1
|
Calabro FJ, Parr AC, Sydnor VJ, Hetherington H, Prasad KM, Ibrahim TS, Sarpal DK, Famalette A, Verma P, Luna B. Leveraging ultra-high field (7T) MRI in psychiatric research. Neuropsychopharmacology 2024; 50:85-102. [PMID: 39251774 PMCID: PMC11525672 DOI: 10.1038/s41386-024-01980-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/21/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Non-invasive brain imaging has played a critical role in establishing our understanding of the neural properties that contribute to the emergence of psychiatric disorders. However, characterizing core neurobiological mechanisms of psychiatric symptomatology requires greater structural, functional, and neurochemical specificity than is typically obtainable with standard field strength MRI acquisitions (e.g., 3T). Ultra-high field (UHF) imaging at 7 Tesla (7T) provides the opportunity to identify neurobiological systems that confer risk, determine etiology, and characterize disease progression and treatment outcomes of major mental illnesses. Increases in scanner availability, regulatory approval, and sequence availability have made the application of UHF to clinical cohorts more feasible than ever before, yet the application of UHF approaches to the study of mental health remains nascent. In this technical review, we describe core neuroimaging methodologies which benefit from UHF acquisition, including high resolution structural and functional imaging, single (1H) and multi-nuclear (e.g., 31P) MR spectroscopy, and quantitative MR techniques for assessing brain tissue iron and myelin. We discuss advantages provided by 7T MRI, including higher signal- and contrast-to-noise ratio, enhanced spatial resolution, increased test-retest reliability, and molecular and neurochemical specificity, and how these have begun to uncover mechanisms of psychiatric disorders. Finally, we consider current limitations of UHF in its application to clinical cohorts, and point to ongoing work that aims to overcome technical hurdles through the continued development of UHF hardware, software, and protocols.
Collapse
Affiliation(s)
- Finnegan J Calabro
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Ashley C Parr
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerie J Sydnor
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Konasale M Prasad
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Tamer S Ibrahim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Deepak K Sarpal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alyssa Famalette
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Piya Verma
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Beatriz Luna
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Ipsiroglu OS, Pandher PK, Hill O, McWilliams S, Braschel M, Edwards K, Friedlander R, Keys E, Kuo C, Lewis MS, Richardson A, Wagner AL, Wensley D. Iron Deficiency and Restless Sleep/Wake Behaviors in Neurodevelopmental Disorders and Mental Health Conditions. Nutrients 2024; 16:3064. [PMID: 39339663 PMCID: PMC11435348 DOI: 10.3390/nu16183064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/30/2024] Open
Abstract
Iron deficiency (ID) and restlessness are associated with sleep/wake-disorders (e.g., restless legs syndrome (RLS)) and neurodevelopmental disorders (attention deficit/hyperactivity and autism spectrum disorders (ADHD; ASD)). However, a standardized approach to assessing ID and restlessness is missing. We reviewed iron status and family sleep/ID history data collected at a sleep/wake behavior clinic under a quality improvement/quality assurance project. Restlessness was explored through patient and parental narratives and a 'suggested clinical immobilization test'. Of 199 patients, 94% had ID, with 43% having a family history of ID. ADHD (46%) and ASD (45%) were common conditions, along with chronic insomnia (61%), sleep-disordered breathing (50%), and parasomnias (22%). In unadjusted analysis, a family history of ID increased the odds (95% CI) of familial RLS (OR: 5.98, p = 0.0002, [2.35-15.2]), insomnia/DIMS (OR: 3.44, p = 0.0084, [1.37-8.64]), and RLS (OR: 7.00, p = 0.01, [1.49-32.93]) in patients with ADHD, and of insomnia/DIMS (OR: 4.77, p = 0.0014, [1.82-12.5]), RLS/PLMS (OR: 5.83, p = 0.009, [1.54-22.1]), RLS (OR: 4.05, p = 0.01, [1.33-12.3]), and familial RLS (OR: 2.82, p = 0.02, [1.17-6.81]) in patients with ASD. ID and restlessness were characteristics of ADHD and ASD, and a family history of ID increased the risk of sleep/wake-disorders. These findings highlight the need to integrate comprehensive blood work and family history to capture ID in children and adolescents with restless behaviors.
Collapse
Affiliation(s)
- Osman S Ipsiroglu
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
- Divisions of Developmental Pediatrics, Child and Adolescent Psychiatry, and Respirology, BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Parveer K Pandher
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Olivia Hill
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Scout McWilliams
- H-Behaviours Research Lab, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Sleep/Wake-Behaviour Clinic, Sleep Program BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Melissa Braschel
- Clinical Research Support Unit, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Katherine Edwards
- Division of Child and Adolescent Psychiatry, BC Children's Hospital, Department of Psychiatry, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Robin Friedlander
- Division of Child and Adolescent Psychiatry, BC Children's Hospital, Department of Psychiatry, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Elizabeth Keys
- School of Nursing, University of British Columbia (Okanagan), Kelowna, BC V1V 1V7, Canada
| | - Calvin Kuo
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marion Suzanne Lewis
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Pacific Autism Family Network, Richmond, BC V7B 1C7, Canada
| | - Anamaria Richardson
- Granville Pediatrics, 205-5678 Granville Street, Vancouver, BC V6M 1X7, Canada
| | - Alexandra L Wagner
- Department of Pediatrics, Division of Neurology, Charité University Hospital Berlin, 10117 Berlin, Germany
| | - David Wensley
- Division of Respirology, BC Children's Hospital, Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| |
Collapse
|
3
|
Fitzpatrick PF, Daubner SC. Biochemical and biophysical approaches to characterization of the aromatic amino acid hydroxylases. Methods Enzymol 2024; 704:345-361. [PMID: 39300655 DOI: 10.1016/bs.mie.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The aromatic amino acid hydroxylases phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase utilize a non-heme iron to catalyze the hydroxylation of the aromatic rings of their amino acid substrates, with a tetrahydropterin serving as the source of the electrons necessary for the monooxygenation reaction. These enzymes have been subjected to a variety of biochemical and biophysical approaches, resulting in a detailed understanding of their structures and mechanism. We summarize here the experimental approaches that have led to this understanding.
Collapse
Affiliation(s)
- Paul F Fitzpatrick
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, TX, United States.
| | - S Colette Daubner
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
4
|
Uzungil V, Luza S, Opazo CM, Mees I, Li S, Ang CS, Williamson NA, Bush AI, Hannan AJ, Renoir T. Phosphoproteomics implicates glutamatergic and dopaminergic signalling in the antidepressant-like properties of the iron chelator deferiprone. Neuropharmacology 2024; 246:109837. [PMID: 38184274 DOI: 10.1016/j.neuropharm.2024.109837] [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: 08/29/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
BACKGROUND Current antidepressants have limitations due to insufficient efficacy and delay before improvement in symptoms. Polymorphisms of the serotonin transporter (5-HTT) gene have been linked to depression (when combined with stressful life events) and altered response to selective serotonergic reuptake inhibitors. We have previously revealed the antidepressant-like properties of the iron chelator deferiprone in the 5-HTT knock-out (KO) mouse model of depression. Furthermore, deferiprone was found to alter neural activity in the prefrontal cortex of both wild-type (WT) and 5-HTT KO mice. METHODS In the current study, we examined the molecular effects of acute deferiprone treatment in the prefrontal cortex of both genotypes via phosphoproteomics analysis. RESULTS In WT mice treated with deferiprone, there were 22 differentially expressed phosphosites, with gene ontology analysis implicating cytoskeletal proteins. In 5-HTT KO mice treated with deferiprone, we found 33 differentially expressed phosphosites. Gene ontology analyses revealed phosphoproteins that were predominantly involved in synaptic and glutamatergic signalling. In a drug-naïve cohort (without deferiprone administration), the analysis revealed 21 differentially expressed phosphosites in 5-HTT KO compared to WT mice. We confirmed the deferiprone-induced increase in tyrosine hydroxylase serine 40 residue phosphorylation (pTH-Ser40) (initially revealed in our phosphoproteomics study) by Western blot analysis, with deferiprone increasing pTH-Ser40 expression in WT and 5-HTT KO mice. CONCLUSION As glutamatergic and synaptic signalling are dysfunctional in 5-HTT KO mice (and are the target of fast-acting antidepressant drugs such as ketamine), these molecular effects may underpin deferiprone's antidepressant-like properties. Furthermore, dopaminergic signalling may also be involved in deferiprone's antidepressant-like properties.
Collapse
Affiliation(s)
- Volkan Uzungil
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Sandra Luza
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne & Melbourne Health, Carlton, VIC, Australia
| | - Carlos M Opazo
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Isaline Mees
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Shanshan Li
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Nicholas A Williamson
- Bio21 Mass Spectrometry and Proteomics Facility, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Anthony J Hannan
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia
| | - Thibault Renoir
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Australia.
| |
Collapse
|
5
|
Sousa RAL, Yehia A, Abulseoud OA. Attenuation of ferroptosis as a potential therapeutic target for neuropsychiatric manifestations of post-COVID syndrome. Front Neurosci 2023; 17:1237153. [PMID: 37554293 PMCID: PMC10405289 DOI: 10.3389/fnins.2023.1237153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/10/2023] [Indexed: 08/10/2023] Open
Abstract
Coronavirus disease-19 (COVID-19), caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), is associated with the persistence of pre-existing or the emergence of new neurological and psychiatric manifestations as a part of a multi-system affection known collectively as "post-COVID syndrome." Cognitive decline is the most prominent feature among these manifestations. The underlying neurobiological mechanisms remain under intense investigation. Ferroptosis is a form of cell death that results from the excessive accumulation of intracellular reactive iron, which mediates lipid peroxidation. The accumulation of lipid-based reactive oxygen species (ROS) and the impairment of glutathione peroxidase 4 (GPX4) activity trigger ferroptosis. The COVID-19-associated cytokine storm enhances the levels of circulating pro-inflammatory cytokines and causes immune-cell hyper-activation that is tightly linked to iron dysregulation. Severe COVID-19 presents with iron overload as one of the main features of its pathogenesis. Iron overload promotes a state of inflammation and immune dysfunction. This is well demonstrated by the strong association between COVID-19 severity and high levels of ferritin, which is a well-known inflammatory and iron overload biomarker. The dysregulation of iron, the high levels of lipid peroxidation biomarkers, and the inactivation of GPX4 in COVID-19 patients make a strong case for ferroptosis as a potential mechanism behind post-COVID neuropsychiatric deficits. Therefore, here we review the characteristics of iron and the attenuation of ferroptosis as a potential therapeutic target for neuropsychiatric post-COVID syndrome.
Collapse
Affiliation(s)
- Ricardo A. L. Sousa
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Asmaa Yehia
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Osama A. Abulseoud
- Department of Psychiatry and Psychology, Mayo Clinic Arizona, Phoenix, AZ, United States
- Department of Neuroscience, Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine, Phoenix, AZ, United States
| |
Collapse
|
6
|
Fiani D, Engler S, Fields S, Calarge CA. Iron Deficiency in Attention-Deficit Hyperactivity Disorder, Autism Spectrum Disorder, Internalizing and Externalizing Disorders, and Movement Disorders. Child Adolesc Psychiatr Clin N Am 2023; 32:451-467. [PMID: 37147046 DOI: 10.1016/j.chc.2022.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
This article reviews the role of iron in brain development and function, with a focus on the association between iron deficiency (ID) and neuropsychiatric conditions. First, we describe how ID is defined and diagnosed. Second, the role of iron in brain development and function is summarized. Third, we review current findings implicating ID in a number of neuropsychiatric conditions in children and adolescents, including attention deficit hyperactivity disorder and other disruptive behavior disorders, depressive and anxiety disorders, autism spectrum disorder, movement disorders, and other situations relevant to mental health providers. Last, we discuss the impact of psychotropic medication on iron homeostasis.
Collapse
Affiliation(s)
- Dimitri Fiani
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, 8080 N Stadium Dr. Ste 180.35, Houston, TX 77054, USA. https://twitter.com/dimitrifiani
| | - Solangia Engler
- Department of Psychological & Brain Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Sherecce Fields
- Department of Psychological & Brain Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Chadi Albert Calarge
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, 8080 N Stadium Dr. Ste 180.35, Houston, TX 77054, USA; Department of Pediatrics, Baylor College of Medicine, 1102 Bates Avenue, Ste 790, Houston, TX 77030, USA.
| |
Collapse
|
7
|
Fitzpatrick PF. The aromatic amino acid hydroxylases: Structures, catalysis, and regulation of phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase. Arch Biochem Biophys 2023; 735:109518. [PMID: 36639008 DOI: 10.1016/j.abb.2023.109518] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023]
Abstract
The aromatic amino acid hydroxylases phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase are non-heme iron enzymes that catalyze key physiological reactions. This review discusses the present understanding of the common catalytic mechanism of these enzymes and recent advances in understanding the relationship between their structures and their regulation.
Collapse
Affiliation(s)
- Paul F Fitzpatrick
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229, USA.
| |
Collapse
|
8
|
Topiwala A, Wang C, Ebmeier KP, Burgess S, Bell S, Levey DF, Zhou H, McCracken C, Roca-Fernández A, Petersen SE, Raman B, Husain M, Gelernter J, Miller KL, Smith SM, Nichols TE. Associations between moderate alcohol consumption, brain iron, and cognition in UK Biobank participants: Observational and mendelian randomization analyses. PLoS Med 2022; 19:e1004039. [PMID: 35834561 PMCID: PMC9282660 DOI: 10.1371/journal.pmed.1004039] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/01/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Brain iron deposition has been linked to several neurodegenerative conditions and reported in alcohol dependence. Whether iron accumulation occurs in moderate drinkers is unknown. Our objectives were to investigate evidence in support of causal relationships between alcohol consumption and brain iron levels and to examine whether higher brain iron represents a potential pathway to alcohol-related cognitive deficits. METHODS AND FINDINGS Observational associations between brain iron markers and alcohol consumption (n = 20,729 UK Biobank participants) were compared with associations with genetically predicted alcohol intake and alcohol use disorder from 2-sample mendelian randomization (MR). Alcohol intake was self-reported via a touchscreen questionnaire at baseline (2006 to 2010). Participants with complete data were included. Multiorgan susceptibility-weighted magnetic resonance imaging (9.60 ± 1.10 years after baseline) was used to ascertain iron content of each brain region (quantitative susceptibility mapping (QSM) and T2*) and liver tissues (T2*), a marker of systemic iron. Main outcomes were susceptibility (χ) and T2*, measures used as indices of iron deposition. Brain regions of interest included putamen, caudate, hippocampi, thalami, and substantia nigra. Potential pathways to alcohol-related iron brain accumulation through elevated systemic iron stores (liver) were explored in causal mediation analysis. Cognition was assessed at the scan and in online follow-up (5.82 ± 0.86 years after baseline). Executive function was assessed with the trail-making test, fluid intelligence with puzzle tasks, and reaction time by a task based on the "Snap" card game. Mean age was 54.8 ± 7.4 years and 48.6% were female. Weekly alcohol consumption was 17.7 ± 15.9 units and never drinkers comprised 2.7% of the sample. Alcohol consumption was associated with markers of higher iron (χ) in putamen (β = 0.08 standard deviation (SD) [95% confidence interval (CI) 0.06 to 0.09], p < 0.001), caudate (β = 0.05 [0.04 to 0.07], p < 0.001), and substantia nigra (β = 0.03 [0.02 to 0.05], p < 0.001) and lower iron in the thalami (β = -0.06 [-0.07 to -0.04], p < 0.001). Quintile-based analyses found these associations in those consuming >7 units (56 g) alcohol weekly. MR analyses provided weak evidence these relationships are causal. Genetically predicted alcoholic drinks weekly positively associated with putamen and hippocampus susceptibility; however, these associations did not survive multiple testing corrections. Weak evidence for a causal relationship between genetically predicted alcohol use disorder and higher putamen susceptibility was observed; however, this was not robust to multiple comparisons correction. Genetically predicted alcohol use disorder was associated with serum iron and transferrin saturation. Elevated liver iron was observed at just >11 units (88 g) alcohol weekly c.f. <7 units (56 g). Systemic iron levels partially mediated associations of alcohol intake with brain iron. Markers of higher basal ganglia iron associated with slower executive function, lower fluid intelligence, and slower reaction times. The main limitations of the study include that χ and T2* can reflect changes in myelin as well as iron, alcohol use was self-reported, and MR estimates can be influenced by genetic pleiotropy. CONCLUSIONS To the best of our knowledge, this study represents the largest investigation of moderate alcohol consumption and iron homeostasis to date. Alcohol consumption above 7 units weekly associated with higher brain iron. Iron accumulation represents a potential mechanism for alcohol-related cognitive decline.
Collapse
Affiliation(s)
- Anya Topiwala
- Nuffield Department Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Chaoyue Wang
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| | - Klaus P. Ebmeier
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Steven Bell
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Daniel F. Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Celeste McCracken
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | | | - Steffen E. Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kingdom
- Health Data Research UK, London, United Kingdom
- Alan Turing Institute, London, United Kingdom
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Masud Husain
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Karla L. Miller
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| | - Stephen M. Smith
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| | - Thomas E. Nichols
- Nuffield Department Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| |
Collapse
|
9
|
Shahandeh A, Bui BV, Finkelstein DI, Nguyen CTO. Effects of Excess Iron on the Retina: Insights From Clinical Cases and Animal Models of Iron Disorders. Front Neurosci 2022; 15:794809. [PMID: 35185447 PMCID: PMC8851357 DOI: 10.3389/fnins.2021.794809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Iron plays an important role in a wide range of metabolic pathways that are important for neuronal health. Excessive levels of iron, however, can promote toxicity and cell death. An example of an iron overload disorder is hemochromatosis (HH) which is a genetic disorder of iron metabolism in which the body’s ability to regulate iron absorption is altered, resulting in iron build-up and injury in several organs. The retina was traditionally assumed to be protected from high levels of systemic iron overload by the blood-retina barrier. However, recent data shows that expression of genes that are associated with HH can disrupt retinal iron metabolism. Thus, the effects of iron overload on the retina have become an area of research interest, as excessively high levels of iron are implicated in several retinal disorders, most notably age–related macular degeneration. This review is an effort to highlight risk factors for excessive levels of systemic iron build-up in the retina and its potential impact on the eye health. Information is integrated across clinical and preclinical animal studies to provide insights into the effects of systemic iron loading on the retina.
Collapse
Affiliation(s)
- Ali Shahandeh
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - David I. Finkelstein
- Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Christine T. O. Nguyen
- Department of Optometry and Vision Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- *Correspondence: Christine T. O. Nguyen,
| |
Collapse
|
10
|
Xu J, Guan X, Wen J, Wang T, Zhang M, Xu X. Substantia nigra iron affects functional connectivity networks modifying working memory performance in younger adults. Eur J Neurosci 2021; 54:7959-7973. [PMID: 34779047 DOI: 10.1111/ejn.15532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 01/19/2023]
Abstract
Brain iron affects working memory (WM) but the impact of iron content in deep grey matter nuclei on WM networks is unknown. We aimed to test whether deep grey matter nuclei iron concentration can affect resting-state functional connectivity (rsFC) within brain networks modifying WM performance. An N-back WM paradigm was applied in a hundred healthy younger adults. The participants then underwent a resting-state functional magnetic resonance imaging (fMRI) for brain network analysis and quantitative susceptibility mapping (QSM) imaging for assessment of deep grey matter nuclei iron concentration. Higher substantia nigra (SN) iron concentration was associated with lower rsFC between SN and brain regions of the temporal/frontal lobe but with better WM performance after controlling for age, gender and education. A follow-up mediation analysis also indicated that functional connectivity may mediate the link between SN iron and WM performance. Our results suggest that high SN iron concentration may affect communication between the SN and temporal/frontal lobe and is associated with strengthened WM performance in younger adults.
Collapse
Affiliation(s)
- Jingjing Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Wen
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Wang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Personalized Medicine to Improve Treatment of Dopa-Responsive Dystonia-A Focus on Tyrosine Hydroxylase Deficiency. J Pers Med 2021; 11:jpm11111186. [PMID: 34834538 PMCID: PMC8625014 DOI: 10.3390/jpm11111186] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/25/2022] Open
Abstract
Dopa-responsive dystonia (DRD) is a rare movement disorder associated with defective dopamine synthesis. This impairment may be due to the fact of a deficiency in GTP cyclohydrolase I (GTPCHI, GCH1 gene), sepiapterin reductase (SR), tyrosine hydroxylase (TH), or 6-pyruvoyl tetrahydrobiopterin synthase (PTPS) enzyme functions. Mutations in GCH1 are most frequent, whereas fewer cases have been reported for individual SR-, PTP synthase-, and TH deficiencies. Although termed DRD, a subset of patients responds poorly to L-DOPA. As this is regularly observed in severe cases of TH deficiency (THD), there is an urgent demand for more adequate or personalized treatment options. TH is a key enzyme that catalyzes the rate-limiting step in catecholamine biosynthesis, and THD patients often present with complex and variable phenotypes, which results in frequent misdiagnosis and lack of appropriate treatment. In this expert opinion review, we focus on THD pathophysiology and ongoing efforts to develop novel therapeutics for this rare disorder. We also describe how different modeling approaches can be used to improve genotype to phenotype predictions and to develop in silico testing of treatment strategies. We further discuss the current status of mathematical modeling of catecholamine synthesis and how such models can be used together with biochemical data to improve treatment of DRD patients.
Collapse
|
12
|
Xiao G, Zhao M, Liu Z, Du F, Zhou B. Zinc antagonizes iron-regulation of tyrosine hydroxylase activity and dopamine production in Drosophila melanogaster. BMC Biol 2021; 19:236. [PMID: 34732185 PMCID: PMC8564973 DOI: 10.1186/s12915-021-01168-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 10/15/2021] [Indexed: 12/21/2022] Open
Abstract
Background Dopamine (DA) is a neurotransmitter that plays roles in movement, cognition, attention, and reward responses, and deficient DA signaling is associated with the progression of a number of neurological diseases, such as Parkinson’s disease. Due to its critical functions, DA expression levels in the brain are tightly controlled, with one important and rate-limiting step in its biosynthetic pathway being catalyzed by tyrosine hydroxylase (TH), an enzyme that uses iron ion (Fe2+) as a cofactor. A role for metal ions has additionally been associated with the etiology of Parkinson’s disease. However, the way dopamine synthesis is regulated in vivo or whether regulation of metal ion levels is a component of DA synthesis is not fully understood. Here, we analyze the role of Catsup, the Drosophila ortholog of the mammalian zinc transporter SLC39A7 (ZIP7), in regulating dopamine levels. Results We found that Catsup is a functional zinc transporter that regulates intracellular zinc distribution between the ER/Golgi and the cytosol. Loss-of-function of Catsup leads to increased DA levels, and we showed that the increased dopamine production is due to a reduction in zinc levels in the cytosol. Zinc ion (Zn2+) negatively regulates dopamine synthesis through direct inhibition of TH activity, by antagonizing Fe2+ binding to TH, thus rendering the enzyme ineffective or non-functional. Conclusions Our findings uncovered a previously unknown mechanism underlying the control of cellular dopamine expression, with normal levels of dopamine synthesis being maintained through a balance between Fe2+ and Zn2+ ions. The findings also provide support for metal modulation as a possible therapeutic strategy in the treatment of Parkinson’s disease and other dopamine-related diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01168-0.
Collapse
Affiliation(s)
- Guiran Xiao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Mengran Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhihua Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, Anhui, China
| | - Fan Du
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bing Zhou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
13
|
Degremont A, Jain R, Philippou E, Latunde-Dada GO. Brain iron concentrations in the pathophysiology of children with attention deficit/hyperactivity disorder: a systematic review. Nutr Rev 2021; 79:615-626. [PMID: 32974643 DOI: 10.1093/nutrit/nuaa065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CONTEXT Attention deficit/hyperactivity disorder (ADHD) is a neurological disorder associated with iron dysregulation in children. Although previous focus was on examining systemic iron status, brain iron content may be a more reliable biomarker of the disorder. OBJECTIVE This systematic review examines whether children with ADHD have lower serum as well as brain iron concentrations, compared with healthy control subjects (HCS). DATA SOURCES A systematic literature search was conducted in Medline via PubMed, the Cochrane Library, Web of Science, Embase. and Ovid for papers published between 2000 and June 7, 2019. DATA EXTRACTION Studies were included if the mean difference of iron concentration, measured as serum iron, serum ferritin, or brain iron, between children with ADHD and HCS was an outcome measure. DATA ANALYSIS Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Risks of bias within and between studies were assessed using the quality assessment tools of the National Institutes of Health. Of 599 records screened, 20 case-control studies met the inclusion criteria. In 10 of 18 studies in which serum ferritin concentration was assessed, and 2 of 10 studies that assessed serum iron, a significant difference between children with ADHD and HCS was observed. Results of systemic iron levels were inconsistent. In 3 studies in which brain iron concentration was assessed, a statistically significant, lower thalamic iron concentration was found in children with ADHD than in HCS. CONCLUSION The evidence, though limited, reveals that brain iron rather than systemic iron levels may be more associated with the pathophysiology of ADHD in children. Larger, longitudinal, magnetic resonance imaging studies are needed to examine any correlations of iron deficiency in specific brain regions and symptoms of ADHD.
Collapse
Affiliation(s)
- Alexia Degremont
- Department of Nutrition and Dietetics, King's College London, London, UK
| | - Rishika Jain
- Department of Nutrition and Dietetics, King's College London, London, UK
| | - Elena Philippou
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | | |
Collapse
|
14
|
Longitudinal Development of Brain Iron Is Linked to Cognition in Youth. J Neurosci 2020; 40:1810-1818. [PMID: 31988059 DOI: 10.1523/jneurosci.2434-19.2020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/19/2019] [Accepted: 01/04/2020] [Indexed: 11/21/2022] Open
Abstract
Brain iron is vital to multiple aspects of brain function, including oxidative metabolism, myelination, and neurotransmitter synthesis. Atypical iron concentration in the basal ganglia is associated with neurodegenerative disorders in aging and cognitive deficits. However, the normative development of brain iron concentration in adolescence and its relationship to cognition are less well understood. Here, we address this gap in a longitudinal sample of 922 humans aged 8-26 years at the first visit (M = 15.1, SD = 3.72; 336 males, 486 females) with up to four multiecho T2* scans each. Using this sample of 1236 imaging sessions, we assessed the longitudinal developmental trajectories of tissue iron in the basal ganglia. We quantified tissue iron concentration using R2* relaxometry within four basal ganglia regions, including the caudate, putamen, nucleus accumbens, and globus pallidus. The longitudinal development of R2* was modeled using generalized additive mixed models (GAMMs) with splines to capture linear and nonlinear developmental processes. We observed significant increases in R2* across all regions, with the greatest and most prolonged increases occurring in the globus pallidus and putamen. Further, we found that the developmental trajectory of R2* in the putamen is significantly related to individual differences in cognitive ability, such that greater cognitive ability is increasingly associated with greater iron concentration through late adolescence and young-adulthood. Together, our results suggest a prolonged period of basal ganglia iron enrichment that extends into the mid-twenties, with diminished iron concentration associated with poorer cognitive ability during late adolescence.SIGNIFICANCE STATEMENT Brain tissue iron is essential to healthy brain function. Atypical basal ganglia tissue iron levels have been linked to impaired cognition in iron deficient children and adults with neurodegenerative disorders. However, the normative developmental trajectory of basal ganglia iron concentration during adolescence and its association with cognition are less well understood. In the largest study of tissue iron development yet reported, we characterize the developmental trajectory of tissue iron concentration across the basal ganglia during adolescence and provide evidence that diminished iron content is associated with poorer cognitive performance even in healthy youth. These results highlight the transition from adolescence to adulthood as a period of dynamic maturation of tissue iron concentration in the basal ganglia.
Collapse
|
15
|
De La Fuente-Ortega E, Plaza-Briceño W, Vargas-Robert S, Haeger P. Prenatal Ethanol Exposure Misregulates Genes Involved in Iron Homeostasis Promoting a Maladaptation of Iron Dependent Hippocampal Synaptic Transmission and Plasticity. Front Pharmacol 2019; 10:1312. [PMID: 31787896 PMCID: PMC6855190 DOI: 10.3389/fphar.2019.01312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/15/2019] [Indexed: 01/05/2023] Open
Abstract
Prenatal ethanol exposure (PAE) induces behavioral maladptations in offspring, including a deficit in memory formation which is part of the umbrella sign of fetal alcohol spectrum disorder. Clinical and preclinical studies have shown that iron depletion exacerbates cognitive problems in offspring exposed to ethanol in utero and that PAE promotes dysregulation in brain iron homeostasis. However, the mechanisms underlying brain iron dysregulation and neuronal activity defects in adolescent offspring of PAE are unclear and poorly understand. Here, we used a PAE rat model to analyze messenger RNA (mRNA) and protein expression of iron homeostasis genes such as transferrin receptor (TfR), divalent metal transporter (DMT1), ferroportin (FPN1), and ferritin (FT) in brain areas associated with memory formation such as the prefrontal cortex (PFC), ventral tegmental area, and hippocampus. Interestingly, we found that 21 day old PAE rats have higher mRNA expression of DMT1 in the PFC, and TfR in the hippocampus, compared to control animals. In contrast FPN has lower mRNA expression in the PFC, and FT and FPN1 have lower expression in the hippocampus. In agreement with these results, we found a 1.5–2 fold increase of TfR and DMT1 protein levels both in the hippocampus and the PFC. Additionally, using an electrophysiological approach, we found that in hippocampal slices from PAE rats, iron treatment decreased long-term potentiation (LTP), but not AMPAR basal transmission (AMPAR fEPSP). In contrast, in control slices Fe-NTA did not affect LTP but decreased significantly the AMPAR fEPSP. Meanwhile, iron chelation with deferiprone decreased AMPAR transmission in PAE and control slices and decreased LTP only in controls slices. These results suggest that PAE affects iron homeostasis of specific brain areas—PFC and hippocampus—which could be involved in maladaptive cognition observed in this animal model.
Collapse
Affiliation(s)
- Erwin De La Fuente-Ortega
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Wladimir Plaza-Briceño
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Sofía Vargas-Robert
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Paola Haeger
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| |
Collapse
|
16
|
Adisetiyo V, Gray KM, Jensen JH, Helpern JA. Brain iron levels in attention-deficit/hyperactivity disorder normalize as a function of psychostimulant treatment duration. Neuroimage Clin 2019; 24:101993. [PMID: 31479897 PMCID: PMC6726915 DOI: 10.1016/j.nicl.2019.101993] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/09/2019] [Accepted: 08/24/2019] [Indexed: 02/07/2023]
Abstract
Brain iron homeostasis is a dopamine-related mechanism that may be modified with long-term psychostimulant treatment in attention-deficit/hyperactivity disorder (ADHD). We previously reported that while medication-naïve youth with ADHD have reduced brain iron compared to controls and psychostimulant-medicated patients, no differences were detected between the latter groups. In this follow-up study, we examined whether the duration of psychostimulant treatment correlates with the degree of iron normalization. Brain iron was indexed with MRI using an advanced method called magnetic field correlation (MFC) imaging and the conventional R2* proton transverse relaxation rate method. MFC was acquired in 30 psychostimulant-medicated youth with comorbid-free ADHD and 29 age-matched controls (all males). R2* was acquired in a subset of these individuals. Region-of-interest analyses for MFC and R2* group differences and within-group correlations with age and years of psychostimulant treatment were conducted in the globus pallidus (GP), putamen (PUT), caudate nucleus (CN), thalamus (THL) and red nucleus. No significant MFC and R2* group differences were detected. However, while all regional MFC and R2* significantly increased with age in the control group, MFC and R2* increased in the GP, PUT, CN and THL with psychostimulant treatment duration in the ADHD group to a greater degree than with age. Our findings suggest that while youth with ADHD may have less prominent age-related brain iron increases than that seen in typical development, long-term use of psychostimulant medications may compensate through a normalizing effect on basal ganglia iron. Longitudinal studies following ADHD patients before and after long-term psychostimulant treatment are needed to confirm these findings.
Collapse
Affiliation(s)
- Vitria Adisetiyo
- Medical University of South Carolina, Department of Neuroscience, Charleston, SC, USA.
| | - Kevin M Gray
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, Charleston, SC, USA
| | - Jens H Jensen
- Medical University of South Carolina, Department of Neuroscience, Charleston, SC, USA; Medical University of South Carolina, Department of Radiology and Radiological Science, Charleston, SC, USA
| | - Joseph A Helpern
- Medical University of South Carolina, Department of Neuroscience, Charleston, SC, USA; Medical University of South Carolina, Department of Radiology and Radiological Science, Charleston, SC, USA
| |
Collapse
|
17
|
Ye Q, Trivedi M, Zhang Y, Böhlke M, Alsulimani H, Chang J, Maher T, Deth R, Kim J. Brain iron loading impairs DNA methylation and alters GABAergic function in mice. FASEB J 2019; 33:2460-2471. [PMID: 30277817 PMCID: PMC6338660 DOI: 10.1096/fj.201801116rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022]
Abstract
Iron deficiency is closely associated with altered GABA metabolism and affective behavior. While mutation in the hemochromatosis ( HFE) gene disrupts iron homeostasis and promotes oxidative stress that increases the risk of neurodegeneration, it is largely unknown whether HFE mutation modifies GABAergic homeostasis and emotional behavior. The goal of our study was to investigate the impact of HFE on GABAergic neurochemistry and redox-epigenetic regulation in the brain using H67D HFE-mutant mice that recapitulates the H63D-HFE mutation in humans. H67D mice displayed elevated redox-active iron levels in the brain by 32% compared to age-matched wild-type mice. Moreover, the H67D brain had increased isoprostane and decreased glutathione, indicating elevated oxidative stress. Additionally, the H67D brain had decreased global methylation and attenuated DNA methyltransferase (DNMT) activity. Direct addition of iron to purified DNMT in vitro decreased enzyme activity in a concentration-dependent manner. Last, H67D mice exhibited decreased anxiety-like behavior, which was associated with increased expression of the GABAA receptor α2 subunits by 93%, and these changes were also observed in H67D mice fed a low-iron diet. Taken together, our results suggest a putative role of HFE in regulating labile iron status in the brain, and mutation in H67D perturbs redox-methylation status, contributing to GABAergic dysfunction.-Ye, Q., Trivedi, M., Zhang, Y., Böhlke, M., Alsulimani, H., Chang, J., Maher, T., Deth, R., Kim, J. Brain iron loading impairs DNA methylation and alters GABAergic function in mice.
Collapse
Affiliation(s)
- Qi Ye
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Malav Trivedi
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, Florida, USA; and
| | - Yiting Zhang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Mark Böhlke
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Science (MCPHS) University, Boston, Massachusetts, USA
| | - Helal Alsulimani
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - JuOae Chang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Timothy Maher
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Science (MCPHS) University, Boston, Massachusetts, USA
| | - Richard Deth
- Department of Pharmaceutical Sciences, Nova Southeastern University, Fort Lauderdale, Florida, USA; and
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Adisetiyo V, McGill CE, DeVries WH, Jensen JH, Hanlon CA, Helpern JA. Elevated Brain Iron in Cocaine Use Disorder as Indexed by Magnetic Field Correlation Imaging. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2018; 4:579-588. [PMID: 30581153 DOI: 10.1016/j.bpsc.2018.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/26/2018] [Accepted: 11/15/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Iron homeostasis is a critical biological process that may be disrupted in cocaine use disorder (CUD). In the brain, iron is required for neural processes involved in addiction and can be lethal to cells if unbound, especially in excess. Moreover, recent studies have implicated elevated brain iron in conditions of prolonged psychostimulant exposure. Thus, the purpose of this study was to examine iron in basal ganglia reward regions of individuals with CUD using an advanced imaging method called magnetic field correlation (MFC) imaging. METHODS MFC imaging was acquired in 19 non-treatment-seeking individuals with CUD and 19 healthy control individuals (both male and female). Region-of-interest analyses for MFC group differences and within-group correlations with age and years of cocaine use were conducted in the globus pallidus internal segment (GPi), globus pallidus external segment, putamen, caudate nucleus, thalamus, and red nucleus. RESULTS Individuals with CUD had significantly elevated MFC compared with control individuals within the GPi. In control individuals, MFC significantly increased with age in the GPi, globus pallidus external segment, putamen, and caudate nucleus. Conversely, there were no significant MFC within-group correlations in the CUD group. CONCLUSIONS Individuals with CUD have excess iron in the GPi, as indexed by MFC, and lack the age-related gradual iron deposition seen in normal aging. Because the globus pallidus is critical for the transition of goal-directed behavior to compulsive behavior, significantly elevated iron in the GPi may contribute to the persistence of CUD. These findings implicate dysregulation of brain iron homeostasis in CUD and support pursuing this new line of research.
Collapse
Affiliation(s)
- Vitria Adisetiyo
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| | - Corinne E McGill
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - William H DeVries
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Jens H Jensen
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, South Carolina
| | - Colleen A Hanlon
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Joseph A Helpern
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, South Carolina; Department of Neurology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
19
|
Yien YY, Shi J, Chen C, Cheung JTM, Grillo AS, Shrestha R, Li L, Zhang X, Kafina MD, Kingsley PD, King MJ, Ablain J, Li H, Zon LI, Palis J, Burke MD, Bauer DE, Orkin SH, Koehler CM, Phillips JD, Kaplan J, Ward DM, Lodish HF, Paw BH. FAM210B is an erythropoietin target and regulates erythroid heme synthesis by controlling mitochondrial iron import and ferrochelatase activity. J Biol Chem 2018; 293:19797-19811. [PMID: 30366982 DOI: 10.1074/jbc.ra118.002742] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 09/11/2018] [Indexed: 01/01/2023] Open
Abstract
Erythropoietin (EPO) signaling is critical to many processes essential to terminal erythropoiesis. Despite the centrality of iron metabolism to erythropoiesis, the mechanisms by which EPO regulates iron status are not well-understood. To this end, here we profiled gene expression in EPO-treated 32D pro-B cells and developing fetal liver erythroid cells to identify additional iron regulatory genes. We determined that FAM210B, a mitochondrial inner-membrane protein, is essential for hemoglobinization, proliferation, and enucleation during terminal erythroid maturation. Fam210b deficiency led to defects in mitochondrial iron uptake, heme synthesis, and iron-sulfur cluster formation. These defects were corrected with a lipid-soluble, small-molecule iron transporter, hinokitiol, in Fam210b-deficient murine erythroid cells and zebrafish morphants. Genetic complementation experiments revealed that FAM210B is not a mitochondrial iron transporter but is required for adequate mitochondrial iron import to sustain heme synthesis and iron-sulfur cluster formation during erythroid differentiation. FAM210B was also required for maximal ferrochelatase activity in differentiating erythroid cells. We propose that FAM210B functions as an adaptor protein that facilitates the formation of an oligomeric mitochondrial iron transport complex, required for the increase in iron acquisition for heme synthesis during terminal erythropoiesis. Collectively, our results reveal a critical mechanism by which EPO signaling regulates terminal erythropoiesis and iron metabolism.
Collapse
Affiliation(s)
- Yvette Y Yien
- From the Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, .,the Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jiahai Shi
- the Whitehead Institute and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Caiyong Chen
- the Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jesmine T M Cheung
- the Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095
| | - Anthony S Grillo
- the Department of Chemistry and Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Rishna Shrestha
- the Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Liangtao Li
- the Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Xuedi Zhang
- From the Department of Biological Sciences, University of Delaware, Newark, Delaware 19716
| | - Martin D Kafina
- the Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Paul D Kingsley
- the Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York 14642
| | - Matthew J King
- the Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Julien Ablain
- the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hojun Li
- the Whitehead Institute and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Leonard I Zon
- the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,the Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, and
| | - James Palis
- the Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York 14642
| | - Martin D Burke
- the Department of Chemistry and Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Daniel E Bauer
- the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,the Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Stuart H Orkin
- the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,the Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Carla M Koehler
- the Department of Chemistry and Biochemistry, UCLA, Los Angeles, California 90095
| | - John D Phillips
- the Division of Hematology and Hematologic Malignancy, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Jerry Kaplan
- the Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Diane M Ward
- the Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84112
| | - Harvey F Lodish
- the Whitehead Institute and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Barry H Paw
- the Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115.,the Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02115, and
| |
Collapse
|
20
|
Darki F, Klingberg T. Functional differentiation between convergence and non-convergence zones of the striatum in children. Neuroimage 2018; 173:384-393. [PMID: 29501552 DOI: 10.1016/j.neuroimage.2018.02.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 11/16/2022] Open
Abstract
Most cortical areas send projections to the striatum. In some parts of the striatum, the connections converge from several cortical areas. It is unknown whether the convergence and non-convergence zones of the striatum differ functionally. Here, we used diffusion-weighted magnetic resonance imaging and probabilistic fiber tracking to parcellate the striatum based on its connections to dorsolateral prefrontal, parietal and orbitofrontal cortices in two different datasets (children aged 6-7 years and adults). In both samples, quantitative susceptibility mapping (QSM) values were significantly correlated with working memory (WM) in convergence zones, but not in non-convergence zones. In children, this was also true for mean diffusivity, MD. The association of MD to WM specifically in the convergent zone was replicated in the Pediatric Imaging, Neurocognition, and Genetics (PING) dataset for 135 children aged 6-9 years. QSM data was not available in the PING dataset, and the association to QSM still needs to be replicated. These results suggest that connectivity-based segments of the striatum exhibit functionally different characteristics. The association between convergence zones and WM performance might relate to a role in integrating and coordinating activity in different cortical areas.
Collapse
Affiliation(s)
- Fahimeh Darki
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | - Torkel Klingberg
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | -
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
21
|
Darki F, Nemmi F, Möller A, Sitnikov R, Klingberg T. Quantitative susceptibility mapping of striatum in children and adults, and its association with working memory performance. Neuroimage 2016; 136:208-14. [DOI: 10.1016/j.neuroimage.2016.04.065] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 04/21/2016] [Accepted: 04/26/2016] [Indexed: 01/13/2023] Open
|
22
|
|
23
|
Bellingham SA, Guo B, Hill AF. The secret life of extracellular vesicles in metal homeostasis and neurodegeneration. Biol Cell 2015; 107:389-418. [PMID: 26032945 DOI: 10.1111/boc.201500030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022]
Abstract
Biologically active metals such as copper, zinc and iron are fundamental for sustaining life in different organisms with the regulation of cellular metal homeostasis tightly controlled through proteins that coordinate metal uptake, efflux and detoxification. Many of the proteins involved in either uptake or efflux of metals are localised and function on the plasma membrane, traffic between intracellular compartments depending upon the cellular metal environment and can undergo recycling via the endosomal pathway. The biogenesis of exosomes also occurs within the endosomal system, with several major neurodegenerative disease proteins shown to be released in association with these vesicles, including the amyloid-β (Aβ) peptide in Alzheimer's disease and the infectious prion protein involved in Prion diseases. Aβ peptide and the prion protein also bind biologically active metals and are postulated to play important roles in metal homeostasis. In this review, we will discuss the role of extracellular vesicles in Alzheimer's and Prion diseases and explore their potential contribution to metal homeostasis.
Collapse
Affiliation(s)
- Shayne A Bellingham
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Belinda Guo
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia
| | - Andrew F Hill
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia.,Bio21 Molecular Science and Biotechnology Institute, Parkville, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Adisetiyo V, Helpern JA. Brain iron: a promising noninvasive biomarker of attention-deficit/hyperactivity disorder that warrants further investigation. Biomark Med 2015; 9:403-6. [DOI: 10.2217/bmm.15.9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Vitria Adisetiyo
- Center for Biomedical Imaging, Department of Radiology & Radiological Science, Medical University of South Carolina, 96 Jonathan Lucas Street MSC 323, Charleston, SC 29425, USA
| | - Joseph A Helpern
- Center for Biomedical Imaging, Department of Radiology & Radiological Science, Medical University of South Carolina, 96 Jonathan Lucas Street MSC 323, Charleston, SC 29425, USA
- Department of Neuroscience, Medical University of South Carolina, 96 Jonathan Lucas Street MSC 323, Charleston, SC 29425, USA
| |
Collapse
|
25
|
Abstract
Restless legs syndrome is a common sensorimotor disorder characterized by an urge to move, and associated with uncomfortable sensations in the legs (limbs). Restless legs syndrome can lead to sleep-onset or sleep-maintenance insomnia, and occasionally excessive daytime sleepiness, all leading to significant morbidity. Brain iron deficiency and dopaminergic neurotransmission abnormalities play a central role in the pathogenesis of this disorder, along with other nondopaminergic systems, although the exact mechanisms are still. Intensive care unit patients are especially vulnerable to have unmasking or exacerbation of restless legs syndrome because of sleep deprivation, circadian rhythm disturbance, immobilization, iron deficiency, and use of multiple medications that can antagonize dopamine.
Collapse
Affiliation(s)
- Saiprakash B Venkateshiah
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Octavian C Ioachimescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
26
|
Hyacinthe C, De Deurwaerdere P, Thiollier T, Li Q, Bezard E, Ghorayeb I. Blood withdrawal affects iron store dynamics in primates with consequences on monoaminergic system function. Neuroscience 2015; 290:621-35. [DOI: 10.1016/j.neuroscience.2015.01.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/24/2015] [Accepted: 01/26/2015] [Indexed: 10/24/2022]
|
27
|
Adisetiyo V, Jensen JH, Tabesh A, Deardorff RL, Fieremans E, Di Martino A, Gray KM, Castellanos FX, Helpern JA. Multimodal MR imaging of brain iron in attention deficit hyperactivity disorder: a noninvasive biomarker that responds to psychostimulant treatment? Radiology 2014; 272:524-32. [PMID: 24937545 DOI: 10.1148/radiol.14140047] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE To comprehensively assess brain iron levels in typically developing control subjects and patients with attention deficit hyperactivity disorder (ADHD) when psychostimulant medication history is accounted for. MATERIALS AND METHODS This prospective study was approved by the institutional review board, and informed consent was obtained. Brain iron was indexed noninvasively by using magnetic resonance (MR) imaging relaxation rates (R2, R2*, R2') and magnetic field correlation (MFC) in the globus pallidus, putamen, caudate nucleus, and thalamus for 22 patients with ADHD (12 medication-naïve patients and 10 with a history of psychostimulant treatment) and 27 control subjects (age range, 8-18 years). Serum iron measures were also collected. Subgroup differences were analyzed with data-appropriate omnibus tests followed by post hoc pairwise comparisons; false discovery rate correction was conducted to control for multiple comparisons. RESULTS Medication-naïve ADHD patients had significantly lower striatal and thalamic MFC indexes of brain iron than did control subjects (putamen, P = .012; caudate nucleus, P = .008; thalamus, P = .012) and psychostimulant-medicated ADHD patients (putamen, P = .006; caudate nucleus, P = .010; thalamus, P = .021). Conversely, the MFC indexes in medicated patients were comparable to those in control subjects. No significant differences were detected with R2, R2*, R2', or serum measures. CONCLUSION Lower MFC indexes of striatal and thalamic brain iron in medication-naïve ADHD patients and lack of differences in psychostimulant-medicated patients suggest that MFC indexes of brain iron may represent a noninvasive diagnostic biomarker that responds to psychostimulant treatment.
Collapse
Affiliation(s)
- Vitria Adisetiyo
- From the Center for Biomedical Imaging (V.A., J.H.J., A.T., R.L.D., J.A.H.), Department of Radiology and Radiological Science (V.A., J.H.J., A.T., R.L.D., J.A.H.), Department of Psychiatry and Behavioral Sciences (K.M.G.), and Department of Neurosciences (J.A.H.), Medical University of South Carolina, 68 President St, Bioengineering Building, 2nd Floor, Charleston, SC 29425; Center for Biomedical Imaging, Department of Radiology (E.F., F.X.C.), Child Study Center, Department of Child and Adolescent Psychiatry (A.D.M., F.X.C.), and Department of Physiology and Neuroscience (F.X.C.), New York University School of Medicine, New York, NY; and Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY (F.X.C.)
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Izumi Y, Ezumi M, Takada-Takatori Y, Akaike A, Kume T. Endogenous Dopamine Is Involved in the Herbicide Paraquat-Induced Dopaminergic Cell Death. Toxicol Sci 2014; 139:466-78. [DOI: 10.1093/toxsci/kfu054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
29
|
Krzyaniak MD, Eser BE, Ellis HR, Fitzpatrick PF, McCracken J. Pulsed EPR study of amino acid and tetrahydropterin binding in a tyrosine hydroxylase nitric oxide complex: evidence for substrate rearrangements in the formation of the oxygen-reactive complex. Biochemistry 2013; 52:8430-41. [PMID: 24168553 DOI: 10.1021/bi4010914] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tyrosine hydroxylase is a nonheme iron enzyme found in the nervous system that catalyzes the hydroxylation of tyrosine to form l-3,4-dihydroxyphenylalanine, the rate-limiting step in the biosynthesis of the catecholamine neurotransmitters. Catalysis requires the binding of three substrates: tyrosine, tetrahydrobiopterin, and molecular oxygen. We have used nitric oxide as an O₂ surrogate to poise Fe(II) at the catalytic site in an S = 3/2, {FeNO}⁷ form amenable to EPR spectroscopy. ²H-electron spin echo envelope modulation was then used to measure the distance and orientation of specifically deuterated substrate tyrosine and cofactor 6-methyltetrahydropterin with respect to the magnetic axes of the {FeNO}⁷ paramagnetic center. Our results show that the addition of tyrosine triggers a conformational change in the enzyme that reduces the distance from the {FeNO}⁷ center to the closest deuteron on 6,7-²H-6-methyltetrahydropterin from >5.9 Å to 4.4 ± 0.2 Å. Conversely, the addition of 6-methyltetrahydropterin to enzyme samples treated with 3,5-²H-tyrosine resulted in reorientation of the magnetic axes of the S = 3/2, {FeNO}⁷ center with respect to the deuterated substrate. Taken together, these results show that the coordination of both substrate and cofactor direct the coordination of NO to Fe(II) at the active site. Parallel studies of a quaternary complex of an uncoupled tyrosine hydroxylase variant, E332A, show no change in the hyperfine coupling to substrate tyrosine and cofactor 6-methyltetrahydropterin. Our results are discussed in the context of previous spectroscopic and X-ray crystallographic studies done on tyrosine hydroxylase and phenylalanine hydroxylase.
Collapse
Affiliation(s)
- Matthew D Krzyaniak
- Department of Chemistry, Michigan State University , East Lansing, Michigan 48824, United States
| | | | | | | | | |
Collapse
|
30
|
Romero MDSC, Pliego-Rivero FB, Altamirano BM, Otero GA. Effect of postlactation iron deficiency on the composition of fatty acids of whole brain myelin. Nutr Neurosci 2013. [DOI: 10.1179/147683010x12611460764606] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
31
|
Hare D, Ayton S, Bush A, Lei P. A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci 2013; 5:34. [PMID: 23874300 PMCID: PMC3715022 DOI: 10.3389/fnagi.2013.00034] [Citation(s) in RCA: 281] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/25/2013] [Indexed: 12/12/2022] Open
Abstract
Iron is the most abundant transition metal within the brain, and is vital for a number of cellular processes including neurotransmitter synthesis, myelination of neurons, and mitochondrial function. Redox cycling between ferrous and ferric iron is utilized in biology for various electron transfer reactions essential to life, yet this same chemistry mediates deleterious reactions with oxygen that induce oxidative stress. Consequently, there is a precise and tightly controlled mechanism to regulate iron in the brain. When iron is dysregulated, both conditions of iron overload and iron deficiencies are harmful to the brain. This review focuses on how iron metabolism is maintained in the brain, and how an alteration to iron and iron metabolism adversely affects neurological function.
Collapse
Affiliation(s)
- Dominic Hare
- The Florey Institute of Neuroscience and Mental Health, University of MelbourneVIC, Australia
- Elemental Bio-imaging Facility, University of TechnologySydney, NSW, Australia
| | - Scott Ayton
- The Florey Institute of Neuroscience and Mental Health, University of MelbourneVIC, Australia
| | - Ashley Bush
- The Florey Institute of Neuroscience and Mental Health, University of MelbourneVIC, Australia
| | - Peng Lei
- The Florey Institute of Neuroscience and Mental Health, University of MelbourneVIC, Australia
| |
Collapse
|
32
|
Pichler I, Del Greco M. F, Gögele M, Lill CM, Bertram L, Do CB, Eriksson N, Foroud T, Myers RH, Nalls M, Keller MF, Benyamin B, Whitfield JB, Pramstaller PP, Hicks AA, Thompson JR, Minelli C. Serum iron levels and the risk of Parkinson disease: a Mendelian randomization study. PLoS Med 2013; 10:e1001462. [PMID: 23750121 PMCID: PMC3672214 DOI: 10.1371/journal.pmed.1001462] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 04/24/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although levels of iron are known to be increased in the brains of patients with Parkinson disease (PD), epidemiological evidence on a possible effect of iron blood levels on PD risk is inconclusive, with effects reported in opposite directions. Epidemiological studies suffer from problems of confounding and reverse causation, and mendelian randomization (MR) represents an alternative approach to provide unconfounded estimates of the effects of biomarkers on disease. We performed a MR study where genes known to modify iron levels were used as instruments to estimate the effect of iron on PD risk, based on estimates of the genetic effects on both iron and PD obtained from the largest sample meta-analyzed to date. METHODS AND FINDINGS We used as instrumental variables three genetic variants influencing iron levels, HFE rs1800562, HFE rs1799945, and TMPRSS6 rs855791. Estimates of their effect on serum iron were based on a recent genome-wide meta-analysis of 21,567 individuals, while estimates of their effect on PD risk were obtained through meta-analysis of genome-wide and candidate gene studies with 20,809 PD cases and 88,892 controls. Separate MR estimates of the effect of iron on PD were obtained for each variant and pooled by meta-analysis. We investigated heterogeneity across the three estimates as an indication of possible pleiotropy and found no evidence of it. The combined MR estimate showed a statistically significant protective effect of iron, with a relative risk reduction for PD of 3% (95% CI 1%-6%; p = 0.001) per 10 µg/dl increase in serum iron. CONCLUSIONS Our study suggests that increased iron levels are causally associated with a decreased risk of developing PD. Further studies are needed to understand the pathophysiological mechanism of action of serum iron on PD risk before recommendations can be made.
Collapse
Affiliation(s)
- Irene Pichler
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Fabiola Del Greco M.
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Martin Gögele
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Christina M. Lill
- Neuropsychiatric Genetics Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Neurology, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Lars Bertram
- Neuropsychiatric Genetics Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Chuong B. Do
- 23andMe, Inc., Mountain View, California, United States of America
| | | | - Tatiana Foroud
- Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Richard H. Myers
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | | | - Michael Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Margaux F. Keller
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biological Anthropology, Temple University, Philadelphia, Pennsylvania, United States of America
| | | | | | - Beben Benyamin
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
- Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - John B. Whitfield
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | - Peter P. Pramstaller
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
- Department of Neurology, General Central Hospital, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Andrew A. Hicks
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - John R. Thompson
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Cosetta Minelli
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
33
|
Davies KM, Hare DJ, Cottam V, Chen N, Hilgers L, Halliday G, Mercer JFB, Double KL. Localization of copper and copper transporters in the human brain. Metallomics 2013; 5:43-51. [PMID: 23076575 DOI: 10.1039/c2mt20151h] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Disturbances in brain copper result in rare and severe neurological disorders and may play a role in the pathogenesis and progression of multiple neurodegenerative diseases. Our current understanding of mammalian brain copper transport is based on model systems outside the central nervous system and no data are available regarding copper transport systems in the human brain. To address this deficit, we quantified regional copper concentrations and examined the distribution and cellular localization of the copper transport proteins Copper transporter 1, Atox1, ATP7A, and ATP7B in multiple regions of the human brain using inductively coupled plasma-mass spectrometry, Western blot and immunohistochemistry. We identified significant relationships between copper transporter levels and brain copper concentrations, supporting a role for these proteins in copper transport in the human brain. Interestingly, the substantia nigra contained twice as much copper than that in other brain regions, suggesting an important role for copper in this brain region. Furthermore, ATP7A levels were significantly greater in the cerebellum, compared with other brain regions, supporting an important role for ATP7A in cerebellar neuronal health. This study provides novel data regarding copper regulation in the human brain, critical to understand the mechanisms by which brain copper levels can be altered, leading to neurological disease.
Collapse
Affiliation(s)
- Katherine M Davies
- Neuroscience Research Australia and The University of New South Wales, Randwick, NSW 2031, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Effect of ascorbic acid deficiency on catecholamine synthesis in adrenal glands of SMP30/GNL knockout mice. Eur J Nutr 2013; 53:177-85. [DOI: 10.1007/s00394-013-0515-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Accepted: 03/06/2013] [Indexed: 12/13/2022]
|
35
|
|
36
|
Lin Q, Feng J, Zhao X, Zhang G, Wang W. Expression and function of ferroportin 1 in O-2A progenitor cells. Anat Rec (Hoboken) 2012; 296:108-16. [PMID: 23117987 DOI: 10.1002/ar.22610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Accepted: 08/22/2012] [Indexed: 11/09/2022]
Abstract
Iron plays a crucial role in the survival, differentiation, and myelin formation of oligodendrocyte lineages. However, the regulation mechanism of iron homeostasis in oligodendrocytes remains unclear. Recently, much research has focused on Ferroportin 1 (FPN1), an iron exporter protein. First, about 95% pure primary rat O-2A progenitor cells were obtained by shaking methods in our laboratory. The expression of FPN1 mRNA and protein in O-2A progenitor cells were determined by reverse transcription-PCR and western blot. In addition, the localization of FPN1 at the cell membrane, in the cytoplasm and in processes was assayed by double-labeling immunofluorescence. A time-dependent increase of iron efflux from O-2A progenitor cells was confirmed by the calcein-indicated iron efflux assay. However, the same cells treated with FPN1 antibody showed no obvious change in iron release. For further confirmation, overexpression of FPN1 in O-2A progenitor cells was transduced with lentivirus. The release of iron in O-2A progenitor cells was dramatically increased by the overexpressed FPN1 when compared with that of the control group. Both ferritin (Ft) and transferrin receptor (TfR) are routinely used as indicators of labile iron pool. Cells pretreated with FPN1 antibody upregulated Ft and downregulated TfR protein level, while the opposite results occurred in the FPN1 overexpressing cells. Determination of Ft and TfR indirectly indicated that FPN1 might contribute to iron release from O-2A progenitor cells. We suggested that expression of FPN1 in O-2A progenitor cells might play a critical role in iron efflux from these cells.
Collapse
Affiliation(s)
- Qing Lin
- Department of Human Anatomy, Histology and Embryology, School of Preclinical Medicine, Fujian Medical University, Fuzhou, People's Republic of China
| | | | | | | | | |
Collapse
|
37
|
Ivanova B, Spiteller M. Coordination ability of bradykinin with ZnII- and AgI-metal ions – Experimental and theoretical study. Inorganica Chim Acta 2012. [DOI: 10.1016/j.ica.2012.06.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
38
|
Allard MM, Sonk JA, Heeg MJ, McGarvey BR, Schlegel HB, Verani CN. Bioinspired Five-Coordinate Iron(III) Complexes for Stabilization of Phenoxyl Radicals. Angew Chem Int Ed Engl 2011; 51:3178-82. [DOI: 10.1002/anie.201103233] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 11/09/2011] [Indexed: 11/09/2022]
|
39
|
Allard MM, Sonk JA, Heeg MJ, McGarvey BR, Schlegel HB, Verani CN. Bioinspired Five-Coordinate Iron(III) Complexes for Stabilization of Phenoxyl Radicals. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201103233] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
40
|
Wang S, Lasagna M, Daubner SC, Reinhart GD, Fitzpatrick PF. Fluorescence spectroscopy as a probe of the effect of phosphorylation at serine 40 of tyrosine hydroxylase on the conformation of its regulatory domain. Biochemistry 2011; 50:2364-70. [PMID: 21302933 DOI: 10.1021/bi101844p] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phosphorylation of Ser40 in the regulatory domain of tyrosine hydroxylase activates the enzyme by increasing the rate constant for dissociation of inhibitory catecholamines from the active site by 3 orders of magnitude. To probe the changes in the structure of the N-terminal domain upon phosphorylation, individual phenylalanine residues at positions 14, 34, and 74 were replaced with tryptophan in a form of the protein in which the endogenous tryptophans had all been mutated to phenylalanine (W(3)F TyrH). The steady-state fluorescence anisotropy of F74W W(3)F TyrH was unaffected by phosphorylation, but the anisotropies of both F14W and F34W W(3)F TyrH increased significantly upon phosphorylation. The fluorescence of the single tryptophan residue at position 74 was less readily quenched by acrylamide than those at the other two positions; fluorescence increased the rate constant for quenching of the residues at positions 14 and 34 but did not affect that for the residue at position 74. Frequency domain analyses were consistent with phosphorylation having no effect on the amplitude of the rotational motion of the indole ring at position 74, resulting in a small increase in the rotational motion of the residue at position 14 and resulting in a larger increase in the rotational motion of the residue at position 34. These results are consistent with the local environment at position 74 being unaffected by phosphorylation, that at position 34 becoming much more flexible upon phosphorylation, and that at position 14 becoming slightly more flexible upon phosphorylation. The results support a model in which phosphorylation at Ser40 at the N-terminus of the regulatory domain causes a conformational change to a more open conformation in which the N-terminus of the protein no longer inhibits dissociation of a bound catecholamine from the active site.
Collapse
Affiliation(s)
- Shanzhi Wang
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | | | | | | | | |
Collapse
|
41
|
Li J, Dangott LJ, Fitzpatrick PF. Regulation of phenylalanine hydroxylase: conformational changes upon phenylalanine binding detected by hydrogen/deuterium exchange and mass spectrometry. Biochemistry 2010; 49:3327-35. [PMID: 20307070 DOI: 10.1021/bi1001294] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Phenylalanine acts as an allosteric activator of the tetrahydropterin-dependent enzyme phenylalanine hydroxylase. Hydrogen/deuterium exchange monitored by mass spectrometry has been used to gain insight into local conformational changes accompanying activation of rat phenylalanine hydroxylase by phenylalanine. Peptides in the regulatory and catalytic domains that lie in the interface between these two domains show large increases in the extent of deuterium incorporation from solvent in the presence of phenylalanine. In contrast, the effects of phenylalanine on the exchange kinetics of a mutant enzyme lacking the regulatory domain are limited to peptides surrounding the binding site for the amino acid substrate. These results support a model in which the N-terminus of the protein acts as an inhibitory peptide, with phenylalanine binding causing a conformational change in the regulatory domain that alters the interaction between the catalytic and regulatory domains.
Collapse
Affiliation(s)
- Jun Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station,Texas 77843-2128, USA
| | | | | |
Collapse
|
42
|
Holowatz LA, Kenney WL. Peripheral mechanisms of thermoregulatory control of skin blood flow in aged humans. J Appl Physiol (1985) 2010; 109:1538-44. [PMID: 20413421 DOI: 10.1152/japplphysiol.00338.2010] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human skin blood flow is controlled via dual innervation from the sympathetic nervous system. Reflex cutaneous vasoconstriction and vasodilation are both impaired with primary aging, rendering the aged more vulnerable to hypothermia and cardiovascular complications from heat-related illness. Age-related alterations in the thermoregulatory control of skin blood flow occur at multiple points along the efferent arm of the reflex, including 1) diminished sympathetic outflow, 2) altered presynaptic neurotransmitter synthesis, 3) reduced vascular responsiveness, and 4) impairments in downstream (endothelial and vascular smooth muscle) second-messenger signaling. This mechanistic review highlights some of the recent findings in the area of aging and the thermoregulatory control of skin blood flow.
Collapse
Affiliation(s)
- Lacy A Holowatz
- Department of Kinesiology, The Pennsylvania State University, Noll Laboratory, University Park, PA 16802, USA.
| | | |
Collapse
|
43
|
Eser BE, Fitzpatrick PF. Measurement of intrinsic rate constants in the tyrosine hydroxylase reaction. Biochemistry 2010; 49:645-52. [PMID: 20025246 DOI: 10.1021/bi901874e] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tyrosine hydroxylase (TyrH) is a pterin-dependent mononuclear non-heme aromatic amino acid hydroxylase that catalyzes the conversion of tyrosine to dihydroxyphenylalanine (DOPA). Chemical quench analyses of the enzymatic reaction show a burst of DOPA formation, followed by a linear rate equal to the k(cat) value at both 5 and 30 degrees C. The effects of increasing solvent viscosity confirm that k(cat) is approximately 84% limited by diffusion, most probably due to slow product release, and that tyrosine has a commitment to catalysis of 0.45. The effect of viscosity on the k(cat)/K(m) for 6-methyltetrahydropterin is greater than the theoretical limit, consistent with the coupling of pterin binding to the movement of a surface loop. The absorbance changes in the spectrum of the tetrahydropterin during the first turnover, the kinetics of DOPA formation during the first turnover, and the previously described kinetics for formation and decay of the Fe(IV)O intermediate [Eser, B. E., Barr, E. W., Frantom, P. A., Saleh, L., Bollinger, J. M., Jr., Krebs, C., and Fitzpatrick, P. F. (2007) J. Am. Chem. Soc. 129, 11334-11335] were analyzed globally, yielding a single set of rate constants for the TyrH reaction. Reversible binding of oxygen is followed by formation of Fe(IV)O and 4a-hydroxypterin with a rate constant of 13 s(-1) at 5 degrees C. Transfer of oxygen from Fe(IV)O to tyrosine to form DOPA follows with a rate constant of 22 s(-1). Release of DOPA and/or the 4a-hydroxypterin with a rate constant of 0.86 s(-1) completes the turnover.
Collapse
Affiliation(s)
- Bekir E Eser
- Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | | |
Collapse
|
44
|
Savica R, Grossardt BR, Carlin JM, Icen M, Bower JH, Ahlskog JE, Maraganore DM, Steensma DP, Rocca WA. Anemia or low hemoglobin levels preceding Parkinson disease: a case-control study. Neurology 2009; 73:1381-7. [PMID: 19858460 DOI: 10.1212/wnl.0b013e3181bd80c1] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE It has been suggested that anemia may be a risk factor for dementia, for restless legs syndrome, and for Parkinson disease (PD). Thus, we investigated the association of anemia with the subsequent risk of PD using a case-control study design. METHODS We used the medical records-linkage system of the Rochester Epidemiology Project to identify 196 subjects who developed PD in Olmsted County, Minnesota, from 1976 through 1995. Each incident case was matched by age (+/-1 year) and sex to a general population control. We reviewed the complete medical records of cases and controls in the system to detect anemia defined using the World Health Organization criteria. RESULTS Anemia was more common in the history of cases than of controls (odds ratio 2.00, 95% confidence interval 1.31-3.06, p = 0.001). The association remained significant after adjustment for cigarette smoking, exposure to pesticides, or hysterectomy (in women). The association was not significantly different between men and women, or between PD patients with or without rest tremor. Analyses stratified by time of onset of anemia showed a greater association for anemia that started 20 to 29 years before the onset of PD. Hemoglobin levels were slightly but consistently lower in cases than in controls across all ages. CONCLUSIONS Our results support an association between anemia experienced early in life and the later development of Parkinson disease. The interpretation of this association remains uncertain.
Collapse
Affiliation(s)
- R Savica
- Department of Neurology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Haahr LT, Jensen KP, Boesen J, Christensen HEM. Experimentally calibrated computational chemistry of tryptophan hydroxylase: trans influence, hydrogen-bonding, and 18-electron rule govern O2-activation. J Inorg Biochem 2009; 104:136-45. [PMID: 19939457 DOI: 10.1016/j.jinorgbio.2009.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 10/09/2009] [Accepted: 10/16/2009] [Indexed: 10/20/2022]
Abstract
Insight into the nature of oxygen activation in tryptophan hydroxylase has been obtained from density functional computations. Conformations of O(2)-bound intermediates have been studied with oxygen trans to glutamate and histidine, respectively. An O(2)-adduct with O(2)trans to histidine (O(his)) and a peroxo intermediate with peroxide trans to glutamate (P(glu)) were found to be consistent (0.57-0.59mm/s) with experimental Mössbauer isomer shifts (0.55mm/s) and had low computed free energies. The weaker trans influence of histidine is shown to give rise to a bent O(2) coordination mode with O(2) pointing towards the cofactor and a more activated O-O bond (1.33A) than in O(glu) (1.30A). It is shown that the cofactor can hydrogen bond to O(2) and activate the O-O bond further (from 1.33 to 1.38A). The O(his) intermediate leads to a ferryl intermediate (F(his)) with an isomer shift of 0.34mm/s, also consistent with the experimental value (0.25mm/s) which we propose as the structure of the hydroxylating intermediate, with the tryptophan substrate well located for further reaction 3.5A from the ferryl group. Based on the optimized transition states, the activation barriers for the two paths (glu and his) are similar, so a two-state scenario involving O(his) and P(glu) is possible. A structure of the activated deoxy state which is high-spin implies that the valence electron count has been lowered from 18 to 16 (glutamate becomes bidentate), giving a "green light" that invites O(2)-binding. Our mechanism of oxygen activation in tryptophan hydroxylase does not require inversion of spin, which may be an important observation.
Collapse
Affiliation(s)
- Laerke T Haahr
- Technical University of Denmark, DTU Chemistry, Kemitorvet 207, 2800 Kgs. Lyngby, DK, Denmark
| | | | | | | |
Collapse
|
46
|
Grote L, Leissner L, Hedner J, Ulfberg J. A randomized, double-blind, placebo controlled, multi-center study of intravenous iron sucrose and placebo in the treatment of restless legs syndrome. Mov Disord 2009; 24:1445-52. [PMID: 19489063 DOI: 10.1002/mds.22562] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Iron deficiency may exacerbate symptoms in the Restless Legs Syndrome (RLS). We investigated the effect of intravenous iron sucrose or placebo on symptoms in patients with RLS and mild to moderate iron deficit. Sixty patients with primary RLS (seven males, age 46 (9) years, S-ferritin < or =45 microg/L) recruited from a cohort of 231 patients were randomly assigned in a 12-months double-blind, multi-centre study of iron sucrose 1000 mg (n = 29) or saline (n = 31). The primary efficacy variable was the RLS severity scale (IRLS) score at week 11. Median IRLS score decreased from 24 to 7 (week 11) after iron sucrose and from 26 to 17 after placebo (P = 0.123, N.S. for between treatment comparison). The corresponding scores at week 7 were 12 and 20 in the two groups (P = 0.017). Drop out rate because of lack of efficacy at 12 months was 19/31 after placebo and 5/29 patients after iron sucrose (Kaplan-Meier estimate, log rank test P = 0.0006) suggesting an iron induced superior long term RLS symptom control. Iron sucrose was well tolerated. This study showed a lack of superiority of iron sucrose at 11 weeks but found evidence that iron sucrose reduced RLS symptoms both in the acute phase (7 weeks) and during long-term follow up in patients with variable degree of iron deficiency. Further studies on target patient groups, dosing and dosing intervals are warranted before iron sucrose could be considered for treatment of iron deficient patients with RLS.
Collapse
Affiliation(s)
- Ludger Grote
- Sleep Disorders Center, Department of Pulmonary Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
47
|
Chow MS, Eser BE, Wilson SA, Hodgson KO, Hedman B, Fitzpatrick PF, Solomon EI. Spectroscopy and kinetics of wild-type and mutant tyrosine hydroxylase: mechanistic insight into O2 activation. J Am Chem Soc 2009; 131:7685-98. [PMID: 19489646 DOI: 10.1021/ja810080c] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tyrosine hydroxylase (TH) is a pterin-dependent nonheme iron enzyme that catalyzes the hydroxylation of L-tyr to L-DOPA in the rate-limiting step of catecholamine neurotransmitter biosynthesis. We have previously shown that the Fe(II) site in phenylalanine hydroxylase (PAH) converts from six-coordinate (6C) to five-coordinate (5C) only when both substrate + cofactor are bound. However, steady-state kinetics indicate that TH has a different co-substrate binding sequence (pterin + O(2) + L-tyr) than PAH (L-phe + pterin + O(2)). Using X-ray absorption spectroscopy (XAS), and variable-temperature-variable-field magnetic circular dichroism (VTVH MCD) spectroscopy, we have investigated the geometric and electronic structure of the wild-type (WT) TH and two mutants, S395A and E332A, and their interactions with substrates. All three forms of TH undergo 6C --> 5C conversion with tyr + pterin, consistent with the general mechanistic strategy established for O(2)-activating nonheme iron enzymes. We have also applied single-turnover kinetic experiments with spectroscopic data to evaluate the mechanism of the O(2) and pterin reactions in TH. When the Fe(II) site is 6C, the two-electron reduction of O(2) to peroxide by Fe(II) and pterin is favored over individual one-electron reactions, demonstrating that both a 5C Fe(II) and a redox-active pterin are required for coupled O(2) reaction. When the Fe(II) is 5C, the O(2) reaction is accelerated by at least 2 orders of magnitude. Comparison of the kinetics of WT TH, which produces Fe(IV)=O + 4a-OH-pterin, and E332A TH, which does not, shows that the E332 residue plays an important role in directing the protonation of the bridged Fe(II)-OO-pterin intermediate in WT to productively form Fe(IV)=O, which is responsible for hydroxylating L-tyr to L-DOPA.
Collapse
Affiliation(s)
- Marina S Chow
- Department of Chemistry, Stanford University, Stanford, California 94305, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Wang S, Sura GR, Dangott LJ, Fitzpatrick PF. Identification by hydrogen/deuterium exchange of structural changes in tyrosine hydroxylase associated with regulation. Biochemistry 2009; 48:4972-9. [PMID: 19371093 PMCID: PMC2730116 DOI: 10.1021/bi9004254] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The activity of tyrosine hydroxylase is regulated by reversible phosphorylation of serine residues in an N-terminal regulatory domain and catecholamine inhibition at the active site. Catecholamines such as dopamine bind very tightly to the resting enzyme; phosphorylation of Ser40 decreases the affinity for catecholamines by 3 orders of magnitude. The effects of dopamine binding and phosphorylation of Ser40 on the kinetics of deuterium incorporation into peptide bonds were examined by mass spectrometry. When dopamine is bound, three peptic peptides show significantly slower deuterium incorporation, 35-41 and 42-71 in the regulatory domain and 295-299 in the catalytic domain. In the phosphorylated enzyme, peptide 295-299 shows more rapid incorporation of deuterium, while 35-41 and 42-71 can not be detected. These results are consistent with tyrosine hydroxylase existing in two different conformations. In the closed conformation, the regulatory domain lies across the active site loop containing residues 295-298; this is stabilized when dopamine is bound in the active site. In the open conformation, the regulatory domain has moved out of the active site, allowing substrate access; this conformation is favored by phosphorylation of Ser40.
Collapse
Affiliation(s)
- Shanzhi Wang
- Departments of Biochemistry and Biophysics Texas A&M University, College Station TX 77843-2128
| | - Giri R. Sura
- Departments of Biochemistry and Biophysics Texas A&M University, College Station TX 77843-2128
| | - Lawrence J. Dangott
- Protein Chemistry Laboratory Texas A&M University, College Station TX 77843-2128
| | - Paul F. Fitzpatrick
- Departments of Biochemistry and Biophysics Texas A&M University, College Station TX 77843-2128
- Department of Chemistry Texas A&M University, College Station TX 77843-2128
| |
Collapse
|
49
|
Windahl MS, Petersen CR, Christensen HEM, Harris P. Crystal structure of tryptophan hydroxylase with bound amino acid substrate. Biochemistry 2008; 47:12087-94. [PMID: 18937498 DOI: 10.1021/bi8015263] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tryptophan hydroxylase (TPH) is a mononuclear non-heme iron enzyme, which catalyzes the reaction between tryptophan, O 2, and tetrahydrobiopterin (BH 4) to produce 5-hydroxytryptophan and 4a-hydroxytetrahydrobiopterin. This is the first and rate-limiting step in the biosynthesis of the neurotransmitter and hormone serotonin (5-hydroxytryptamine). We have determined the 1.9 A resolution crystal structure of the catalytic domain (Delta1-100/Delta415-445) of chicken TPH isoform 1 (TPH1) in complex with the tryptophan substrate and an iron-bound imidazole. This is the first structure of any aromatic amino acid hydroxylase with bound natural amino acid substrate. The iron coordination can be described as distorted trigonal bipyramidal coordination with His273, His278, and Glu318 (partially bidentate) and one imidazole as ligands. The tryptophan stacks against Pro269 with a distance of 3.9 A between the iron and the tryptophan Czeta3 atom that is hydroxylated. The binding of tryptophan and maybe the imidazole has caused the structural changes in the catalytic domain compared to the structure of the human TPH1 without tryptophan. The structure of chicken TPH1 is more compact, and the loops of residues Leu124-Asp139 and Ile367-Thr369 close around the active site. Similar structural changes are seen in the catalytic domain of phenylalanine hydroxylase (PAH) upon binding of substrate analogues norleucine and thienylalanine to the PAH.BH 4 complex. In fact, the chicken TPH1.Trp.imidazole structure resembles the PAH.BH 4.thienylalanine structure more (root-mean-square deviation for Calpha atoms of 0.90 A) than the human TPH1 structure (root-mean-square deviation of 1.47 A).
Collapse
Affiliation(s)
- Michael S Windahl
- Department of Basic Sciences and Environment, Faculty of Life Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg C, Denmark
| | | | | | | |
Collapse
|
50
|
Panay AJ, Fitzpatrick PF. Kinetic isotope effects on aromatic and benzylic hydroxylation by Chromobacterium violaceum phenylalanine hydroxylase as probes of chemical mechanism and reactivity. Biochemistry 2008; 47:11118-24. [PMID: 18817418 DOI: 10.1021/bi801295w] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Phenylalanine hydroxylase from Chromobacterium violaceum (CvPheH) is a non-heme iron monooxygenase that catalyzes the hydroxylation of phenylalanine to tyrosine. In this study, we used deuterium kinetic isotope effects to probe the chemical mechanisms of aromatic and benzylic hydroxylation to compare the reactivities of bacterial and eukaryotic aromatic amino acid hydroxylases. The (D) k cat value for the reaction of CvPheH with [(2)H 5]phenylalanine is 1.2 with 6-methyltetrahydropterin and 1.4 with 6,7-dimethyltetrahydropterin. With the mutant enzyme I234D, the (D) k cat value decreases to 0.9 with the latter pterin; this is likely to be the intrinsic effect for addition of oxygen to the amino acid. The isotope effect on the subsequent tautomerization of a dienone intermediate was determined to be 5.1 by measuring the retention of deuterium in tyrosine produced from partially deuterated phenylalanine; this large isotope effect is responsible for the normal effect on k cat. The isotope effect for hydroxylation of the methyl group of 4-CH 3-phenylalanine, obtained from the partitioning of benzylic and aromatic hydroxylation products, is 10. The temperature dependence of this isotope effect establishes the contribution of hydrogen tunneling to benzylic hydroxylation by this enzyme. The results presented here provide evidence that the reactivities of the prokaryotic and eukaryotic hydroxylases are similar and further define the reactivity of the iron center for the family of aromatic amino acid hydroxylases.
Collapse
Affiliation(s)
- Aram J Panay
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843-2128, USA
| | | |
Collapse
|