1
|
Ngo M, Dao T, Hoang T, Nguyen U, Stenman J, Duong H, Ho T. Accurate quantification of cell-free Ceruloplasmin mRNA as a biomarker for early detection of hepatocellular carcinoma. Sci Rep 2025; 15:14660. [PMID: 40287496 PMCID: PMC12033234 DOI: 10.1038/s41598-025-99302-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Accurate and early detection of hepatocellular carcinoma (HCC) is critical for improving patient outcomes. Current biomarkers like AFP have limited sensitivity, necessitating novel diagnostic markers. A novel semi-nested RT-PCR assay was developed to quantify circulating Ceruloplasmin (CP) mRNA in peripheral blood. This method co-amplifies CP mRNA and an internal control (IC) gene, followed by DNA melting analysis to distinguish and quantify CP mRNA. CP mRNA levels were significantly higher in the HCC group (median: 3.37) compared to both the CLD group (0.24, p = 0.0066) and the HD group (0.17, p < 0.0001). Further analysis using ROC curves highlighted the diagnostic performance of the assay. For differentiating HCC from CLD, the area under the ROC curve (AUC) was 0.704, with 50.98% sensitivity and 95.24% specificity. In comparison to HD, the AUC was 0.812, with 74.51% sensitivity and 80.65% specificity. Against the combined control group (CLD and HD), the AUC was 0.768, with 50.98% sensitivity and 96.15% specificity. Additionally, in 59.1% of HCC cases with AFP levels below 20 ng/mL, CP mRNA levels were elevated, indicating that CP mRNA could help detect a substantial proportion of AFP-negative HCC cases. This study, the first comprehensive clinical investigation of cell-free CP mRNA for HCC diagnosis, demonstrates its potential as a sensitive and specific non-invasive biomarker. Further validation in larger cohorts is needed to confirm its clinical utility.
Collapse
Affiliation(s)
- Minh Ngo
- Department of Gastroenterology and Hepatology, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
- Radiology Center, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Trang Dao
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, Hanoi, Vietnam
| | - Trang Hoang
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, Hanoi, Vietnam
| | - Ung Nguyen
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, Hanoi, Vietnam
| | - Jakob Stenman
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Huy Duong
- Department of Gastroenterology and Hepatology, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Tho Ho
- Department of Genomics and Cytogenetics, Institute of Biomedicine and Pharmacy (IBP), Vietnam Military Medical University, Hanoi, Vietnam.
- Department of Microbiology, 103 Military Hospital, Vietnam Military Medical University, Hanoi, Vietnam.
| |
Collapse
|
2
|
Guan L, Wang Y, Lin L, Zou Y, Qiu L. Variations in Blood Copper and Possible Mechanisms During Pregnancy. Biol Trace Elem Res 2024; 202:429-441. [PMID: 37777692 DOI: 10.1007/s12011-023-03716-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/24/2023] [Indexed: 10/02/2023]
Abstract
Copper (Cu), an essential trace element, is crucial for both the mother and fetus. Currently, an increasing number of studies have focused on blood copper levels during pregnancy. Studies have found that blood copper levels in pregnant women are higher than those in reproductive-age women, but the trend, mainly in the 2nd and 3rd trimester, is still controversial. Most studies showed that blood copper levels gradually increased during pregnancy, while some studies found that blood copper levels remained stable or even decreased in the 3rd trimester. The possible mechanisms of variations in blood copper during pregnancy include the influence of estrogen (hepatic uptake and excretion, ceruloplasmin synthesis, maternal-fetal transport, etc.), the interaction of other trace elements (Fe, Zn, etc.) and other factors. Among them, maternal-fetal copper transport caused by elevated estrogen may be the main reason for the inconsistencies observed in the 2nd and 3rd trimester during pregnancy. However, there are some mechanisms require further investigation. In the future, the trend and mechanisms of blood copper during pregnancy should be explored more deeply to help doctors better monitor copper status and detect copper abnormalities in time.
Collapse
Affiliation(s)
- Lihua Guan
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Yifei Wang
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Liling Lin
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Yutong Zou
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China
| | - Ling Qiu
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Science, Beijing, 100730, People's Republic of China.
| |
Collapse
|
3
|
Hefter H, Kruschel TS, Novak M, Rosenthal D, Luedde T, Meuth SG, Albrecht P, Hartmann CJ, Samadzadeh S. Differences in the Time Course of Recovery from Brain and Liver Dysfunction in Conventional Long-Term Treatment of Wilson Disease. J Clin Med 2023; 12:4861. [PMID: 37510976 PMCID: PMC10381896 DOI: 10.3390/jcm12144861] [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: 05/03/2023] [Revised: 06/30/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The aim of this study was to demonstrate that both neurological and hepatic symptoms respond to copper chelation therapy in Wilson disease (WD). However, the time course of their recovery is different. METHODS Eighteen patients with neurological WD from a single specialized center who had been listed for liver transplantation during the last ten years and two newly diagnosed homozygous twins were recruited for this retrospective study. The mean duration of conventional treatment was 7.3 years (range: 0.25 to 36.2 years). A custom Wilson disease score with seven motor items, three non-motor items, and 33 biochemical parameters of the blood and urine, as well as the MELD score, was determined at various checkup visits during treatment. These data were extracted from the charts of the patients. RESULTS Treatment was initiated with severity-dependent doses (≥900 mg) of D-penicillamine (DPA) or triethylene-tetramin-dihydrochloride (TRIEN). The motor score improved in 10 and remained constant in 8 patients. Worsening of neurological symptoms was observed only in two patients who developed comorbidities (myasthenia gravis or hemispheric stroke). The neurological symptoms continuously improved over the years until the majority of patients became only mildly affected. In contrast to this slow recovery of the neurological symptoms, the MELD score and liver enzymes had already started to improve after 1 month and rapidly improved over the next 6 months in 19 patients. The cholinesterase levels continued to increase significantly (p < 0.0074) even further. One patient whose MELD score indicated further progression of liver disease received an orthotopic liver transplantation 3 months after the diagnosis of WD and the onset of DPA treatment. CONCLUSIONS Neurological and hepatic symptoms both respond to copper chelation therapy. For patients with acute liver failure, the first 4 months are critical. This is the time span in which patients have to wait either for a donor organ or until significant improvement has occurred under conventional therapy. For patients with severe neurological symptoms, it is important that they are treated with fairly high doses over several years.
Collapse
Affiliation(s)
- Harald Hefter
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Theodor S Kruschel
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Max Novak
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Dietmar Rosenthal
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Tom Luedde
- Departments of Gastroenterology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Sven G Meuth
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Philipp Albrecht
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
- Department of Neurology, Kliniken Maria Hilf GmbH Mönchengladbach, 41063 Mönchengladbach, Germany
| | - Christian J Hartmann
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
| | - Sara Samadzadeh
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, D-40225 Düsseldorf, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Unverstät zu Berlin, Experimental and Clinical Research Center, 13125 Berlin, Germany
- Department of Regional Health Research and Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
- Department of Neurology, Slagelse Hospital, 4200 Slagelse, Denmark
| |
Collapse
|
4
|
Gromadzka G, Przybyłkowski A, Litwin T, Karpińska A. Antioxidant Capacity Is Decreased in Wilson's Disease and Correlates to Liver Function. Biol Trace Elem Res 2023; 201:1582-1587. [PMID: 35524917 DOI: 10.1007/s12011-022-03277-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/01/2022] [Indexed: 11/27/2022]
Abstract
The metabolic disorder Wilson's disease (WD) is caused by copper accumulation in the tissues due to a biallelic pathogenic mutation of the gene ATP7B, encoding intracellular copper transporter ATPase-7B. As copper is a redox active metal; aberrations in its homeostasis may create favourable conditions for superoxide-yielding redox cycling and oxidative damage to the cells. We tried to characterise antioxidant defence in WD patients and to evaluate whether it is related to liver function. The blood glutathione concentration, the activity of manganese-SOD (MnSOD), catalase (Cat), glutathione peroxidase, and glutathione S-transferase glutathione (GST), and serum antioxidant potential (AOP-450) were measured in WD treatment-naive patients and healthy controls and correlated with clinical data. The blood glutathione concentration, the activity of MnSOD, Cat, glutathione peroxidase, and GST and AOP-450 are significantly decreased in WD patients. There was a positive correlation of AOP-450 with AST. Moreover, the Cat and GST activity as well as AOP-450 strongly correlated with parameters of synthetic liver function. MnSOD activity correlated positively with ALT and AST.The blood glutathione concentration, the activity of MnSOD, Cat, glutathione peroxidase, and GST and AOP-450 are significantly decreased in WD patients. There was a positive correlation of AOP-450 with AST. Moreover, the Cat and GST activity as well as AOP-450 strongly correlated with parameters of synthetic liver function. MnSOD activity correlated positively with ALT and AST. Liver injury in course of WD is linked with decreased antioxidant capacity.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Faculty of Medicine, Cardinal Stefan Wyszyński University in Warsaw, Collegium Medicum, Wóycickiego 1/3, 01-938, Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University in Warsaw, Banacha 1a, 02-097, Warsaw, Poland.
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957, Warsaw, Poland
| | - Agata Karpińska
- Department of Clinical and Experimental Pharmacology, Medical University of Warsaw, Żwirki i Wigury 81, 02-091, Warsaw, Poland
| |
Collapse
|
5
|
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
|
6
|
Hefter H, Arslan M, Kruschel TS, Novak M, Rosenthal D, Meuth SG, Albrecht P, Hartmann CJ, Samadzadeh S. Pseudocholinesterase as a Biomarker for Untreated Wilson's Disease. Biomolecules 2022; 12:biom12121791. [PMID: 36551217 PMCID: PMC9775970 DOI: 10.3390/biom12121791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/24/2022] [Accepted: 11/26/2022] [Indexed: 12/02/2022] Open
Abstract
The aim of this study was to demonstrate that pseudocholinesterase (CHE) serum level is a useful diagnostic biomarker for untreated Wilson's disease (WD). Between 2013 and 2019, about 75 patients were referred to the outpatient department of the University of Düsseldorf with suspected Wilson's disease. In 31 patients with suspected Wilson's disease (WD-SUS-group), WD was excluded by means of investigations other than analysis of blood and urine. A total of 27 parameters of blood and urine in these 31 patients were compared to those of 20 de novo patients with manifest WD (WD-DEF-group), which parameter showed the highest significance level of difference between the WD-DEF-group and the WD-SUS-group. Thereafter, receiver operating characteristics (ROC-curves) were analyzed to evaluate which parameter showed the largest area under the curve (AUC) to detect WD. Finally, a logistic regression analysis was performed to analyze which combination of parameters allowed the best classification of the 51 patients either into the WD-DEF-group or into the WD-SUS-group. CHE showed the highest significance level for a difference between the WD-DEF- and WD-SUS-group, had the highest AUC, and, in combination with ceruloplasmin, allowed 100% correct classification. Without CHE, no other combination of parameters reached this level of correct classification. After the initiation of treatment, which regularly results in an improvement in CHE, the high diagnostic accuracy of this biomarker was lost. Cholinesterase turns out to be an excellent biomarker for differentiation between untreated de novo patients with manifest WD and heterozygotic gene carriers.
Collapse
Affiliation(s)
- Harald Hefter
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-811-7025
| | - Max Arslan
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
- Departments of Anesthesiology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Theodor S. Kruschel
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Max Novak
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Dietmar Rosenthal
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Sven G. Meuth
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Philipp Albrecht
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Christian J. Hartmann
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Sara Samadzadeh
- Departments of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
- Experimental and Clinical Research Center, Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
7
|
Pal A, Cerchiaro G, Rani I, Ventriglia M, Rongioletti M, Longobardi A, Squitti R. Iron in Alzheimer's Disease: From Physiology to Disease Disabilities. Biomolecules 2022; 12:1248. [PMID: 36139084 PMCID: PMC9496246 DOI: 10.3390/biom12091248] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
Reactive oxygen species (ROS) play a key role in the neurodegeneration processes. Increased oxidative stress damages lipids, proteins, and nucleic acids in brain tissue, and it is tied to the loss of biometal homeostasis. For this reason, attention has been focused on transition metals involved in several biochemical reactions producing ROS. Even though a bulk of evidence has uncovered the role of metals in the generation of the toxic pathways at the base of Alzheimer's disease (AD), this matter has been sidelined by the advent of the Amyloid Cascade Hypothesis. However, the link between metals and AD has been investigated in the last two decades, focusing on their local accumulation in brain areas known to be critical for AD. Recent evidence revealed a relation between iron and AD, particularly in relation to its capacity to increase the risk of the disease through ferroptosis. In this review, we briefly summarize the major points characterizing the function of iron in our body and highlight why, even though it is essential for our life, we have to monitor its dysfunction, particularly if we want to control our risk of AD.
Collapse
Affiliation(s)
- Amit Pal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Kalyani 741245, West Bengal, India
| | - Giselle Cerchiaro
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André 09210-580, SP, Brazil
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar University (MMU), Mullana, Ambala 133203, Haryana, India
| | - Mariacarla Ventriglia
- Fatebenefratelli Foundation for Health Research and Education, AFaR Division, 00186 Rome, Italy
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy
| | - Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Rosanna Squitti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy
| |
Collapse
|
8
|
Samadzadeh S, Kruschel T, Novak M, Kallenbach M, Hefter H. Different Response Behavior to Therapeutic Approaches in Homozygotic Wilson's Disease Twins with Clinical Phenotypic Variability: Case Report and Literature Review. Genes (Basel) 2022; 13:1217. [PMID: 35885998 PMCID: PMC9318625 DOI: 10.3390/genes13071217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/17/2022] Open
Abstract
Background: Wilson’s disease (WD) is an autosomal-recessive disorder of copper deposition caused by pathogenic variants in the copper-transporting ATP7B gene. There is not a clear correlation between genotype and phenotype in WD regarding symptom manifestations. This is supported by the presentation of genetically identical WD twins with phenotypic discordance and different response behavior to WD-specific therapy. Case Presentation: One of the female homozygous twins (age: 26 yrs) developed writing, speaking, swallowing and walking deficits which led to in-patient examination without conclusive results but recommended genetic testing. Both sisters were tested and were heterozygous for the C.2304dupC;p(Met769Hisf*26) and the C.3207C>A;p(His1069Gln) mutation. Self-medication of the affected sibling with 450 mg D-penicillamine (DPA) did not prevent further deterioration. She developed a juvenile parkinsonian syndrome and became wheelchair-bound and anarthric. A percutaneous endoscopic gastrostomy was applied. Her asymptomatic sister helped her with her daily life. Despite the immediate increase of the DPA dose (up to 1800 mg within 3 weeks) in the severely affected patient and the initiation of DPA therapy (up to 600 mg within 2 weeks) in the asymptomatic patient after the first visit in our institution, liver function tests further deteriorated in both patients. After 2 months, the parkinsonian patient started to improve and walk again, but experienced several falls, broke her right shoulder and underwent two necessary surgical interventions. With further consequent copper elimination therapy, liver dysfunction improved in both patients, without need for orthotopic liver transplantation (LTX) in the severely affected patient. Her excellent recovery of liver and brain dysfunction was only transiently interrupted by the development of a nephrotic syndrome which disappeared after switching to Cuprior®. Unfortunately, she died from fulminant pneumonia. Conclusion: Despite identical genetic disposition, WD symptom presentations may develop differently in monozygotic twins, and they may need to be placed on a very different therapeutical regimen. The underlying gene-environment interaction is unclear so far.
Collapse
Affiliation(s)
- Sara Samadzadeh
- Department of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (S.S.); (T.K.); (M.N.)
| | - Theodor Kruschel
- Department of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (S.S.); (T.K.); (M.N.)
| | - Max Novak
- Department of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (S.S.); (T.K.); (M.N.)
| | - Michael Kallenbach
- Department of Gastroenterology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany;
| | - Harald Hefter
- Department of Neurology, University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany; (S.S.); (T.K.); (M.N.)
| |
Collapse
|
9
|
Genome-wide association and Mendelian randomization study of blood copper levels and 213 deep phenotypes in humans. Commun Biol 2022; 5:405. [PMID: 35501403 PMCID: PMC9061855 DOI: 10.1038/s42003-022-03351-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 04/10/2022] [Indexed: 11/29/2022] Open
Abstract
Metal elements are present in the human body, and their levels in the blood have important impacts on health. In this study, 2488 Chinese individuals were included in a genome-wide association study of 21 serum metal levels, with approximately 179,000 East Asian individuals in a bidirectional two-sample Mendelian randomization (MR) analysis, and 628,000 Europeans in a two-sample MR analysis. We identified two single nucleotide polymorphisms (SNPs) rs35691438 and rs671 that were significantly associated with serum copper levels (SCLs). The bidirectional two-sample MR analysis in the East Asian population showed that gamma-glutamyl transpeptidase levels have a causal effect on SCLs. SCLs have causal effects on six outcomes, namely risks of esophageal varix, glaucoma, sleep apnea syndrome, and systemic lupus erythematosus, white blood cell count, and usage of drugs affecting bone structure and mineralization. The two-sample MR analyses in the European population showed causal effects of erythrocyte copper levels on risks of carpal tunnel syndrome and compression fracture. Our results provide original insights into the causal relationship between blood metal levels and multiple human phenotypes. A genome-wide association study in a Chinese population identifies SNPs associated with serum copper levels. Mendelian randomization analysis reveals causal effects on multiple human phenotypes in East Asian and European populations.
Collapse
|
10
|
Ceruloplasmin as Redox Marker Related to Heart Failure Severity. Int J Mol Sci 2021; 22:ijms221810074. [PMID: 34576235 PMCID: PMC8467566 DOI: 10.3390/ijms221810074] [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: 08/01/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
This study examined ceruloplasmin levels in patients with HFrEF, depending on cardiopulmonary exercise testing (CPET) parameters; a correlation was found between ceruloplasmin (CER) and iron and hepatic status, inflammatory and redox biomarkers. A group of 552 patients was divided according to Weber’s classification: there were 72 (13%) patients in class A (peak VO2 > 20 mL/kg/min), 116 (21%) patients in class B (peak VO2 16–20 mL/kg/min), 276 (50%) patients in class C (peak VO2 10–15.9 mL/kg/min) and 88 (16%) patients in class D (peak VO2 < 10 mL/kg/min). A higher concentration of CER was found in patients with peak VO2 < 16 mL/kg/min and VE/CO2 slope > 45 compared to patients with VE/CO2 slope < 45 (escectively CER 30.6 mg/dL and 27.5 mg/dL). A significantly positive correlation was found between ceruloplasmin and NYHA class, RV diameter, NT-proBNP, uric acid, total protein, fibrinogen and hepatic enzymes. CER was positively correlated with both total oxidant status (TOS), total antioxidant capacity (TAC) and malondialdehyde. A model constructed to predict CER concentration indicated that TOS, malondialdehyde and alkaline phosphatase were independent predictive variables (R2 0.14, p < 0.001). CER as a continuous variable was an independent predictor of pVO2 ≤ 12 mL/kg/min after adjustment for sex, age and BMI. These results provide the basis of a new classification to encourage the determination of CER as a useful biomarker in HFrEF.
Collapse
|
11
|
Ma L, Gholam Azad M, Dharmasivam M, Richardson V, Quinn RJ, Feng Y, Pountney DL, Tonissen KF, Mellick GD, Yanatori I, Richardson DR. Parkinson's disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies. Redox Biol 2021; 41:101896. [PMID: 33799121 PMCID: PMC8044696 DOI: 10.1016/j.redox.2021.101896] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
A plethora of studies indicate that iron metabolism is dysregulated in Parkinson's disease (PD). The literature reveals well-documented alterations consistent with established dogma, but also intriguing paradoxical observations requiring mechanistic dissection. An important fact is the iron loading in dopaminergic neurons of the substantia nigra pars compacta (SNpc), which are the cells primarily affected in PD. Assessment of these changes reveal increased expression of proteins critical for iron uptake, namely transferrin receptor 1 and the divalent metal transporter 1 (DMT1), and decreased expression of the iron exporter, ferroportin-1 (FPN1). Consistent with this is the activation of iron regulator protein (IRP) RNA-binding activity, which is an important regulator of iron homeostasis, with its activation indicating cytosolic iron deficiency. In fact, IRPs bind to iron-responsive elements (IREs) in the 3ꞌ untranslated region (UTR) of certain mRNAs to stabilize their half-life, while binding to the 5ꞌ UTR prevents translation. Iron loading of dopaminergic neurons in PD may occur through these mechanisms, leading to increased neuronal iron and iron-mediated reactive oxygen species (ROS) generation. The "gold standard" histological marker of PD, Lewy bodies, are mainly composed of α-synuclein, the expression of which is markedly increased in PD. Of note, an atypical IRE exists in the α-synuclein 5ꞌ UTR that may explain its up-regulation by increased iron. This dysregulation could be impacted by the unique autonomous pacemaking of dopaminergic neurons of the SNpc that engages L-type Ca+2 channels, which imparts a bioenergetic energy deficit and mitochondrial redox stress. This dysfunction could then drive alterations in iron trafficking that attempt to rescue energy deficits such as the increased iron uptake to provide iron for key electron transport proteins. Considering the increased iron-loading in PD brains, therapies utilizing limited iron chelation have shown success. Greater therapeutic advancements should be possible once the exact molecular pathways of iron processing are dissected.
Collapse
Affiliation(s)
- L Ma
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Gholam Azad
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - M Dharmasivam
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - V Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - R J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - Y Feng
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - D L Pountney
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - K F Tonissen
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - G D Mellick
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia
| | - I Yanatori
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - D R Richardson
- School of Environment and Science, Griffith University Nathan, Brisbane, Queensland, Australia; Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan.
| |
Collapse
|
12
|
Abstract
Iron is essential for a variety of physiological processes. Hepatic iron overload acts as a trigger for the progression of hepatic steatosis to nonalcoholic steatohepatitis and hepatocellular carcinoma. In the present study, we aimed to study the effects of iron overload on cellular responses in hepatocytes. Rat primary hepatocytes (RPH), mouse primary hepatocytes (MPH), HepG2 human hepatoma cells and Hepa1-6 mouse hepatoma cells were treated with FeCl3. Treatment with FeCl3 effectively increased iron accumulation in primary hepatocytes. Expression levels of molecules involved in cellular signaling such as AMPK pathway, TGF-β family pathway, and MAP kinase pathway were decreased by FeCl3 treatment in RPH. Cell viability in response to FeCl3 treatment was decreased in RPH but not in HepG2 and Hepa1-6 cells. Treatment with FeCl3 also decreased expression level of LC-3B, a marker of autophagy in RPH but not in liver-derived cell lines. Ultrastructural observations revealed that cell death resembling ferroptosis and necrosis was induced upon FeCl3 treatment in RPH. The expression level of genes involved in iron transport varied among different liver-derived cells- iron is thought to be efficiently incorporated as free Fe2+ in primary hepatocytes, whereas transferrin-iron is the main route for iron uptake in HepG2 cells. The present study reveals specific cellular responses in different liver-derived cells as a consequence of iron overload.
Collapse
|
13
|
Bagwe-Parab S, Kaur G. Molecular targets and therapeutic interventions for iron induced neurodegeneration. Brain Res Bull 2020; 156:1-9. [DOI: 10.1016/j.brainresbull.2019.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/14/2019] [Accepted: 12/17/2019] [Indexed: 01/17/2023]
|
14
|
Asthana M, Syiemlieh I, Kumar A, Lal RA. Direct oxidation of alcohols catalysed by heterometallic complex [CuNi(bz)3(bpy)2]ClO4 to aldehydes and ketones mediated by hydrogen peroxide as a terminal oxidant. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2019.119286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
15
|
Brissot P, Bernard DG, Brissot E, Loréal O, Troadec MB. Rare anemias due to genetic iron metabolism defects. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2018; 777:52-63. [PMID: 30115430 DOI: 10.1016/j.mrrev.2018.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/05/2018] [Accepted: 06/21/2018] [Indexed: 01/19/2023]
Abstract
Anemia is defined by a deficiency of hemoglobin, an iron-rich protein that binds oxygen in the blood. It can be due to multiple causes, either acquired or genetic. Alterations of genes involved in iron metabolism may be responsible, usually at a young age, for rare forms of chronic and often severe congenital anemia. These diseases encompass a variety of sideroblastic anemias, characterized by the presence of ring sideroblasts in the bone marrow. Clinical expression of congenital sideroblastic anemia is either monosyndromic (restricted to hematological lineages) or polysyndromic (with systemic expression), depending on whether iron metabolism, and especially heme synthesis, is directly or indirectly affected. Beside sideroblastic anemias, a number of other anemias can develop due to mutations of key proteins acting either on cellular iron transport (such as the DMT1 transporter), plasma iron transport (transferrin), and iron recycling (ceruloplasmin). Contrasting with the aforementioned entities which involve compartmental, and sometimes, systemic iron excess, the iron refractory iron deficiency anemia (IRIDA) corresponds to a usually severe anemia with whole body iron deficiency related to chronic increase of plasma hepcidin, the systemic negative regulator of plasma iron. Once clinically suggested, these diseases are confirmed by genetic testing in specialized laboratories.
Collapse
Affiliation(s)
- Pierre Brissot
- INSERM, Univ Rennes, INRA, Institut NUMECAN (Nutrition, Metabolisms and Cancer), UMR_S 1241, F-35000 Rennes, France.
| | - Delphine G Bernard
- UMR 1078 "Génétique, Génomique Fonctionnelle et Biotechnologies", INSERM, Univ. Brest, EFS, IBSAM, Brest, France
| | - Eolia Brissot
- Sorbonne Universités, UPMC Univ. Paris 06, AP-HP, Centre de recherche Saint-Antoine, UMR-S938, Paris, France; Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, APHP, Paris, France
| | - Olivier Loréal
- INSERM, Univ Rennes, INRA, Institut NUMECAN (Nutrition, Metabolisms and Cancer), UMR_S 1241, F-35000 Rennes, France
| | - Marie-Bérengère Troadec
- Univ. Rennes, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F- 35000 Rennes, France.
| |
Collapse
|
16
|
Sarkar J, Potdar AA, Saidel GM. Whole-body iron transport and metabolism: Mechanistic, multi-scale model to improve treatment of anemia in chronic kidney disease. PLoS Comput Biol 2018; 14:e1006060. [PMID: 29659573 PMCID: PMC5919696 DOI: 10.1371/journal.pcbi.1006060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/26/2018] [Accepted: 02/27/2018] [Indexed: 02/04/2023] Open
Abstract
Iron plays vital roles in the human body including enzymatic processes, oxygen-transport via hemoglobin and immune response. Iron metabolism is characterized by ~95% recycling and minor replenishment through diet. Anemia of chronic kidney disease (CKD) is characterized by a lack of synthesis of erythropoietin leading to reduced red blood cell (RBC) formation and aberrant iron recycling. Treatment of CKD anemia aims to normalize RBC count and serum hemoglobin. Clinically, the various fluxes of iron transport and accumulation are not measured so that changes during disease (e.g., CKD) and treatment are unknown. Unwanted iron accumulation in patients is known to lead to adverse effects. Current whole-body models lack the mechanistic details of iron transport related to RBC maturation, transferrin (Tf and TfR) dynamics and assume passive iron efflux from macrophages. Hence, they are not predictive of whole-body iron dynamics and cannot be used to design individualized patient treatment. For prediction, we developed a mechanistic, multi-scale computational model of whole-body iron metabolism incorporating four compartments containing major pools of iron and RBC generation process. The model accounts for multiple forms of iron in vivo, mechanisms involved in iron uptake and release and their regulation. Furthermore, the model is interfaced with drug pharmacokinetics to allow simulation of treatment dynamics. We calibrated our model with experimental and clinical data from peer-reviewed literature to reliably simulate CKD anemia and the effects of current treatment involving combination of epoietin-alpha and iron dextran. This in silico whole-body model of iron metabolism predicts that a year of treatment can potentially lead to 90% downregulation of ferroportin (FPN) levels, 15-fold increase in iron stores with only a 20% increase in iron flux from the reticulo-endothelial system (RES). Model simulations quantified unmeasured iron fluxes, previously unknown effects of treatment on FPN-level and iron stores in the RES. This mechanistic whole-body model can be the basis for future studies that incorporate iron metabolism together with related clinical experiments. Such an approach could pave the way for development of effective personalized treatment of CKD anemia.
Collapse
Affiliation(s)
- Joydeep Sarkar
- Pricewaterhouse Coopers LLP, New York, NY, United States of America
| | - Alka A. Potdar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Gerald M. Saidel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- * E-mail:
| |
Collapse
|
17
|
Abstract
Although substantial improvements have been made in majority of cardiac disorders, heart failure (HF) remains a major health problem, with both increasing incidence and prevalence over the past decades. For that reason, the number of potential biomarkers that could contribute to diagnosis and treatment of HF patients is, almost exponentially, increasing over the recent years. The biomarkers that are, at the moment, more or less ready for use in everyday clinical practice, reflect different pathophysiological processes present in HF. In this review, seven groups of biomarkers associated to myocardial stretch (mid-regional proatrial natriuretic peptide, MR-proANP), myocyte injury (high-sensitive troponins, hs-cTn; heart-type fatty acid-binding protein, H-FABP; glutathione transferase P1, GSTP1), matrix remodeling (galectin-3; soluble isoform of suppression of tumorigenicity 2, sST2), inflammation (growth differentiation factor-15, GDF-15), renal dysfunction (neutrophil gelatinase-associated lipocalin, NGAL; kidney injury molecule-1, KIM-1), neurohumoral activation (adrenomedullin, MR-proADM; copeptin), and oxidative stress (ceruloplasmin; myeloperoxidase, MPO; 8-hydroxy-2'-deoxyguanosine, 8-OHdG; thioredoxin 1, Trx1) in HF will be overviewed. It is important to note that clinical value of individual biomarkers within the single time points in both diagnosis and outcome prediction in HF is limited. Hence, the future of biomarker application in HF lies in the multimarker panel strategy, which would include specific combination of biomarkers that reflect different pathophysiological processes underlying HF.
Collapse
|
18
|
Abstract
Iron is a crucial component of heme- and iron-sulfur clusters, involved in vital cellular functions such as oxygen transport, DNA synthesis, and respiration. Both excess and insufficient levels of iron and heme-precursors cause human disease, such as iron-deficiency anemia, hemochromatosis, and porphyrias. Hence, their levels must be tightly regulated, requiring a complex network of transporters and feedback mechanisms. The use of zebrafish to study these pathways and the underlying genetics offers many advantages, among others their optical transparency, ex-vivo development and high genetic and physiological conservations. This chapter first reviews well-established methods, such as large-scale mutagenesis screens that have led to the initial identification of a series of iron and heme transporters and the generation of a variety of mutant lines. Other widely used techniques are based on injection of RNA, including complementary morpholino knockdown and gene overexpression. In addition, we highlight several recently developed approaches, most notably endonuclease-based gene knockouts such as TALENs or the CRISPR/Cas9 system that have been used to study how loss of function can induce human disease phenocopies in zebrafish. Rescue by chemical complementation with iron-based compounds or small molecules can subsequently be used to confirm causality of the genetic defect for the observed phenotype. All together, zebrafish have proven to be - and will continue to serve as an ideal model to advance our understanding of the pathogenesis of human iron and heme-related diseases and to develop novel therapies to treat these conditions.
Collapse
Affiliation(s)
| | - Barry H. Paw
- Brigham & Women’s Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Dana-Farber Cancer Institute, Boston, MA, United States
- Boston Children’s Hospital, Boston, MA, United States
| |
Collapse
|
19
|
Susnea I, Weiskirchen R. Trace metal imaging in diagnostic of hepatic metal disease. MASS SPECTROMETRY REVIEWS 2016; 35:666-686. [PMID: 25677057 DOI: 10.1002/mas.21454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/25/2014] [Accepted: 12/02/2014] [Indexed: 06/04/2023]
Abstract
The liver is the most central organ and the largest gland of the body that influences and controls a variety of metabolic and catabolic processes. It produces inconceivable many essential proteins, is responsible for the recovery of various food components, degrades toxins, mediates the bile production, and is involved in the excretion of unwanted metabolites. Several of these anabolic or catabolic functions of the liver depend on trace elements. These are either integral part of enzymes, cofactors, or act as chemical catalysts. Therefore, a lack of trace elements can lead to organ failure or systemic illness. Conversely, excessive hepatic trace element deposition resulting from genetic disorders, intoxication, extensive dietary supply, or long-term parenteral nutrition may cause hepatic inflammation, fibrosis, cirrhosis, and even hepatocellular carcinoma. Although specific serum parameters currently allow rough assessment of metal deficit and excess, the precise quantification of hepatic metal content in liver is presently only possible by different titration or staining techniques of biopsy specimens. Recently, novel innovative metal imaging techniques were developed that are on the way to replace these traditional methods. In the present review, we summarize the function of different trace elements in liver health and disease and discuss the present knowledge on how quantitative biometal imaging techniques such as synchrotron X-ray fluorescence microscopy, secondary ion mass spectrometry, and laser ablation inductively coupled plasma mass spectrometry enrich diagnostics in the detection and quantification of hepatic metal disorders. We will further discuss sample preparation, sensitivity, spatial resolution, specificity, quantification strategies, and potential future applications of metal bioimaging in experimental research and clinical daily routine. © 2015 Wiley Periodicals, Inc. Mass Spec Rev 35:666-686, 2016.
Collapse
Affiliation(s)
- Iuliana Susnea
- Central Institute of Engineering, Electronics and Analytics (ZEA-3), Forschungszentrum Jülich, D-52425, Jülich, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH University Hospital Aachen, D-52074, Aachen, Germany.
| |
Collapse
|
20
|
Freestone D, Denoyer D, Jakab M, Leigh Ackland M, Cater MA, Michalczyk A. Ceruloplasmin is regulated by copper and lactational hormones in PMC42-LA mammary epithelial cell culture models. Metallomics 2016; 8:941-50. [PMID: 27426449 DOI: 10.1039/c6mt00086j] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ceruloplasmin (Cp) is a multicopper ferroxidase that is considered to be an important source of copper in milk for normal neonatal development. We investigated the expression, subcellular localization and secretion of Cp in PMC42-LA cell culture models representative of resting, lactating and suckled human mammary epithelia. Both secreted Cp (sCp) and plasma membrane associated glycosylphosphatidylinositol-linked Cp (GPI-Cp) were expressed in PMC42-LA cells. In all three epithelial models (resting, lactating and suckled), the expression and secretion of copper-bound, ferroxidase active, Cp (holo-Cp) was dependent on media copper concentration. In low copper (bathocuproinedisulphonic acid/d-penicillamine treated models) there was greater than a 2-fold decrease in holo-Cp expression and secretion, which was mirrored by a 2-fold increase in the expression and secretion of copper-free Cp protein (apo-Cp). Cell surface biotinylation studies revealed that the state of PMC42-LA cell differentiation (functionality), and the level of extracellular copper, had no significant effect on the level of plasma membrane bound GPI-Cp. Quantitative real time PCR analyses determined that there was no significant (P > 0.05) difference in Cp mRNA levels across all copper conditions investigated (0, 5, 50 μM). However, there was a significant (P < 0.05) increase (∼2-fold) in Cp mRNA in both the lactating and suckled models in comparison to the resting model. Furthermore, the Cp mRNA increase in response to PMC42-LA differentiation corresponded with more secreted Cp protein, both apo and holo forms, indicating a link between function and Cp requirement. Our results provide significant insight on the regulation of Cp expression and secretion in lactation and copper incorporation into milk.
Collapse
Affiliation(s)
- David Freestone
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, 221 Burwood Highway, Burwood, Victoria 3125, Australia.
| | | | | | | | | | | |
Collapse
|
21
|
Linder MC. Ceruloplasmin and other copper binding components of blood plasma and their functions: an update. Metallomics 2016; 8:887-905. [PMID: 27426697 DOI: 10.1039/c6mt00103c] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We know that blood plasma contains many proteins and also other components that bind copper. The largest contributor to copper in the plasma is ceruloplasmin, which accounts for 40-70 percent. Apart from ceruloplasmin and albumin, most of these components have not been studied extensively, and even for ceruloplasmin and albumin, much remains to be discovered. New components with new functions, and new functions of known components are emerging, some warranting reconsideration of earlier findings. The author's laboratory has been actively involved in research on this topic. This review summarizes and updates our knowledge of the nature and functions of ceruloplasmin and the other known and emerging copper-containing molecules (principally proteins) in this fluid, to better understand how they contribute to copper homeostasis and consider their potential significance to health and disease.
Collapse
Affiliation(s)
- M C Linder
- California State University, Fullerton, CA, USA.
| |
Collapse
|
22
|
Iron metabolism and related genetic diseases: A cleared land, keeping mysteries. J Hepatol 2016; 64:505-515. [PMID: 26596411 DOI: 10.1016/j.jhep.2015.11.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022]
Abstract
Body iron has a very close relationship with the liver. Physiologically, the liver synthesizes transferrin, in charge of blood iron transport; ceruloplasmin, acting through its ferroxidase activity; and hepcidin, the master regulator of systemic iron. It also stores iron inside ferritin and serves as an iron reservoir, both protecting the cell from free iron toxicity and ensuring iron delivery to the body whenever needed. The liver is first in line for receiving iron from the gut and the spleen, and is, therefore, highly exposed to iron overload when plasma iron is in excess, especially through its high affinity for plasma non-transferrin bound iron. The liver is strongly involved when iron excess is related either to hepcidin deficiency, as in HFE, hemojuvelin, hepcidin, and transferrin receptor 2 related haemochromatosis, or to hepcidin resistance, as in type B ferroportin disease. It is less involved in the usual (type A) form of ferroportin disease which targets primarily the macrophagic system. Hereditary aceruloplasminemia raises important pathophysiological issues in light of its peculiar organ iron distribution.
Collapse
|
23
|
Myeloperoxidase-Related Chlorination Activity Is Positively Associated with Circulating Ceruloplasmin in Chronic Heart Failure Patients: Relationship with Neurohormonal, Inflammatory, and Nutritional Parameters. BIOMED RESEARCH INTERNATIONAL 2015; 2015:691693. [PMID: 26539521 PMCID: PMC4619849 DOI: 10.1155/2015/691693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/04/2015] [Indexed: 12/20/2022]
Abstract
Rationale. Heart failure (HF) is accompanied by the development of an imbalance between oxygen- and nitric oxide-derived free radical production leading to protein nitration. Both chlorinating and peroxidase cycle of Myeloperoxidase (MPO) contribute to oxidative and nitrosative stress and are involved in tyrosine nitration of protein. Ceruloplasmin (Cp) has antioxidant function through its ferroxidase I (FeOxI) activity and has recently been proposed as a physiological defense mechanism against MPO inappropriate actions. Objective. We investigated the relationship between plasma MPO-related chlorinating activity, Cp and FeOxI, and nitrosative stress, inflammatory, neurohormonal, and nutritional biomarkers in HF patients. Methods and Results. In chronic HF patients (n = 81, 76 ± 9 years, NYHA Class II (26); Class III (29); Class IV (26)) and age-matched controls (n = 17, 75 ± 11 years, CTR), plasma MPO chlorinating activity, Cp, FeOxI, nitrated protein, free Malondialdehyde, BNP, norepinephrine, hsCRP, albumin, and prealbumin were measured. Plasma MPO chlorinating activity, Cp, BNP, norepinephrine, and hsCRP were increased in HF versus CTR. FeOxI, albumin, and prealbumin were decreased in HF. MPO-related chlorinating activity was positively related to Cp (r = 0.363, P < 0.001), nitrated protein, hsCRP, and BNP and inversely to albumin. Conclusions. Plasma MPO chlorinated activity is increased in elderly chronic HF patients and positively associated with Cp, inflammatory, neurohormonal, and nitrosative parameters suggesting a role in HF progression.
Collapse
|
24
|
Tfp1 is required for ion homeostasis, fluconazole resistance and N-Acetylglucosamine utilization in Candida albicans. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2731-44. [PMID: 26255859 DOI: 10.1016/j.bbamcr.2015.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 11/23/2022]
Abstract
The vacuolar-type H+-ATPase (V-ATPase) is crucial for the maintenance of ion homeostasis. Dysregulation of ion homeostasis affects various aspects of cellular processes. However, the importance of V-ATPase in Candida albicans is not totally clear. In this study, we demonstrated the essential roles of V-ATPase through Tfp1, a putative V-ATPase subunit. Deletion of TFP1 led to generation of an iron starvation signal and reduced total iron content, which was associated with mislocalization of Fet34p that was finally due to disorders in copper homeostasis. Furthermore, the tfp1∆/∆ mutant exhibited weaker growth and lower aconitase activity on nonfermentable carbon sources, and iron or copper addition partially rescued the growth defect. In addition, the tfp1∆/∆ mutant also showed elevated cytosolic calcium levels in normal or low calcium medium that were relevant to calcium release from vacuole. Kinetics of cytosolic calcium response to an alkaline pulse and VCX1 (VCX1 encodes a putative vacuolar Ca2+/H+ exchanger) overexpression assays indicated that the cytosolic calcium status was in relation to Vcx1 activity. Spot assay and concentration-kill curve demonstrated that the tfp1∆/∆ mutant was hypersensitive to fluconazole, which was attributed to reduced ergosterol biosynthesis and CDR1 efflux pump activity, and iron/calcium dysregulation. Interestingly, carbon source utilization tests found the tfp1∆/∆ mutant was defective for growth on N-Acetylglucosamine (GlcNAc) plate, which was associated with ATP depletion due to the decreased ability to catabolize GlcNAc. Taken together, our study gives new insights into functions of Tfp1, and provides the potential to better exploit V-ATPase as an antifungal target.
Collapse
|
25
|
Cavey T, Ropert M, de Tayrac M, Bardou-Jacquet E, Island ML, Leroyer P, Bendavid C, Brissot P, Loréal O. Mouse genetic background impacts both on iron and non-iron metals parameters and on their relationships. Biometals 2015; 28:733-43. [PMID: 26041486 DOI: 10.1007/s10534-015-9862-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/20/2015] [Indexed: 12/17/2022]
Abstract
Iron is reported to interact with other metals. In addition, it has been shown that genetic background may impact iron metabolism. Our objective was to characterize, in mice of three genetic backgrounds, the links between iron and several non-iron metals. Thirty normal mice (C57BL/6, Balb/c and DBA/2; n = 10 for each group), fed with the same diet, were studied. Quantification of iron, zinc, cobalt, copper, manganese, magnesium and rubidium was performed by ICP/MS in plasma, erythrocytes, liver and spleen. Transferrin saturation was determined. Hepatic hepcidin1 mRNA level was evaluated by quantitative RT-PCR. As previously reported, iron parameters were modulated by genetic background with significantly higher values for plasma iron parameters and liver iron concentration in DBA/2 and Balb/c strains. Hepatic hepcidin1 mRNA level was lower in DBA/2 mice. No iron parameter was correlated with hepcidin1 mRNA levels. Principal component analysis of the data obtained for non-iron metals indicated that metals parameters stratified the mice according to their genetic background. Plasma and tissue metals parameters that are dependent or independent of genetic background were identified. Moreover, relationships were found between plasma and tissue content of iron and some other metals parameters. Our data: (i) confirms the impact of the genetic background on iron parameters, (ii) shows that genetic background may also play a role in the metabolism of non-iron metals, (iii) identifies links between iron and other metals parameters which may have implications in the understanding and, potentially, the modulation of iron metabolism.
Collapse
|
26
|
Yang J, Li X, Yang R, Yu X, Yu C, Qian Y, Yu Y. Susceptibility-Weighted Imaging Manifestations in the Brain of Wilson's Disease Patients. PLoS One 2015; 10:e0125100. [PMID: 25915414 PMCID: PMC4411130 DOI: 10.1371/journal.pone.0125100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/20/2015] [Indexed: 12/25/2022] Open
Abstract
Purpose It is well known that patients with Wilson’s disease (WD) suffer copper metabolism disorder. However, recent studies point to an additional iron metabolism disorder in WD patients. The purpose of our study was to examine susceptibility-weighted imaging (SWI) manifestations of WD in the brains of WD patients. Methods A total of 33 patients with WD and 18 normal controls underwent conventional MRI (Magnetic resonance imaging) and SWI. The phase values were measured on SWI-filtered phase images of the bilateral head of the caudate nuclei, globus pallidus, putamen, thalamus, substantia nigra, and red nucleus. Student’s t-tests were used to compare the phase values between WD groups and normal controls. Results The mean phase values for the bilateral head of the caudate nuclei, globus pallidus, putamen, thalamus, substantia nigra, and red nucleus were significantly lower than those in the control group (P < 0.001), and bilateral putamen was most strongly affected. Conclusions There is paramagnetic mineralization deposition in brain gray nuclei of WD patients and SWI is an effective method to evaluate these structures.
Collapse
Affiliation(s)
- Jinjing Yang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaohu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renmin Yang
- Department of Neurology, The Hospital Affiliated of Anhui College of TCM, Hefei, China
| | - Xuen Yu
- Department of Neurology, The Hospital Affiliated of Anhui College of TCM, Hefei, China
| | - Changliang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yinfeng Qian
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Iron transport across the blood-brain barrier: development, neurovascular regulation and cerebral amyloid angiopathy. Cell Mol Life Sci 2014; 72:709-27. [PMID: 25355056 DOI: 10.1007/s00018-014-1771-4] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/10/2014] [Accepted: 10/23/2014] [Indexed: 12/14/2022]
Abstract
There are two barriers for iron entry into the brain: (1) the brain-cerebrospinal fluid (CSF) barrier and (2) the blood-brain barrier (BBB). Here, we review the literature on developmental iron accumulation by the brain, focusing on the transport of iron through the brain microvascular endothelial cells (BMVEC) of the BBB. We review the iron trafficking proteins which may be involved in the iron flux across BMVEC and discuss the plausible mechanisms of BMVEC iron uptake and efflux. We suggest a model for how BMVEC iron uptake and efflux are regulated and a mechanism by which the majority of iron is trafficked across the developing BBB under the direct guidance of neighboring astrocytes. Thus, we place brain iron uptake in the context of the neurovascular unit of the adult brain. Last, we propose that BMVEC iron is involved in the aggregation of amyloid-β peptides leading to the progression of cerebral amyloid angiopathy which often occurs prior to dementia and the onset of Alzheimer's disease.
Collapse
|
28
|
Koch A, Kumar A, De AK, Phukan A, Lal RA. Synthesis, characterization and reactivity of trinuclear Cu(II) complexes derived from disalicylaldehyde malonoyldihydrazone. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2014; 129:103-113. [PMID: 24727168 DOI: 10.1016/j.saa.2014.02.202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 02/25/2014] [Accepted: 02/27/2014] [Indexed: 06/03/2023]
Abstract
Three new homotrinuclear copper(II) complexes [Cu3(slmh)(μ-Cl)2(CH3OH)3]⋅0.5CH3OH (1), [Cu3(slmh)(NO3)2(CH3OH)5]⋅1.5CH3OH (2) and [Cu3(slmh)(μ-ClO4)2(CH3OH)3]⋅2CH3OH (3) from disalicylaldehyde malonoyldihydrazone have been synthesized and characterized. The composition of the complexes has been established on the basis of data obtained from analytical and thermoanalytical data. The structure of the complexes has been discussed in the light of molar conductance, electronic, FT-IR and far-IR spectral data, magnetic moment and EPR spectral studies. The molar conductance values for the complexes in DMSO solution indicate that all of them are non-electrolyte. The magnetic moment values for the complexes suggest considerable metal-metal intramolecular interaction between metal ions in the structural unit of the complexes. The EPR spectral features reveal that at RT, the ground state for the complexes is a mixture of the quartet state (S=3/2) and doublet state (S=1/2). At lower temperature, the ground state for the complexes is dx(2)-y(2) with considerable contribution from dz(2) orbital. Dihydrazone ligand is present in enol form in all of the complexes. The complexes have distorted square pyramidal stereochemistry. The electron transfer reactions of the complexes have been investigated by cyclic voltammetry. Hydrogen peroxide mediated oxidation of benzyl alcohol catalyzed by complex 1 has been studied.
Collapse
Affiliation(s)
- Angira Koch
- Department of Chemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Arvind Kumar
- Department of Chemistry, Faculty of Science and Agriculture, The University of West-Indies, St. Augustine, Trinidad and Tobago
| | - Arjun K De
- Department of Science and Humanities, Tripura Institute of Technology, Narsingarh 799009, Tripura, India
| | - Arnab Phukan
- Department of Chemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Ram A Lal
- Department of Chemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India.
| |
Collapse
|
29
|
Iron homeostasis in breast cancer. Cancer Lett 2014; 347:1-14. [DOI: 10.1016/j.canlet.2014.01.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/16/2013] [Accepted: 01/24/2014] [Indexed: 02/08/2023]
|
30
|
Cabassi A, Binno SM, Tedeschi S, Ruzicka V, Dancelli S, Rocco R, Vicini V, Coghi P, Regolisti G, Montanari A, Fiaccadori E, Govoni P, Piepoli M, de Champlain J. Low serum ferroxidase I activity is associated with mortality in heart failure and related to both peroxynitrite-induced cysteine oxidation and tyrosine nitration of ceruloplasmin. Circ Res 2014; 114:1723-32. [PMID: 24687133 DOI: 10.1161/circresaha.114.302849] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
RATIONALE Ceruloplasmin antioxidant function is mainly related to its ferroxidase I (FeOxI) activity, which influences iron-dependent oxidative and nitrosative radical species generation. Peroxynitrite, whose production is increased in heart failure (HF), can affect ceruloplasmin antioxidant function through amino acid modification. OBJECTIVE We investigated the relationship between FeOxI and ceruloplasmin tyrosine and cysteine modification and explored in a cohort of patients with HF the potential clinical relevance of serum FeOxI. METHODS AND RESULTS In patients with chronic HF (n=96, 76 ± 9 years; New York Heart Association class, 2.9 ± 0.8) and age-matched controls (n=35), serum FeOxI, FeOxII, ceruloplasmin, nitrotyrosine-bound ceruloplasmin, B-type natriuretic peptide, norepinephrine, and high-sensitivity C-reactive protein were measured, and the patients were followed up for 24 months. Ceruloplasmin, B-type natriuretic peptide, norepinephrine, and high-sensitivity C-reactive protein were increased in HF versus controls. FeOxI was decreased in HF (-20%) and inversely related to nitrotyrosine-bound ceruloplasmin (r, -0.305; P=0.003). In HF, FeOxI lower tertile had a mortality rate doubled compared with middle-higher tertiles. FeOxI emerged as a mortality predictor (hazard ratio, 2.95; 95% confidence intervals [1.29-6.75]; P=0.011) after adjustment for age, sex, hypertension, smoking, sodium level, estimated glomerular filtration rate, and high-sensitivity C-reactive protein. In experimental settings, peroxynitrite incubation of serum samples and isolated purified ceruloplasmin reduced FeOxI activity while increasing ceruloplasmin tyrosine nitration and cysteine thiol oxidation. Reduced glutathione prevented peroxynitrite-induced FeOxI drop, tyrosine nitration, and cysteine oxidation; flavonoid(-)-epicatechin, which prevented ceruloplasmin tyrosine nitration but not cysteine oxidation, partially impeded peroxynitrite-induced FeOxI drop. CONCLUSIONS Reduced activity of serum FeOxI is associated with ceruloplasmin nitration and reduced survival in patients with HF. Both ceruloplasmin tyrosine nitration and cysteine thiol oxidation may be operant in vivo in peroxynitrite-induced FeOxI activity inhibition.
Collapse
Affiliation(s)
- Aderville Cabassi
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.).
| | - Simone Maurizio Binno
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Stefano Tedeschi
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Valerie Ruzicka
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Simona Dancelli
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Rossana Rocco
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Vanni Vicini
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Pietro Coghi
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Giuseppe Regolisti
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Alberto Montanari
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Enrico Fiaccadori
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Paolo Govoni
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Massimo Piepoli
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| | - Jacques de Champlain
- From the Cardiorenal Research Unit (A.C., S.M.B., S.T., V.R., S.D., R.R., P.C., G.R., A.M., E.F.), Laboratory of Experimental Physiopathology (A.C., V.R., S.D., P.C., A.M.), and Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy; Histology and Embriology Unit, Department of Biomedical, Biotechnological and Translational Sciences (S.Bi.Bi.T), University of Parma Medical School, Parma, Italy (P.G.); Cardiology Clinic, Azienda Ospedaliera-Universitaria di Parma, Parma, Italy (V.V.); Cardiology Department, Guglielmo da Saliceto Polichirurgico Hospital, Piacenza, Italy (M.P.); and Institut de Recherches Cliniques de Montréal, University of Montreal, Montreal, Quebec, Canada (J.d.C.)
| |
Collapse
|
31
|
Honarmand Ebrahimi K, Dienemann C, Hoefgen S, Than ME, Hagedoorn PL, Hagen WR. The amyloid precursor protein (APP) does not have a ferroxidase site in its E2 domain. PLoS One 2013; 8:e72177. [PMID: 23977245 PMCID: PMC3747053 DOI: 10.1371/journal.pone.0072177] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/02/2013] [Indexed: 02/07/2023] Open
Abstract
The ubiquitous 24-meric iron-storage protein ferritin and multicopper oxidases such as ceruloplasmin or hephaestin catalyze oxidation of Fe(II) to Fe(III), using molecular oxygen as oxidant. The ferroxidase activity of these proteins is essential for cellular iron homeostasis. It has been reported that the amyloid precursor protein (APP) also has ferroxidase activity. The activity is assigned to a ferroxidase site in the E2 domain of APP. A synthetic 22-residue peptide that carries the putative ferroxidase site of E2 domain (FD1 peptide) has been claimed to encompass the same activity. We previously tested the ferroxidase activity of the synthetic FD1 peptide but we did not observe any activity above the background oxidation of Fe(II) by molecular oxygen. Here we used isothermal titration calorimetry to study Zn(II) and Fe(II) binding to the natural E2 domain of APP, and we employed the transferrin assay and oxygen consumption measurements to test the ferroxidase activity of the E2 domain. We found that this domain neither in the presence nor in the absence of the E1 domain binds Fe(II) and it is not able to catalyze the oxidation of Fe(II). Binding of Cu(II) to the E2 domain did not induce ferroxidase activity contrary to the presence of redox active Cu(II) centers in ceruloplasmin or hephaestin. Thus, we conclude that E2 or E1 domains of APP do not have ferroxidase activity and that the potential involvement of APP as a ferroxidase in the pathology of Alzheimer’s disease must be re-evaluated.
Collapse
|
32
|
Multi-copper oxidases and human iron metabolism. Nutrients 2013; 5:2289-313. [PMID: 23807651 PMCID: PMC3738974 DOI: 10.3390/nu5072289] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 05/29/2013] [Accepted: 06/06/2013] [Indexed: 01/13/2023] Open
Abstract
Multi-copper oxidases (MCOs) are a small group of enzymes that oxidize their substrate with the concomitant reduction of dioxygen to two water molecules. Generally, multi-copper oxidases are promiscuous with regards to their reducing substrates and are capable of performing various functions in different species. To date, three multi-copper oxidases have been detected in humans—ceruloplasmin, hephaestin and zyklopen. Each of these enzymes has a high specificity towards iron with the resulting ferroxidase activity being associated with ferroportin, the only known iron exporter protein in humans. Ferroportin exports iron as Fe2+, but transferrin, the major iron transporter protein of blood, can bind only Fe3+ effectively. Iron oxidation in enterocytes is mediated mainly by hephaestin thus allowing dietary iron to enter the bloodstream. Zyklopen is involved in iron efflux from placental trophoblasts during iron transfer from mother to fetus. Release of iron from the liver relies on ferroportin and the ferroxidase activity of ceruloplasmin which is found in blood in a soluble form. Ceruloplasmin, hephaestin and zyklopen show distinctive expression patterns and have unique mechanisms for regulating their expression. These features of human multi-copper ferroxidases can serve as a basis for the precise control of iron efflux in different tissues. In this manuscript, we review the biochemical and biological properties of the three human MCOs and discuss their potential roles in human iron homeostasis.
Collapse
|
33
|
Cornock R, Gambling L, Langley-Evans SC, McArdle HJ, McMullen S. The effect of feeding a low iron diet prior to and during gestation on fetal and maternal iron homeostasis in two strains of rat. Reprod Biol Endocrinol 2013; 11:32. [PMID: 23635304 PMCID: PMC3654972 DOI: 10.1186/1477-7827-11-32] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/09/2013] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Iron deficiency anaemia during pregnancy is a global problem, with short and long term consequences for maternal and child health. Animal models have demonstrated that the developing fetus is vulnerable to maternal iron restriction, impacting on postnatal metabolic and blood pressure regulation. Whilst long-term outcomes are similar across different models, the commonality in mechanistic events across models is unknown. This study examined the impact of iron deficiency on maternal and fetal iron homeostasis in two strains of rat. METHODS Wistar (n=20) and Rowett Hooded Lister (RHL, n=19) rats were fed a control or low iron diet for 4 weeks prior to and during pregnancy. Tissues were collected at day 21 of gestation for analysis of iron content and mRNA/protein expression of regulatory proteins and transporters. RESULTS A reduction in maternal liver iron content in response to the low iron diet was associated with upregulation of transferrin receptor expression and a reduction in hepcidin expression in the liver of both strains, which would be expected to promote increased iron absorption across the gut and increased turnover of iron in the liver. Placental expression of transferrin and DMT1+IRE were also upregulated, indicating adaptive responses to ensure availability of iron to the fetus. There were considerable differences in hepatic maternal and fetal iron content between strains. The higher quantity of iron present in livers from Wistar rats was not explained by differences in expression of intestinal iron transporters, and may instead reflect greater materno-fetal transfer in RHL rats as indicated by increased expression of placental iron transporters in this strain. CONCLUSIONS Our findings demonstrate substantial differences in iron homeostasis between two strains of rat during pregnancy, with variable impact of iron deficiency on the fetus. Whilst common developmental processes and pathways have been observed across different models of nutrient restriction during pregnancy, this study demonstrates differences in maternal adaptation which may impact on the trajectory of the programmed response.
Collapse
Affiliation(s)
- Ruth Cornock
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Lorraine Gambling
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, AB21 9SB, UK
| | - Simon C Langley-Evans
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| | - Harry J McArdle
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, AB21 9SB, UK
| | - Sarah McMullen
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD, UK
| |
Collapse
|
34
|
Borthakur R, Asthana M, Kumar A, Koch A, Lal RA. Solvent free selective oxidation of alcohols catalyzed by a trinuclear complex with a dicopper(ii)–monozinc(ii) centre using hydrogen peroxide as an oxidant. RSC Adv 2013. [DOI: 10.1039/c3ra44970j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
35
|
Loréal O, Bardou-Jacquet E, Jouanolle AM, Gandon Y, Deugnier Y, Brissot P, Ropert M. Métabolisme du fer et outils diagnostiques pour le clinicien. Rev Med Interne 2012; 33 Suppl 1:S3-9. [DOI: 10.1016/j.revmed.2012.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
36
|
Du F, Qian ZM, Gong Q, Zhu ZJ, Lu L, Ke Y. The iron regulatory hormone hepcidin inhibits expression of iron release as well as iron uptake proteins in J774 cells. J Nutr Biochem 2012; 23:1694-700. [PMID: 22560353 DOI: 10.1016/j.jnutbio.2011.12.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 11/29/2011] [Accepted: 12/02/2011] [Indexed: 10/28/2022]
Abstract
The mechanism by which hepcidin controls cellular iron release protein ferroportin 1 (Fpn1) in macrophages has been well established. However, little is known about the effects of hepcidin on cellular iron uptake proteins. Here, we demonstrated for the first time that hepcidin can significantly inhibit the expression of transferrin receptor 1 (TfR1) and divalent metal transporter 1 in addition to Fpn1, and therefore reduce transferrin-bound iron and non-transferrin-bound iron uptake and also iron release in J774 macrophages. Analysis of mechanisms using the iron-depleted cells showed that hepcidin has a direct inhibitory effect on all iron transport proteins we examined. Further studies demonstrated that the down-regulation of TfR1 induced by hepcidin is associated with cyclic adenosine monophosphate (cAMP) and protein kinase A (PKA), probably being mediated by the cAMP-PKA pathway in J774 macrophages.
Collapse
Affiliation(s)
- Fang Du
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | | | | | |
Collapse
|
37
|
Pfeiffenberger J, Gotthardt DN, Herrmann T, Seessle J, Merle U, Schirmacher P, Stremmel W, Weiss KH. Iron metabolism and the role of HFE gene polymorphisms in Wilson disease. Liver Int 2012; 32:165-70. [PMID: 22098612 DOI: 10.1111/j.1478-3231.2011.02661.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 09/09/2011] [Indexed: 12/12/2022]
Abstract
UNLABELLED Wilson disease (WD) is a rare inherited disorder of copper metabolism, which can lead to severe liver failure and to a variety of neuropsychiatric symptoms. Previous animal studies and case reports suggest that hepatic iron overload and alterations in iron processing are associated with WD. The aim of this study was the assessment of iron metabolism and of the frequency of the most common HFE gene polymorphisms in WD patients. PATIENTS AND METHODS Data from 143 patients with WD were analysed. Clinical presentation, liver function and iron metabolism parameters were recorded. Blood samples of the patients were analysed for HFE gene alterations (H63D; C282Y). Twenty-seven liver biopsies of these patients were studied with regard to iron content and fibrosis score. RESULTS Contrary to previous reports of HFE gene polymorphisms in WD patients, in our cohort the allele frequencies (C282Y: 2.1%; H63D: 7.3%) were in line with frequencies obtained for general population. Male WD patients with decreased serum ceruloplasmin (Cp), showed increased serum ferritin levels. Hepatic iron content was normal in most cases. DISCUSSION Male patients with very low Cp serum concentrations showed slightly elevated median serum ferritin concentrations, probably related to lack of ferroxidase acitivity. However, in consideration of absolute numbers of ferritin concentrations, these changes seem to be of minor clinical relevance.
Collapse
Affiliation(s)
- Jan Pfeiffenberger
- Department of Gastroenterology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Marques L, Auriac A, Willemetz A, Banha J, Silva B, Canonne-Hergaux F, Costa L. Immune cells and hepatocytes express glycosylphosphatidylinositol-anchored ceruloplasmin at their cell surface. Blood Cells Mol Dis 2011; 48:110-20. [PMID: 22178061 DOI: 10.1016/j.bcmd.2011.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 10/21/2011] [Accepted: 11/09/2011] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ceruloplasmin is a positive acute-phase protein with both anti- and pro-oxidant activities, thus having still unclear physiological functions in inflammatory processes. Importantly, ceruloplasmin has been implicated in iron metabolism due to its ferroxidase activity, assisting ferroportin on cellular iron efflux. Ceruloplasmin can be expressed as a secreted or as a membrane glycosylphosphatidylinositol-anchored protein (GPI-ceruloplasmin), this latter one being reported as expressed mostly in the brain. DESIGN AND METHODS We studied the expression of both ceruloplasmin isoforms in human peripheral blood lymphocytes, monocytes, mouse macrophages and human hepatocarcinoma cell line HepG2, using immunofluorescence and immunoblotting techniques. Co-localization of ceruloplasmin and ferroportin was also investigated by immunofluorescence in mouse macrophages. RESULTS Ceruloplasmin was detected by immunoblotting and immunofluorescence in membrane and cytosol of all cell types. The cell surface ceruloplasmin was identified as the GPI-isoform and localized in lipid rafts from monocytes, macrophages and HepG2 cells. In macrophages, increased expression levels and co-localization of ferroportin and GPI-ceruloplasmin in cell surface lipid rafts were observed after iron treatment. Such iron upregulation of ceruloplasmin was not observed in HepG2. CONCLUSIONS Our results revealed an unexpected ubiquitous expression of the GPI-ceruloplasmin isoform in immune and hepatic cells. Different patterns of regulation of ceruloplasmin in these cells may reflect distinct physiologic functions of this oxidase. In macrophages, GPI-ceruloplasmin and ferroportin likely interact in lipid rafts to export iron from cells. Precise knowledge about ceruloplasmin isoforms expression and function in various cell types will help to clarify the role of ceruloplasmin in many diseases related to iron metabolism, inflammation and oxidative biology.
Collapse
Affiliation(s)
- Liliana Marques
- National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | | | | | | | | | | | | |
Collapse
|
39
|
Strickland NJ, Matsha T, Erasmus RT, Zaahl MG. Molecular analysis of Ceruloplasmin in a South African cohort presenting with oesophageal cancer. Int J Cancer 2011; 131:623-32. [DOI: 10.1002/ijc.26418] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 08/23/2011] [Indexed: 12/15/2022]
|
40
|
Jolliff JS, Mahan DC. Effect of injected and dietary iron in young pigs on blood hematology and postnatal pig growth performance. J Anim Sci 2011; 89:4068-80. [PMID: 21873542 DOI: 10.2527/jas.2010-3736] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The relationship of injected Fe doses on blood hematology and pig growth performance during both preweaning and postweaning periods was studied. In Exp. 1, the effect of BW of 347 pigs injected with 200 mg of Fe (dextran) intramuscularly (i.m.) at birth on hemoglobin (Hb) and percent hematocrit (Hct) at weaning was assessed. As BW increased there was a decline (P < 0.01) in Hb and Hct. In Exp. 2, Fe injection doses and timing of injected Fe on blood hematology and pig growth were evaluated. Injections were as follows: 1) 200 mg of Fe at birth; 2) 300 mg of Fe at birth; or 3) 200 mg of Fe at birth + 100 mg of Fe at d 10. A total of 269 pigs were allotted within litter to 3 treatments. The 2 greater quantities of injected Fe (i.e., 300 or 200 + 100 mg of Fe) had similar but greater (P < 0.05) Hb and Hct values than pigs receiving 200 mg of Fe, but growth rates were similar at weaning. The effects of injecting 200 mg of Fe at birth and either saline or 100 mg of Fe at 10 d of age were investigated in Exp. 3. Weaned pigs of each group were fed diets with 0, 80, or 160 mg/kg of added Fe for 35 d as a 2 × 3 factorial arrangement with 12 replicates (n = 360 pigs) in a randomized complete block design (RCB). The innate Fe contents of diets averaged 200 mg/kg. The greater Fe injection group (200 + 100 mg) had greater (P < 0.01) Hb and Hct values through 14 d postweaning (P < 0.05) and greater (P < 0.01) Hct values through 21 d postweaning. As dietary Fe increased, Hb was greater only at d 14 (P < 0.05 4), whereas Hct increased linearly to d 35 (P < 0.01) postweaning. Dietary Fe resulted in linear increases (P < 0.01) in ADG from d 21 to 35 and d 0 to 35. In Exp. 4, 3 dietary Fe (80, 160, and 240 mg/kg of diet), 2 injected Fe treatments (200 or 300 mg of Fe) at birth, and birth BW (<1.5 or ≥1.5 kg) were evaluated as a 2 × 2 × 3 factorial arrangement of treatments in a RCB design with 6 replicates (n = 280 pigs). The 300 mg of Fe injection group had lighter BW in both birth BW groups, with a birth BW × injected Fe interaction (P < 0.01). This resulted in the lighter birth BW pigs receiving 200 mg of Fe having greater BW gains to 240 mg/kg of dietary Fe, whereas light birth BW pigs injected with 300 mg of Fe plateaued at 160 mg/kg of Fe. Pigs in the heavy birth BW group injected with 200 or 300 mg of Fe at birth responded similarly to dietary Fe postweaning. These results indicate that blood Hb and Hct were affected by pig BW at weaning, but the additional 100 mg of Fe i.m. at 10 d of age increased blood hematology and that Fe injected preweaning affected initial postweaning performance.
Collapse
Affiliation(s)
- J S Jolliff
- The Ohio State University and The Ohio Agricultural Research and Development Center, Columbus, Ohio 43210-1095, USA
| | | |
Collapse
|
41
|
Plasma Ceruloplasmin Ferroxidase Activity Correlates with the Nigral Sonographic Area in Parkinson’s Disease Patients: A Pilot Study. Neurochem Res 2011; 36:2111-5. [DOI: 10.1007/s11064-011-0535-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2011] [Indexed: 10/18/2022]
|
42
|
Schulpis KH, Papastamataki M, Stamou H, Papassotiriou I, Margeli A. The effect of diet on total antioxidant status, ceruloplasmin, transferrin and ferritin serum levels in phenylketonuric children. Acta Paediatr 2010; 99:1565-70. [PMID: 20491710 DOI: 10.1111/j.1651-2227.2010.01890.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To investigate the effect of diet on total antioxidative status (TAS), transferrin, ferritin and ceruloplasmin serum levels in phenylketonuric (PKU) children. PATIENTS AND METHODS Seventeen poorly controlled PKU children underwent clinical and laboratory examinations before, 'off diet', and 60 days after adhering to their special diet 'on diet', whereas controls (N = 24) were examined once. Blood chemistry was performed with the appropriate methodologies. RESULTS Phenylalanine levels differed significantly among the examined groups. Lipids and lipoproteins were higher in 'off diet' than in 'on diet' group, except of high density lipoprotein and apolipoprotein AI that remained unaffected. Total antioxidative status (386 ± 30 vs 204 ± 23 μmol/L, p < 0.001), ferritin (48.2 ± 2.3 vs 33.0 ± 2.8 μg/L, p < 0.001) and ceruloplasmin (40.02 ± 2.5 vs 25.5 ± 2.8 mg/dL, p < 0.001) levels were significantly higher in 'on diet' patients' group compared to 'off diet' one. The low lipoprotein and the high TAS and ferritin levels in patients with PKU 'on diet' may be related to the vegetarian diet and the rich in iron formula supplementation. CONCLUSIONS The low ferritin levels found in 'off diet' patients with PKU may be attributed to a decreased liver production of ceruloplasmin, which evaluation may be a useful tool for the follow-up of patients with PKU.
Collapse
|
43
|
Tapryal N, Mukhopadhyay C, Mishra MK, Das D, Biswas S, Mukhopadhyay CK. Glutathione synthesis inhibitor butathione sulfoximine regulates ceruloplasmin by dual but opposite mechanism: Implication in hepatic iron overload. Free Radic Biol Med 2010; 48:1492-500. [PMID: 20211720 DOI: 10.1016/j.freeradbiomed.2010.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 01/10/2010] [Accepted: 02/19/2010] [Indexed: 12/01/2022]
Abstract
Glutathione (GSH) depletion is often detected in chronic pathological conditions like hepatitis C infection, alcohol consumption or xenobiotic assault with simultaneous reactive oxygen species (ROS) generation and hepatic iron overload. However, relation between GSH depletion and regulators of iron homeostasis is not clear so far. To determine that hepatic HepG2 cells were treated with GSH synthesis inhibitor butathione sulfoximine (BSO) and a dual regulation of ceruloplasmin (Cp) that involves in hepatic iron release was detected unlike other iron homeostasis regulators. BSO treatment that caused marginal GSH deficiency increased Cp synthesis due to increased transcription mediated by activator protein (AP)-1-binding site. In higher GSH deficiency (> 40 %) with increased ROS generation, Cp expression was decreased due to promotion of Cp mRNA decay mediated by 3'untranslated region (3'UTR) as found by transfecting chimera of chloramphenicol acetyl transferase (CAT) gene with Cp 3'UTR. RNA gel shift assay showed significant reduction in 3'UTR binding protein complex in similar condition. Decreased CAT expression and RNA-protein complex binding are reversed by pretreatment with antioxidant N-acetyl cysteine suggesting 3'UTR binding protein complex is redox-sensitive. This unique and opposite regulation of Cp provides a mechanism of hepatic iron-deposition during glutathione deficiency detected in chronic pathological conditions.
Collapse
Affiliation(s)
- Nisha Tapryal
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi - 110 067, India
| | | | | | | | | | | |
Collapse
|
44
|
Yeager MP, Coleman RA. In silico evidence for glutathione- and iron-related pathogeneses in Parkinson's disease. J Neurosci Methods 2010; 188:151-64. [DOI: 10.1016/j.jneumeth.2010.01.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 01/25/2010] [Accepted: 01/29/2010] [Indexed: 12/20/2022]
|
45
|
Johnstone D, Milward EA. Molecular genetic approaches to understanding the roles and regulation of iron in brain health and disease. J Neurochem 2010; 113:1387-402. [PMID: 20345752 DOI: 10.1111/j.1471-4159.2010.06697.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Iron is essential in the brain, yet too much iron can be toxic. Tight regulation of iron in the brain may involve intrinsic mechanisms that control internal homeostasis independent of systemic iron status. Iron abnormalities occur in various neurological disorders, usually with symptoms or neuropathology associated with movement impairment or behavioral disturbances rather than cognitive impairment or dementia. Consistent with this, polymorphisms in the HFE gene, associated with the iron overload disorder hemochromatosis, show stronger associations with the movement disorder amyotrophic lateral sclerosis (motor neuron disease) than with cognitive impairment. Such associations may arise because certain brain regions involved in movement or executive control are particularly iron-rich, notably the basal ganglia, and may be highly reliant on iron. Various mechanisms, including iron redistribution causing functional iron deficiency, lysosomal and mitochondrial abnormalities or oxidative damage, could underlie iron-related neuropathogenesis. Clarifying how iron contributes causatively to neurodegeneration may improve treatment options in a range of neurodegenerative disorders. This review considers how modern molecular genetic approaches can be applied to resolve the complex molecular systems and pathways by which brain iron homeostasis is regulated and the molecular changes that occur with iron dyshomeostasis and neuropathogenesis.
Collapse
Affiliation(s)
- Daniel Johnstone
- School of Biomedical Sciences and Pharmacy and Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| | | |
Collapse
|
46
|
Sorbello O, Sini M, Civolani A, Demelia L. HFE gene mutations and Wilson's disease in Sardinia. Dig Liver Dis 2010; 42:216-9. [PMID: 19640812 DOI: 10.1016/j.dld.2009.06.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Revised: 06/14/2009] [Accepted: 06/17/2009] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hypocaeruloplasminaemia can lead to tissue iron storage in Wilson's disease and the possibility of iron overload in long-term overtreated patients should be considered. The HFE gene encodes a protein that is intimately involved in intestinal iron absorption. AIMS The aim of this study was to determine the prevalence of the HFE gene mutation, its role in iron metabolism of Wilson's disease patients and the interplay of therapy in copper and iron homeostasis. METHODS The records of 32 patients with Wilson's disease were reviewed for iron and copper indices, HFE gene mutations and liver biopsy. RESULTS Twenty-six patients were negative for HFE gene mutations and did not present significant alterations of iron metabolism. The HFE mutation was significantly associated with increased hepatic iron content (P<0.02) and transferrin saturation index (P<0.03). After treatment period, iron indices were significantly decreased only in HFE gene wild-type. CONCLUSIONS The HFE gene mutations may be an addictional factor in iron overload in Wilson's disease. Our results showed that an adjustment of dosage of drugs could prevent further iron overload induced by overtreatment only in patients HFE wild-type.
Collapse
Affiliation(s)
- Orazio Sorbello
- Department of Gastroenterology, Azienda Ospedaliero - Universitaria of Cagliari, Italy.
| | | | | | | |
Collapse
|
47
|
Wang SM, Fu LJ, Duan XL, Crooks DR, Yu P, Qian ZM, Di XJ, Li J, Rouault TA, Chang YZ. Role of hepcidin in murine brain iron metabolism. Cell Mol Life Sci 2009; 67:123-33. [PMID: 19898775 DOI: 10.1007/s00018-009-0167-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/27/2009] [Accepted: 09/29/2009] [Indexed: 12/21/2022]
Abstract
Brain iron homeostasis is maintained by a balance of both iron uptake and release, and accumulating evidence has revealed that brain iron concentrations increase with aging. Hepcidin, an iron regulatory hormone produced by hepatocytes in response to inflammatory stimuli, iron, and hypoxia, has been shown to be the long-sought hormone responsible for the regulation of body iron balance and recycling in mammals. In this study, we report that hepcidin is widely expressed in the murine brain. In cerebral cortex, hippocampus and striatum, hepcidin mRNA levels increased with aging. Injection of hepcidin into the lateral cerebral ventricle resulted in decreased Fpn1 protein levels in cerebral cortex, hippocampus, and striatum. Additionally, treatment of primary cultured neurons with hepcidin caused decreased neuronal iron release and Fpn1 protein levels. Together, our data provide further evidence that hepcidin may be involved in the regulation of brain iron metabolism.
Collapse
Affiliation(s)
- S-M Wang
- Laboratory of Molecular Iron Metabolism, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Collins JF, Hua P, Lu Y, Ranganathan PN. Alternative splicing of the Menkes copper Atpase (Atp7a) transcript in the rat intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 2009; 297:G695-707. [PMID: 19679821 PMCID: PMC2763807 DOI: 10.1152/ajpgi.00203.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The intestinal Menkes copper Atpase (Atp7a) gene is strongly induced by iron deficiency in the rat intestine. We sought to develop an in vitro model to understand the mechanism of this induction by performing molecular studies in native rat intestine and in intestinal epithelial (IEC-6) cells. IEC-6 cells express Atp7a, and induction was noted with iron deprivation. 5' Rapid amplification of cDNA ends and PCR experiments revealed three splice variants in rat intestine and IEC-6 cells; all variants were strongly induced during iron deprivation (five- to sevenfold). The splice variants presumably encode proteins that would either contain the extreme NH(2) terminus of the protein (containing copper binding domain 1) or not. We thus hypothesized that more than one version of Atp7a protein exists. Antibodies against this NH(2)-terminal region of the protein were developed (named N-term) and used along with previously reported antibodies (against more COOH-terminal regions, termed 54-10) to perform immunoblotting and immunolocalization studies. Results with the 54-10 antiserum revealed an Atp7a protein variant of approximately 190 kDa that localized to the trans-Golgi network of IEC-6 cells and trafficked to the plasma membrane with copper loading. Using the N-term antiserum, however, we noted protein of approximately 97 and 64 kDa. The 97-kDa protein was cytosolic and nuclear, whereas the 64-kDa protein was nuclear specific. Immunolocalization analyses with the N-term antiserum showed strong staining of nuclei in IEC-6 and Caco-2 cells and in rat intestine. We conclude that novel Atp7a protein variants may exist in rat and human intestinal epithelial cells, with different intracellular locations and potentially distinct physiological functions.
Collapse
Affiliation(s)
- James F. Collins
- Food Science and Human Nutrition Department, Institute for Food and Agricultural Sciences, University of Florida, Gainesville, Florida
| | - Ping Hua
- Food Science and Human Nutrition Department, Institute for Food and Agricultural Sciences, University of Florida, Gainesville, Florida
| | - Yan Lu
- Food Science and Human Nutrition Department, Institute for Food and Agricultural Sciences, University of Florida, Gainesville, Florida
| | - P. N. Ranganathan
- Food Science and Human Nutrition Department, Institute for Food and Agricultural Sciences, University of Florida, Gainesville, Florida
| |
Collapse
|
49
|
Hongsachart P, Huang-Liu R, Sinchaikul S, Pan FM, Phutrakul S, Chuang YM, Yu CJ, Chen ST. Glycoproteomic analysis of WGA-bound glycoprotein biomarkers in sera from patients with lung adenocarcinoma. Electrophoresis 2009; 30:1206-20. [PMID: 19294700 DOI: 10.1002/elps.200800405] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Differential protein expression profiles in the serum samples from patients with lung adenocarcinoma may be associated with glycosylation during cancer development. In this study, we used various glycoproteomic approaches to investigate the different glycoproteomic profiles of human normal and lung adenocarcinoma serum samples and to investigate putative altered glycoprotein biomarkers. In our preliminary screening, FITC-labeled lectin staining was used for the detection of specific glycoprotein profiles. wheat germ agglutinin (WGA) lectin had the highest level of specific binding to glycoproteins in both samples. We enriched for glycoproteins in the serum samples using WGA lectin affinity and then performed co-immunoprecipitation with anti-haptoglobin and 2-DE, 2-D difference in-gel electrophoresis and MS analyses. From these analyses, we identified 39 differentially expressed proteins, including 27 up-regulated proteins and 12 down-regulated proteins. Bioinformatics tools were used to search for protein ontology, category classifications and prediction of glycosylation sites. In addition, three up-regulated glycoproteins (adiponectin, cerulolasmin and glycosylphosphatidyl-inositol-80) and two down-regulated glycoproteins (cyclin H and Fyn) that were found to be correlated with lung cancer development were validated by Western blot analysis. We suggest that these altered glycoproteins may be useful as biomarkers for lung cancer development and progression.
Collapse
Affiliation(s)
- Piyorot Hongsachart
- Institute of Biological Chemistry and Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kurup RK, Kurup PA. HYPOTHALAMIC DIGOXIN, HEMISPHERIC CHEMICAL DOMINANCE, AND ONCOGENESIS: EVIDENCE FROM MULTIPLE MYELOMA. Int J Neurosci 2009; 113:1719-40. [PMID: 14602544 DOI: 10.1080/00207450390245108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This study assessed the changes in the isoprenoid pathway and its metabolites digoxin, dolichol, and ubiquinone in multiple myeloma. The isoprenoid pathway and digoxin status were also studied for comparison in individuals of differing hemispheric dominance to find out the rote of cerebral dominance in the genesis of multiple myeloma and neoplasms. The following parameters were assessed: isoprenoid pathway metabolites, tyrosine and tryptophan catabolites, glycoconjugate metabolism, RBC membrane composition, and free radical metabolism--in multiple myeloma, as well as in individuals of differing hemispheric dominance. There was elevation in plasma HMG CoA reductase activity, serum digoxin, and dolichol, and a reduction in RBC membrane Na(+)-K+ ATPase activity, serum ubiquinone, and magnesium levels. Serum tryptophan, serotonin, nicotine, strychnine, and quinolinic acid were elevated, while tyrosine, dopamine, noradrenaline, and morphine were decreased. The total serum glycosaminoglycans and glycosaminoglycan fractions, the activity of GAG degrading enzymes and glycohydrolases, carbohydrate residues of glycoproteins, and serum glycolipids were elevated. The RBC membrane glycosaminoglycans, hexose, and fucose residues of glycoproteins, cholesterol, and phospholipids were reduced. The activity of all free-radical scavenging enzymes, concentration of glutathione, iron binding capacity, and ceruloplasmin decreased significantly, while the concentration of lipid peroxidation products and nitric oxide increased. Hyperdigoxinemia-related altered intracellular Ca++/Mg++ ratios mediated oncogene activation, dolichol-induced altered glycoconjugate metabolism, and ubiquinone deficiency-related mitochondrial dysfunction can contribute to the pathogenesis of multiple myeloma. The biochemical patterns obtained in multiple myeloma are similar to those obtained in left-handed/right hemispheric chemically dominant individuals by the dichotic listening test. But all the patients with multiple myeloma were right-handed/left hemispheric dominant by the dichotic listening test. Hemispheric chemical dominance has no correlation with handedness or the dichotic listening test. Multiple myeloma occurs in right hemispheric chemically dominant individuals and is a reflection of altered brain function.
Collapse
Affiliation(s)
- Ravi Kumar Kurup
- Department of Neurology, Medical College Hospital, Trivandrum, Kerala, India
| | | |
Collapse
|