOI: 10.2174/1874192401711010033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/10/2017] [Accepted: 02/27/2017] [Indexed: 01/08/2023] Open
Abstract
Increased number of patients affected by metabolic syndrome (MS) has prompted the necessity of better understanding what is involved in such syndrome. Nevertheless, the establishment of promising therapies depends on the knowledge about the interaction of molecules within MS. In such context, Nitric Oxide (NO) emerges from a bulk of works relating its roles on aspects of MS, including cardiovascular diseases, their symptoms and comorbidities, which are thought to be triggered by similar sources. NO, nitric oxide synthase and enzymatic chains are keys for those disease and symptoms processes. NO has been separately described as part of hypertensive, ischemic and pain signaling. Although there are similar pathways likely shared for generating cardiovascular symptoms such angina, they are barely associated to NO in literature. The present review aims to clarify the patterns of NO alteration in metabolic syndrome directly concerned to cardiovascular symptoms, especially angina.
Collapse
Affiliation(s)
- Carolina Baraldi Araujo Restini
- Biotechnology Dept. (Lab: Cardiorenal Pharmacology)/Medical School, University of Ribeirao Preto (UNAERP), Ribeirão Preto-SP, Brazil
| | - Leticia Gonçalves
- Biotechnology Dept. (Lab: Cardiorenal Pharmacology)/Medical School, University of Ribeirao Preto (UNAERP), Ribeirão Preto-SP, Brazil
| |
Collapse
|
20
|
Islam ASM, Bhowmick R, Pal K, Katarkar A, Chaudhuri K, Ali M. A Smart Molecule for Selective Sensing of Nitric Oxide: Conversion of NO to HSNO; Relevance of Biological HSNO Formation. Inorg Chem 2017; 56:4324-4331. [DOI: 10.1021/acs.inorgchem.6b02787] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Abu Saleh Musha Islam
- Department of Chemistry, Jadavpur University, 188 Raja S.C. Mallick Road, Kolkata 700 032, India
| | - Rahul Bhowmick
- Department of Chemistry, Jadavpur University, 188 Raja S.C. Mallick Road, Kolkata 700 032, India
| | - Kaberi Pal
- Department of Chemistry, Jadavpur University, 188 Raja S.C. Mallick Road, Kolkata 700 032, India
| | - Atul Katarkar
- Molecular & Human Genetics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mallick Road, Kolkata 700 032, India
| | - Keya Chaudhuri
- Molecular & Human Genetics Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mallick Road, Kolkata 700 032, India
| | - Mahammad Ali
- Department of Chemistry, Jadavpur University, 188 Raja S.C. Mallick Road, Kolkata 700 032, India
| |
Collapse
|
21
|
Leecharoenkiat K, Lithanatudom P, Sornjai W, Smith DR. Iron dysregulation in beta-thalassemia. ASIAN PAC J TROP MED 2016; 9:1035-1043. [PMID: 27890361 DOI: 10.1016/j.apjtm.2016.07.035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 08/01/2016] [Accepted: 10/14/2016] [Indexed: 11/24/2022] Open
Abstract
Iron deficiency anemia and iron overload conditions affect more than one billion people worldwide. Iron homeostasis involves the regulation of cells that export iron into the plasma and cells that utilize or store iron. The cellular iron balance in humans is primarily mediated by the hepcidin-ferroportin axis. Ferroportin is the sole cellular iron export protein, and its expression is regulated transcriptionally, post-transcriptionally and post-translationally. Hepcidin, a hormone produced by liver cells, post-translationally regulates ferroportin expression on iron exporting cells by binding with ferroportin and promoting its internalization by endocytosis and subsequent degradation by lysosomes. Dysregulation of iron homeostasis leading to iron deposition in vital organs is the main cause of death in beta-thalassemia patients. Beta-thalassemia patients show marked hepcidin suppression, ineffective erythropoiesis, anemia and iron overload. Beta-thalassemia is common in the Mediterranean region, Southeast Asia and the Indian subcontinent, and the focus of this review is to provide an update on the factors mediating hepcidin related iron dysregulation in beta-thalassemia disease. Understanding this process may pave the way for new treatments to ameliorate iron overloading and improve the long term prognosis of these patients.
Collapse
Affiliation(s)
- Kamonlak Leecharoenkiat
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, 154 Rama 4 Road, Bangkok 10330, Thailand
| | - Pathrapol Lithanatudom
- Department of Biology, Faculty of Science, Chiang Mai University, 239 Huaykaew Road, Amphur Muang, Chiang Mai 50200, Thailand
| | - Wannapa Sornjai
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Duncan R Smith
- Molecular Pathology Laboratory, Institute of Molecular Biosciences, Mahidol University, 25/25 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand.
| |
Collapse
|
22
|
Mutchler SM, Straub AC. Compartmentalized nitric oxide signaling in the resistance vasculature. Nitric Oxide 2015; 49:8-15. [PMID: 26028569 DOI: 10.1016/j.niox.2015.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) was first described as a bioactive molecule through its ability to stimulate soluble guanylate cyclase, but the revelation that NO was the endothelium derived relaxation factor drove the field to its modern state. The wealth of research conducted over the past 30 years has provided us with a picture of how diverse NO signaling can be within the vascular wall, going beyond simple vasodilation to include such roles as signaling through protein S-nitrosation. This expanded view of NO's actions requires highly regulated and compartmentalized production. Importantly, resistance arteries house multiple proteins involved in the production and transduction of NO allowing for efficient movement of the molecule to regulate vascular tone and reactivity. In this review, we focus on the many mechanisms regulating NO production and signaling action in the vascular wall, with a focus on the control of endothelial nitric oxide synthase (eNOS), the enzyme responsible for synthesizing most of the NO within these confines. We also explore how cross talk between the endothelium and smooth muscle in the microcirculation can modulate NO signaling, illustrating that this one small molecule has the capability to produce a plethora of responses.
Collapse
Affiliation(s)
- Stephanie M Mutchler
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
23
|
Sudden sensorineural hearing loss and polymorphisms in iron homeostasis genes: new insights from a case-control study. BIOMED RESEARCH INTERNATIONAL 2015; 2015:834736. [PMID: 25789325 PMCID: PMC4348611 DOI: 10.1155/2015/834736] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/15/2014] [Accepted: 01/06/2015] [Indexed: 12/19/2022]
Abstract
Background. Even if various pathophysiological events have been proposed as explanations, the putative cause of sudden hearing loss remains unclear. Objectives. To investigate and to reveal associations (if any) between the main iron-related gene variants and idiopathic sudden sensorineural hearing loss. Study Design. Case-control study. Materials and Methods. A total of 200 sudden sensorineural hearing loss patients (median age 63.65 years; range 10-92) were compared with 400 healthy control subjects. The following genetic variants were investigated: the polymorphism c.-8CG in the promoter of the ferroportin gene (FPN1; SLC40A1), the two isoforms C1 and C2 (p.P570S) of the transferrin protein (TF), the amino acidic substitutions p.H63D and p.C282Y in the hereditary hemochromatosis protein (HFE), and the polymorphism c.-582AG in the promoter of the HEPC gene, which encodes the protein hepcidin (HAMP). Results. The homozygous genotype c.-8GG of the SLC40A1 gene revealed an OR for ISSNHL risk of 4.27 (CI 95%, 2.65-6.89; P = 0.001), being overrepresented among cases. Conclusions. Our study indicates that the homozygous genotype FPN1 -8GG was significantly associated with increased risk of developing sudden hearing loss. These findings suggest new research should be conducted in the field of iron homeostasis in the inner ear.
Collapse
|
24
|
Zhao GY, DI DH, Wang B, Zhang P, Xu YJ. Iron regulates the expression of ferroportin 1 in the cultured hFOB 1.19 osteoblast cell line. Exp Ther Med 2014; 8:826-830. [PMID: 25120608 PMCID: PMC4113530 DOI: 10.3892/etm.2014.1823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 06/11/2014] [Indexed: 12/16/2022] Open
Abstract
Iron metabolism is tightly regulated in osteoblasts, and ferroportin 1 (FPN1) is the only identified iron exporter in mammals to date. In the present study, the regulation of FNP1 in human osteoblasts was investigated following various iron treatments. The human osteoblast cell line hFOB 1.19 was treated with ferric ammonium citrate (FAC) or desferrioxamine (DFO) of various concentrations. The intracellular iron ion levels were measured using a confocal laser scanning microscope. In addition, the mRNA and protein expression levels of FPN1 were detected by quantitative polymerase chain reaction, western blot analysis and immunofluorescence. The results demonstrated that increasing iron concentrations via FAC treatment increased the expression of FPN1. By contrast, decreasing the iron concentration by DFO treatment decreased FNP1 expression levels. In addition to demonstrating that the FNP1 expression changed according to the iron concentration, the observations indicated that changes in FPN1 expression may contribute to the maintenance of the intracellular iron balance in osteoblasts.
Collapse
Affiliation(s)
- Guo-Yang Zhao
- Department of Orthopedics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Dong-Hua DI
- Department of Orthopedics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Bo Wang
- Department of Orthopedics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Peng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - You-Jia Xu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
25
|
Chen Q, Xiao DS. Long-term aerobic exercise increases redox-active iron through nitric oxide in rat hippocampus. Nitric Oxide 2013; 36:1-10. [PMID: 24184442 DOI: 10.1016/j.niox.2013.10.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/10/2013] [Accepted: 10/22/2013] [Indexed: 12/30/2022]
Abstract
Adult hippocampus is highly vulnerable to iron-induced oxidative stress. Aerobic exercise has been proposed to reduce oxidative stress but the findings in the hippocampus are conflicting. This study aimed to observe the changes of redox-active iron and concomitant regulation of cellular iron homeostasis in the hippocampus by aerobic exercise, and possible regulatory effect of nitric oxide (NO). A randomized controlled study was designed in the rats with swimming exercise treatment (for 3 months) and/or an unselective inhibitor of NO synthase (NOS) (L-NAME) treatment. The results from the bleomycin-detectable iron assay showed additional redox-active iron in the hippocampus by exercise treatment. The results from nonheme iron content assay, combined with the redox-active iron content, showed increased storage iron content by exercise treatment. NOx (nitrate plus nitrite) assay showed increased NOx content by exercise treatment. The results from the Western blot assay showed decreased ferroportin expression, no changes of TfR1 and DMT1 expressions, increased IRP1 and IRP2 expression, increased expressions of eNOS and nNOS rather than iNOS. In these effects of exercise treatment, the increased redox-active iron content, storage iron content, IRP1 and IRP2 expressions were completely reversed by L-NAME treatment, and decreased ferroportin expression was in part reversed by L-NAME. L-NAME treatment completely inhibited increased NOx and both eNOS and nNOS expression in the hippocampus. Our findings suggest that aerobic exercise could increase the redox-active iron in the hippocampus, indicating an increase in the capacity to generate hydroxyl radicals through the Fenton reactions, and aerobic exercise-induced iron accumulation in the hippocampus might mainly result from the role of the endogenous NO.
Collapse
Affiliation(s)
- Qian Chen
- Department of Histology and Embryology, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - De-Sheng Xiao
- Department of Preventive Medicine, School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
26
|
Redox activation of Nrf2 & NF-κB: a double end sword? Cell Signal 2013; 25:2548-57. [PMID: 23993959 DOI: 10.1016/j.cellsig.2013.08.007] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 08/23/2013] [Indexed: 12/20/2022]
Abstract
Moderate concentrations of reactive oxygen species (ROS) are produced by diverse sources under physiological conditions. At such low levels, these molecules may act as upstream mediators of relevant signaling pathways; however an increase in their concentration with respect to the antioxidant system activity, changes their redox signaling function into a deleterious role. Thus, cell health depends, at least in part, on redox balance. This review includes global aspects of oxygen chemistry, ROS generation, antioxidant system, and redox signaling. It is also focused on the description of two relevant redox-sensitive transcription factors: nuclear factor erythroid 2-related factor 2 (Nrf2), which may be a potential target to confer cell protection, and nuclear factor κB (NF-κB), which is involved in deleterious effects in the cell. Finally, recent findings on the interplay between both factors for the development of different pathologies are discussed.
Collapse
|
27
|
Brabin L, Brabin BJ, Gies S. Influence of iron status on risk of maternal or neonatal infection and on neonatal mortality with an emphasis on developing countries. Nutr Rev 2013; 71:528-40. [PMID: 23865798 PMCID: PMC3721428 DOI: 10.1111/nure.12049] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Infection is a major cause of neonatal death in developing countries. This review investigates whether host iron status affects the risk of maternal and/or neonatal infection, potentially contributing to neonatal death, and summarizes the iron acquisition mechanisms described for pathogens causing stillbirth, preterm birth, and congenital infection. In vitro evidence shows that iron availability influences the severity and chronicity of infections that cause these negative outcomes of pregnancy. In vivo evidence is lacking, as relevant studies of maternal iron supplementation have not assessed the effect of iron status on the risk of maternal and/or neonatal infection. Reducing iron-deficiency anemia among women is beneficial and should improve the iron stores of babies; moreover, there is evidence that iron status in young children predicts the risk of malaria and, possibly, the risk of invasive bacterial diseases. Caution with maternal iron supplementation is indicated in iron-replete women who may be at high risk of exposure to infection, although distinguishing between iron-replete and iron-deficient women is currently difficult in developing countries, where a point-of-care test is needed. Further research is indicated to investigate the risk of infection relative to iron status in mothers and babies in order to avoid iron intervention strategies that may result in detrimental birth outcomes in some groups of women.
Collapse
Affiliation(s)
- Loretta Brabin
- Academic Unit of Obstetrics & Gynaecology, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
28
|
Hepcidin regulates intrarenal iron handling at the distal nephron. Kidney Int 2013; 84:756-66. [PMID: 23615502 DOI: 10.1038/ki.2013.142] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 01/15/2013] [Accepted: 02/14/2013] [Indexed: 02/08/2023]
Abstract
Hepcidin, the key regulatory hormone of iron homeostasis, and iron carriers such as transferrin receptor1 (TFR1), divalent metal transporter1 (DMT1), and ferroportin (FPN) are expressed in kidney. Whether hepcidin plays an intrinsic role in the regulation of renal iron transport is unknown. Here, we analyzed the renal handling of iron in hemochromatosis Hepc(-/-) and Hjv(-/-) mouse models, as well as in phenylhydrazine (PHZ)-treated mice. We found a marked medullary iron deposition in the kidneys of Hepc(-/-) mice, and iron leak in the urine. The kidneys of Hepc(-/-) mice exhibited a concomitant decrease in TFR1 and increase in ferritin and FPN expression. Increased FPN abundance was restricted to the thick ascending limb (TAL). DMT1 protein remained unaffected despite a significant decrease of its mRNA level, suggesting that DMT1 protein is stabilized in the absence of hepcidin. Treatment of kidney sections from Hepc(-/-) mice with hepcidin decreased DMT1 protein, an effect confirmed in renal cell lines where hepcidin markedly decreased (55)Fe transport. In the kidneys of Hjv(-/-) mice exhibiting low hepcidin expression, the iron overload was similar to that in the kidneys of Hepc(-/-) mice. However, in PHZ mice, iron accumulation resulting from hemoglobin leak was detected in the proximal tubule. Thus, kidneys exhibit a tissue-specific handling of iron that depends on the extra iron source. Hepcidin may control the expression of iron transporters to prevent renal iron overload.
Collapse
|
29
|
Chiabrando D, Fiorito V, Marro S, Silengo L, Altruda F, Tolosano E. Cell-specific regulation of Ferroportin transcription following experimentally-induced acute anemia in mice. Blood Cells Mol Dis 2013; 50:25-30. [DOI: 10.1016/j.bcmd.2012.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 08/03/2012] [Accepted: 08/03/2012] [Indexed: 12/17/2022]
|
30
|
Abstract
Ferroportin (FPN) is the sole iron export membrane protein identified in mammals that is abundantly expressed on absorptive enterocytes and macrophages, and is essential for physiological regulation of cellular iron. The expression of FPN is positively induced by cellular iron and is suppressed by liver hepcidin in response to either increased systemic iron or inflammatory stimuli. Hepcidin binds to cell surface FPN inducing FPN internalization followed by lysosomal degradation of the protein and consequently iron efflux from macrophages is blocked and there is suboptimal iron absorption by duodenal enterocytes. Dozens of FPN gene mutations have been identified in different ethnic populations and some of the mutations are associated with autosomal dominant iron overload disorder described as FPN disease or hemochromatosis type 4 that is distinct from hereditary hemochromatosis due to HFE mutations. Clinical manifestations of iron overload FPN disease can be classified into two groups according to whether there is selective macrophage iron loading or parenchymal and reticuloendothelial iron accumulation. There is evidence suggesting that altered hepcidin-FPN interaction can modulate host's response to infection. Resistance to hepcidin promotes iron egress from cells and this inhibits growth of intracellular pathogens. Conversely, iron retention due to loss of iron export activity by mutated FPN results in intracellular iron accumulation and a permissive environment for intracellular pathogens.
Collapse
Affiliation(s)
- Ishmael Kasvosve
- Department of Medical Laboratory Sciences Faculty of Health Sciences, University of Botswana Private Bag UB 00712, Gaborone, Botswana.
| |
Collapse
|
31
|
Naz N, Malik IA, Sheikh N, Ahmad S, Khan S, Blaschke M, Schultze F, Ramadori G. Ferroportin-1 is a 'nuclear'-negative acute-phase protein in rat liver: a comparison with other iron-transport proteins. J Transl Med 2012; 92:842-56. [PMID: 22469696 DOI: 10.1038/labinvest.2012.52] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Liver is the central organ of iron metabolism. During acute-phase-response (APR), serum iron concentration rapidly decreases. The current study aimed to compare expression and localization of iron transport protein ferroportin-1 (Fpn-1) and of other iron import proteins after experimental tissue damage induced by injecting turpentine oil in the hind limbs of rats and mice. Serum and spleen iron concentration decreased with an increase in total liver, cytoplasmic and nuclear iron concentration. In liver, mRNA amount of Fpn-1, Fpn-1a, Fpn-1b, HFE, hemojuvelin (HJV) and hephaestin (heph) genes showed a rapid decrease. Hepcidin, divalent metal transporter-1 (DMT-1), transferrin (Tf) and Tf-receptor-1 (TfR1), TfR-2 (TfR2) gene expression was increased. Western blot analysis of liver tissue lysate confirmed the changes observed at mRNA level. In spleen, a rapid decrease in gene expression of Fpn-1, Fpn-1a, Fpn-1b, DMT-1, Tf, TfR1 and TfR2, and an increase in hepcidin was observed. Immunohistochemistry of DMT-1 and TfR2 were mainly detected in the nucleus of rat liver and spleen, whereas TfR1 was clearly localized in the plasma membrane. Fpn-1 was mostly found in the nuclei of liver cells, whereas in spleen, the protein was mainly detected in the cell membrane. Western blot analysis of liver fractions confirmed immunohistochemical results. In livers of wild-type mice, gene expression of Fpn-1, Fpn-1a and Fpn-1b was downregulated, whereas hepcidin gene expression was increased. In contrast, these changes were less pronounced in IL-6ko-mice. Cytokine (IL-6, IL-1b and TNF-a) treatment of rat hepatocytes showed a downregulation of Fpn-1, Fpn-1a and Fpn-1b, and upregulation of hepcidin gene expression. Moreover, western blot analysis of cell lysate of IL-6-treated hepatocytes detected, as expected, an increase of a2-macroglobulin (positive acute-phase protein), whereas albumin (negative acute-phase protein) and Fpn-1 were downregulated. Our results demonstrate that liver behaves as a 'sponge' for iron under acute-phase conditions, and Fpn-1 behaves as a negative acute-phase protein in rat hepatocytes mainly, but not exclusively, because of the effect of IL-6. These changes could explain iron retention in the cytoplasm and in the nucleus of hepatocytes during APR.
Collapse
Affiliation(s)
- Naila Naz
- Division of Gastroenterology and Endocrinology, Department of Internal Medicine, University Hospital, Georg-August-University, Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Toll-like receptor signal adaptor protein MyD88 is required for sustained endotoxin-induced acute hypoferremic response in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2340-50. [PMID: 22497726 DOI: 10.1016/j.ajpath.2012.01.046] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 01/24/2012] [Accepted: 01/30/2012] [Indexed: 12/24/2022]
Abstract
Hypoferremia, associated with immune system activation, involves a marked reduction in the levels of circulating iron, coupled with iron sequestration within macrophages. Toll-like receptor (TLR) signaling plays an important role in the development of the hypoferremic response, but how downstream signaling events affect genes involved in iron metabolism is incompletely understood. We investigated the involvement of MyD88-dependent (MyD88) and MyD88-independent (TRIF) TLR signaling in the development of hypoferremia. Using MyD88-deficient and TRIF-deficient mice, we show that MyD88 and TRIF signaling pathways are critical for up-regulation by lipopolysaccharide (LPS) of the iron regulator hepcidin. In addition, MyD88 signaling is required for the induction of lipocalin 2 secretion and iron sequestration in the spleen. Activation of TLR4 and TLR3 signaling through LPS and polyinosinic:polycytidylic acid [poly(I:C)] treatments resulted in rapid down-regulation of HFE protein [encoded by the hemochromatosis gene (Hfe)] and ferroportin [encoded by solute carrier family 40 (iron-regulated transporter), member 1 (Slc40a1)] expression in the spleen, independent of MyD88 or TRIF signaling and proinflammatory cytokine production. However, lack of MyD88 signaling significantly impaired the hypoferremic response triggered by LPS, indicating that ferroportin and HFE protein down-regulation alone are insufficient to maintain hypoferremia. The extent of the hypoferremic response was found to be limited by initial, basal iron levels. Together, these results suggest that targeting specific TLR signaling pathways by affecting the function of adaptor molecules may provide new strategies to counteract iron sequestration within macrophages during inflammation.
Collapse
|
33
|
Förstermann U, Sessa WC. Nitric oxide synthases: regulation and function. Eur Heart J 2012; 33:829-37, 837a-837d. [PMID: 21890489 PMCID: PMC3345541 DOI: 10.1093/eurheartj/ehr304] [Citation(s) in RCA: 2666] [Impact Index Per Article: 222.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 07/14/2011] [Accepted: 07/28/2011] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO), the smallest signalling molecule known, is produced by three isoforms of NO synthase (NOS; EC 1.14.13.39). They all utilize l-arginine and molecular oxygen as substrates and require the cofactors reduced nicotinamide-adenine-dinucleotide phosphate (NADPH), flavin adenine dinucleotide (FAD), flavin mononucleotide (FMN), and (6R-)5,6,7,8-tetrahydrobiopterin (BH(4)). All NOS bind calmodulin and contain haem. Neuronal NOS (nNOS, NOS I) is constitutively expressed in central and peripheral neurons and some other cell types. Its functions include synaptic plasticity in the central nervous system (CNS), central regulation of blood pressure, smooth muscle relaxation, and vasodilatation via peripheral nitrergic nerves. Nitrergic nerves are of particular importance in the relaxation of corpus cavernosum and penile erection. Phosphodiesterase 5 inhibitors (sildenafil, vardenafil, and tadalafil) require at least a residual nNOS activity for their action. Inducible NOS (NOS II) can be expressed in many cell types in response to lipopolysaccharide, cytokines, or other agents. Inducible NOS generates large amounts of NO that have cytostatic effects on parasitic target cells. Inducible NOS contributes to the pathophysiology of inflammatory diseases and septic shock. Endothelial NOS (eNOS, NOS III) is mostly expressed in endothelial cells. It keeps blood vessels dilated, controls blood pressure, and has numerous other vasoprotective and anti-atherosclerotic effects. Many cardiovascular risk factors lead to oxidative stress, eNOS uncoupling, and endothelial dysfunction in the vasculature. Pharmacologically, vascular oxidative stress can be reduced and eNOS functionality restored with renin- and angiotensin-converting enzyme-inhibitors, with angiotensin receptor blockers, and with statins.
Collapse
Affiliation(s)
- Ulrich Förstermann
- Department of Pharmacology, Johannes Gutenberg University Medical Center, 55101 Mainz, Germany.
| | | |
Collapse
|
34
|
Gu J, Xu H, Han Y, Dai W, Hao W, Wang C, Gu N, Xu H, Cao J. The internalization pathway, metabolic fate and biological effect of superparamagnetic iron oxide nanoparticles in the macrophage-like RAW264.7 cell. SCIENCE CHINA-LIFE SCIENCES 2011; 54:793-805. [PMID: 21922429 DOI: 10.1007/s11427-011-4215-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 03/29/2011] [Indexed: 12/29/2022]
Abstract
The potential applications of superparamagnetic iron oxide nanoparticles (SPIONs) in several nanomedical fields have attracted intense interest based on the cell-nano interaction. However, the mechanisms underlying cell uptake, the intracellular trail, final fate and the biological effects of SPIONs have not yet been clearly elucidated. Here, we showed that multiple endocytic pathways were involved in the internalization process of SPIONs in the RAW264.7 macrophage. The internalized SPIONs were biocompatible and used three different metabolic pathways: The SPIONs were distributed to daughter cells during mitosis; they were degraded in the lysosome and free iron was released into the intracellular iron metabolic pool; and, the intact SPIONs were potentially exocytosed out of the cells. The internalized SPIONs did not induce cell damage but affected iron metabolism, inducing the upregulation of ferritin light chain at both the mRNA and protein levels and ferroportin 1 at the mRNA level. These results may contribute to the development of nanobiology and to the safe use of SPIONs in medicine when administered as a contrast medium or a drug delivery tool.
Collapse
Affiliation(s)
- Jingli Gu
- Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Carroll CB, Zeissler ML, Chadborn N, Gibson K, Williams G, Zajicek JP, Morrison KE, Hanemann CO. Changes in iron-regulatory gene expression occur in human cell culture models of Parkinson's disease. Neurochem Int 2011; 59:73-80. [PMID: 21672570 DOI: 10.1016/j.neuint.2011.05.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 05/26/2011] [Indexed: 12/23/2022]
Abstract
BACKGROUND Neuronal iron accumulation is thought to be relevant to the pathogenesis of Parkinson's disease (PD), although the mechanism remains elusive. We hypothesized that neuronal iron uptake may be stimulated by functional mitochondrial iron deficiency. OBJECTIVE To determine firstly whether the mitochondrial toxin, 1-methyl-4-phenylpyridinium iodide (MPP(+)), results in upregulation of iron-import proteins and transporters of iron into the mitochondria, and secondly whether similar changes in expression are induced by toxins with different mechanisms of action. METHODS We used quantitative PCR and Western blotting to investigate expression of the iron importers, divalent metal transporter, transferrin receptor 1 and 2 (TfR1 and TfR2) and mitoferrin-2 and the iron exporter ferroportin in differentiated SH-SY5Y cells exposed to three different toxins relevant to PD, MPP(+), paraquat (a free radical generator) and lactacystin (an inhibitor of the ubiquitin-proteasome system (UPS)). RESULTS MPP(+) resulted in increased mRNA and protein levels of genes involved in cellular iron import and transport into the mitochondria. Similar changes occurred following exposure to paraquat, another inducer of oxidative stress. Lactacystin also resulted in increased TfR1 mRNA levels, although the other changes were not found. CONCLUSION Our results support the hypothesis of a functional mitochondrial iron deficit driving neuronal iron uptake but also suggest that differences exist in neuronal iron handling induced by different toxins.
Collapse
Affiliation(s)
- C B Carroll
- Department of Clinical Neurobiology, Peninsula College of Medicine and Dentistry, University of Plymouth, John Bull Building, Tamar Science Park, Plymouth, Devon, PL6 8BU, UK.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Brasse-Lagnel C, Karim Z, Letteron P, Bekri S, Bado A, Beaumont C. Intestinal DMT1 cotransporter is down-regulated by hepcidin via proteasome internalization and degradation. Gastroenterology 2011; 140:1261-1271.e1. [PMID: 21199652 DOI: 10.1053/j.gastro.2010.12.037] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 12/07/2010] [Accepted: 12/20/2010] [Indexed: 12/16/2022]
Abstract
BACKGROUNDS & AIMS The mechanism by which hepcidin regulates iron export from macrophages has been well established and is believed to involve degradation of ferroportin. However, in the small intestine, hepcidin's mechanisms of action are not known. We studied human polarized intestinal (Caco-2/TC7) cells and mouse duodenal segments, ex vivo, to investigate the molecular mechanisms by which hepcidin down-regulates intestinal transepithelial iron transport. METHODS Iron transport was analyzed using ⁵⁵FeNTA. Expression of Divalent Metal Transporter 1 (DMT1) and ferroportin was evaluated by reverse-transcription quantitative polymerase chain reaction and immunoblotting. Videomicroscopy analysis was performed on live cells that expressed either DMT1 or ferroportin fused to green fluorescent protein. RESULTS In Caco-2/TC7 cells, physiologic doses of hepcidin (50-1000 nmol/L) inhibited transport of ⁵⁵Fe in a dose-dependent manner; a half-maximum effect was observed at 75-100 nmol/L. However, 200 nmol/L hepcidin induced a significant decrease in DMT1 protein expression but no change in ferroportin protein levels, unlike macrophages. This result was confirmed ex vivo in isolated duodenal segments: 200 nmol/L hepcidin induced a significant reduction in iron transport and DMT1 protein levels but no change in ferroportin levels. In Caco-2/TC7 cells, the effect of hepcidin on the DMT1 protein level was completely abolished in the presence of a proteasome inhibitor (MG-132); DMT1 ubiquitination was induced by the addition of hepcidin. CONCLUSIONS An acute increase in hepcidin concentration reduces intestinal iron absorption through ubiquitin-dependent proteasome degradation of DMT1.
Collapse
Affiliation(s)
- Carole Brasse-Lagnel
- INSERM U773, Centre de Recherche Biomédicale Bichat Beaujon CRB3, Université Paris Diderot, site Bichat, Paris, France.
| | | | | | | | | | | |
Collapse
|
37
|
Recalcati S, Minotti G, Cairo G. Iron regulatory proteins: from molecular mechanisms to drug development. Antioxid Redox Signal 2010; 13:1593-616. [PMID: 20214491 DOI: 10.1089/ars.2009.2983] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Eukaryotic cells require iron for survival but, as an excess of poorly liganded iron can lead to the catalytic production of toxic radicals that can damage cell structures, regulatory mechanisms have been developed to maintain appropriate cell and body iron levels. The interactions of iron responsive elements (IREs) with iron regulatory proteins (IRPs) coordinately regulate the expression of the genes involved in iron uptake, use, storage, and export at the post-transcriptional level, and represent the main regulatory network controlling cell iron homeostasis. IRP1 and IRP2 are similar (but not identical) proteins with partially overlapping and complementary functions, and control cell iron metabolism by binding to IREs (i.e., conserved RNA stem-loops located in the untranslated regions of a dozen mRNAs directly or indirectly related to iron metabolism). The discovery of the presence of IREs in a number of other mRNAs has extended our knowledge of the influence of the IRE/IRP regulatory network to new metabolic pathways, and it has been recently learned that an increasing number of agents and physiopathological conditions impinge on the IRE/IRP system. This review focuses on recent findings concerning the IRP-mediated regulation of iron homeostasis, its alterations in disease, and new research directions to be explored in the near future.
Collapse
Affiliation(s)
- Stefania Recalcati
- Department of Human Morphology and Biomedical Sciences Città Studi, University of Milan, Milano, Italy
| | | | | |
Collapse
|
38
|
Intestinal ferritin H is required for an accurate control of iron absorption. Cell Metab 2010; 12:273-82. [PMID: 20816093 DOI: 10.1016/j.cmet.2010.08.003] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 05/08/2010] [Accepted: 06/16/2010] [Indexed: 12/26/2022]
Abstract
To maintain appropriate body iron levels, iron absorption by the proximal duodenum is thought to be controlled by hepcidin, a polypeptide secreted by hepatocytes in response to high serum iron. Hepcidin limits basolateral iron efflux from the duodenal epithelium by binding and downregulating the intestinal iron exporter ferroportin. Here, we found that mice with an intestinal ferritin H gene deletion show increased body iron stores and transferrin saturation. As expected for iron-loaded animals, the ferritin H-deleted mice showed induced liver hepcidin mRNA levels and reduced duodenal expression of DMT1 and DcytB mRNA. In spite of these feedback controls, intestinal ferroportin protein and (59)Fe absorption were increased more than 2-fold in the deleted mice. Our results demonstrate that hepcidin-mediated regulation alone is insufficient to restrict iron absorption and that intestinal ferritin H is also required to limit iron efflux from intestinal cells.
Collapse
|
39
|
Ferroportin and erythroid cells: an update. Adv Hematol 2010; 2010. [PMID: 20827391 PMCID: PMC2935194 DOI: 10.1155/2010/404173] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 04/08/2010] [Accepted: 06/23/2010] [Indexed: 12/21/2022] Open
Abstract
In recent years there have been major advances in our knowledge of the regulation of iron metabolism that have had implications for understanding the pathophysiology of some human disorders like beta-thalassemia and other iron overload diseases. However, little is known about the relationship among ineffective erythropoiesis, the role of iron-regulatory genes, and tissue iron distribution in beta-thalassemia. The principal aim of this paper is an update about the role of Ferroportin during human normal and pathological erythroid differentiation. Particular attention will be given to beta-thalassemia and other diseases with iron overload. Recent discoveries indicate that there is a potential for therapeutic intervention in beta-thalassemia by means of manipulating iron metabolism.
Collapse
|
40
|
Crosstalk between Iron Metabolism and Erythropoiesis. Adv Hematol 2010; 2010:605435. [PMID: 20631898 PMCID: PMC2902017 DOI: 10.1155/2010/605435] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 03/25/2010] [Indexed: 01/19/2023] Open
Abstract
Iron metabolism and erythropoiesis are inextricably linked. The majority of iron extracted from circulation daily is used for hemoglobin synthesis. In the last 15 years, major advances have been made in understanding the pathways regulating iron metabolism. Hepcidin is a key regulator of iron absorption and recycling and is itself regulated by erythropoiesis. While several viable candidates have been proposed, elucidating the “erythroid regulator” of hepcidin continues to generate significant experimental activity in the field. Although the mechanism responsible for sensing iron demand for erythropoiesis is still incompletely understood, evaluating diseases in which disordered erythropoiesis and/or iron metabolism are showcased has resulted in a more robust appreciation of potential candidates coordinated erythroid iron demand with regulators of iron supply. We present data drawn from four different conditions—iron deficiency, congenital hypotransferrinemia, beta-thalassemia, and hereditary hemochromatosis—both in human and non-human models of disease, together suggesting that erythroid iron demand exerts a stronger influence on circulating iron supply than systemic iron stores. Greater understanding of the interplay between the key factors involved in the regulation of iron metabolism and erythropoiesis will help develop more effective therapies for disorders of iron overload, iron deficiency, and hemoglobin synthesis.
Collapse
|
41
|
Wang H, Duan X, Liu J, Zhao H, Liu Y, Chang Y. Nitric Oxide contributes to the regulation of iron metabolism in skeletal muscle in vivo and in vitro. Mol Cell Biochem 2010; 342:87-94. [PMID: 20411304 DOI: 10.1007/s11010-010-0471-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 04/12/2010] [Indexed: 12/13/2022]
Abstract
Nitric Oxide (NO) plays an important role in iron redistribution during exercise, while its molecular regulatory mechanism is still not clear. Our present studies were to investigate the effects of NO on iron metabolism and to elucidate the regulatory mechanism of iron transport in skeletal muscle both in vivo and in vitro. One group of male Wistar rats (300 +/- 10 g) were subjected to an exercise of 30 min on a treadmill for 5 weeks (exercise group, EG, 6 rats) and the other one was placed on the treadmill without running (control group, CG, 6 rats). The cultured L6 rat skeletal muscle cells were treated with either 0.5 mM SNAP (NO donor) or not for 24 h, and their iron release and intake amount were examined by measuring radiolabelled (55)Fe. The results showed: (1) The NO content (CG, 1.09 +/- 0.18 micromol/g vs. EG, 1.49 +/- 0.17 micromol/g) and non-heme iron in gastrocnemius (CG, 118.35 +/- 11.41 microg/g vs. EG, 216.65 +/- 11.10 microg/g) of EG were significantly increased compared with CG. (2) The expression of DMT1 (IRE) and TfR1 of EG was increased. (3) The iron intake was increased in L6 cells treated with SNAP (P < 0.01). (4) Western blot results showed the protein level of both TfR1 and DMT1 (IRE) in SNAP cells were up-regulated, while the expression of FPN1 was down-regulated (P < 0.05). The data suggested that the induced elevation of NO level by exercise lead to the up-regulation of both TfR1 and DMT1 (IRE), which in turn increasing the iron absorption in skeletal muscle.
Collapse
Affiliation(s)
- Haitao Wang
- College of Physical Education, Hebei Normal University, Shijiazhuang, Hebei Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
42
|
Yin Z, Jiang H, Lee ESY, Ni M, Erikson KM, Milatovic D, Bowman AB, Aschner M. Ferroportin is a manganese-responsive protein that decreases manganese cytotoxicity and accumulation. J Neurochem 2009; 112:1190-8. [PMID: 20002294 DOI: 10.1111/j.1471-4159.2009.06534.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Although manganese (Mn) is an essential trace element for human development and growth, chronic exposure to excessive Mn levels can result in psychiatric and motor disturbances, referred to as manganism. However, there are no known mechanism(s) for efflux of excess Mn from mammalian cells. Here, we test the hypothesis that the cytoplasmic iron (Fe) exporter ferroportin (Fpn) may also function as a Mn exporter to attenuate Mn toxicity. Using an inducible human embryonic kidney (HEK293T) cell model, we examined the influence of Fpn expression on Mn-induced cytotoxicity and intracellular Mn concentrations. We found that induction of an Fpn-green fluorescent protein fusion protein in HEK293T cells was cytoprotective against several measures of Mn toxicity, including Mn-induced cell membrane leakage and Mn-induced reductions in glutamate uptake. Fpn-green fluorescent protein mediated cytoprotection correlated with decreased Mn accumulation following Mn exposure. Thus, Fpn expression reduces Mn toxicity concomitant with reduced Mn accumulation. To determine if mammalian cells may utilize Fpn in response to increased intracellular Mn concentrations and toxicity, we assessed endogenous Fpn levels in Mn-exposed HEK293T cells and in mouse brain in vivo. We find that 6 h of Mn exposure in HEK293T cells is associated with a significant increase in Fpn levels. Furthermore, mice exposed to Mn showed an increase in Fpn levels in both the cerebellum and cortex. Collectively, these results indicate that (i) Mn exposure promotes Fpn protein expression, (ii) Fpn expression reduces net Mn accumulation, and (iii) reduces cytotoxicity associated with exposure to this metal.
Collapse
Affiliation(s)
- Zhaobao Yin
- Departments of Pediatrics and Pharmacology, and Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Important advances in our understanding of iron metabolism have been made during the past 10 years, highlighting the mechanisms by which dysregulated iron homeostasis leads to hematologic, metabolic, and neurodegenerative diseases. In particular, the discovery of hepcidin and its fundamental role as the hormonal peptide regulating iron metabolism has delineated the organization of the complex network of proteins that regulates iron metabolism within the body. Maintenance of iron homeostasis is the consequence of tight coordination between iron absorption from the diet by enterocytes, and iron recycling by macrophages following degradation of senescent erythrocytes. Thus, any perturbation of these processes leads to a wide spectrum of diseases, ranging from iron deficiency anemia to iron overload. This review will focus particularly on the mechanisms involved in iron recycling by macrophages and summarize the pathological conditions perturbing this process.
Collapse
Affiliation(s)
- Carole Beaumont
- INSERM U773, Centre de Recherche Biomédicale Bichat Beaujon, Université Paris Diderot Paris, UFR de Médecine site Bichat, Paris, France.
| | | |
Collapse
|
44
|
Zhang DL, Hughes RM, Ollivierre-Wilson H, Ghosh MC, Rouault TA. A ferroportin transcript that lacks an iron-responsive element enables duodenal and erythroid precursor cells to evade translational repression. Cell Metab 2009; 9:461-73. [PMID: 19416716 PMCID: PMC2685206 DOI: 10.1016/j.cmet.2009.03.006] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2008] [Revised: 01/28/2009] [Accepted: 03/12/2009] [Indexed: 01/13/2023]
Abstract
Ferroportin (FPN1), the sole characterized mammalian iron exporter, has an iron-responsive element (IRE) in its 5' untranslated region, which ensures that its translation is repressed by iron regulatory proteins (IRPs) in iron-deficient conditions to maintain cellular iron content. However, here we demonstrate that duodenal epithelial and erythroid precursor cells utilize an alternative upstream promoter to express a FPN1 transcript, FPN1B, which lacks the IRE and is not repressed in iron-deficient conditions. The FPN1B transcript encodes ferroportin with an identical open reading frame and contributes significantly to ferroportin protein expression in erythroid precursors and likely also in the duodenum of iron-starved animals. The identification of FPN1B reveals how FPN1 expression can bypass IRP-dependent repression in intestinal iron uptake, even when cells throughout the body are iron deficient. In erythroid precursor cells, we hypothesize that FPN1B expression enhances real-time sensing of systemic iron status and facilitates restriction of erythropoiesis in response to low systemic iron.
Collapse
Affiliation(s)
- De-Liang Zhang
- Molecular Medicine Program, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
45
|
Hower V, Mendes P, Torti FM, Laubenbacher R, Akman S, Shulaev V, Torti SV. A general map of iron metabolism and tissue-specific subnetworks. MOLECULAR BIOSYSTEMS 2009; 5:422-43. [PMID: 19381358 PMCID: PMC2680238 DOI: 10.1039/b816714c] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Iron is required for survival of mammalian cells. Recently, understanding of iron metabolism and trafficking has increased dramatically, revealing a complex, interacting network largely unknown just a few years ago. This provides an excellent model for systems biology development and analysis. The first step in such an analysis is the construction of a structural network of iron metabolism, which we present here. This network was created using CellDesigner version 3.5.2 and includes reactions occurring in mammalian cells of numerous tissue types. The iron metabolic network contains 151 chemical species and 107 reactions and transport steps. Starting from this general model, we construct iron networks for specific tissues and cells that are fundamental to maintaining body iron homeostasis. We include subnetworks for cells of the intestine and liver, tissues important in iron uptake and storage, respectively, as well as the reticulocyte and macrophage, key cells in iron utilization and recycling. The addition of kinetic information to our structural network will permit the simulation of iron metabolism in different tissues as well as in health and disease.
Collapse
Affiliation(s)
- Valerie Hower
- Department of Cancer Biology, Wake Forest University School of Medicine, Medical Center Blvd, Winston Salem, NC 27157, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Aydemir F, Jenkitkasemwong S, Gulec S, Knutson MD. Iron loading increases ferroportin heterogeneous nuclear RNA and mRNA levels in murine J774 macrophages. J Nutr 2009; 139:434-8. [PMID: 19141705 PMCID: PMC2646225 DOI: 10.3945/jn.108.094052] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The transmembrane protein ferroportin is highly expressed in tissue macrophages, where it mediates iron export into the bloodstream. Although ferroportin expression can be controlled post-transcriptionally through a 5' iron-responsive element in its mRNA, various studies have documented increased ferroportin mRNA levels in response to iron, suggesting transcriptional regulation. We studied the effect of iron loading on levels of macrophage ferroportin mRNA, as well as heterogeneous nuclear RNA (hnRNA), the immediate product of ferroportin gene transcription. J774 cells, a mouse macrophage cell line, were incubated for 0, 3, 6, 9, 12, and 24 h in medium supplemented or not with 200 mumol/L iron. Quantitative RT-PCR was used to measure steady-state levels of ferroportin mRNA and hnRNA. Ferroportin mRNA levels increased by 12 h after iron treatment, reaching 6 times the control levels after 24 h. Changes in ferroportin mRNA levels were paralleled by similar changes in the levels of ferroportin hnRNA. Time course studies of ferroportin mRNA and hnRNA abundance after incubating cells with the transcriptional inhibitor actinomycin D revealed that ferroportin mRNA has a half-life of approximately 4 h and that iron loading does not stabilize ferroportin mRNA or hnRNA. Collectively, these data are consistent with the hypothesis that iron increases macrophage ferroportin mRNA levels by inducing transcription of the ferroportin gene.
Collapse
Affiliation(s)
- Fikret Aydemir
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | | | | | | |
Collapse
|
47
|
Park BY, Chung J. Effects of various metal ions on the gene expression of iron exporter ferroportin-1 in J774 macrophages. Nutr Res Pract 2008; 2:317-21. [PMID: 20016736 PMCID: PMC2788185 DOI: 10.4162/nrp.2008.2.4.317] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 11/07/2008] [Accepted: 11/28/2008] [Indexed: 01/11/2023] Open
Abstract
Macrophages play a key role in iron metabolism by recycling iron through erythrophagocytosis. Ferroportin-1 (FPN1) is a transporter protein that is known to mediate iron export from macrophages. Since divalent metals often interact with iron metabolism, we examined if divalent metals could regulate the expression of FPN1 in macrophages. J774 macrophage cells were treated with copper, manganese, zinc, or cobalt at 10, 50, or 100 microM for 16 to 24 h. Then, FPN1 mRNA and protein levels were determined by quantitative real-time PCR and Western blot analyses, respectively. In addition, effects of divalent metals on FPN1 promoter activity were examined by luciferase reporter assays. Results showed that copper significantly increased FPN1 mRNA levels in a dose-dependent manner. The copper-induced expression of FPN1 mRNA was associated with a corresponding increase in FPN1 protein levels. Also, copper directly stimulated the activity of FPN1 promoter-driven reporter construct. In contrast, manganese and zinc had no effect on the FPN1 gene expression in J774 cells. Interestingly, cobalt treatment in J774 cells decreased FPN1 protein levels without affecting FPN1 mRNA levels. In conclusion, our study results demonstrate that divalent metals differentially regulate FPN1 expression in macrophages and indicate a potential interaction of divalent metals with the FPN1-mediated iron export in macrophages.
Collapse
Affiliation(s)
- Bo-Yeon Park
- Department of Food & Nutrition, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Korea
| | | |
Collapse
|
48
|
Van Zandt KE, Sow FB, Florence WC, Zwilling BS, Satoskar AR, Schlesinger LS, Lafuse WP. The iron export protein ferroportin 1 is differentially expressed in mouse macrophage populations and is present in the mycobacterial-containing phagosome. J Leukoc Biol 2008; 84:689-700. [PMID: 18586980 DOI: 10.1189/jlb.1107781] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Intracellular pathogens, including Mycobacterium tuberculosis, obtain iron from the host for their survival. Ferroportin 1 (FPN1; SLC40A1) is the sole iron exporter from mammalian cells and is expressed in the duodenum and macrophages. In the present study, we show that FPN1 mRNA levels in the mouse macrophage cell line RAW264.7 are synergistically induced by treatment with live or gamma-irradiated M. tuberculosis and IFN-gamma. FPN1 mRNA levels were also induced by Mycobacterium avium and IFN-gamma in RAW264.7 cells and the mouse alveolar macrophage cell line AMJ2-C8. Treatment of mouse resident peritoneal macrophages with M. tuberculosis and IFN-gamma resulted in a sixfold increase in FPN1 mRNA expression. In contrast, M. tuberculosis and IFN-gamma inhibited FPN1 mRNA expression in bone marrow-derived macrophages and lung macrophages, which have high basal levels of FPN1 mRNA expression. Using confocal microscopy, FPN1 protein localized rapidly to M. tuberculosis phagosomes after infection in RAW264.7 macrophages. In RAW264.7 cells expressing wild-type natural resistance-associated macrophage protein 1 (Nramp1(Gly169)), FPN1 and Nramp1 partially colocalized in late endosomes/lysosomes prior to infection. After 2 h of infection, Nramp1 and FPN1 were present in M. tuberculosis phagosomes. Our studies provide evidence for transcriptional regulation of FPN1 by pathogenic mycobacteria and IFN-gamma, which is dependent on the macrophage type. The trafficking of FPN1 to the M. tuberculosis phagosome suggests that it is involved in regulating iron availability to the mycobacteria in this locale.
Collapse
Affiliation(s)
- Kristopher E Van Zandt
- Department of Microbiology, The Ohio State University, 333 W. 10th Ave., Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Sequential regulation of ferroportin expression after erythrophagocytosis in murine macrophages: early mRNA induction by haem, followed by iron-dependent protein expression. Biochem J 2008; 411:123-31. [PMID: 18072938 DOI: 10.1042/bj20071474] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tissue macrophages play an essential role in iron recycling through the phagocytosis of senescent RBCs (red blood cells). Following haem catabolism by HO1 (haem oxygenase 1), they recycle iron back into the plasma through the iron exporter Fpn (ferroportin). We previously described a cellular model of EP (erythrophagocytosis), based on primary cultures of mouse BMDMs (bone-marrow-derived macrophages) and aged murine RBCs, and showed that EP induces changes in the expression profiles of Fpn and HO1. In the present paper, we demonstrate that haem derived from human or murine RBCs or from an exogenous source of haem led to marked transcriptional activation of the Fpn and HO1 genes. Iron released from haem catabolism subsequently stimulated the Fpn mRNA and protein expression associated with localization of the transporter at the cell surface, which probably promotes the export of iron into the plasma. These findings highlight a dual mechanism of Fpn regulation in BMDMs, characterized by early induction of the gene transcription predominantly mediated by haem, followed by iron-mediated post-transcriptional regulation of the exporter.
Collapse
|
50
|
Chua ACG, Graham RM, Trinder D, Olynyk JK. The regulation of cellular iron metabolism. Crit Rev Clin Lab Sci 2008; 44:413-59. [PMID: 17943492 DOI: 10.1080/10408360701428257] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
While iron is an essential trace element required by nearly all living organisms, deficiencies or excesses can lead to pathological conditions such as iron deficiency anemia or hemochromatosis, respectively. A decade has passed since the discovery of the hemochromatosis gene, HFE, and our understanding of hereditary hemochromatosis (HH) and iron metabolism in health and a variety of diseases has progressed considerably. Although HFE-related hemochromatosis is the most widespread, other forms of HH have subsequently been identified. These forms are not attributed to mutations in the HFE gene but rather to mutations in genes involved in the transport, storage, and regulation of iron. This review is an overview of cellular iron metabolism and regulation, describing the function of key proteins involved in these processes, with particular emphasis on the liver's role in iron homeostasis, as it is the main target of iron deposition in pathological iron overload. Current knowledge on their roles in maintaining iron homeostasis and how their dysregulation leads to the pathogenesis of HH are discussed.
Collapse
Affiliation(s)
- Anita C G Chua
- School of Medicine and Pharmacology, University of Western Australia, Fremantle, Western Australia, Australia
| | | | | | | |
Collapse
|