1
|
Ding T, Shen W, Tao W, Peng J, Pan M, Qi X, Feng W, Wei N, Zheng S, Jin H. Curcumol ameliorates alcohol and high-fat diet-induced fatty liver disease via modulation of the Ceruloplasmin/iron overload/mtDNA signaling pathway. J Nutr Biochem 2025; 136:109807. [PMID: 39549858 DOI: 10.1016/j.jnutbio.2024.109807] [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: 07/17/2024] [Revised: 09/28/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
Fatty liver disease (FLD), a chronic liver disease characterized by excessive lipid deposition, is affecting more and more people worldwide owing to the increasing global incidence of obesity and heavy alcohol consumption. However, there is still no effective strategy for prevention or treatment of alcohol and high-fat diet (HFD)-induced FLD. The purpose of this study was to investigate the effect of curcumol on alcohol and HFD-induced FLD and the underlying molecular mechanisms. The results showed that curcumol ameliorated alcohol and HFD-induced hepatocyte injury in vivo and in vitro, and the mechanism might be related to its up-regulation of ceruloplasmin and subsequent alleviation of iron overload. Moreover, curcumol inhibited alcohol and HFD-induced mitochondrial damage and mtDNA release in hepatocytes by modulating iron overload. Furthermore, curcumol's inhibition of mtDNA release could suppress the activation of cGAS-STING and subsequent inflammation, and this phenomenon could be reversed by cGAS overexpression. Notably, alcohol and HFD-induced mtDNA release from hepatocytes contributed to HSC activation and this effect could be weakened by curcumol. In conclusion, these findings elucidated that curcumol ameliorated alcohol and HFD-induced FLD via modulating ceruloplasmin/iron overload/mtDNA signaling pathway, which lead to the inhibition of inflammation and HSCs activation.
Collapse
Affiliation(s)
- Tingting Ding
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Wanqing Shen
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Wenhui Tao
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Junlu Peng
- Department of Digestive surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Meijun Pan
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Xiaoyu Qi
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Wanyu Feng
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Na Wei
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
| | - Shuguo Zheng
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China; Laboratory of Pharmacology of Chinese Medicine, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China.
| | - Huanhuan Jin
- Department of Pharmacology, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China; Laboratory of Pharmacology of Chinese Medicine, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
2
|
Qiu L, Frazer DM, Hu M, Song R, Liu X, Qin X, Ma J, Zhou J, Tan Z, Ren F, Collins JF, Wang X. Mechanism and regulation of iron absorption throughout the life cycle. J Adv Res 2025:S2090-1232(25)00002-5. [PMID: 39814221 DOI: 10.1016/j.jare.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Iron plays a crucial role through various life stages of human. Iron homeostasis is primarily regulated by iron absorption which is mediated via divalent metal-ion transporter 1 (DMT1), and iron export protein ferroportin (FPN), as there is no active pathway for iron excretion from the body. Recent studies have shown that the magnitude of iron absorption changes through various life stages to meet changing iron requirements. AIM OF REVIEW This review aims to provide an overview of recent researches on the regulation of iron absorption throughout mammalian life cycle, with the potential to reveal novel molecules and pathways at special stage of life. Such insights may pave the way for new treatments for disorders associated with aberrant iron homeostasis in the future. KEY SCIENTIFIC CONCEPTS OF REVIEW This review first summarize the mechanism and regulation of iron absorption throughout various life stages, highlighting that regulatory mechanisms have developed to precisely align iron absorption to iron requirements. In adults, iron absorption is enhanced when body is deficient of iron, conversely, iron absorption is reduced when iron demand decreases via systemic regulator Hepcidin and cellular regulation. In the elderly, age-related inflammation, hormonal changes, and chronic diseases may affect the production of Hepcidin, affecting iron absorption. In infants, intestinal iron absorption and its regulatory mechanism are different from that in adults and there might be an alternative pathway independent of DMT1 and FPN due to high iron absorption. Unique to the fetus, iron is absorbed from maternal stores for its own use through the placenta and is regulated by maternal iron status. This review also proposes directions for further studies, offering promising avenues for developing new treatments for disorders associated with aberrant iron homeostasis.
Collapse
Affiliation(s)
- Lili Qiu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston 4029 Australia
| | - Mengxiao Hu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Rui Song
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Xiaoxue Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China
| | - Xiyu Qin
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Jie Ma
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Jun Zhou
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Zidi Tan
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China
| | - Fazheng Ren
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China
| | - James F Collins
- Food Science & Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | - Xiaoyu Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083 China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083 China.
| |
Collapse
|
3
|
Ferretti S, Zanella I. The Underestimated Role of Iron in Frontotemporal Dementia: A Narrative Review. Int J Mol Sci 2024; 25:12987. [PMID: 39684697 DOI: 10.3390/ijms252312987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The term frontotemporal dementia (FTD) comprises a group of neurodegenerative disorders characterized by the progressive degeneration of the frontal and temporal lobes of the brain with language impairment and changes in cognitive, behavioral and executive functions, and in some cases motor manifestations. A high proportion of FTD cases are due to genetic mutations and inherited in an autosomal-dominant manner with variable penetrance depending on the implicated gene. Iron is a crucial microelement that is involved in several cellular essential functions in the whole body and plays additional specialized roles in the central nervous system (CNS) mainly through its redox-cycling properties. Such a feature may be harmful under aerobic conditions, since it may lead to the generation of highly reactive hydroxyl radicals. Dysfunctions of iron homeostasis in the CNS are indeed involved in several neurodegenerative disorders, although it is still challenging to determine whether the dyshomeostasis of this essential but harmful metal is a direct cause of neurodegeneration, a contributor factor or simply a consequence of other neurodegenerative mechanisms. Unlike many other neurodegenerative disorders, evidence of the dysfunction in brain iron homeostasis in FTD is still scarce; nonetheless, the recent literature intriguingly suggests its possible involvement. The present review aims to summarize what is currently known about the contribution of iron dyshomeostasis in FTD based on clinical, imaging, histological, biochemical and molecular studies, further suggesting new perspectives and offering new insights for future investigations on this underexplored field of research.
Collapse
Affiliation(s)
- Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Medical Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
4
|
Pelucchi S, Risca G, Lanzafame C, Scollo CM, Garofalo A, Martinez D, Mariani R, Botti M, Capitoli G, Rossi F, Casati M, Piperno A, Galimberti S. Reference Values of Ceruloplasmin across the Adult Age Range in a Large Italian Healthy Population. J Appl Lab Med 2024; 9:1053-1063. [PMID: 39239918 DOI: 10.1093/jalm/jfae098] [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] [Received: 04/29/2024] [Accepted: 07/29/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Ceruloplasmin (Cp) is the most important serum copper transport protein playing a key role in the binding of iron to transferrin. It is a positive acute-phase response protein and the first-level diagnostic marker for Wilson disease and aceruloplasminemia. However, standardization of Cp measurement has not been successful, and assay specific reference levels of Cp are required. METHODS From May 2019 to July 2022, we enrolled 1706 consecutive healthy Italian blood donors (1285 men and 421 women, 18 to 65 years) to identify the reference intervals of serum Cp through quantile regression and evaluate the relationship of Cp with age, sex, iron, and metabolic status through linear regression. RESULTS We found that mean serum Cp was influenced by sex and slightly by age. The lower reference Cp value rose slightly with increasing age in both men and women. The upper reference value increased, reaching a plateau of about 25 mg/dL around 25 years in men, while in women it initially increased to around 45 mg/dL in young adults to fall sharply below 30 mg/dL for adults after their fifties. CONCLUSIONS We showed that the normal reference curves of serum Cp vary according to sex in a large population of healthy adults. While the lower reference values did not appear to be influenced by age and sex, the upper ones differed according to sex and age showing a particularly high variability in women, possibly reflecting different hormonal status.
Collapse
Affiliation(s)
- Sara Pelucchi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Giulia Risca
- Department of Medicine and Surgery, Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, University of Milan-Bicocca, Monza, Italy
| | - Corradina Lanzafame
- Immunotransfusional Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Andrea Garofalo
- Department of Laboratory Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Davide Martinez
- Department of Laboratory Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Raffaella Mariani
- SSD Rare Diseases-European Reference Network for Rare Hematological Diseases-EuroBloodNet-Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Mara Botti
- SSD Rare Diseases-European Reference Network for Rare Hematological Diseases-EuroBloodNet-Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Capitoli
- Department of Medicine and Surgery, Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, University of Milan-Bicocca, Monza, Italy
| | - Fabio Rossi
- Immunotransfusional Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Marco Casati
- Department of Laboratory Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Stefania Galimberti
- Department of Medicine and Surgery, Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, University of Milan-Bicocca, Monza, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| |
Collapse
|
5
|
Kopeć Z, Starzyński RR, Lenartowicz M, Grzesiak M, Opiela J, Smorąg Z, Gajda B, Nicpoń J, Ogłuszka M, Wang X, Mazgaj R, Stankiewicz A, Płonka W, Pirga-Niemiec N, Herman S, Lipiński P. Comparison of Molecular Potential for Iron Transfer across the Placenta in Domestic Pigs with Varied Litter Sizes and Wild Boars. Int J Mol Sci 2024; 25:9638. [PMID: 39273585 PMCID: PMC11395084 DOI: 10.3390/ijms25179638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Neonatal iron deficiency anemia is prevalent among domestic pigs but does not occur in the offspring of wild boar. The main causes of this disorder in piglets of modern pig breeds are paucity of hepatic iron stores, high birth weight, and rapid growth. Replenishment of fetal iron stores is a direct result of iron transfer efficiency across the placenta. In this study, we attempted to investigate the molecular potential of iron transfer across the placenta as a possible cause of differences between wild boar and Polish Large White (PLW) offspring. Furthermore, by analyzing placentas from PLW gilts that had litters of different sizes, we aimed to elucidate the impact of the number of fetuses on placental ability to transport iron. Using RNA sequencing, we examined the expression of iron-related genes in the placentas from wild boar and PLW gilts. We did not reveal significant differences in the expression of major iron transporters among all analyzed placentas. However, in wild boar placentas, we found higher expression of copper-dependent ferroxidases such as ceruloplasmin, zyklopen, and hephaestin, which facilitate iron export to the fetal circulation. We also determined a close co-localization of ceruloplasmin and zyklopen with ferroportin, the only iron exporter.
Collapse
Affiliation(s)
- Zuzanna Kopeć
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Rafał Radosław Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Małgorzata Lenartowicz
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, 31-007 Kraków, Poland
| | - Małgorzata Grzesiak
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Jagiellonian University, 31-007 Krakow, Poland
| | - Jolanta Opiela
- National Research Institute of Animal Production, 32-083 Balice, Poland
| | - Zdzisław Smorąg
- National Research Institute of Animal Production, 32-083 Balice, Poland
| | - Barbara Gajda
- National Research Institute of Animal Production, 32-083 Balice, Poland
| | - Jakub Nicpoń
- Department of Surgery, Faculty of Veterinary Sciences, Wrocław University of Environmental and Life Sciences, 50-375 Wrocław, Poland
| | - Magdalena Ogłuszka
- Department of Genomics, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Xiuying Wang
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Rafał Mazgaj
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Adrian Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Wiktoria Płonka
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, 31-007 Kraków, Poland
| | - Natalia Pirga-Niemiec
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, 31-007 Kraków, Poland
| | - Sylwia Herman
- Laboratory of Genetics and Evolution, Institute of Zoology and Biomedical Research, Jagiellonian University, 31-007 Kraków, Poland
| | - Paweł Lipiński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| |
Collapse
|
6
|
Zhang DD. Ironing out the details of ferroptosis. Nat Cell Biol 2024; 26:1386-1393. [PMID: 38429476 DOI: 10.1038/s41556-024-01361-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/22/2024] [Indexed: 03/03/2024]
Abstract
Ferroptosis, spurred by excess labile iron and lipid peroxidation, is implicated in various diseases. Advances have been made in comprehending the lipid-peroxidation side of ferroptosis, but the exact role of iron in driving ferroptosis remains unknown. Although iron overload is characterized in multiple disease states, the potential role of ferroptosis within them remains undefined. This overview focuses on the 'ferro' side of ferroptosis, highlighting iron dysregulation in human diseases and potential therapeutic strategies targeting iron regulation and metabolism.
Collapse
Affiliation(s)
- Donna D Zhang
- Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
| |
Collapse
|
7
|
Kharel Z, Kharel H, Phatak PD. Diagnosing aceruloplasminemia: navigating through red herrings. Ann Hematol 2024; 103:2173-2176. [PMID: 38637332 DOI: 10.1007/s00277-024-05743-7] [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: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
A 58-year-old female was found to have hyperferritinemia (Serum ferritin:1683 ng/mL) during work-up for mild normocytic anemia. Transferrin saturation(TSAT) was low-normal. Magnetic resonance imaging (MRI) abdomen showed evidence of hepatic iron deposition. Liver biopsy showed 4 + hepatic iron deposition without any evidence of steatosis or fibrosis. Quantitative liver iron was elevated at 348.3 µmol/g dry liver weight [Reference range(RR): 3-33 µmol/g dry liver weight]. She was presumptively diagnosed with tissue iron overload, cause uncertain. A diagnosis of ferroportin disease (FD) was considered, but the pattern of iron distribution in the liver, mainly within the hepatic parenchyma (rather than in the hepatic Kupffer cells seen in FD), and the presence of anemia (uncommon in FD) made this less likely. She was treated with intermittent phlebotomy for over a decade with poor tolerance due to worsening normocytic to microcytic anemia. A trial of deferasirox was done but it was discontinued after a month due to significant side effects. During the course of treatment, her ferritin level decreased. Over the past 1.5 years, she developed progressively worsening neurocognitive decline. MRI brain showed areas of susceptibility involving basal ganglia, midbrain and cerebellum raising suspicion for metabolic deposition disease. Neuroimaging findings led to testing for serum copper and ceruloplasmin levels which were both found to be severely low. Low serum copper, ceruloplasmin levels and neuroimaging findings led us to consider Wilson disease however prior liver biopsy showing elevated hepatic iron rather than hepatic copper excluded the diagnosis of Wilson disease. After shared decision making, ceruloplasmin gene analysis was not pursued due to patient's preference and prohibitive cost of testing. The diagnosis of aceruloplasminemia was ultimately made. The biochemical triad of hyperferritinemia, low-normal TSAT and microcytic anemia should raise the possibility of aceruloplasminemia. Since neurological manifestations are rare in most inherited iron overload syndromes, neurological symptoms in a patient with tissue iron overload should prompt consideration of aceruloplasminemia as a differential diagnosis.
Collapse
Affiliation(s)
- Zeni Kharel
- Division of Hematology/Oncology, Department of Medicine, Rochester General Hospital, Rochester, NY, USA.
| | - Himal Kharel
- Department of Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Pradyumna D Phatak
- Division of Hematology/Oncology, Department of Medicine, Rochester General Hospital, Rochester, NY, USA
| |
Collapse
|
8
|
Mannella V, Chaabane L, Canu T, Zanardi A, Raia S, Conti A, Ferrini B, Caricasole A, Musco G, Alessio M. Lipid dysmetabolism in ceruloplasmin-deficient mice revealed both in vivo and ex vivo by MRI, MRS and NMR analyses. FEBS Open Bio 2024; 14:258-275. [PMID: 37986139 PMCID: PMC10839333 DOI: 10.1002/2211-5463.13740] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023] Open
Abstract
Ceruloplasmin (Cp) is a ferroxidase that plays a role in cellular iron homeostasis and is mainly expressed in the liver and secreted into the blood. Cp is also produced by adipose tissue, which releases it as an adipokine. Although a dysfunctional interaction of iron with the metabolism of lipids has been associated with several metabolic diseases, the role of Cp in adipose tissue metabolism and in the interplay between hepatocytes and adipocytes has been poorly investigated. We previously found that Cp-deficient (CpKO) mice become overweight and demonstrate adipose tissue accumulation together with liver steatosis during aging, suggestive of lipid dysmetabolism. In the present study, we investigated the lipid alterations which occur during aging in adipose tissue and liver of CpKO and wild-type mice both in vivo and ex vivo. During aging of CpKO mice, we observed adipose tissue accumulation and liver lipid deposition, both of which are associated with macrophage infiltration. Liver lipid deposition was characterized by accumulation of triglycerides, fatty acids and ω-3 fatty acids, as well as by a switch from unsaturated to saturated fatty acids, which is characteristic of lipid storage. Liver steatosis was preceded by iron deposition and macrophage infiltration, and this was observed to be already occurring in younger CpKO mice. The accumulation of ω-3 fatty acids, which can only be acquired through diet, was associated with body weight increase in CpKO mice despite food intake being equal to that of wild-type mice, thus underlining the alterations in lipid metabolism/catabolism in Cp-deficient animals.
Collapse
Affiliation(s)
- Valeria Mannella
- COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Linda Chaabane
- Preclinical Imaging, Experimental Imaging CentreIRCCS‐San Raffaele HospitalMilanoItaly
- Present address:
LC, Euro‐BioImaging ERIC, Med‐Hub section, Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TorinoItaly
- Present address:
SR, Deloitte & Touche SpAMilanoItaly
| | - Tamara Canu
- Preclinical Imaging, Experimental Imaging CentreIRCCS‐San Raffaele HospitalMilanoItaly
| | - Alan Zanardi
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Sara Raia
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
- Present address:
LC, Euro‐BioImaging ERIC, Med‐Hub section, Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TorinoItaly
- Present address:
SR, Deloitte & Touche SpAMilanoItaly
| | - Antonio Conti
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Barbara Ferrini
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| | - Andrea Caricasole
- Department of Research & Innovation, Kedrion S.p.A.Loc BolognanaGallicanoItaly
| | - Giovanna Musco
- Biomolecular Nuclear Magnetic Resonance, Division of Genetics and Cell BiologyIRCCS‐San Raffaele HospitalMilanoItaly
| | - Massimo Alessio
- Proteome Biochemistry, COSR‐Centre for Omics SciencesIRCCS‐San Raffaele HospitalMilanoItaly
| |
Collapse
|
9
|
Pan S, Hale AT, Lemieux ME, Raval DK, Garton TP, Sadler B, Mahaney KB, Strahle JM. Iron homeostasis and post-hemorrhagic hydrocephalus: a review. Front Neurol 2024; 14:1287559. [PMID: 38283681 PMCID: PMC10811254 DOI: 10.3389/fneur.2023.1287559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/21/2023] [Indexed: 01/30/2024] Open
Abstract
Iron physiology is regulated by a complex interplay of extracellular transport systems, coordinated transcriptional responses, and iron efflux mechanisms. Dysregulation of iron metabolism can result in defects in myelination, neurotransmitter synthesis, and neuronal maturation. In neonates, germinal matrix-intraventricular hemorrhage (GMH-IVH) causes iron overload as a result of blood breakdown in the ventricles and brain parenchyma which can lead to post-hemorrhagic hydrocephalus (PHH). However, the precise mechanisms by which GMH-IVH results in PHH remain elusive. Understanding the molecular determinants of iron homeostasis in the developing brain may lead to improved therapies. This manuscript reviews the various roles iron has in brain development, characterizes our understanding of iron transport in the developing brain, and describes potential mechanisms by which iron overload may cause PHH and brain injury. We also review novel preclinical treatments for IVH that specifically target iron. Understanding iron handling within the brain and central nervous system may provide a basis for preventative, targeted treatments for iron-mediated pathogenesis of GMH-IVH and PHH.
Collapse
Affiliation(s)
- Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew T. Hale
- Department of Neurosurgery, University of Alabama at Birmingham School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mackenzie E. Lemieux
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Dhvanii K. Raval
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Thomas P. Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Brooke Sadler
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Hematology and Oncology, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Kelly B. Mahaney
- Department of Neurosurgery, Stanford University School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pediatrics, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
10
|
Jormakka M. Structural insights into ferroportin mediated iron transport. Biochem Soc Trans 2023; 51:BST20230594. [PMID: 38115725 DOI: 10.1042/bst20230594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
Iron is a vital trace element for almost all organisms, and maintaining iron homeostasis is critical for human health. In mammals, the only known gatekeeper between intestinally absorbed iron and circulatory blood plasma is the membrane transporter ferroportin (Fpn). As such, dysfunction of Fpn or its regulation is a key driver of iron-related pathophysiology. This review focuses on discussing recent insights from high-resolution structural studies of the Fpn protein family. While these studies have unveiled crucial details of Fpn regulation and structural architecture, the associated functional studies have also at times provided conflicting data provoking more questions than answers. Here, we summarize key findings and illuminate important remaining questions and contradictions.
Collapse
Affiliation(s)
- Mika Jormakka
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
Srivastava NK, Mukherjee S, Mishra VN. One advantageous reflection of iron metabolism in context of normal physiology and pathological phases. Clin Nutr ESPEN 2023; 58:277-294. [PMID: 38057018 DOI: 10.1016/j.clnesp.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/05/2023] [Accepted: 10/09/2023] [Indexed: 12/08/2023]
Abstract
PURPOSE (BACKGROUND) The presented review is an updating of Iron metabolism in context of normal physiology and pathological phases. Iron is one of the vital elements in humans and associated into proteins as a component of heme (e.g. hemoglobin, myoglobin, cytochromes proteins, myeloperoxidase, nitric oxide synthetases), iron sulfur clusters (e.g. respiratory complexes I-III, coenzyme Q10, mitochondrial aconitase, DNA primase), or other functional groups (e.g. hypoxia inducible factor prolyl hydroxylases). All these entire iron-containing proteins ar e needed for vital cellular and organismal functions together with oxygen transport, mitochondrial respiration, intermediary and xenobiotic metabolism, nucleic acid replication and repair, host defense, and cell signaling. METHODS (METABOLIC STRATEGIES) Cells have developed metabolic strategies to import and employ iron safely. Regulatory process of iron uptake, storage, intracellular trafficking and utilization is vital for the maintenance of cellular iron homeostasis. Cellular iron utilization and intracellular iron trafficking pathways are not well established and very little knowledge about this. The predominant organs, which are associated in the metabolism of iron, are intestine, liver, bone marrow and spleen. Iron is conserved, recycled and stored. The reduced bioavailability of iron in humans has developed extremely efficient mechanisms for iron conservation. Prominently, the losses of iron cannot considerably enhance through physiologic mechanisms, even if iron intake and stores become excessive. Loss of iron is balanced or maintained from dietary sources. RESULTS (OUTCOMES) Numerous physiological abnormalities are associated with impaired iron metabolism. These abnormalities are appeared in the form of several diseases. There are duodenal ulcer, inflammatory bowel disease, sideroblastic anaemia, congenital dyserythropoietic anemias and low-grade myelodysplastic syndromes. Hereditary hemochromatosis and anaemia are two chronic diseases, which are responsible for disturbing the iron metabolism in various tissues, including the spleen and the intestine. Impairment in hepatic hepcidin synthesis is responsible for chronic liver disease, which is grounding from alcoholism or viral hepatitis. This condition directs to iron overload that can cause further hepatic damage. Iron has important role in several infectious diseases are tuberculosis, malaria trypanosomatid diseases and acquired immunodeficiency syndrome (AIDS). Iron is also associated with Systemic lupus erythematosus [SLE], cancer, Alzheimer's disease (AD) and post-traumatic epilepsy. CONCLUSION Recently, numerous research studies are gradually more dedicated in the field of iron metabolism, but a number of burning questions are still waiting for answer. Cellular iron utilization and intracellular iron trafficking pathways are not well established and very little knowledge about this. Increased information of the physiology of iron homeostasis will support considerate of the pathology of iron disorders and also make available the support to advance treatment.
Collapse
Affiliation(s)
- Niraj Kumar Srivastava
- School of Sciences (SOS), Indira Gandhi National Open University (IGNOU), New Delhi, 110068, India.
| | | | - Vijaya Nath Mishra
- Department of Neurology, Institute of Medical Sciences (IMS), Banaras Hindu University (BHU), Varanasi, 221005, UP, India
| |
Collapse
|
12
|
Shan C, Liang Y, Wang K, Li P. Noncoding RNAs in cancer ferroptosis: From biology to clinical opportunity. Biomed Pharmacother 2023; 165:115053. [PMID: 37379641 DOI: 10.1016/j.biopha.2023.115053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
Ferroptosis is a recently discovered pattern of programmed cell death that is nonapoptotic and irondependent. It is involved in lipid peroxidation dependent on reactive oxygen species. Ferroptosis has been verified to play a crucial regulatory role in a variety of pathological courses of disease, in particularly cancer. Emerging research has highlighted the potential of ferroptosis in tumorigenesis, cancer development and resistance to chemotherapy. However, the regulatory mechanism of ferroptosis remains unclear, which limits the application of ferroptosis in cancer treatment. Noncoding RNAs (ncRNAs) are noncoding transcripts that regulate gene expression in various ways to affect the malignant phenotypes of cancer cells. At present, the biological function and underlying regulatory mechanism of ncRNAs in cancer ferroptosis have been partially elucidated. Herein, we summarize the current knowledge of the central regulatory network of ferroptosis, with a focus on the regulatory functions of ncRNAs in cancer ferroptosis. The clinical application and prospects of ferroptosis-related ncRNAs in cancer diagnosis, prognosis and anticancer therapies are also discussed. Elucidating the function and mechanism of ncRNAs in ferroptosis, along with assessing the clinical significance of ferroptosis-related ncRNAs, provides new perspectives for understanding cancer biology and treatment approaches, which may benefit numerous cancer patients in the future.
Collapse
Affiliation(s)
- Chan Shan
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Kun Wang
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
13
|
Sangkhae V, Fisher AL, Ganz T, Nemeth E. Iron Homeostasis During Pregnancy: Maternal, Placental, and Fetal Regulatory Mechanisms. Annu Rev Nutr 2023; 43:279-300. [PMID: 37253681 PMCID: PMC10723031 DOI: 10.1146/annurev-nutr-061021-030404] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Pregnancy entails a large negative balance of iron, an essential micronutrient. During pregnancy, iron requirements increase substantially to support both maternal red blood cell expansion and the development of the placenta and fetus. As insufficient iron has long been linked to adverse pregnancy outcomes, universal iron supplementation is common practice before and during pregnancy. However, in high-resource countries with iron fortification of staple foods and increased red meat consumption, the effects of too much iron supplementation during pregnancy have become a concern because iron excess has also been linked to adverse pregnancy outcomes. In this review, we address physiologic iron homeostasis of the mother, placenta, and fetus and discuss perturbations in iron homeostasis that result in pathological pregnancy. As many mechanistic regulatory systems have been deduced from animal models, we also discuss the principles learned from these models and how these may apply to human pregnancy.
Collapse
Affiliation(s)
- Veena Sangkhae
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| | - Allison L Fisher
- Endocrine Unit and Nephrology Division, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas Ganz
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| | - Elizabeta Nemeth
- Center for Iron Disorders, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA;
| |
Collapse
|
14
|
Anderson BD, Lee T, Bell B, Song Y, Dunaief JL. Low ceruloplasmin levels exacerbate retinal degeneration in a hereditary hemochromatosis model. Dis Model Mech 2023; 16:dmm050226. [PMID: 37439255 PMCID: PMC10354715 DOI: 10.1242/dmm.050226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/09/2023] [Indexed: 07/14/2023] Open
Abstract
In a previous report, a 39-year-old patient with high serum iron levels from hereditary hemochromatosis (HH) was diagnosed with a form of retinal degeneration called bull's eye maculopathy. This is atypical for patients with HH, so it was theorized that the low serum levels of ferroxidase ceruloplasmin (CP) of this patient coupled with the high iron levels led to the retinal degeneration. CP, by oxidizing iron from its ferrous to ferric form, helps prevent the oxidative damage caused by ferrous iron. To test this, a hepcidin knockout (KO) mouse model of HH was combined with Cp KO to test whether the combination would lead to more severe retinal degeneration. Monthly in vivo retinal images were acquired and, after 11 months, mice were euthanized for further analyses. Both heterozygous and homozygous Cp KO increased the rate and severity of retinal degeneration. These results demonstrate the protective role of CP, which is most likely owing to its ferroxidase activity. The findings suggest that CP levels may influence the severity of retinal degeneration, especially in individuals with high serum iron.
Collapse
Affiliation(s)
- Brandon D. Anderson
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Timothy Lee
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Brent Bell
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ying Song
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joshua L. Dunaief
- FM Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
15
|
Helman SL, Zhou J, Fuqua BK, Lu Y, Collins JF, Chen H, Vulpe CD, Anderson GJ, Frazer DM. The biology of mammalian multi-copper ferroxidases. Biometals 2023; 36:263-281. [PMID: 35167013 PMCID: PMC9376197 DOI: 10.1007/s10534-022-00370-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/04/2022] [Indexed: 12/24/2022]
Abstract
The mammalian multicopper ferroxidases (MCFs) ceruloplasmin (CP), hephaestin (HEPH) and zyklopen (ZP) comprise a family of conserved enzymes that are essential for body iron homeostasis. Each of these enzymes contains six biosynthetically incorporated copper atoms which act as intermediate electron acceptors, and the oxidation of iron is associated with the four electron reduction of dioxygen to generate two water molecules. CP occurs in both a secreted and GPI-linked (membrane-bound) form, while HEPH and ZP each contain a single C-terminal transmembrane domain. These enzymes function to ensure the efficient oxidation of iron so that it can be effectively released from tissues via the iron export protein ferroportin and subsequently bound to the iron carrier protein transferrin in the blood. CP is particularly important in facilitating iron release from the liver and central nervous system, HEPH is the major MCF in the small intestine and is critical for dietary iron absorption, and ZP is important for normal hair development. CP and HEPH (and possibly ZP) function in multiple tissues. These proteins also play other (non-iron-related) physiological roles, but many of these are ill-defined. In addition to disrupting iron homeostasis, MCF dysfunction perturbs neurological and immune function, alters cancer susceptibility, and causes hair loss, but, despite their importance, how MCFs co-ordinately maintain body iron homeostasis and perform other functions remains incompletely understood.
Collapse
Affiliation(s)
- Sheridan L Helman
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jie Zhou
- Department of Physiological Sciences, University of Florida, Gainsville, FL, USA
| | - Brie K Fuqua
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yan Lu
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia
- Mucosal Immunology Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James F Collins
- Food Science and Human Nutrition Department, University of Florida, Gainsville, FL, USA
| | - Huijun Chen
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Christopher D Vulpe
- Department of Physiological Sciences, University of Florida, Gainsville, FL, USA
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, 300 Herston Road, Brisbane, QLD, 4006, Australia.
- School of Chemistry and Molecular Bioscience, University of Queensland, Brisbane, Australia.
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
16
|
Yu L, Yousuf S, Yousuf S, Yeh J, Biggins SW, Morishima C, Shyu I, O’Shea-Stone G, Eilers B, Waldum A, Copié V, Burkhead J. Copper deficiency is an independent risk factor for mortality in patients with advanced liver disease. Hepatol Commun 2023; 7:e0076. [PMID: 36809345 PMCID: PMC9949837 DOI: 10.1097/hc9.0000000000000076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 11/17/2022] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND AND AIM Copper is an essential trace metal serving as a cofactor in innate immunity, metabolism, and iron transport. We hypothesize that copper deficiency may influence survival in patients with cirrhosis through these pathways. METHODS We performed a retrospective cohort study involving 183 consecutive patients with cirrhosis or portal hypertension. Copper from blood and liver tissues was measured using inductively coupled plasma mass spectrometry. Polar metabolites were measured using nuclear magnetic resonance spectroscopy. Copper deficiency was defined by serum or plasma copper below 80 µg/dL for women or 70 µg/dL for men. RESULTS The prevalence of copper deficiency was 17% (N=31). Copper deficiency was associated with younger age, race, zinc and selenium deficiency, and higher infection rates (42% vs. 20%, p=0.01). Serum copper correlated positively with albumin, ceruloplasmin, hepatic copper, and negatively with IL-1β. Levels of polar metabolites involved in amino acids catabolism, mitochondrial transport of fatty acids, and gut microbial metabolism differed significantly according to copper deficiency status. During a median follow-up of 396 days, mortality was 22.6% in patients with copper deficiency compared with 10.5% in patients without. Liver transplantation rates were similar (32% vs. 30%). Cause-specific competing risk analysis showed that copper deficiency was associated with a significantly higher risk of death before transplantation after adjusting for age, sex, MELD-Na, and Karnofsky score (HR: 3.40, 95% CI, 1.18-9.82, p=0.023). CONCLUSIONS In advanced cirrhosis, copper deficiency is relatively common and is associated with an increased infection risk, a distinctive metabolic profile, and an increased risk of death before transplantation.
Collapse
Affiliation(s)
- Lei Yu
- Department of Medicine, Division of Gastroenterology and Center for Liver Investigation Fostering Discovery, University of Washington, Seattle, Washington, USA
| | - Sarim Yousuf
- College of Medicine, Ziauddin University, Karachi, Pakistan
| | - Shahrukh Yousuf
- Department of Medicine, Division of Gastroenterology and Center for Liver Investigation Fostering Discovery, University of Washington, Seattle, Washington, USA
| | - Jeffrey Yeh
- Department of Medicine, Division of Gastroenterology and Center for Liver Investigation Fostering Discovery, University of Washington, Seattle, Washington, USA
| | - Scott W. Biggins
- Department of Medicine, Division of Gastroenterology and Center for Liver Investigation Fostering Discovery, University of Washington, Seattle, Washington, USA
| | - Chihiro Morishima
- Department of Lab Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Irene Shyu
- Department of Lab Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Galen O’Shea-Stone
- Department of Chemistry, Montana State University, Bozeman, Montana, USA
| | - Brian Eilers
- Department of Chemistry, Montana State University, Bozeman, Montana, USA
| | - Annie Waldum
- Department of Chemistry, Montana State University, Bozeman, Montana, USA
| | - Valérie Copié
- Department of Chemistry, Montana State University, Bozeman, Montana, USA
| | - Jason Burkhead
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, Alaska, USA
| |
Collapse
|
17
|
Hereditary Hyperferritinemia. Int J Mol Sci 2023; 24:ijms24032560. [PMID: 36768886 PMCID: PMC9917042 DOI: 10.3390/ijms24032560] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/26/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Ferritin is a ubiquitous protein that is present in most tissues as a cytosolic protein. The major and common role of ferritin is to bind Fe2+, oxidize it and sequester it in a safe form in the cell, and to release iron according to cellular needs. Ferritin is also present at a considerably low proportion in normal mammalian sera and is relatively iron poor compared to tissues. Serum ferritin might provide a useful and convenient method of assessing the status of iron storage, and its measurement has become a routine laboratory test. However, many additional factors, including inflammation, infection, metabolic abnormalities, and malignancy-all of which may elevate serum ferritin-complicate interpretation of this value. Despite this long history of clinical use, fundamental aspects of the biology of serum ferritin are still unclear. According to the high number of factors involved in regulation of ferritin synthesis, secretion, and uptake, and in its central role in iron metabolism, hyperferritinemia is a relatively common finding in clinical practice and is found in a large spectrum of conditions, both genetic and acquired, associated or not with iron overload. The diagnostic strategy to reveal the cause of hyperferritinemia includes family and personal medical history, biochemical and genetic tests, and evaluation of liver iron by direct or indirect methods. This review is focused on the forms of inherited hyperferritinemia with or without iron overload presenting with normal transferrin saturation, as well as a step-by-step approach to distinguish these forms to the acquired forms, common and rare, of isolated hyperferritinemia.
Collapse
|
18
|
Multicopper oxidases with laccase-ferroxidase activity: Classification and study of ferroxidase activity determinants in a member from Heterobasidion annosum s. l.. Comput Struct Biotechnol J 2023; 21:1041-1053. [PMID: 36733701 PMCID: PMC9880977 DOI: 10.1016/j.csbj.2023.01.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/24/2023] Open
Abstract
Multi-copper oxidases (MCO) share a common molecular architecture and the use of copper ions as cofactors to reduce O2 to H2O, but show high sequence heterogeneity and functional diversity. Many new emerging MCO genes are wrongly annotated as laccases, the largest group of MCOs, with the widest range of biotechnological applications (particularly those from basidiomycete fungi) due to their ability to oxidise aromatic compounds and lignin. Thus, comprehensive studies for a better classification and structure-function characterisation of MCO families are required. Laccase-ferroxidases (LAC-FOXs) constitute a separate and unexplored group of MCOs with proposed dual features between laccases and ferroxidases. We aim to better define this cluster and the structural determinants underlying putative hybrid activity. We performed a phylogenetic analysis of the LAC-FOXs from basidiomycete fungi, that resulted in two subgroups. This division seemed to correlate with the presence or absence of some of the three acidic residues responsible for ferroxidase activity in Fet3p from Saccharomyces cerevisiae. One of these LAC-FOXs (with only one of these residues) from the fungus Heterobasidion annosum s. l. (HaLF) was synthesised, heterologously expressed and characterised to evaluate its catalytic activity. HaLF oxidised typical laccase substrates (phenols, aryl amines and N-heterocycles), but no Fe (II). The enzyme was subjected to site-directed mutagenesis to determine the key residues that confer ferroxidase activity. The mutated HaLF variant with full restoration of the three acidic residues exhibited efficient ferroxidase activity, while it partially retained the wide-range oxidative activity of the native enzyme associated to laccases sensu stricto.
Collapse
|
19
|
Thepsuwan P, Bhattacharya A, Song Z, Hippleheuser S, Feng S, Wei X, Das NK, Sierra M, Wei J, Fang D, Huang YMM, Zhang K, Shah YM, Sun S. Hepatic SEL1L-HRD1 ER-associated degradation regulates systemic iron homeostasis via ceruloplasmin. Proc Natl Acad Sci U S A 2023; 120:e2212644120. [PMID: 36595688 PMCID: PMC9926173 DOI: 10.1073/pnas.2212644120] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/18/2022] [Indexed: 01/05/2023] Open
Abstract
Iron homeostasis is critical for cellular and organismal function and is tightly regulated to prevent toxicity or anemia due to iron excess or deficiency, respectively. However, subcellular regulatory mechanisms of iron remain largely unexplored. Here, we report that SEL1L-HRD1 protein complex of endoplasmic reticulum (ER)-associated degradation (ERAD) in hepatocytes controls systemic iron homeostasis in a ceruloplasmin (CP)-dependent, and ER stress-independent, manner. Mice with hepatocyte-specific Sel1L deficiency exhibit altered basal iron homeostasis and are sensitized to iron deficiency while resistant to iron overload. Proteomics screening for a factor linking ERAD deficiency to altered iron homeostasis identifies CP, a key ferroxidase involved in systemic iron distribution by catalyzing iron oxidation and efflux from tissues. Indeed, CP is highly unstable and a bona fide substrate of SEL1L-HRD1 ERAD. In the absence of ERAD, CP protein accumulates in the ER and is shunted to refolding, leading to elevated secretion. Providing clinical relevance of these findings, SEL1L-HRD1 ERAD is responsible for the degradation of a subset of disease-causing CP mutants, thereby attenuating their pathogenicity. Together, this study uncovers the role of SEL1L-HRD1 ERAD in systemic iron homeostasis and provides insights into protein misfolding-associated proteotoxicity.
Collapse
Affiliation(s)
- Pattaraporn Thepsuwan
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Asmita Bhattacharya
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48105
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Stephen Hippleheuser
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Shaobin Feng
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Xiaoqiong Wei
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48105
| | - Nupur K. Das
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48105
| | - Mariana Sierra
- Department of Physics and Astronomy, Wayne State University, Detroit, MI48201
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Yu-ming M. Huang
- Department of Physics and Astronomy, Wayne State University, Detroit, MI48201
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI48201
| | - Yatrik M. Shah
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48105
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI48109
| | - Shengyi Sun
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI48201
| |
Collapse
|
20
|
Raia S, Conti A, Zanardi A, Ferrini B, Scotti GM, Gilberti E, De Palma G, David S, Alessio M. Ceruloplasmin-Deficient Mice Show Dysregulation of Lipid Metabolism in Liver and Adipose Tissue Reduced by a Protein Replacement. Int J Mol Sci 2023; 24:ijms24021150. [PMID: 36674661 PMCID: PMC9863737 DOI: 10.3390/ijms24021150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
Ceruloplasmin is a ferroxidase that plays a role in iron homeostasis; its deficiency fosters inter alia iron accumulation in the liver, which expresses the soluble form of the protein secreted into the bloodstream. Ceruloplasmin is also secreted by the adipose tissue, but its role in adipocytes has been poorly investigated. We hypothesized that ceruloplasmin might have a role in iron/lipid interplay. We investigated iron/lipid dysmetabolism in the liver and adipose tissue of the ceruloplasmin-deficient mouse (CpKO) model of aceruloplasminemia and evaluated the effectiveness of ceruloplasmin replacement. We found that CpKO mice were overweight, showing adipose tissue accumulation, liver iron deposition and steatosis. In the adipose tissue of CpKO mice, iron homeostasis was not altered. Conversely, the levels of adiponectin and leptin adipokines behaved opposite to the wild-type. Increased macrophage infiltration was observed in adipose tissue and liver of CpKO mice, indicating tissue inflammation. The treatment of CpKO mice with ceruloplasmin limited liver iron accumulation and steatosis without normalizing the expression of iron homeostasis-related proteins. In the CpKO mice, the protein replacement limited macrophage infiltration in both adipose and hepatic tissues reduced the level of serum triglycerides, and partially recovered adipokines levels in the adipose tissue. These results underline the link between iron and lipid dysmetabolism in ceruloplasmin-deficient mice, suggesting that ceruloplasmin in adipose tissue has an anti-inflammatory role rather than a role in iron homeostasis. Furthermore, these data also indicate that ceruloplasmin replacement therapy may be effective at a systemic level.
Collapse
Affiliation(s)
- Sara Raia
- Proteome Biochemistry, COSR-Centre for Omics Sciences, IRCCS-San Raffaele Hospital, 20132 Milan, Italy
| | - Antonio Conti
- Proteome Biochemistry, COSR-Centre for Omics Sciences, IRCCS-San Raffaele Hospital, 20132 Milan, Italy
| | - Alan Zanardi
- Proteome Biochemistry, COSR-Centre for Omics Sciences, IRCCS-San Raffaele Hospital, 20132 Milan, Italy
| | - Barbara Ferrini
- Proteome Biochemistry, COSR-Centre for Omics Sciences, IRCCS-San Raffaele Hospital, 20132 Milan, Italy
| | - Giulia Maria Scotti
- COSR-Centre for Omics Sciences, IRCCS-San Raffaele Hospital, 20132 Milan, Italy
| | - Enrica Gilberti
- Unit of Occupational Health and Industrial Hygiene, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Giuseppe De Palma
- Unit of Occupational Health and Industrial Hygiene, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Samuel David
- Center for Research in Neuroscience, The Research Institute of The McGill University Health Center, Montreal, QC H3G 1A4, Canada
| | - Massimo Alessio
- Proteome Biochemistry, COSR-Centre for Omics Sciences, IRCCS-San Raffaele Hospital, 20132 Milan, Italy
- Correspondence:
| |
Collapse
|
21
|
Schilsky ML, Roberts EA, Bronstein JM, Dhawan A, Hamilton JP, Rivard AM, Washington MK, Weiss KH, Zimbrean PC. A multidisciplinary approach to the diagnosis and management of Wilson disease: 2022 Practice Guidance on Wilson disease from the American Association for the Study of Liver Diseases. Hepatology 2022:01515467-990000000-00207. [PMID: 36151586 DOI: 10.1002/hep.32801] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Michael L Schilsky
- Medicine and Surgery , Yale University School of Medicine , New Haven , Connecticut , USA
| | - Eve A Roberts
- Paediatrics, Medicine, Pharmacology and Toxicology , University of Toronto , Toronto , Ontario , Canada
| | - Jeff M Bronstein
- Neurology , University of California Los Angeles , Los Angeles , California , USA
| | - Anil Dhawan
- Paediatric Liver, GI and Nutrition Centre and MowatLabs , King's College Hospital , London , UK
| | - James P Hamilton
- Medicine , Johns Hopkins University School of Medicine , Baltimore , Maryland , USA
| | - Anne Marie Rivard
- Food and Nutrition Services , Yale New Haven Hospital , New Haven , Connecticut , USA
| | - Mary Kay Washington
- Pathology, Immunology and Microbiology , Vanderbilt University Medical Center , Nashville , Tennessee , USA
| | | | - Paula C Zimbrean
- Psychiatry , Yale University School of Medicine , New Haven , Connecticut , USA
| |
Collapse
|
22
|
Iron Metabolism and Ferroptosis in Physiological and Pathological Pregnancy. Int J Mol Sci 2022; 23:ijms23169395. [PMID: 36012659 PMCID: PMC9409111 DOI: 10.3390/ijms23169395] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/13/2022] Open
Abstract
Iron is a vital element in nearly every living organism. During pregnancy, optimal iron concentration is essential for both maternal health and fetal development. As the barrier between the mother and fetus, placenta plays a pivotal role in mediating and regulating iron transport. Imbalances in iron metabolism correlate with severe adverse pregnancy outcomes. Like most other nutrients, iron exhibits a U-shaped risk curve. Apart from iron deficiency, iron overload is also dangerous since labile iron can generate reactive oxygen species, which leads to oxidative stress and activates ferroptosis. In this review, we summarized the molecular mechanism and regulation signals of placental iron trafficking under physiological conditions. In addition, we revealed the role of iron metabolism and ferroptosis in the view of preeclampsia and gestational diabetes mellitus, which may bring new insight to the pathogenesis and treatment of pregnancy-related diseases.
Collapse
|
23
|
Zaugg J, Solenthaler F, Albrecht C. Materno-fetal iron transfer and the emerging role of ferroptosis pathways. Biochem Pharmacol 2022; 202:115141. [PMID: 35700759 DOI: 10.1016/j.bcp.2022.115141] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 11/28/2022]
Abstract
A successful pregnancy and the birth of a healthy baby depend to a great extent on the controlled supply of essential nutrients via the placenta. Iron is essential for mitochondrial energy supply and oxygen distribution via the blood. However, its high reactivity requires tightly regulated transport processes. Disturbances of maternal-fetal iron transfer during pregnancy can aggravate or lead to severe pathological consequences for the mother and the fetus with lifelong effects. Furthermore, high intracellular iron levels due to disturbed gestational iron homeostasis have recently been associated with the non-apoptotic cell death pathway called ferroptosis. Therefore, the investigation of transplacental iron transport mechanisms, their physiological regulation and potential risks are of high clinical importance. The present review summarizes the current knowledge on principles and regulatory mechanisms underlying materno-fetal iron transport and gives insight into common pregnancy conditions in which iron homeostasis is disturbed. Moreover, the significance of the newly emerging ferroptosis pathway and its impact on the regulation of placental iron homeostasis, oxidative stress and gestational diseases will be discussed.
Collapse
Affiliation(s)
- Jonas Zaugg
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Switzerland
| | - Fabia Solenthaler
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research (NCCR) TransCure, University of Bern, Switzerland.
| |
Collapse
|
24
|
Liu Y, Baumann B, Song Y, Zhang K, Sterling JK, Lakhal-Littleton S, Kozmik Z, Su G, Dunaief JL. Minimal effect of conditional ferroportin KO in the neural retina implicates ferrous iron in retinal iron overload and degeneration. Exp Eye Res 2022; 218:108988. [PMID: 35202704 PMCID: PMC9050912 DOI: 10.1016/j.exer.2022.108988] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/09/2023]
Abstract
Iron-induced oxidative stress can cause or exacerbate retinal degenerative diseases. Retinal iron overload has been reported in several mouse disease models with systemic or neural retina-specific knockout (KO) of homologous ferroxidases ceruloplasmin (Cp) and hephaestin (Heph). Cp and Heph can potentiate ferroportin (Fpn) mediated cellular iron export. Here, we used retina-specific Fpn KO mice to test the hypothesis that retinal iron overload in Cp/Heph DKO mice is caused by impaired iron export from neurons and glia. Surprisingly, there was no indication of retinal iron overload in retina-specific Fpn KO mice: the mRNA levels of transferrin receptor in the retina were not altered at 7-10-months age. Consistent with this, levels and localization of ferritin light chain were unchanged. To "stress the system", we injected iron intraperitoneally into Fpn KO mice with or without Cp KO. Only mice with both retina-specific Fpn KO and Cp KO had modestly elevated retinal iron levels. These results suggest that impaired iron export through Fpn is not sufficient to explain the retinal iron overload in Cp/Heph DKO mice. An increase in the levels of retinal ferrous iron caused by the absence of these ferroxidases, followed by uptake into cells by ferrous iron importers, is most likely necessary.
Collapse
Affiliation(s)
- Yingrui Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, Jilin, China,F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA, USA
| | - Bailey Baumann
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA, USA
| | - Ying Song
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA, USA
| | - Kevin Zhang
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA, USA
| | - Jacob K. Sterling
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA, USA
| | - Samira Lakhal-Littleton
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Zbynek Kozmik
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Academy of Sciences of the Czech Republic (ASCR), Prague, Czech Republic
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, Jilin, China,Co-correspondence authors: Correspondence to: Joshua L. Dunaief, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA 19104, USA. Tel: +1 215 898 5235, , Guanfang Su, Department of Ophthalmology, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, Jilin, 130041, China. Tel: +86 13843091955,
| | - Joshua L. Dunaief
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA, USA,Co-correspondence authors: Correspondence to: Joshua L. Dunaief, F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd, Philadelphia, PA 19104, USA. Tel: +1 215 898 5235, , Guanfang Su, Department of Ophthalmology, The Second Hospital of Jilin University, No. 218 Ziqiang Street, Changchun, Jilin, 130041, China. Tel: +86 13843091955,
| |
Collapse
|
25
|
Zhang KR, Baumann B, Song Y, Sterling J, Erler EA, Guttha S, Kozmik Z, Dunaief JL. Conditional knockout of hephaestin in the neural retina disrupts retinal iron homeostasis. Exp Eye Res 2022; 218:109028. [PMID: 35271829 PMCID: PMC9050911 DOI: 10.1016/j.exer.2022.109028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/24/2022] [Accepted: 03/02/2022] [Indexed: 11/18/2022]
Abstract
Iron accumulation has been implicated in degenerative retinal diseases. It can catalyze the production of damaging reactive oxygen species. Previous work has demonstrated iron accumulation in multiple retinal diseases, including age-related macular degeneration and diabetic retinopathy. In mice, systemic knockout of the ferroxidases ceruloplasmin (Cp) and hephaestin (Heph), which oxidize iron, results in retinal iron accumulation and iron-induced degeneration. To determine the role of Heph in the retina, we generated a neural retina-specific Heph knockout on a background of systemic Cp knockout. This resulted in elevated neural retina iron. Conversely, retinal ganglion cells had elevated transferrin receptor and decreased ferritin, suggesting diminished iron levels. The retinal degeneration observed in systemic Cp-/-, Heph-/- mice did not occur. These findings indicate that Heph has a local role in regulating neural retina iron homeostasis, but also suggest that preserved Heph function in either the RPE or systemically mitigates the degeneration phenotype observed in the systemic Cp-/-, Heph-/- mice.
Collapse
Affiliation(s)
- Kevin R Zhang
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Bailey Baumann
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Ying Song
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Jacob Sterling
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Elizabeth A Erler
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Samyuktha Guttha
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| | - Zbynek Kozmik
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, ASCR, v. v. i. Vídeňská 1083, 142 20, Prague 4, Czech Republic.
| | - Joshua L Dunaief
- F.M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, Perelman School of Medicine at the University of Pennsylvania, 305 Stellar-Chance Laboratory, 422 Curie Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
26
|
Naskar A, Stezin A, Dharmappa A, Hegde S, Philip M, Kamble N, Saini J, Sandhya K, Tatu U, Yadav R, Pal PK, Alladi PA. Fibrinogen and Complement Factor H Are Promising CSF Protein Biomarkers for Parkinson's Disease with Cognitive Impairment─A Proteomics-ELISA-Based Study. ACS Chem Neurosci 2022; 13:1030-1045. [PMID: 35200010 DOI: 10.1021/acschemneuro.2c00019] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) with cognitive impairment (PDCI) is essentially diagnosed through clinical and neuropsychological examinations. There is a need to identify biomarkers to foresee cognitive decline in them. We performed label-free unbiased nontargeted proteomics (Q-TOF LC/MS-MS) on the CSF of non-neurological control; PDCI; PD; and normal pressure hydrocephalus (NPH) patients, followed by targeted ELISA for validation. Of the 281 proteins identified, 42 were differentially altered in PD, PDCI, and NPH. With a certain overlap, 28 proteins were altered in PDCI and 25 proteins were altered in NPH. Five significantly upregulated proteins in PDCI were fibrinogen, gelsolin, complement factor-H, and apolipoproteins A-I and A-IV, whereas carnosine dipeptidase-1, carboxypeptidase-E, dickkopf-3, and secretogranin-3 precursor proteins were downregulated. Those uniquely altered in NPH were the insulin-like growth factor-binding protein, ceruloplasmin, α-1 antitrypsin, VGF nerve growth factor, and neural cell adhesion molecule L1-like protein. The ELISA-derived protein concentrations correlated with neuropsychological scores of certain cognitive domains. In PDCI, the Wisconsin card sorting percentile correlated negatively with fibrinogen. Intraperitoneal injection of native fibrinogen caused motor deficits in C57BL/6J mice as assessed by the pole test. Thus, a battery of proteins such as fibrinogen-α-chain, CFAH, and APOA-I/APOA-IV alongside neuropsychological assessment could be reliable biomarkers to distinguish PDCI and NPH.
Collapse
Affiliation(s)
- Aditi Naskar
- Department of Clinical Psychopharmacology & Neurotoxicology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Albert Stezin
- Department of Clinical Neurosciences, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Arpitha Dharmappa
- Department of Clinical Psychology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Shantala Hegde
- Department of Clinical Psychology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Mariamma Philip
- Department of Biostatistics, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Nitish Kamble
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Jitender Saini
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - K. Sandhya
- Department of Anaesthesiology, Bangalore Medical College and Research Institute, Bengaluru 560002, India
| | - Utpal Tatu
- Department of Biochemistry, Indian Institute of Science, Bengaluru 560012, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| | - Phalguni Anand Alladi
- Department of Clinical Psychopharmacology & Neurotoxicology, National Institute of Mental Health and Neuro Sciences, Bengaluru 560029, India
| |
Collapse
|
27
|
The (Bio)Chemistry of Non-Transferrin-Bound Iron. Molecules 2022; 27:molecules27061784. [PMID: 35335148 PMCID: PMC8951307 DOI: 10.3390/molecules27061784] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
In healthy individuals, virtually all blood plasma iron is bound by transferrin. However, in several diseases and clinical conditions, hazardous non-transferrin-bound iron (NTBI) species occur. NTBI represents a potentially toxic iron form, being a direct cause of oxidative stress in the circulating compartment and tissue iron loading. The accumulation of these species can cause cellular damage in several organs, namely, the liver, spleen, and heart. Despite its pathophysiological relevance, the chemical nature of NTBI remains elusive. This has precluded its use as a clinical biochemical marker and the development of targeted therapies. Herein, we make a critical assessment of the current knowledge of NTBI speciation. The currently accepted hypotheses suggest that NTBI is mostly iron bound to citric acid and iron bound to serum albumin, but the chemistry of this system remains fuzzy. We explore the complex chemistry of iron complexation by citric acid and its implications towards NTBI reactivity. Further, the ability of albumin to bind iron is revised and the role of protein post-translational modifications on iron binding is discussed. The characterization of the NTBI species structure may be the starting point for the development of a standardized analytical assay, the better understanding of these species’ reactivity or the identification of NTBI uptake mechanisms by different cell types, and finally, to the development of new therapies.
Collapse
|
28
|
Abstract
Zn2+ ions are essential in many physiological processes, including enzyme catalysis, protein structural stabilization, and the regulation of many proteins. The affinities of proteins for Zn2+ ions span several orders of magnitude, with catalytic Zn2+ ions generally held more tightly than structural or regulatory ones. Metal carrier proteins, most of which are not specific for Zn2+, bind these ions with a broad range of affinities that overlap those of catalytic, structural, and regulatory Zn2+ ions and are thought to be responsible for distributing the metal through most cells, tissues, and fluid compartments. While little is known about how many proteins obtain or release these ions, there is now considerable experimental evidence suggesting that metal carrier proteins may be responsible for transferring metals to and from some Zn2+-dependent proteins, thus serving as a major regulatory factor for them. In this review, the biological roles of Zn2+ and structures of Zn2+ binding sites are examined, and experimental evidence demonstrating the direct participation of metal carrier proteins in enzyme regulation is discussed. Mechanisms of metal ion transfer are also offered, and the potential physiological significance of this phenomenon is explored.
Collapse
|
29
|
Niu L, Zhou Y, Lu L, Su A, Guo X. Ceruloplasmin Deficiency Impaired Brain Iron Metabolism and Behavior in Mice. Cell Biochem Biophys 2022; 80:385-393. [PMID: 35147903 DOI: 10.1007/s12013-022-01061-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/28/2022]
Abstract
Iron accumulation is an important cause of various brain diseases. As a ferroxidase, ceruloplasmin (Cp) plays a key role in iron homeostasis and its abnormal activity leads to iron accumulation. However, the detailed biological function of Cp in brain iron homeostasis needs to be investigated. In this study, Cp knockout mice were prepared and the changes in iron content and protein expression related to iron metabolism were detected. The results showed that iron accumulation occurred in multiple tissues and organs of Cp knockout mice, but there was no obvious change in brain tissues. However, Cp deficiency affected the expression of many iron metabolism-related proteins in midbrain, such as DMT1+IRE, heavy chain ferritin (H-ferritin) and light chain ferritin (L-ferritin). Cp deficiency also impaired the behavioral ability of mice, including weakened exercise ability and reduced motor coordination. In vitro cell experiment indicated that the sensitivity of Cp knockout neuron and astrocyte to hypoxia was higher than that of wild type, which means Cp deficiency leads to the damage of cell self-protection. All these results confirm that Cp exerts a protective effect on the brain by regulating iron metabolism.
Collapse
Affiliation(s)
- Lijing Niu
- Institute of Medicine, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, China.
| | - Yi Zhou
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li Lu
- Institute of Medicine, Hebei Provincial Center for Disease Control and Prevention, Shijiazhuang, China
| | - Aixia Su
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Xiaoqiang Guo
- Department of Sport Human Sciences, Hebei Sport University, Shijiazhuang, China. .,Xiaobei Medical Research Institute, Department of Physical Education, Shijiazhuang Vocational College of Technology, Shijiazhuang, China.
| |
Collapse
|
30
|
Shahandeh A, Bui BV, Finkelstein DI, Nguyen CTO. Effects of Excess Iron on the Retina: Insights From Clinical Cases and Animal Models of Iron Disorders. Front Neurosci 2022; 15:794809. [PMID: 35185447 PMCID: PMC8851357 DOI: 10.3389/fnins.2021.794809] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Iron plays an important role in a wide range of metabolic pathways that are important for neuronal health. Excessive levels of iron, however, can promote toxicity and cell death. An example of an iron overload disorder is hemochromatosis (HH) which is a genetic disorder of iron metabolism in which the body’s ability to regulate iron absorption is altered, resulting in iron build-up and injury in several organs. The retina was traditionally assumed to be protected from high levels of systemic iron overload by the blood-retina barrier. However, recent data shows that expression of genes that are associated with HH can disrupt retinal iron metabolism. Thus, the effects of iron overload on the retina have become an area of research interest, as excessively high levels of iron are implicated in several retinal disorders, most notably age–related macular degeneration. This review is an effort to highlight risk factors for excessive levels of systemic iron build-up in the retina and its potential impact on the eye health. Information is integrated across clinical and preclinical animal studies to provide insights into the effects of systemic iron loading on the retina.
Collapse
Affiliation(s)
- Ali Shahandeh
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Christine T. O. Nguyen,
| |
Collapse
|
31
|
Silva AM, Moniz T, de Castro B, Rangel M. Human transferrin: An inorganic biochemistry perspective. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
Monteith AJ, Skaar EP. The impact of metal availability on immune function during infection. Trends Endocrinol Metab 2021; 32:916-928. [PMID: 34483037 PMCID: PMC8516721 DOI: 10.1016/j.tem.2021.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022]
Abstract
Nutrient transition metals are required cofactors for many proteins to perform functions necessary for life. As such, the concentration of nutrient metals is carefully maintained to retain critical biological processes while limiting toxicity. During infection, invading bacterial pathogens must acquire essential metals, such as zinc, manganese, iron, and copper, from the host to colonize and cause disease. To combat this, the host exploits the essentiality and toxicity of nutrient metals by producing factors that limit metal availability, thereby starving pathogens or accumulating metals in excess to intoxicate the pathogen in a process termed 'nutritional immunity'. As a result of inflammation, a heterogeneous environment containing both metal-replete and -deplete niches is created, in which nutrient metal availability may have an underappreciated role in regulating immune cell function during infection. How the host manipulates nutrient metal availability during infection, and the downstream effects that nutrient metals and metal-sequestering proteins have on immune cell function, are discussed in this review.
Collapse
Affiliation(s)
- Andrew J Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, & Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
33
|
Helman SL, Wilkins SJ, McKeating DR, Perkins AV, Whibley PE, Cuffe JSM, Simmons DG, Fuqua BK, Vulpe CD, Wallace DF, O'Callaghan JL, Pelzer ES, Anderson GJ, Frazer DM. The Placental Ferroxidase Zyklopen Is Not Essential for Iron Transport to the Fetus in Mice. J Nutr 2021; 151:2541-2550. [PMID: 34114013 DOI: 10.1093/jn/nxab174] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/09/2021] [Accepted: 05/07/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The ferroxidase zyklopen (Zp) has been implicated in the placental transfer of iron to the fetus. However, the evidence for this is largely circumstantial. OBJECTIVES This study aimed to determine whether Zp is essential for placental iron transfer. METHODS A model was established using 8- to 12-wk-old pregnant C57BL/6 mice on standard rodent chow in which Zp was knocked out in the fetus and fetal components of the placenta. Zp was also disrupted in the entire placenta using global Zp knockout mice. Inductively coupled plasma MS was used to measure total fetal iron, an indicator of the amount of iron transferred by the placenta to the fetus, at embryonic day 18.5 of gestation. Iron transporter expression in the placenta was measured by Western blotting, and the expression of Hamp1, the gene encoding the iron regulatory hormone hepcidin, was determined in fetal liver by real-time PCR. RESULTS There was no change in the amount of iron transferred to the fetus when Zp was disrupted in either the fetal component of the placenta or the entire placenta. No compensatory changes in the expression of the iron transport proteins transferrin receptor 1 or ferroportin were observed, nor was there any change in fetal liver Hamp1 mRNA. Hephl1, the gene encoding Zp, was expressed mainly in the maternal decidua of the placenta and not in the nutrient-transporting syncytiotrophoblast. Disruption of Zp in the whole placenta resulted in a 26% increase in placental size (P < 0.01). CONCLUSIONS Our data indicate that Zp is not essential for the efficient transfer of iron to the fetus in mice and is localized predominantly in the maternal decidua. The increase in placental size observed when Zp is knocked out in the entire placenta suggests that this protein may play a role in placental development.
Collapse
Affiliation(s)
- Sheridan L Helman
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,Faculty of Medicine, The University of Queensland, St. Lucia, Australia
| | - Sarah J Wilkins
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Daniel R McKeating
- School of Medical Science, Griffith University, Gold Coast Campus, Southport, Australia
| | - Anthony V Perkins
- School of Medical Science, Griffith University, Gold Coast Campus, Southport, Australia
| | - Page E Whibley
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - James S M Cuffe
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - David G Simmons
- School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - Brie K Fuqua
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Christopher D Vulpe
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Daniel F Wallace
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| | - Jessica L O'Callaghan
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| | - Elise S Pelzer
- School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| | - Gregory J Anderson
- Iron Metabolism Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Chemistry and Molecular Bioscience, The University of Queensland, St. Lucia, Australia
| | - David M Frazer
- Molecular Nutrition Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia.,School of Biomedical Sciences, Queensland University of Technology, Gardens Point, Australia
| |
Collapse
|
34
|
Santiago González DA, Cheli VT, Rosenblum SL, Denaroso G, Paez PM. Ceruloplasmin deletion in myelinating glial cells induces myelin disruption and oxidative stress in the central and peripheral nervous systems. Redox Biol 2021; 46:102118. [PMID: 34474395 PMCID: PMC8408659 DOI: 10.1016/j.redox.2021.102118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023] Open
Abstract
Ceruloplasmin (Cp) is a ferroxidase enzyme that is essential for cell iron efflux and has been postulated to have a neuroprotective role. During the myelination process, oligodendrocytes (OLs) and Schwann cells (SCs) express high levels of Cp, but the role of this enzyme in glial cell development and function is completely unknown. To define the function of Cp in the myelination of the central and peripheral nervous systems, we have conditionally knocked-out Cp specifically in OLs and SCs during early postnatal development as well as in aged mice. Cp ablation in early OLs (postnatal day 2, P2) significantly affects the differentiation of these cells and the synthesis of myelin through the first four postnatal weeks. The total number of mature myelinating OLs was reduced, and the density of apoptotic OLs was increased. These changes were accompanied with reductions in the percentage of myelinated axons and increases in the g-ratio of myelinated fibers. Cp ablation in young myelinating OLs (P30 or P60) did not affect myelin synthesis and/or OL numbers, however, Cp loss in aged OLs (8 months) induced cell iron overload, apoptotic cell death, brain oxidative stress, neurodegeneration and myelin disruption. Furthermore, Cp deletion in SCs affected postnatal SC development and myelination and produced motor coordination deficits as well as oxidative stress in young and aged peripheral nerves. Together, our data indicate that Cp ferroxidase activity is essential for OLs and SCs maturation during early postnatal development and iron homeostasis in matured myelinating cells. Additionally, our results suggest that Cp expression in myelinating glial cells is crucial to prevent oxidative stress and neurodegeneration in the central and peripheral nervous systems. Cp activity is essential for the development and function of myelinating glial cell. Cp ablation delays oligodendrocyte and Schwann cell maturation. Cp deletion interrupts the myelination of the central and peripheral nervous systems. Cp deletion in aged oligodendrocytes induces cell dead and brain oxidative stress.
Collapse
Affiliation(s)
- D A Santiago González
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, USA
| | - V T Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, USA
| | - S L Rosenblum
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, USA
| | - G Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, USA
| | - P M Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
35
|
Abstract
Significance: Iron is an essential element required for growth and proper functioning of the body. However, an excess of labile ferrous iron increases the risk of oxidative stress-induced injury due to the high reactivity of the unpaired reactive electrons of both ferrous iron and oxygen. This high reactivity can be exemplified in the outside world by one of its consequences, rust formation. In cells, this redox-active iron is involved in the formation of lipid radicals. Recent Advances: Defect or insufficient membrane-protective mechanisms can result in iron-catalyzed excessive lipid peroxidation and subsequent cell death, now conceptualized as ferroptosis. Growing reports propose the detrimental role of iron and ferroptosis in many experimental disease models such as ischemia-reperfusion, acute and chronic organ injuries. Critical Issues: This review first provides a snapshot of iron metabolism, followed by a brief introduction of the molecular mechanisms of ferroptosis, as an iron-dependent lipid peroxidation-driven mode of cell death. Upon describing how iron dysbiosis affects ferroptosis induction, we elaborate on the detrimental role of the iron-ferroptosis axis in several diseases. Future Directions: Despite compelling findings suggesting a role of ferroptosis in experimental animal models, the exact contribution of ferroptosis in human contexts still needs further investigation. Development of reliable ferroptosis biomarkers will be an important step in characterizing ferroptosis in human disease. This can provide therapeutic opportunities aiming at targeting ferroptosis in human diseases. Antioxid. Redox Signal. 35, 487-509.
Collapse
Affiliation(s)
- Behrouz Hassannia
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Samya Van Coillie
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium.,Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
36
|
Chen F, Han B, Meng Y, Han Y, Liu B, Zhang B, Chang Y, Cao P, Fan Y, Tan K. Ceruloplasmin correlates with immune infiltration and serves as a prognostic biomarker in breast cancer. Aging (Albany NY) 2021; 13:20438-20467. [PMID: 34413268 PMCID: PMC8436892 DOI: 10.18632/aging.203427] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022]
Abstract
Breast-invasive carcinoma (BRCA) is the most frequent and malignant tumor in females. Ceruloplasmin (CP) is a multifunctional molecule involved in iron metabolism, but its expression profile, prognostic potential and relationship with immune cell infiltration in BRCA are unknown. Ceruloplasmin mRNA and protein expression was significantly decreased in BRCA patients according to the Oncomine, UALCAN, GEPIA and TCGA databases. Ceruloplasmin expression was strongly correlated with various clinicopathological features of BRCA patients. BRCA patients with high ceruloplasmin expression exhibited shorter survival times than those with low ceruloplasmin expression based on the Kaplan-Meier plotter and PrognoScan databases. GO and KEGG analyses and GSEA revealed a strong correlation between ceruloplasmin and various immune-related pathways. Ceruloplasmin expression was significantly associated with the infiltration of immune cells into tumor sites by analyzing the TIMER and CIBERSORT. Additionally, ceruloplasmin was positively correlated with immune checkpoints in BRCA. These findings suggest that low ceruloplasmin expression correlates with a favorable prognosis and tumor immune cell infiltration in BRCA patients. Ceruloplasmin may serve as a therapeutic target and predict the efficacy of immunotherapy for BRCA.
Collapse
Affiliation(s)
- Fei Chen
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bihui Han
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yanxiu Meng
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yu Han
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bing Liu
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Bo Zhang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yanzhong Chang
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Pengxiu Cao
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Yumei Fan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| | - Ke Tan
- Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University, Shijiazhuang 050024, Hebei, China
| |
Collapse
|
37
|
Lakhal-Littleton S. Advances in understanding the crosstalk between mother and fetus on iron utilization. Semin Hematol 2021; 58:153-160. [PMID: 34389107 DOI: 10.1053/j.seminhematol.2021.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/16/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022]
Abstract
A full-term pregnancy comes with significant demand for iron. Not meeting this demand has adverse effects on maternal health and on the intrauterine and postnatal development of the infant. In the infant, some of these adverse effects cannot be reversed by postnatal iron supplementation, highlighting the need to tackle iron deficiency in utero. Achieving this requires sound understanding of the pathways that govern iron transfer at the fetomaternal interface. Two pathways are emerging as key players in this context; the hepcidin/ferroportin axis pathway and the iron regulatory protein (IRPs) pathway. In late gestation, suppression of maternal hepcidin, by as yet unknown factors, is required for increasing iron availability to the growing fetus. In the placenta, the rate of iron uptake by transferrin receptor TfR1 at the apical/maternal side and of iron release by ferroportin FPN at the basal/fetal side is controlled by IRP1. In fetal hepatocytes, build up of fetal iron stores requires post-translational inhibition of FPN by the cell-autonomous action of hepcidin. In the fetal liver, FPN is also subject to additional control at the transcriptional level, possibly by the action of hypoxia-inducible factor HIF2α. The rates of apical iron uptake and basal iron release in the placenta are modulated according to iron availability in the maternal blood and the placenta's own needs. This placental modulation ensures that the amount of iron delivered to the fetal circulation is maintained within a normal range, even in the face of mild maternal iron deficiency or overload. However, when maternal iron deficiency or overload are extreme, placental modulation is not sufficient to maintain normal iron supply to the fetus, resulting in fetal iron deficiency and overload respectively. Thus, the rate of iron transfer at the fetomaternal interface is subject to several regulatory signals operating simultaneously in the maternal liver, the placenta and the fetal liver. These regulatory signals act in concert to maintain normal iron supply to the fetus within a wide range of maternal iron states, but fail to do so when maternal iron deficiency or overload are extreme. The limitations of existing experimental models must be overcome if we are to gain better understanding of the role of these regulatory signals in normal and complicated pregnancy. Ultimately, that understanding could help identify better markers of fetal iron demand and underpin novel iron replacement strategies to treat maternal and fetal iron deficiency.
Collapse
|
38
|
Camaschella C, Pagani A. Mendelian inheritance of anemia due to disturbed iron homeostasis. Semin Hematol 2021; 58:175-181. [PMID: 34389109 DOI: 10.1053/j.seminhematol.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/16/2021] [Accepted: 05/31/2021] [Indexed: 02/01/2023]
Abstract
Genetic disorders that affect proteins involved in maintaining iron balance may lead to Mendelian anemias. They may be classified as defects of intestinal iron absorption, iron transport in the circulation, iron uptake and utilization by maturing erythroid cells, iron recycling by macrophages and systemic regulation of iron homeostasis. All these Mendelian anemias are rare disorders, prevalently recessive, characterized by microcytic and hypochromic red blood cells. Advances in our knowledge of iron metabolism and its systemic regulation on one side have facilitated the identification of novel iron related anemias, while on the other the study of the affected patients and of the corresponding animal models have contributed to our understanding of iron trafficking and regulation.
Collapse
Affiliation(s)
- Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.
| | - Alessia Pagani
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
39
|
Puchkova LV, Kiseleva IV, Polishchuk EV, Broggini M, Ilyechova EY. The Crossroads between Host Copper Metabolism and Influenza Infection. Int J Mol Sci 2021; 22:ijms22115498. [PMID: 34071094 PMCID: PMC8197124 DOI: 10.3390/ijms22115498] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Three main approaches are used to combat severe viral respiratory infections. The first is preemptive vaccination that blocks infection. Weakened or dead viral particles, as well as genetic constructs carrying viral proteins or information about them, are used as an antigen. However, the viral genome is very evolutionary labile and changes continuously. Second, chemical agents are used during infection and inhibit the function of a number of viral proteins. However, these drugs lose their effectiveness because the virus can rapidly acquire resistance to them. The third is the search for points in the host metabolism the effect on which would suppress the replication of the virus but would not have a significant effect on the metabolism of the host. Here, we consider the possibility of using the copper metabolic system as a target to reduce the severity of influenza infection. This is facilitated by the fact that, in mammals, copper status can be rapidly reduced by silver nanoparticles and restored after their cancellation.
Collapse
Affiliation(s)
- Ludmila V. Puchkova
- International Research Laboratory of Trace Elements Metabolism, ADTS Institute, RC AFMLCS, ITMO University, 197101 St. Petersburg, Russia;
| | - Irina V. Kiseleva
- Department of Virology, Institute of Experimental Medicine, 197376 St. Petersburg, Russia;
| | | | - Massimo Broggini
- Istituto di Ricerche Farmacologiche “Mario Negri”, IRCCS, 20156 Milan, Italy;
| | - Ekaterina Yu. Ilyechova
- International Research Laboratory of Trace Elements Metabolism, ADTS Institute, RC AFMLCS, ITMO University, 197101 St. Petersburg, Russia;
- Department of Molecular Genetics, Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- Correspondence: ; Tel.: +7-921-760-5274
| |
Collapse
|
40
|
Zacchi P, Belmonte B, Mangogna A, Morello G, Scola L, Martorana A, Borelli V. The Ferroxidase Hephaestin in Lung Cancer: Pathological Significance and Prognostic Value. Front Oncol 2021; 11:638856. [PMID: 34094919 PMCID: PMC8170403 DOI: 10.3389/fonc.2021.638856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/26/2021] [Indexed: 12/30/2022] Open
Abstract
Hephaestin (HEPH) belongs to a group of exocytoplasmic ferroxidases which contribute to cellular iron homeostasis by favouring its export. Down-regulation of HEPH expression, possibly by stimulating cell proliferation due to an increase in iron availability, has shown to correlate with poor survival in breast cancer. The lung is particularly sensitive to iron-induced oxidative stress, given the high oxygen tension present, however, HEPH distribution in lung cancer and its influence on prognosis have not been investigated yet. In this study we explored the prognostic value of HEPH and its expression pattern in the most prevalent histotypes of lung cancers, namely lung adenocarcinoma and lung squamous cell carcinoma. In silico analyses, based on UALCAN, Gene Expression Profiling Interactive Analysis (GEPIA) and Kaplan–Meier plotter bioinformatics, revealed a significant correlation between higher levels of HEPH expression and favorable prognosis, in both cancer histotypes. Moreover, TIMER web platform showed a statistically significant association between HEPH expression and cell elements belonging to the tumor microenvironment identified as endothelial cells and a subpopulation of cancer-associated fibroblasts, further confirmed by double immunohistochemical labeling with cell type specific markers. Taken together, these data shed a light on the complex mechanisms of local iron handling lung cancer can exploit to support tumorigenesis.
Collapse
Affiliation(s)
- Paola Zacchi
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Gaia Morello
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Letizia Scola
- Clinical Pathology, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Anna Martorana
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Violetta Borelli
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
41
|
A Nonsense Variant in Hephaestin Like 1 ( HEPHL1) Is Responsible for Congenital Hypotrichosis in Belted Galloway Cattle. Genes (Basel) 2021; 12:genes12050643. [PMID: 33926013 PMCID: PMC8147104 DOI: 10.3390/genes12050643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/31/2022] Open
Abstract
Genodermatosis such as hair disorders mostly follow a monogenic mode of inheritance. Congenital hypotrichosis (HY) belong to this group of disorders and is characterized by abnormally reduced hair since birth. The purpose of this study was to characterize the clinical phenotype of a breed-specific non-syndromic form of HY in Belted Galloway cattle and to identify the causative genetic variant for this recessive disorder. An affected calf born in Switzerland presented with multiple small to large areas of alopecia on the limbs and on the dorsal part of the head, neck, and back. A genome-wide association study using Swiss and US Belted Galloway cattle encompassing 12 cases and 61 controls revealed an association signal on chromosome 29. Homozygosity mapping in a subset of cases refined the HY locus to a 1.5 Mb critical interval and subsequent Sanger sequencing of protein-coding exons of positional candidate genes revealed a stop gain variant in the HEPHL1 gene that encodes a multi-copper ferroxidase protein so-called hephaestin like 1 (c.1684A>T; p.Lys562*). A perfect concordance between the homozygous presence of this most likely pathogenic loss-of-function variant and the HY phenotype was found. Genotyping of more than 700 purebred Swiss and US Belted Galloway cattle showed the global spread of the mutation. This study provides a molecular test that will permit the avoidance of risk matings by systematic genotyping of relevant breeding animals. This rare recessive HEPHL1-related form of hypotrichosis provides a novel large animal model for similar human conditions. The results have been incorporated in the Online Mendelian Inheritance in Animals (OMIA) database (OMIA 002230-9913).
Collapse
|
42
|
Culbertson EM, Khan AA, Muchenditsi A, Lutsenko S, Sullivan DJ, Petris MJ, Cormack BP, Culotta VC. Changes in mammalian copper homeostasis during microbial infection. Metallomics 2021; 12:416-426. [PMID: 31976503 DOI: 10.1039/c9mt00294d] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Animals carefully control homeostasis of Cu, a metal that is both potentially toxic and an essential nutrient. During infection, various shifts in Cu homeostasis can ensue. In mice infected with Candida albicans, serum Cu progressively rises and at late stages of infection, liver Cu rises, while kidney Cu declines. The basis for these changes in Cu homeostasis was poorly understood. We report here that the progressive rise in serum Cu is attributable to liver production of the multicopper oxidase ceruloplasmin (Cp). Through studies using Cp-/- mice, we find this elevated Cp helps recover serum Fe levels at late stages of infection, consistent with a role for Cp in loading transferrin with Fe. Cp also accounts for the elevation in liver Cu seen during infection, but not for the fluctuations in kidney Cu. The Cu exporting ATPase ATP7B is one candidate for kidney Cu control, but we find no change in the pattern of kidney Cu loss during infection of Atp7b-/- mice, implying alternative mechanisms. To test whether fungal infiltration of kidney tissue was required for kidney Cu loss, we explored other paradigms of infection. Infection with the intravascular malaria parasite Plasmodium berghei caused a rise in serum Cu and decrease in kidney Cu similar to that seen with C. albicans. Thus, dynamics in kidney Cu homeostasis appear to be a common feature among vastly different infection paradigms. The implications for such Cu homeostasis control in immunity are discussed.
Collapse
Affiliation(s)
- Edward M Culbertson
- The Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Aslam A Khan
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Abigael Muchenditsi
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Svetlana Lutsenko
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David J Sullivan
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Michael J Petris
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA
| | - Brendan P Cormack
- Department of Molecular Biology and Genetics, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valeria C Culotta
- The Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
43
|
Culbertson EM, Culotta VC. Copper in infectious disease: Using both sides of the penny. Semin Cell Dev Biol 2021; 115:19-26. [PMID: 33423931 DOI: 10.1016/j.semcdb.2020.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/28/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
The transition metal Cu is an essential micronutrient that serves as a co-factor for numerous enzymes involved in redox and oxygen chemistry. However, Cu is also a potentially toxic metal, especially to unicellular microbes that are in direct contact with their environment. Since 400 BCE, Cu toxicity has been leveraged for its antimicrobial properties and even today, Cu based materials are being explored as effective antimicrobials against human pathogens spanning bacteria, fungi, and viruses, including the SARS-CoV-2 agent of the 2019-2020 pandemic. Given that Cu has the double-edged property of being both highly toxic and an essential micronutrient, it plays an active and complicated role at the host-pathogen interface. Humans have evolved methods of incorporating Cu into innate and adaptive immune processes and both sides of the penny (Cu toxicity and Cu as a nutrient) are employed. Here we review the evolution of Cu in biology and its multi-faceted roles in infectious disease, from the viewpoints of the microbial pathogens as well as the animal hosts they infect.
Collapse
Affiliation(s)
- Edward M Culbertson
- The Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Valeria C Culotta
- The Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
44
|
Venkataramani V. Iron Homeostasis and Metabolism: Two Sides of a Coin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1301:25-40. [PMID: 34370286 DOI: 10.1007/978-3-030-62026-4_3] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Iron is an ancient, essential and versatile transition metal found in almost all living organisms on Earth. This fundamental trace element is used in the synthesis of heme and iron-sulfur (Fe-S) containing proteins and other vital cofactors that are involved in respiration, redox reactions, catalysis, DNA synthesis and transcription. At the same time, the ability of iron to cycle between its oxidized, ferric (Fe3+) and its reduced, ferrous (Fe2+) state contributes to the production of free radicals that can damage biomolecules, including proteins, lipids and DNA. In particular, the regulated non-apoptotic cell death ferroptosis is driven by Fe2+-dependent lipid peroxidation that can be prevented by iron chelation or genetic inhibition of cellular iron uptake. Therefore, iron homeostasis must be tightly regulated to avoid iron toxicity. This review provides an overview of the origin and chemistry of iron that makes it suitable for a variety of biological functions and addresses how organisms evolved various strategies, including their scavenging and antioxidant machinery, to manage redox-associated drawbacks. Finally, key mechanisms of iron metabolism are highlighted in human diseases and model organisms, underlining the perils of dysfunctional iron handlings.
Collapse
Affiliation(s)
- Vivek Venkataramani
- Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany.
| |
Collapse
|
45
|
Lelièvre P, Sancey L, Coll JL, Deniaud A, Busser B. The Multifaceted Roles of Copper in Cancer: A Trace Metal Element with Dysregulated Metabolism, but Also a Target or a Bullet for Therapy. Cancers (Basel) 2020; 12:E3594. [PMID: 33271772 PMCID: PMC7760327 DOI: 10.3390/cancers12123594] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
In the human body, copper (Cu) is a major and essential player in a large number of cellular mechanisms and signaling pathways. The involvement of Cu in oxidation-reduction reactions requires close regulation of copper metabolism in order to avoid toxic effects. In many types of cancer, variations in copper protein levels have been demonstrated. These variations result in increased concentrations of intratumoral Cu and alterations in the systemic distribution of copper. Such alterations in Cu homeostasis may promote tumor growth or invasiveness or may even confer resistance to treatments. Once characterized, the dysregulated Cu metabolism is pinpointing several promising biomarkers for clinical use with prognostic or predictive capabilities. The altered Cu metabolism in cancer cells and the different responses of tumor cells to Cu are strongly supporting the development of treatments to disrupt, deplete, or increase Cu levels in tumors. The metallic nature of Cu as a chemical element is key for the development of anticancer agents via the synthesis of nanoparticles or copper-based complexes with antineoplastic properties for therapy. Finally, some of these new therapeutic strategies such as chelators or ionophores have shown promising results in a preclinical setting, and others are already in the clinic.
Collapse
Affiliation(s)
- Pierre Lelièvre
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
| | - Lucie Sancey
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
| | - Jean-Luc Coll
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
| | - Aurélien Deniaud
- Univ. Grenoble Alpes, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie des Métaux, 38000 Grenoble, France
| | - Benoit Busser
- Institute for Advanced Biosciences, UGA INSERM U1209 CNRS UMR5309, 38700 La Tronche, France; (P.L.); (L.S.); (J.-L.C.)
- Department of Clinical Biochemistry, Grenoble Alpes University Hospital, 38043 Grenoble, France
| |
Collapse
|
46
|
Wimmer I, Scharler C, Kadowaki T, Hillebrand S, Scheiber-Mojdehkar B, Ueda S, Bradl M, Berger T, Lassmann H, Hametner S. Iron accumulation in the choroid plexus, ependymal cells and CNS parenchyma in a rat strain with low-grade haemolysis of fragile macrocytic red blood cells. Brain Pathol 2020; 31:333-345. [PMID: 33220123 PMCID: PMC8018038 DOI: 10.1111/bpa.12920] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/16/2020] [Accepted: 11/18/2020] [Indexed: 12/03/2022] Open
Abstract
Iron accumulation in the CNS is associated with many neurological diseases via amplification of inflammation and neurodegeneration. However, experimental studies on iron overload are challenging, since rodents hardly accumulate brain iron in contrast to humans. Here, we studied LEWzizi rats, which present with elevated CNS iron loads, aiming to characterise choroid plexus, ependymal, CSF and CNS parenchymal iron loads in conjunction with altered blood iron parameters and, thus, signifying non‐classical entry sites for iron into the CNS. Non‐haem iron in formalin‐fixed paraffin‐embedded tissue was detected via DAB‐enhanced Turnbull Blue stainings. CSF iron levels were determined via atomic absorption spectroscopy. Ferroportin and aquaporin‐1 expression was visualised using immunohistochemistry. The analysis of red blood cell indices and serum/plasma parameters was based on automated measurements; the fragility of red blood cells was manually determined by the osmotic challenge. Compared with wild‐type animals, LEWzizi rats showed strongly increased iron accumulation in choroid plexus epithelial cells as well as in ependymal cells of the ventricle lining. Concurrently, red blood cell macrocytosis, low‐grade haemolysis and significant haemoglobin liberation from red blood cells were apparent in the peripheral blood of LEWzizi rats. Interestingly, elevated iron accumulation was also evident in kidney proximal tubules, which share similarities with the blood–CSF barrier. Our data underscore the importance of iron gateways into the CNS other than the classical route across microvessels in the CNS parenchyma. Our findings of pronounced choroid plexus iron overload in conjunction with peripheral iron overload and increased RBC fragility in LEWzizi rats may be seminal for future studies of human diseases, in which similar constellations are found.
Collapse
Affiliation(s)
- Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna, Austria.,Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Cornelia Scharler
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Taro Kadowaki
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria.,Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Sophie Hillebrand
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | | | - Shuichi Ueda
- Department of Histology and Neurobiology, Dokkyo Medical University, Tochigi, Japan
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Simon Hametner
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria.,Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Jankovska E, Lipcseyova D, Svrdlikova M, Pavelcova M, Kubala Havrdova E, Holada K, Petrak J. Quantitative proteomic analysis of cerebrospinal fluid of women newly diagnosed with multiple sclerosis. Int J Neurosci 2020; 132:724-734. [PMID: 33059501 DOI: 10.1080/00207454.2020.1837801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The lack of reliable diagnostic and/or prognostic biomarkers for multiple sclerosis (MS) is the major obstacle to timely and accurate patient diagnosis in MS patients. To identify new proteins associated with MS we performed a detailed proteomic analysis of cerebrospinal fluid (CSF) of patients newly diagnosed with relapsing-remitting MS (RRMS) and healthy controls. MATERIAL Reflecting significantly higher prevalence of MS in women we included only women patients and controls in the study. To eliminate a potential effect of therapy on the CSF composition, only the therapy-naïve patients were included. METHODS Pooled CSF samples were processed in a technical duplicate, and labeled with stable-isotope coded TMT tags. To maximize the proteome coverage, peptide fractionation using 2D-LC preceded mass analysis using Orbitrap Fusion Tribrid Mass Spectrometer. Differential concentration of selected identified proteins between patients and controls was verified using specific antibodies. RESULTS Of the identified 900 CSF proteins, we found 69 proteins to be differentially abundant between patients and controls. In addition to several proteins identified as differentially abundant in MS patients previously, we observed several linked to MS for the first time, namely eosinophil-derived neurotoxin and Nogo receptor. CONCLUSIONS Our data confirm differential abundance of several previously proposed protein markers, and provide indirect support for involvement of copper-iron disbalance in MS. Most importantly, we identified two new differentially abundant CSF proteins that seem to be directly connected with myelin loss and axonal damage via TLR2 signaling and Nogo-receptor pathway in women newly diagnosed with RRMS.
Collapse
Affiliation(s)
- Eliska Jankovska
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Denisa Lipcseyova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Michaela Svrdlikova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Miluse Pavelcova
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva Kubala Havrdova
- Department of Neurology and Center for Clinical Neuroscience, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Karel Holada
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Petrak
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| |
Collapse
|
48
|
Altamura S, Marques O, Colucci S, Mertens C, Alikhanyan K, Muckenthaler MU. Regulation of iron homeostasis: Lessons from mouse models. Mol Aspects Med 2020; 75:100872. [DOI: 10.1016/j.mam.2020.100872] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
|
49
|
D’Mello SR, Kindy MC. Overdosing on iron: Elevated iron and degenerative brain disorders. Exp Biol Med (Maywood) 2020; 245:1444-1473. [PMID: 32878460 PMCID: PMC7553095 DOI: 10.1177/1535370220953065] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPACT STATEMENT Brain degenerative disorders, which include some neurodevelopmental disorders and age-associated diseases, cause debilitating neurological deficits and are generally fatal. A large body of emerging evidence indicates that iron accumulation in neurons within specific regions of the brain plays an important role in the pathogenesis of many of these disorders. Iron homeostasis is a highly complex and incompletely understood process involving a large number of regulatory molecules. Our review provides a description of what is known about how iron is obtained by the body and brain and how defects in the homeostatic processes could contribute to the development of brain diseases, focusing on Alzheimer's disease and Parkinson's disease as well as four other disorders belonging to a class of inherited conditions referred to as neurodegeneration based on iron accumulation (NBIA) disorders. A description of potential therapeutic approaches being tested for each of these different disorders is provided.
Collapse
Affiliation(s)
| | - Mark C Kindy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
- James A. Haley Veterans Affairs Medical Center, Tampa, FL 33612, USA
| |
Collapse
|
50
|
Iron overload and its impact on outcome of patients with hematological diseases. Mol Aspects Med 2020; 75:100868. [PMID: 32620237 DOI: 10.1016/j.mam.2020.100868] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 01/19/2023]
Abstract
Systemic iron overload (SIO) is a common challenge in patients with hematological diseases and develops as a result of ineffective erythropoiesis, multiple red blood cell (RBC) transfusions and disease-specific therapies. Iron homeostasis is tightly regulated as there is no physiological pathway to excrete iron from the body. Excess iron is, therefore, stored in tissues like liver, heart and bone marrow and can lead to progressive organ damage. The presence of free iron in the form of non-transferrin bound iron (NTBI) is especially detrimental. Reactive oxygen species can also cause stromal damage in the bone marrow and promote leukemic cell growth in vitro. In acute leukemias and myelodysplastic syndromes outcome is worse in patients with SIO compared to patients without. Especially in patients undergoing allogeneic HSCT presence of NTBI before or during transplant has been shown to negatively affect non-relapse mortality and overall survival. Although the mechanisms, of how these effects are mediated by SIO are not very well understood monitoring of iron status by serum markers and imaging techniques is, therefore, mandatory especially in these patients. Whether peri-interventional iron chelation may improve outcome of these patients is part of current clinical research.
Collapse
|