1
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Hosseinpour Mashkani SM, Bishop DP, Westerhausen MT, Adlard PA, Golzan SM. Alterations in zinc, copper, and iron levels in the retina and brain of Alzheimer's disease patients and the APP/PS1 mouse model. Metallomics 2024; 16:mfae053. [PMID: 39520546 PMCID: PMC11630249 DOI: 10.1093/mtomcs/mfae053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Transition metals like copper (Cu), iron (Fe), and zinc (Zn) are vital for normal central nervous system function and are also linked to neurodegeneration, particularly in the onset and progression of Alzheimer's disease (AD). Their alterations in AD, identified prior to amyloid plaque aggregation, offer a unique target for staging pre-amyloid AD. However, analysing their levels in the brain is extremely challenging, necessitating the development of alternative approaches. Here, we utilized laser ablation-inductively coupled plasma-mass spectrometry and solution nebulization-inductively coupled plasma-mass spectrometry to quantitatively measure Cu, Fe, and Zn concentrations in the retina and hippocampus samples obtained from human donors (i.e. AD and healthy controls), and in the amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of AD and wild-type (WT) controls, aged 9 and 18 months. Our findings revealed significantly elevated Cu, Fe, and Zn levels in the retina (*P < .05, P < .01, and P < .001) and hippocampus (*P < .05, *P < .05, and *P < .05) of human AD samples compared to healthy controls. Conversely, APP/PS1 mouse models exhibited notably lower metal levels in the same regions compared to WT mice-Cu, Fe, and Zn levels in the retina (**P < .01, *P < .05, and *P < .05) and hippocampus (**P < .01, **P < .01, and *P < .05). The contrasting metal profiles in human and mouse samples, yet similar patterns within each species' retina and brain, suggest the retina mirrors cerebral metal dyshomoeostasis in AD. Our findings lay the groundwork for staging pre-AD pathophysiology through assessment of transition metal levels in the retina.
Collapse
Affiliation(s)
- Seyed Mostafa Hosseinpour Mashkani
- Institute for Biomedical Materials and Devices, School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - David P Bishop
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Mika T Westerhausen
- Hyphenated Mass Spectrometry Laboratory, School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne 3010, Australia
| | - S Mojtaba Golzan
- Vision Science Group (Orthoptics Discipline), Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
3
|
Suryana E, Rowlands BD, Bishop DP, Finkelstein DI, Double KL. Empirically derived formulae for calculation of age- and region-related levels of iron, copper and zinc in the adult C57BL/6 mouse brain. Neurobiol Aging 2024; 136:34-43. [PMID: 38301453 DOI: 10.1016/j.neurobiolaging.2024.01.003] [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: 05/18/2023] [Revised: 12/05/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
Metal dyshomeostasis is associated with neurodegenerative disorders, cancers and vascular disease. We report the effects of age (range: 3 to 18 months) on regional copper, iron and zinc levels in the brain of the C57BL/6 mouse, a widely used inbred strain with a permissive background allowing maximal expression of mutations in models that recapitulate these disorders. We present formulae that can be used to determine regional brain metal concentrations in the C57BL/6 mouse at any age in the range of three to eighteen months of life. Copper levels in the C57BL/6 mouse adult brain were highest in the striatum and cerebellum and increased with age, excepting the cortex and hippocampus. Regional iron levels increased linearly with age in all brain regions, while regional zinc concentrations became more homogeneous with age. Knockdown of the copper transporter Ctr1 reduced brain copper, but not iron or zinc, concentrations in a regionally-dependent manner. These findings demonstrate biometals in the brain change with age in a regionally-dependent manner. These data and associated formulae have implications for improving design and interpretation of a wide variety of studies in the C57BL/6 mouse.
Collapse
Affiliation(s)
- E Suryana
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - B D Rowlands
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - D P Bishop
- School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - D I Finkelstein
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - K L Double
- Brain and Mind Centre and School of Medical Sciences (Neuroscience), Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|
4
|
Billings JL, Hilton JBW, Liddell JR, Hare DJ, Crouch PJ. Fundamental Neurochemistry Review: Copper availability as a potential therapeutic target in progressive supranuclear palsy: Insight from other neurodegenerative diseases. J Neurochem 2023; 167:337-346. [PMID: 37800457 DOI: 10.1111/jnc.15978] [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: 05/16/2023] [Revised: 08/21/2023] [Accepted: 09/08/2023] [Indexed: 10/07/2023]
Abstract
Since the first description of Parkinson's disease (PD) over two centuries ago, the recognition of rare types of atypical parkinsonism has introduced a spectrum of related PD-like diseases. Among these is progressive supranuclear palsy (PSP), a neurodegenerative condition that clinically differentiates through the presence of additional symptoms uncommon in PD. As with PD, the initial symptoms of PSP generally present in the sixth decade of life when the underpinning neurodegeneration is already significantly advanced. The causal trigger of neuronal cell loss in PSP is unknown and treatment options are consequently limited. However, converging lines of evidence from the distinct neurodegenerative conditions of PD and amyotrophic lateral sclerosis (ALS) are beginning to provide insights into potential commonalities in PSP pathology and opportunity for novel therapeutic intervention. These include accumulation of the high abundance cuproenzyme superoxide dismutase 1 (SOD1) in an aberrant copper-deficient state, associated evidence for altered availability of the essential micronutrient copper, and evidence for neuroprotection using compounds that can deliver available copper to the central nervous system. Herein, we discuss the existing evidence for SOD1 pathology and copper imbalance in PSP and speculate that treatments able to provide neuroprotection through manipulation of copper availability could be applicable to the treatment of PSP.
Collapse
Affiliation(s)
- Jessica L Billings
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - James B W Hilton
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, New South Wales, Australia
| | - Jeffrey R Liddell
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Dominic J Hare
- School of Mathematical and Physical Sciences, University of Technology Sydney, Broadway, Ultimo, New South Wales, Australia
| | - Peter J Crouch
- Department of Anatomy and Physiology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
MELEK İM, KUŞ B, KAPTAN Z, PETEKKAYA E. Correlation of metal ions with specific brain region volumes in neurodegenerative diseases. Turk J Med Sci 2023; 53:1465-1475. [PMID: 38812995 PMCID: PMC10763799 DOI: 10.55730/1300-0144.5714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/26/2023] [Accepted: 08/26/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim There are reports stating that deteriorations in metal homeostasis in neurodegenerative diseases promote abnormal protein accumulation. In this study, the serum metal levels in Alzheimer's disease (AD) and Parkinson's disease (PD) and its relationship with the cortical regions of the brain were investigated. Materials and methods The patients were divided into 3 groups consisting of the AD group, PD group, and healthy control group (n = 15 for each). The volumes of specific brain regions were measured over the participants' 3-dimensional magnetic resonance images, and they were compared across the groups. Copper, zinc, iron, and ferritin levels in the serums were determined, and their correlations with the brain region volumes were examined. Results The volumes of left hippocampus and right substantia nigra were lower in the AD and PD groups, while the volume of the left nucleus caudatus (CdN) and bilateral insula were lower in the AD group compared to the control group. Serum zinc levels were lower in the AD and PD groups, while the iron level was lower in the PD group in comparison to the control group. In addition, the serum ferritin level was higher in the AD group than in the control group. Serum zinc and copper levels in the AD group were positively correlated with the volumes of the right entorhinal cortex, thalamus, CdN, and insula. Serum zinc and copper levels in the PD group showed a negative correlation with the left nucleus accumbens (NAc), right putamen, and right insula volumes. While the serum ferritin level in the PD group displayed a positive correlation with the bilateral CdN, putamen, and NAc, as well as the right hippocampus and insula volumes, no area was detected that showed a correlation with the serum ferritin level in the AD group. Conclusion A relationship was determined between the serum metal levels in the AD and PD groups and certain brain cortical regions that showed volumetric changes, which can be important for the early diagnosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- İsmet Murat MELEK
- Department of Neurology, Faculty of Medicine, Hatay Mustafa Kemal University, Hatay,
Turkiye
| | - Berna KUŞ
- Department of Biochemistry, Faculty of Medicine, Hatay Mustafa Kemal University, Hatay,
Turkiye
| | - Zülal KAPTAN
- Department of Physiology, Faculty of Medicine, Beykent University, İstanbul,
Turkiye
| | - Emine PETEKKAYA
- Department of Anatomy, Faculty of Medicine, Kastamonu University, Kastamonu,
Turkiye
| |
Collapse
|
6
|
Chakraborty K, Kar S, Rai B, Bhagat R, Naskar N, Seth P, Gupta A, Bhattacharjee A. Copper dependent ERK1/2 phosphorylation is essential for the viability of neurons and not glia. Metallomics 2022; 14:mfac005. [PMID: 35150272 PMCID: PMC8975716 DOI: 10.1093/mtomcs/mfac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 02/10/2022] [Indexed: 01/24/2023]
Abstract
Intracellular copper [Cu(I)] has been hypothesized to play role in the differentiation of the neurons. This necessitates understanding the role of Cu(I) not only in the neurons but also in the glia considering their anatomical proximity, contribution towards ion homeostasis, and neurodegeneration. In this study, we did a systematic investigation of the changes in the cellular copper homeostasis during neuronal and glial differentiation and the pathways triggered by them. Our study demonstrates increased mRNA for the plasma membrane copper transporter CTR1 leading to increased Cu(I) during the neuronal (PC-12) differentiation. ATP7A is retained in the trans-Golgi network (TGN) despite high Cu(I) demonstrating its utilization towards the neuronal differentiation. Intracellular copper triggers pathways essential for neurite generation and ERK1/2 activation during the neuronal differentiation. ERK1/2 activation also accompanies the differentiation of the foetal brain derived neuronal progenitor cells. The study demonstrates that ERK1/2 phosphorylation is essential for the viability of the neurons. In contrast, differentiated C-6 (glia) cells contain low intracellular copper and significant downregulation of the ERK1/2 phosphorylation demonstrating that ERK1/2 activation does not regulate the viability of the glia. But ATP7A shows vesicular localization despite low copper in the glia. In addition to the TGN, ATP7A localizes into RAB11 positive recycling endosomes in the glial neurites. Our study demonstrates the role of copper dependent ERK1/2 phosphorylation in the neuronal viability. Whereas glial differentiation largely involves sequestration of Cu(I) into the endosomes potentially (i) for ready release and (ii) rendering cytosolic copper unavailable for pathways like the ERK1/2 activation.
Collapse
Affiliation(s)
| | - Sumanta Kar
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, India
| | - Bhawana Rai
- Amity Institute of Biotechnology, Amity University, Kolkata, India
| | - Reshma Bhagat
- Molecular and Cellular Neuroscience, Neurovirology Division, National Brain Research Centre, Manesar, India
| | - Nabanita Naskar
- Chemical Sciences Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Pankaj Seth
- Molecular and Cellular Neuroscience, Neurovirology Division, National Brain Research Centre, Manesar, India
| | - Arnab Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, India
| | | |
Collapse
|
7
|
Chin-Chan M, Montes S, Blanco-Álvarez VM, Aguirre-Alarcón HA, Hernández-Rodríguez I, Bautista E. Relevance of biometals during neuronal differentiation and myelination: in vitro and in vivo studies. Biometals 2022; 35:395-427. [DOI: 10.1007/s10534-022-00380-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/27/2022] [Indexed: 12/20/2022]
|
8
|
Probable Reasons for Neuron Copper Deficiency in the Brain of Patients with Alzheimer’s Disease: The Complex Role of Amyloid. INORGANICS 2022. [DOI: 10.3390/inorganics10010006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease is a progressive neurodegenerative disorder that eventually leads the affected patients to die. The appearance of senile plaques in the brains of Alzheimer’s patients is known as a main symptom of this disease. The plaques consist of different components, and according to numerous reports, their main components include beta-amyloid peptide and transition metals such as copper. In this disease, metal dyshomeostasis leads the number of copper ions to simultaneously increase in the plaques and decrease in neurons. Copper ions are essential for proper brain functioning, and one of the possible mechanisms of neuronal death in Alzheimer’s disease is the copper depletion of neurons. However, the reason for the copper depletion is as yet unknown. Based on the available evidence, we suggest two possible reasons: the first is copper released from neurons (along with beta-amyloid peptides), which is deposited outside the neurons, and the second is the uptake of copper ions by activated microglia.
Collapse
|
9
|
Wen MH, Xie X, Huang PS, Yang K, Chen TY. Crossroads between membrane trafficking machinery and copper homeostasis in the nerve system. Open Biol 2021; 11:210128. [PMID: 34847776 PMCID: PMC8633785 DOI: 10.1098/rsob.210128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Imbalanced copper homeostasis and perturbation of membrane trafficking are two common symptoms that have been associated with the pathogenesis of neurodegenerative and neurodevelopmental diseases. Accumulating evidence from biophysical, cellular and in vivo studies suggest that membrane trafficking orchestrates both copper homeostasis and neural functions-however, a systematic review of how copper homeostasis and membrane trafficking interplays in neurons remains lacking. Here, we summarize current knowledge of the general trafficking itineraries for copper transporters and highlight several critical membrane trafficking regulators in maintaining copper homeostasis. We discuss how membrane trafficking regulators may alter copper transporter distribution in different membrane compartments to regulate intracellular copper homeostasis. Using Parkinson's disease and MEDNIK as examples, we further elaborate how misregulated trafficking regulators may interplay parallelly or synergistically with copper dyshomeostasis in devastating pathogenesis in neurodegenerative diseases. Finally, we explore multiple unsolved questions and highlight the existing challenges to understand how copper homeostasis is modulated through membrane trafficking.
Collapse
Affiliation(s)
- Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Xihong Xie
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Karen Yang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
10
|
Lachowicz JI, Lecca LI, Meloni F, Campagna M. Metals and Metal-Nanoparticles in Human Pathologies: From Exposure to Therapy. Molecules 2021; 26:6639. [PMID: 34771058 PMCID: PMC8587420 DOI: 10.3390/molecules26216639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/29/2021] [Accepted: 10/30/2021] [Indexed: 01/13/2023] Open
Abstract
An increasing number of pathologies correlates with both toxic and essential metal ions dyshomeostasis. Next to known genetic disorders (e.g., Wilson's Disease and β-Thalassemia) other pathological states such as neurodegeneration and diabetes are characterized by an imbalance of essential metal ions. Metal ions can enter the human body from the surrounding environment in the form of free metal ions or metal-nanoparticles, and successively translocate to different tissues, where they are accumulated and develop distinct pathologies. There are no characteristic symptoms of metal intoxication, and the exact diagnosis is still difficult. In this review, we present metal-related pathologies with the most common onsets, biomarkers of metal intoxication, and proper techniques of metal qualitative and quantitative analysis. We discuss the possible role of drugs with metal-chelating ability in metal dyshomeostasis, and present recent advances in therapies of metal-related diseases.
Collapse
Affiliation(s)
| | | | | | - Marcello Campagna
- Division of Occupational Medicine, Department of Medical Sciences and Public Health, University of Cagliari, 09048 Monserrato, CA, Italy; (J.I.L.); (L.I.L.); (F.M.)
| |
Collapse
|
11
|
Adamson SXF, Zheng W, Agim ZS, Du S, Fleming S, Shannahan J, Cannon J. Systemic Copper Disorders Influence the Olfactory Function in Adult Rats: Roles of Altered Adult Neurogenesis and Neurochemical Imbalance. Biomolecules 2021; 11:1315. [PMID: 34572528 PMCID: PMC8471899 DOI: 10.3390/biom11091315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Disrupted systemic copper (Cu) homeostasis underlies neurodegenerative diseases with early symptoms including olfactory dysfunction. This study investigated the impact of Cu dyshomeostasis on olfactory function, adult neurogenesis, and neurochemical balance. Models of Cu deficiency (CuD) and Cu overload (CuO) were established by feeding adult rats with Cu-restricted diets plus ip. injection of a Cu chelator (ammonium tetrathiomolybdate) and excess Cu, respectively. CuD reduced Cu levels in the olfactory bulb (OB), subventricular zone (SVZ), rostral migratory stream (RMS), and striatum, while CuO increased Cu levels in these areas. The buried pellet test revealed both CuD and CuO prolonged the latency to uncover food. CuD increased neural proliferation and stem cells in the SVZ and newly differentiated neurons in the OB, whereas CuO caused opposite alterations, suggesting a "switch"-type function of Cu in regulating adult neurogenesis. CuO increased GABA in the OB, while both CuD and CuO reduced DOPAC, HVA, 5-HT and the DA turnover rate in olfactory-associated brain regions. Altered mRNA expression of Cu transport and storage proteins in tested brain areas were observed under both conditions. Together, results support an association between systemic Cu dyshomeostasis and olfactory dysfunction. Specifically, altered adult neurogenesis along the SVZ-RMS-OB pathway and neurochemical imbalance could be the factors that may contribute to olfactory dysfunction.
Collapse
Affiliation(s)
- Sherleen Xue-Fu Adamson
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
- Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN 47907, USA
| | - Zeynep Sena Agim
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Sarah Du
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Sheila Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA;
| | - Jonathan Shannahan
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
| | - Jason Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47907, USA; (S.X.-F.A.); (Z.S.A.); (S.D.); (J.S.)
- Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
12
|
Michalke B, Berthele A, Venkataramani V. Simultaneous Quantification and Speciation of Trace Metals in Paired Serum and CSF Samples by Size Exclusion Chromatography-Inductively Coupled Plasma-Dynamic Reaction Cell-Mass Spectrometry (SEC-DRC-ICP-MS). Int J Mol Sci 2021; 22:8892. [PMID: 34445607 PMCID: PMC8396360 DOI: 10.3390/ijms22168892] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Transition metals play a crucial role in brain metabolism: since they exist in different oxidation states they are involved in ROS generation, but they are also co-factors of enzymes in cellular energy metabolism or oxidative defense. METHODS Paired serum and cerebrospinal fluid (CSF) samples were analyzed for iron, zinc, copper and manganese as well as for speciation using SEC-ICP-DRC-MS. Brain extracts from Mn-exposed rats were additionally analyzed with SEC-ICP-DRC-MS. RESULTS The concentration patterns of transition metal size fractions were correlated between serum and CSF: Total element concentrations were significantly lower in CSF. Fe-ferritin was decreased in CSF whereas a LMW Fe fraction was relatively increased. The 400-600 kDa Zn fraction and the Cu-ceruloplasmin fraction were decreased in CSF, by contrast the 40-80 kDa fraction, containing Cu- and Zn-albumin, relatively increased. For manganese, the α-2-macroglobulin fraction showed significantly lower concentration in CSF, whereas the citrate Mn fraction was enriched. Results from the rat brain extracts supported the findings from human paired serum and CSF samples. CONCLUSIONS Transition metals are strictly controlled at neural barriers (NB) of neurologic healthy patients. High molecular weight species are down-concentrated along NB, however, the Mn-citrate fraction seems to be less controlled, which may be problematic under environmental load.
Collapse
Affiliation(s)
- Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, Helmholtz Center Munich—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Achim Berthele
- Department of Neurology, School of Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany;
| | - Vivek Venkataramani
- Department of Medicine II, Hematology/Oncology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
- Institute of Pathology, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany
| |
Collapse
|
13
|
Ramos P, Pinto E, Santos A, Almeida A. Reference values for trace element levels in the human brain: A systematic review of the literature. J Trace Elem Med Biol 2021; 66:126745. [PMID: 33813265 DOI: 10.1016/j.jtemb.2021.126745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/14/2020] [Accepted: 03/15/2021] [Indexed: 11/25/2022]
Abstract
Some trace elements (TE) are eminently toxic for humans (e.g., Al, Pb, Hg, Cd) and its presence in the central nervous system has been linked to the etiology of neurodegenerative diseases (ND). More recently, the focus has shifted to the potential role of the imbalances on essential TE levels (e.g., Fe, Cu, Zn, Se) within the brain tissue, and they have also been identified as potentially responsible for the cognitive decline associated with normal ageing and the development of some ND, although their definite role remains unclear. Accurately, well-defined reference values for TE levels in human body fluids and tissues are indispensable to identify possible disturbances in individual cases. Moreover, since the brain is a highly heterogeneous organ, with anatomically and physiologically very different areas, a detailed mapping of TE distribution across the brain tissue of normal individuals, with an in-depth analysis of TE levels in the different brain regions, is a mandatory prior work so that the results obtained from patients suffering from ND and other brain diseases can be interpreted. This review aims to compile and summarize the available data regarding TE levels in the different human brain regions of "normal" (non-diseased) individuals in order to contribute to the establishment of robust reference values. Fifty-four studies, published since 1960, were considered. The results showed a great variability between different studies. The potential sources of this variability are discussed. The need for increased harmonization of experimental strategies is highlighted in order to improve the comparability of the data obtained.
Collapse
Affiliation(s)
- Patrícia Ramos
- LAQV / REQUIMTE, Department of Chemical Sciences, Laboratory of Applied Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Edgar Pinto
- LAQV / REQUIMTE, Department of Chemical Sciences, Laboratory of Applied Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal; Department of Environmental Health, School of Health, P.Porto, CISA/Research Center in Environment and Health, 4200-072, Porto, Portugal
| | - Agostinho Santos
- National Institute of Legal Medicine and Forensic Sciences, North Branch, Jardim Carrilho Videira, 4050-167, Porto, Portugal; Faculty of Medicine, University of Porto, Al. Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Agostinho Almeida
- LAQV / REQUIMTE, Department of Chemical Sciences, Laboratory of Applied Chemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
14
|
Scholefield M, Church SJ, Xu J, Patassini S, Roncaroli F, Hooper NM, Unwin RD, Cooper GJS. Widespread Decreases in Cerebral Copper Are Common to Parkinson's Disease Dementia and Alzheimer's Disease Dementia. Front Aging Neurosci 2021; 13:641222. [PMID: 33746735 PMCID: PMC7966713 DOI: 10.3389/fnagi.2021.641222] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/05/2021] [Indexed: 01/24/2023] Open
Abstract
Several studies of Parkinson's disease (PD) have reported dysregulation of cerebral metals, particularly decreases in copper and increases in iron in substantia nigra (SN). However, few studies have investigated regions outside the SN, fewer have measured levels of multiple metals across different regions within the same brains, and there are no currently-available reports of metal levels in Parkinson's disease dementia (PDD). This study aimed to compare concentrations of nine essential metals across nine different brain regions in cases of PDD and controls. Investigated were: primary motor cortex (MCX); cingulate gyrus (CG); primary visual cortex (PVC); hippocampus (HP); cerebellar cortex (CB); SN; locus coeruleus (LC); medulla oblongata (MED); and middle temporal gyrus (MTG), thus covering regions with severe, moderate, or low levels of neuronal loss in PDD. Levels of eight essential metals and selenium were determined using an analytical methodology involving the use of inductively-coupled plasma mass spectrometry (ICP-MS), and compared between cases and controls, to better understand the extent and severity of metal perturbations. Findings were also compared with those from our previous study of sporadic Alzheimer's disease dementia (ADD), which employed equivalent methods, to identify differences and similarities between these conditions. Widespread copper decreases occurred in PDD in seven of nine regions (exceptions being LC and CB). Four PDD-affected regions showed similar decreases in ADD: CG, HP, MTG, and MCX. Decreases in potassium and manganese were present in HP, MTG and MCX; decreased manganese was also found in SN and MED. Decreased selenium and magnesium were present in MCX, and decreased zinc in HP. There was no evidence for increased iron in SN or any other region. These results identify alterations in levels of several metals across multiple regions of PDD brain, the commonest being widespread decreases in copper that closely resemble those in ADD, pointing to similar disease mechanisms in both dementias.
Collapse
Affiliation(s)
- Melissa Scholefield
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stephanie J. Church
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Jingshu Xu
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Stefano Patassini
- Faculty of Science, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Division of Neuroscience and Experimental Psychology, Faculty of Brain and Mental Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Nigel M. Hooper
- Division of Neuroscience & Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Richard D. Unwin
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Stoller Biomarker Discovery Centre & Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, The University of Manchester, Manchester, United Kingdom
| | - Garth J. S. Cooper
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Centre for Advanced Discovery & Experimental Therapeutics, School of Medical Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Faculty of Science, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
15
|
Cilliers K. Trace element alterations in Alzheimer's disease: A review. Clin Anat 2021; 34:766-773. [PMID: 33580904 DOI: 10.1002/ca.23727] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 12/27/2022]
Abstract
Dyshomeostasis of trace elements have been implicated in the progression of Alzheimer's disease (AD), which is characterized by amyloid-β (Aβ) plaques. Trace elements are particularly associated with the Aβ plaques. Metal-protein attenuating compounds have been developed to inhibit metals from binding to Aβ proteins, which result in Aβ termination, in the hope of improving cognitive functioning. However, there are still some contradicting reports. This review aims to first establish which trace elements are increased or decreased in the brains of Alzheimer's patients, and secondly, to review the effectiveness of clinical trials with metal-protein attenuating compounds for AD. Studies have consistently reported unchanged or increased iron, contradicting reports for zinc, decreased copper, unchanged or decreased manganese, inconsistent results for calcium, and magnesium seems to be unaffected. However, varied results have been reported for all trace elements. Clinical trials using metal-protein attenuating compounds to treat AD have also reported varied results. Copper chelators have repeatedly been used in clinical trials, even though few studies report increased brain copper levels in AD patients. Homeostasis of copper levels is important since copper has a vital role in several enzymes, such as cytochrome c, Cu/Zn superoxide dismutase and ceruloplasmin. Dyshomeostasis of copper levels can lead to increased oxidative stress and neuronal loss. Future studies should assess a variety of trace element levels in moderately and severely affected AD patients since there are contradicting reports. This review thus provides some insight into trace element alterations in the brains of individuals with AD.
Collapse
Affiliation(s)
- Karen Cilliers
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, Western Cape, South Africa
| |
Collapse
|
16
|
Cilliers K, Muller CJF. Multi-element Analysis of Brain Regions from South African Cadavers. Biol Trace Elem Res 2021; 199:425-441. [PMID: 32361883 DOI: 10.1007/s12011-020-02158-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022]
Abstract
Trace elements are vital for a variety of functions in the brain. However, an imbalance can result in oxidative stress. It is important to ascertain the normal levels in different brain regions, as such information is still lacking. Therefore, this study aimed to provide baseline trace element concentrations from a South African population, as well as determine trace element differences between sex and brain regions. Samples from the caudate nucleus, putamen, globus pallidus and hippocampus were analysed using inductively coupled plasma mass spectrometry. Aluminium, antimony, arsenic, barium, boron, cadmium, calcium, chromium, cobalt, copper, iron, lead, magnesium, manganese, mercury, molybdenum, nickel, phosphorus, potassium, selenium, silicon, sodium, strontium, vanadium and zinc were assessed. A multiple median regression model was used to determine differences between sex and regions. Twenty-nine male and 13 female cadavers from a Western Cape, South African population were included (mean age 35 years, range 19 to 45). Trace element levels were comparable to those of other populations, although magnesium was considerably lower. While there were no sex differences, significant anatomical regional differences existed; the caudate nucleus and hippocampus were the most similar, and the globus pallidus and hippocampus the most different. In conclusion, this is the first article to report the trace element concentrations of brain regions from a South African population. Low magnesium levels in the brain may be linked to a dietary deficiency, and migraines, depression and epilepsy have been linked to low magnesium levels. Future research should be directed to increase the dietary intake of magnesium.
Collapse
Affiliation(s)
- Karen Cilliers
- Division of Clinical Anatomy, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Western Cape, South Africa.
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Western Cape, South Africa
- Division of Medical Physiology, Faculty of Medicine and Health ScieAnces, Stellenbosch University, Tygerberg, Western Cape, South Africa
| |
Collapse
|
17
|
Wandt VK, Winkelbeiner N, Bornhorst J, Witt B, Raschke S, Simon L, Ebert F, Kipp AP, Schwerdtle T. A matter of concern - Trace element dyshomeostasis and genomic stability in neurons. Redox Biol 2021; 41:101877. [PMID: 33607499 PMCID: PMC7902532 DOI: 10.1016/j.redox.2021.101877] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/18/2020] [Accepted: 01/20/2021] [Indexed: 02/09/2023] Open
Abstract
Neurons are post-mitotic cells in the brain and their integrity is of central importance to avoid neurodegeneration. Yet, the inability of self-replenishment of post-mitotic cells results in the need to withstand challenges from numerous stressors during life. Neurons are exposed to oxidative stress due to high oxygen consumption during metabolic activity in the brain. Accordingly, DNA damage can occur and accumulate, resulting in genome instability. In this context, imbalances in brain trace element homeostasis are a matter of concern, especially regarding iron, copper, manganese, zinc, and selenium. Although trace elements are essential for brain physiology, excess and deficient conditions are considered to impair neuronal maintenance. Besides increasing oxidative stress, DNA damage response and repair of oxidative DNA damage are affected by trace elements. Hence, a balanced trace element homeostasis is of particular importance to safeguard neuronal genome integrity and prevent neuronal loss. This review summarises the current state of knowledge on the impact of deficient, as well as excessive iron, copper, manganese, zinc, and selenium levels on neuronal genome stability. Post-mitotic neurons show an increased vulnerability to oxidative stress. Trace element dyshomeostasis impairs neuronal genome maintenance, affecting DNA damage response as well as DNA repair. The review summarises the effects of excessive and deficient trace element levels neuronal genome stability maintenance.
Collapse
Affiliation(s)
- Viktoria K Wandt
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Nicola Winkelbeiner
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Julia Bornhorst
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstr. 20, 42119, Wuppertal, Germany.
| | - Barbara Witt
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Stefanie Raschke
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany.
| | - Luise Simon
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany.
| | - Anna P Kipp
- TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; Department of Molecular Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743, Jena, Germany.
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; TraceAge - DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany; German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| |
Collapse
|
18
|
Cilliers K, Muller CJF. Effect of Human Immunodeficiency Virus on Trace Elements in the Brain. Biol Trace Elem Res 2021; 199:41-52. [PMID: 32239375 DOI: 10.1007/s12011-020-02129-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/23/2020] [Indexed: 11/27/2022]
Abstract
Comorbidities of human immunodeficiency virus (HIV) include HIV-associated neurocognitive disorder (HAND). Changes in the brain due to HIV include atrophy, hyperintensities, and diffusion changes. However, no research has focused on trace elements concentration changes in the brain due to HIV, as seen in other neurodegenerative diseases. Therefore, the aim of this study was to determine the concentration of several trace elements in the brains of individuals with and without HIV infection. Prior to formalin embalming, blood was drawn and tested in triplicate with Determine HIV-1/2 rapid tests and confirmed with a SD HIV Device 1/2 3.0 rapid HIV Kit. After embalming, tissue was sampled from the caudate nucleus and analyzed using inductively coupled plasma mass spectrometry. A Kruskal-Wallis test was used to determine statistically significant differences between the two groups (p < 0.05). Fifteen HIV-positive and 14 HIV-negative male cadavers were included (mean age 44, range 22 to 61). Cadmium was marginally decreased, possibly due to malnutrition or utilization by the HIV nucleocapsid. Nickel was marginally increased, perhaps due to a reduced capability to remove metals from the body. In conclusion, this article provides the first information on trace element levels in the brains from HIV-infected individuals and postulates that cadmium and nickel may play a role in the pathophysiology of HAND. This information can contribute to finding a treatment for HAND, other than the use of antiretroviral drugs. Future studies should asses the levels of cadmium and nickel in a larger cohort of HIV-infected individuals.
Collapse
Affiliation(s)
- Karen Cilliers
- Division of Clinical Anatomy, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Western Cape, South Africa.
| | - Christo J F Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council, Tygerberg, Western Cape, South Africa
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Western Cape, South Africa
| |
Collapse
|
19
|
Hollings AL, Lam V, Takechi R, Mamo JCL, Reinhardt J, de Jonge MD, Kappen P, Hackett MJ. Revealing differences in the chemical form of zinc in brain tissue using K-edge X-ray absorption near-edge structure spectroscopy. Metallomics 2020; 12:2134-2144. [PMID: 33300524 DOI: 10.1039/d0mt00198h] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Zinc is a prominent trace metal required for normal memory function. Memory loss and cognitive decline during natural ageing and neurodegenerative disease have been associated with altered brain-Zn homeostasis. Yet, the exact chemical pathways through which Zn influences memory function during health, natural ageing, or neurodegenerative disease remain unknown. The gap in the literature may in part be due to the difficulty to simultaneously image, and therefore, study the different chemical forms of Zn within the brain (or biological samples in general). To this extent, we have begun developing and optimising protocols that incorporate X-ray absorption near-edge structure (XANES) spectroscopic analysis of tissue at the Zn K-edge as an analytical tool to study Zn speciation in the brain. XANES is ideally suited for this task as all chemical forms of Zn are detected, the technique requires minimal sample preparation that may otherwise redistribute or alter the chemical form of Zn, and the Zn K-edge has known sensitivity to coordination geometry and ligand type. Herein, we report our initial results where we fit K-edge spectra collected from micro-dissected flash-frozen brain tissue, to a spectral library prepared from standard solutions, to demonstrate differences in the chemical form of Zn that exist between two brain regions, the hippocampus and cerebellum. Lastly, we have used an X-ray microprobe to demonstrate differences in Zn speciation within sub-regions of thin air-dried sections of the murine hippocampus; but, the corresponding results highlight that the chemical form of Zn is easily perturbed by sample preparation such as tissue sectioning or air-drying, which must be a critical consideration for future work.
Collapse
Affiliation(s)
- Ashley L Hollings
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6102, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Mahan B, Antonelli MA, Burckel P, Turner S, Chung R, Habekost M, Jørgensen AL, Moynier F. Longitudinal biometal accumulation and Ca isotope composition of the Göttingen minipig brain. Metallomics 2020; 12:1585-1598. [PMID: 33084720 DOI: 10.1039/d0mt00134a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biometals play a critical role in both the healthy and diseased brain's functioning. They accumulate in the normal aging brain, and are inherent to neurodegenerative disorders and their associated pathologies. A prominent example of this is the brain accumulation of metals such as Ca, Fe and Cu (and more ambiguously, Zn) associated with Alzheimer's disease (AD). The natural stable isotope compositions of such metals have also shown utility in constraining biological mechanisms, and in differentiating between healthy and diseased states, sometimes prior to conventional methods. Here we have detailed the distribution of the biologically relevant elements Mg, P, K, Ca, Fe, Cu and Zn in brain regions of Göttingen minipigs ranging in age from three months to nearly six years, including control animals and both a single- and double-transgenic model of AD (PS1, APP/PS1). Moreover, we have characterized the Ca isotope composition of the brain for the first time. Concentration data track rises in brain biometals with age, namely for Fe and Cu, as observed in the normal ageing brain and in AD, and biometal data point to increased soluble amyloid beta (Aβ) load prior to AD plaque identification via brain imaging. Calcium isotope results define the brain as the isotopically lightest permanent reservoir in the body, indicating that brain Ca dyshomeostasis may induce measurable isotopic disturbances in accessible downstream reservoirs such as biofluids.
Collapse
Affiliation(s)
- Brandon Mahan
- Earth and Environmental Science, James Cook University, Townsville, Queensland 4811, Australia. and Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Michael A Antonelli
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France and Institute of Geochemistry and Petrology, Department of Earth Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Pierre Burckel
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France
| | - Simon Turner
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Roger Chung
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Mette Habekost
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Frédéric Moynier
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France
| |
Collapse
|
21
|
Air Pollution-Related Brain Metal Dyshomeostasis as a Potential Risk Factor for Neurodevelopmental Disorders and Neurodegenerative Diseases. ATMOSPHERE 2020. [DOI: 10.3390/atmos11101098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence links air pollution (AP) exposure to effects on the central nervous system structure and function. Particulate matter AP, especially the ultrafine (nanoparticle) components, can carry numerous metal and trace element contaminants that can reach the brain in utero and after birth. Excess brain exposure to either essential or non-essential elements can result in brain dyshomeostasis, which has been implicated in both neurodevelopmental disorders (NDDs; autism spectrum disorder, schizophrenia, and attention deficit hyperactivity disorder) and neurodegenerative diseases (NDGDs; Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis). This review summarizes the current understanding of the extent to which the inhalational or intranasal instillation of metals reproduces in vivo the shared features of NDDs and NDGDs, including enlarged lateral ventricles, alterations in myelination, glutamatergic dysfunction, neuronal cell death, inflammation, microglial activation, oxidative stress, mitochondrial dysfunction, altered social behaviors, cognitive dysfunction, and impulsivity. Although evidence is limited to date, neuronal cell death, oxidative stress, and mitochondrial dysfunction are reproduced by numerous metals. Understanding the specific contribution of metals/trace elements to this neurotoxicity can guide the development of more realistic animal exposure models of human AP exposure and consequently lead to a more meaningful approach to mechanistic studies, potential intervention strategies, and regulatory requirements.
Collapse
|
22
|
DeBenedictis CA, Raab A, Ducie E, Howley S, Feldmann J, Grabrucker AM. Concentrations of Essential Trace Metals in the Brain of Animal Species-A Comparative Study. Brain Sci 2020; 10:E460. [PMID: 32709155 PMCID: PMC7407190 DOI: 10.3390/brainsci10070460] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/29/2022] Open
Abstract
The essential trace metals iron, zinc, and copper have a significant physiological role in healthy brain development and function. Especially zinc is important for neurogenesis, synaptogenesis, synaptic transmission and plasticity, and neurite outgrowth. Given the key role of trace metals in many cellular processes, it is important to maintain adequate levels in the brain. However, the physiological concentration of trace metals, and in particular zinc, in the human and animal brain is not well described so far. For example, little is known about the trace metal content of the brain of animals outside the class of mammals. Here, we report the concentration of iron, zinc, and copper in fresh brain tissue of different model-species of the phyla Chordata (vertebrates (mammals, fish)), Annelida, Arthropoda (insects), and Mollusca (snails), using inductively coupled plasma mass-spectrometry (ICP-MS). Our results show that the trace metals are present in the nervous system of all species and that significant differences can be detected between species of different phyla. We further show that a region-specific distribution of metals within the nervous system already exists in earthworms, hinting at a tightly controlled metal distribution. In line with this, the trace metal content of the brain of different species does not simply correlate with brain size. We conclude that although the functional consequences of the controlled metal homeostasis within the brain of many species remains elusive, trace metal biology may not only play an important role in the nervous system of mammals but across the whole animal kingdom.
Collapse
Affiliation(s)
- Chiara Alessia DeBenedictis
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.); (E.D.); (S.H.)
- Bernal Institute, University of Limerick, V94T9PX Limerick, Ireland
| | - Andrea Raab
- Trace Element Speciation Laboratory (TESLA), Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, UK; (A.R.); (J.F.)
- Institute of Chemistry, University of Graz, A-8010 Graz, Austria
- Institute of Chemistry, Environmental Analytical Chemistry, University of Graz, 8010 Graz, Austria
| | - Ellen Ducie
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.); (E.D.); (S.H.)
| | - Shauna Howley
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.); (E.D.); (S.H.)
| | - Joerg Feldmann
- Trace Element Speciation Laboratory (TESLA), Department of Chemistry, University of Aberdeen, Aberdeen AB24 3UE, UK; (A.R.); (J.F.)
- Institute of Chemistry, University of Graz, A-8010 Graz, Austria
- Institute of Chemistry, Environmental Analytical Chemistry, University of Graz, 8010 Graz, Austria
| | - Andreas Martin Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland; (C.A.D.); (E.D.); (S.H.)
- Bernal Institute, University of Limerick, V94T9PX Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, V94T9PX Limerick, Ireland
| |
Collapse
|
23
|
Huntington's disease associated resistance to Mn neurotoxicity is neurodevelopmental stage and neuronal lineage dependent. Neurotoxicology 2019; 75:148-157. [PMID: 31545971 DOI: 10.1016/j.neuro.2019.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
Manganese (Mn) is essential for neuronal health but neurotoxic in excess. Mn levels vary across brain regions and neurodevelopment. While Mn requirements during infanthood and childhood are significantly higher than in adulthood, the relative vulnerability to excess extracellular Mn across human neuronal developmental time and between distinct neural lineages is unknown. Neurological disease is associated with changes in brain Mn homeostasis and pathology associated with Mn neurotoxicity is not uniform across brain regions. For example, mutations associated with Huntington's disease (HD) decrease Mn bioavailability and increase resistance to Mn cytotoxicity in human and mouse striatal neuronal progenitors. Here, we sought to compare the differences in Mn cytotoxicity between control and HD human-induced pluripotent stem cells (hiPSCs)-derived neuroprogenitor cells (NPCs) and maturing neurons. We hypothesized that there would be differences in Mn sensitivity between lineages and developmental stages. However, we found that the different NPC lineage specific media substantially influenced Mn cytotoxicity in the hiPSC derived human NPCs and did so consistently even in a non-human cell line. This limited the ability to determine which human neuronal sub-types were more sensitive to Mn. Nonetheless, we compared within neuronal subtypes and developmental stage the sensitivity to Mn cytotoxicity between control and HD patient derived neuronal lineages. Consistent with studies in other striatal model systems the HD genotype was associated with resistance to Mn cytotoxicity in human striatal NPCs. In addition, we report an HD genotype-dependent resistance to Mn cytotoxicity in cortical NPCs and hiPSCs. Unexpectedly, the HD genotype conferred increased sensitivity to Mn in early post-mitotic midbrain neurons but had no effect on Mn sensitivity in midbrain NPCs or post-mitotic cortical neurons. Overall, our data suggest that sensitivity to Mn cytotoxicity is influenced by HD genotype in a human neuronal lineage type and stage of development dependent manner.
Collapse
|
24
|
Zhao X, Liu Y, Zhu G, Liang Y, Liu B, Wu Y, Han M, Sun W, Han Y, Chen G, Jiang J. SIRT1 downregulation mediated Manganese-induced neuronal apoptosis through activation of FOXO3a-Bim/PUMA axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 646:1047-1055. [PMID: 30235590 DOI: 10.1016/j.scitotenv.2018.07.363] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/21/2018] [Accepted: 07/25/2018] [Indexed: 06/08/2023]
Abstract
Manganese (Mn) is an essential trace element. Excessive exposure to Mn may lead to neuronal death and neurodegenerative disorders. Accumulating evidence has shown that silent mating type information regulation 2 homolog 1 (SIRT1) plays a vital role in brain damage. However, whether aberrant SIRT1 levels contribute to Mn-induced neurotoxicity remains unknown. In this study, we report the important role of SIRT1 downregulation during Mn-induced neuronal apoptosis. Mn was found to downregulate SIRT1 protein levels in the rat pheochromocytoma (PC12) cells and mouse brain tissues. Mn enhanced SIRT1 protein degradation and downregulated its gene expression. Furthermore, Mn induced cell apoptosis in a dose-dependent manner both in vitro and in vivo, and resulted in an increase in forkhead box O (FOXO) 3a expression and acetylation. SIRT1 activation by resveratrol clearly attenuated Mn-triggered apoptosis and FOXO3a activation. Mn markedly increased the expression of Bcl-2 interacting mediator of cell death (Bim) and p53-up-regulated modulator of apoptosis (PUMA), whereas downregulation of FOXO3a significantly inhibited their upregulation and subsequent apoptosis. In summary, we determined that Mn downregulated SIRT1 by multiple mechanisms, thus led to apoptosis via activation of the FOXO3a-Bim/PUMA axis in PC12 cells. These findings on the impact of Mn on SIRT1 may lead to an improved understanding of Mn-induced neurotoxicity and provide a molecular target to antagonise Mn-associated neuronal damage.
Collapse
Affiliation(s)
- Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yiming Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Ganlin Zhu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yuanyuan Liang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Bo Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yifan Wu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Muxi Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Wenxing Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Yu Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Gang Chen
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
25
|
Kardos J, Héja L, Simon Á, Jablonkai I, Kovács R, Jemnitz K. Copper signalling: causes and consequences. Cell Commun Signal 2018; 16:71. [PMID: 30348177 PMCID: PMC6198518 DOI: 10.1186/s12964-018-0277-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022] Open
Abstract
Copper-containing enzymes perform fundamental functions by activating dioxygen (O2) and therefore allowing chemical energy-transfer for aerobic metabolism. The copper-dependence of O2 transport, metabolism and production of signalling molecules are supported by molecular systems that regulate and preserve tightly-bound static and weakly-bound dynamic cellular copper pools. Disruption of the reducing intracellular environment, characterized by glutathione shortage and ambient Cu(II) abundance drives oxidative stress and interferes with the bidirectional, copper-dependent communication between neurons and astrocytes, eventually leading to various brain disease forms. A deeper understanding of of the regulatory effects of copper on neuro-glia coupling via polyamine metabolism may reveal novel copper signalling functions and new directions for therapeutic intervention in brain disorders associated with aberrant copper metabolism.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - István Jablonkai
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Richard Kovács
- Institute of Neurophysiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Katalin Jemnitz
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| |
Collapse
|
26
|
Słupski J, Cubała WJ, Górska N, Gałuszko-Węgielnik M, Wiglusz MS. Role of copper in depression. Relationship with ketamine treatment. Med Hypotheses 2018; 119:14-17. [PMID: 30122482 DOI: 10.1016/j.mehy.2018.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/28/2018] [Accepted: 07/14/2018] [Indexed: 01/26/2023]
Abstract
Depression is one of the most common psychiatric issues with a proportion of adults with major depressive disorder who fail to achieve remission with index pharmacological treatment. There are unmet needs in ADT focus on non-monoaminergic agents. Accumulating evidence suggests that the N-Methyl-d-aspartate receptor (NMDAR) plays an important role in the neurobiology and treatment of major depressive disorder. The role of copper ions in pathogenesis and treatment of depression is not fully clarified, however interaction between copper and NMDAR is of prime importance. Release of copper ions inhibits NMDAR and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor function thus protects neurons from glutamatergic excitotoxity. Abnormalities in glutamatergic transmission are the key of glutamate hypothesis of depression. Some authors revealed that NMDARs are also regulated by cellular prion protein (PrPC) and indicated that interactions of copper, glycine and NMDARs subunits are vital for the regulation of the receptor. As NMDAR antagonist ketamine is known to produce rapid antidepressive effect, observation of copper serum levels in patients treated with ketamine may provide important information about connections between NMDAR antagonistic agents and trace elements antagonistic to that receptor. It is necessary to carry out further studies related to copper and ketamine in depression treatment.
Collapse
Affiliation(s)
- Jakub Słupski
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - Wiesław Jerzy Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Górska
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | | |
Collapse
|
27
|
González de San Román E, Bidmon HJ, Malisic M, Susnea I, Küppers A, Hübbers R, Wree A, Nischwitz V, Amunts K, Huesgen PF. Molecular composition of the human primary visual cortex profiled by multimodal mass spectrometry imaging. Brain Struct Funct 2018; 223:2767-2783. [PMID: 29633039 PMCID: PMC5995978 DOI: 10.1007/s00429-018-1660-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 03/29/2018] [Indexed: 12/14/2022]
Abstract
The primary visual cortex (area V1) is an extensively studied part of the cerebral cortex with well-characterized connectivity, cellular and molecular architecture and functions (for recent reviews see Amunts and Zilles, Neuron 88:1086-1107, 2015; Casagrande and Xu, Parallel visual pathways: a comparative perspective. The visual neurosciences, MIT Press, Cambridge, pp 494-506, 2004). In humans, V1 is defined by heavily myelinated fibers arriving from the radiatio optica that form the Gennari stripe in cortical layer IV, which is further subdivided into laminae IVa, IVb, IVcα and IVcβ. Due to this unique laminar pattern, V1 represents an excellent region to test whether multimodal mass spectrometric imaging could reveal novel biomolecular markers for a functionally relevant parcellation of the human cerebral cortex. Here we analyzed histological sections of three post-mortem brains with matrix-assisted laser desorption/ionization mass spectrometry imaging and laser ablation inductively coupled plasma mass spectrometry imaging to investigate the distribution of lipids, proteins and metals in human V1. We identified 71 peptides of 13 different proteins by in situ tandem mass spectrometry, of which 5 proteins show a differential laminar distribution pattern revealing the border between V1 and V2. High-accuracy mass measurements identified 123 lipid species, including glycerolipids, glycerophospholipids and sphingolipids, of which at least 20 showed differential distribution within V1 and V2. Specific lipids labeled not only myelinated layer IVb, but also IVa and especially IVc in a layer-specific manner, but also and clearly separated V1 from V2. Elemental imaging further showed a specific accumulation of copper in layer IV. In conclusion, multimodal mass spectrometry imaging identified novel biomolecular and elemental markers with specific laminar and inter-areal differences. We conclude that mass spectrometry imaging provides a promising new approach toward multimodal, molecule-based cortical parcellation.
Collapse
Affiliation(s)
- Estibaliz González de San Román
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
- Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hans-Jürgen Bidmon
- Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Milena Malisic
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
- Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Iuliana Susnea
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | - Astrid Küppers
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | - Rene Hübbers
- Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine, INM-1, Forschungszentrum Jülich, Jülich, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Volker Nischwitz
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | - Katrin Amunts
- Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Institute of Neuroscience and Medicine, INM-1, Forschungszentrum Jülich, Jülich, Germany.
| | - Pitter F Huesgen
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
28
|
Effect of Age on High T1 Signal Intensity of the Dentate Nucleus and Globus Pallidus in a Large Population Exposed to Gadodiamide. Invest Radiol 2018; 53:214-222. [DOI: 10.1097/rli.0000000000000431] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Xu J, Church SJ, Patassini S, Begley P, Kellett KAB, Vardy ERLC, Unwin RD, Hooper NM, Cooper GJS. Plasma metals as potential biomarkers in dementia: a case-control study in patients with sporadic Alzheimer's disease. Biometals 2018; 31:267-276. [PMID: 29516299 PMCID: PMC5978903 DOI: 10.1007/s10534-018-0089-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/26/2018] [Indexed: 12/17/2022]
Abstract
Sporadic Alzheimer's disease (AD) is a neurodegenerative disorder that causes the most prevalent form of age-related dementia but its pathogenesis remains obscure. Altered regulation of metals, particularly pan-cerebral copper deficiency, and more regionally-localized perturbation of other metals, are prominent in AD brain although data on how these CNS perturbations are reflected in the peripheral bloodstream are inconsistent to date. To assess the potential use of metal dysregulation to generate biomarkers in AD, we performed a case-control study of seven essential metals and selenium, measured by inductively coupled plasma mass-spectrometry, in samples from AD and matched control cases. Metals were sodium, potassium, calcium, magnesium, iron, zinc, and copper. In the whole study-group and in female participants, plasma metal levels did not differ between cases and controls. In males by contrast, there was moderate evidence that zinc levels trended towards increase in AD [10.8 (10.2-11.5)] µmol/L, mean (± 95% CI; P = 0.021) compared with controls [10.2 (9.6-10.4)]. Thus alterations in plasma zinc levels differed between genders in AD. In correlational analysis, there was evidence for an increased number of 'strong' metal co-regulations in AD cases and differential co-modulations of metal pairs: copper-sodium (Rcontrol = - 0.03, RAD = 0.65; P = 0.009), and copper-calcium (Rcontrol = - 0.01, RAD = 0.65; P = 0.01) were significant in AD males, potentially consistent with reported evidence for dysregulation of copper in severely damaged brain regions in AD. In conclusion, our data suggest that the measurement of metals co-regulation in plasma may provide a useful representation of those metal perturbations taking place in the AD brain and therefore might be useful as plasma-based biomarkers.
Collapse
Affiliation(s)
- Jingshu Xu
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Stefano Patassini
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Katherine A B Kellett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Emma R L C Vardy
- Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Nigel M Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Garth J S Cooper
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
- School of Biological Sciences and Maurice Wilkins Centre for Molecular Biodiscovery, Faculty of Science, University of Auckland, Auckland, New Zealand.
- Rm 3.08, Core Technology Facility, Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Grafton Street, Manchester, M13 9NT, UK.
| |
Collapse
|
30
|
Xu J, Church SJ, Patassini S, Begley P, Waldvogel HJ, Curtis MA, Faull RLM, Unwin RD, Cooper GJS. Evidence for widespread, severe brain copper deficiency in Alzheimer's dementia. Metallomics 2017; 9:1106-1119. [PMID: 28654115 DOI: 10.1039/c7mt00074j] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Datasets comprising simultaneous measurements of many essential metals in Alzheimer's disease (AD) brain are sparse, and available studies are not entirely in agreement. To further elucidate this matter, we employed inductively-coupled-plasma mass spectrometry to measure post-mortem levels of 8 essential metals and selenium, in 7 brain regions from 9 cases with AD (neuropathological severity Braak IV-VI), and 13 controls who had normal ante-mortem mental function and no evidence of brain disease. Of the regions studied, three undergo severe neuronal damage in AD (hippocampus, entorhinal cortex and middle-temporal gyrus); three are less-severely affected (sensory cortex, motor cortex and cingulate gyrus); and one (cerebellum) is relatively spared. Metal concentrations in the controls differed among brain regions, and AD-associated perturbations in most metals occurred in only a few: regions more severely affected by neurodegeneration generally showed alterations in more metals, and cerebellum displayed a distinctive pattern. By contrast, copper levels were substantively decreased in all AD-brain regions, to 52.8-70.2% of corresponding control values, consistent with pan-cerebral copper deficiency. This copper deficiency could be pathogenic in AD, since levels are lowered to values approximating those in Menkes' disease, an X-linked recessive disorder where brain-copper deficiency is the accepted cause of severe brain damage. Our study reinforces others reporting deficient brain copper in AD, and indicates that interventions aimed at safely and effectively elevating brain copper could provide a new experimental-therapeutic approach.
Collapse
Affiliation(s)
- Jingshu Xu
- School of Biological Sciences, Faculty of Science, and the Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand. and Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| | - Stefano Patassini
- School of Biological Sciences, Faculty of Science, and the Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand. and Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| | - Henry J Waldvogel
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| | - Garth J S Cooper
- School of Biological Sciences, Faculty of Science, and the Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand. and Centre for Advanced Discovery and Experimental Therapeutics, Central Manchester University Hospitals NHS Foundation Trust (CMFT), Manchester M13 9WL, UK and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand and Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, and Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| |
Collapse
|
31
|
Božić B, Korać J, Stanković DM, Stanić M, Popović-Bijelić A, Bogdanović Pristov J, Spasojević I, Bajčetić M. Mechanisms of redox interactions of bilirubin with copper and the effects of penicillamine. Chem Biol Interact 2017; 278:129-134. [PMID: 29079291 DOI: 10.1016/j.cbi.2017.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/10/2017] [Accepted: 10/22/2017] [Indexed: 10/18/2022]
Abstract
Toxic effects of unconjugated bilirubin (BR) in neonatal hyperbilirubinemia have been related to redox and/or coordinate interactions with Cu2+. However, the development and mechanisms of such interactions at physiological pH have not been resolved. This study shows that BR reduces Cu2+ to Cu1+ in 1:1 stoichiometry. Apparently, BR undergoes degradation, i.e. BR and Cu2+ do not form stable complexes. The binding of Cu2+ to inorganic phosphates, liposomal phosphate groups, or to chelating drug penicillamine, impedes redox interactions with BR. Cu1+ undergoes spontaneous oxidation by O2 resulting in hydrogen peroxide accumulation and hydroxyl radical production. In relation to this, copper and BR induced synergistic oxidative/damaging effects on erythrocytes membrane, which were alleviated by penicillamine. The production of reactive oxygen species by BR and copper represents a plausible cause of BR toxic effects and cell damage in hyperbilirubinemia. Further examination of therapeutic potentials of copper chelators in the treatment of severe neonatal hyperbilirubinemia is needed.
Collapse
Affiliation(s)
- Bojana Božić
- Department of Pharmacology, Clinical Pharmacology and Toxicology, School of Medicine, University of Belgrade, P.O. Box 38, 11000 Belgrade, Serbia
| | - Jelena Korać
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Kneza Višeslava 1, 11000 Belgrade, Serbia
| | - Dalibor M Stanković
- The Vinča Institute of Nuclear Sciences, University of Belgrade, POB 522, 11001 Belgrade, Serbia; Department of Analytical Chemistry, Innovation Center of the Faculty of Chemistry, University of Belgrade, Studentski trg 12-16, Belgrade, 11000, Serbia
| | - Marina Stanić
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Kneza Višeslava 1, 11000 Belgrade, Serbia
| | - Ana Popović-Bijelić
- EPR Laboratory, Faculty of Physical Chemistry, University of Belgrade, Studentski trg 12-16, 11158 Belgrade, Serbia
| | - Jelena Bogdanović Pristov
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Kneza Višeslava 1, 11000 Belgrade, Serbia
| | - Ivan Spasojević
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Kneza Višeslava 1, 11000 Belgrade, Serbia.
| | - Milica Bajčetić
- Department of Pharmacology, Clinical Pharmacology and Toxicology, School of Medicine, University of Belgrade, P.O. Box 38, 11000 Belgrade, Serbia; Clinical Pharmacology Unit, University Children's Hospital, 11000 Belgrade, Serbia
| |
Collapse
|
32
|
Griesbach GS, Masel BE, Helvie RE, Ashley MJ. The Impact of Traumatic Brain Injury on Later Life: Effects on Normal Aging and Neurodegenerative Diseases. J Neurotrauma 2017; 35:17-24. [PMID: 28920532 DOI: 10.1089/neu.2017.5103] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The acute and chronic effects of traumatic brain injury (TBI) have been widely described; however, there is limited knowledge on how a TBI sustained during early adulthood or mid-adulthood will influence aging. Epidemiological studies have explored whether TBI poses a risk for dementia and other neurodegenerative diseases associated with aging. We will discuss the influence of TBI and resulting medical comorbidities such as endocrine, sleep, and inflammatory disturbances on age-related gray and white matter changes and cognitive decline. Post mortem studies examining amyloid, tau, and other proteins will be discussed within the context of neurodegenerative diseases and chronic traumatic encephalopathy. The data support the suggestion that pathological changes triggered by an earlier TBI will have an influence on normal aging processes and will interact with neurodegenerative disease processes rather than the development of a specific disease, such as Alzheimer's or Parkinson's. Chronic neurophysiologic change after TBI may have detrimental effects on neurodegenerative disease.
Collapse
Affiliation(s)
- Grace S Griesbach
- 1 Centre for Neuro Skills Clinical Research and Education Foundation , Bakersfield, California.,2 Department of Neurosurgery, David Geffen School of Medicine at the University of California , Los Angeles, California
| | - Brent E Masel
- 1 Centre for Neuro Skills Clinical Research and Education Foundation , Bakersfield, California.,3 University of Texas Medical Branch , Galveston, Texas
| | - Richard E Helvie
- 1 Centre for Neuro Skills Clinical Research and Education Foundation , Bakersfield, California
| | - Mark J Ashley
- 1 Centre for Neuro Skills Clinical Research and Education Foundation , Bakersfield, California
| |
Collapse
|
33
|
Knauer B, Majka P, Watkins KJ, Taylor AWR, Malamanova D, Paul B, Yu HH, Bush AI, Hare DJ, Reser DH. Whole-brain metallomic analysis of the common marmoset (Callithrix jacchus). Metallomics 2017; 9:411-423. [PMID: 28246661 DOI: 10.1039/c7mt00012j] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Despite the importance of transition metals for normal brain function, relatively little is known about the distribution of these elemental species across the different tissue compartments of the primate brain. In this study, we employed laser ablation-inductively coupled plasma-mass spectrometry on PFA-fixed brain sections obtained from two adult common marmosets. Concurrent cytoarchitectonic, myeloarchitectonic, and chemoarchitectonic measurements allowed for identification of the major neocortical, archaecortical, and subcortical divisions of the brain, and precise localisation of iron, manganese, and zinc concentrations within each division. Major findings across tissue compartments included: (1) differentiation of white matter tracts from grey matter based on manganese and zinc distribution; (2) high iron concentrations in the basal ganglia, cortex, and substantia nigra; (3) co-localization of high concentrations of iron and manganese in the primary sensory areas of the cerebral cortex; and (4) high manganese in the hippocampus. The marmoset has become a model species of choice for connectomic, aging, and transgenic studies in primates, and the application of metallomics to these disciplines has the potential to yield high translational and basic science value.
Collapse
Affiliation(s)
- B Knauer
- Department of Physiology, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia and Research School, Ruhr University Bochum, Bochum, Germany
| | - P Majka
- Laboratory of Neuroinformatics, Department of Neurophysiology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - K J Watkins
- Department of Physiology, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - A W R Taylor
- Department of Physiology, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - D Malamanova
- Department of Physiology, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - B Paul
- School of Earth Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia and The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Hsin-Hao Yu
- Department of Physiology, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - A I Bush
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - D J Hare
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, 3010, Victoria, Australia and Elemental Bio-imaging Facility, University of Technology Sydney, Broadway, NSW, Australia
| | - D H Reser
- Department of Physiology, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia and Graduate Entry Medical Program, School of Rural Health, Monash University, Churchill, Victoria 3842, Australia
| |
Collapse
|
34
|
Liu X, Yang J, Lu C, Jiang S, Nie X, Han J, Yin L, Jiang J. Downregulation of Mfn2 participates in manganese-induced neuronal apoptosis in rat striatum and PC12 cells. Neurochem Int 2017; 108:40-51. [PMID: 28232070 DOI: 10.1016/j.neuint.2017.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 12/24/2022]
Abstract
Manganese (Mn) is a widely distributed trace element that is essential for normal brain function and development. However, chronic exposure to excessive Mn has been known to lead to neuronal loss and manganism, a disease with debilitating motor and cognitive deficits, whose clinical syndrome resembling idiopathic Parkinson's disease (IPD). However, the precise molecular mechanism underlying Mn neurotoxicity remains largely unclear. Accumulating evidence indicates that abnormal mitochondrial functionality is an early and causal event in Mn-induced neurodegeneration and apoptosis. Here, we investigated whether Mitofusin 2 (Mfn2), a highly conserved dynamin-related protein (DRP), played a role in the regulation of Mn-induced neuronal apoptosis. We revealed that Mfn2 was significantly dysregulated in rat striatum and PC12 neuronal-like cells following Mn exposure. Western blot analysis revealed that the expression of Mfn2 was remarkably decreased following different concentrations of Mn exposure. Immunohistochemistry analysis confirmed a remarkable downregulation of Mfn2 in rat striatum after Mn exposure. Immunofluorescent staining showed that Mfn2 was expressed predominantly in neurons, and neuronal loss of Mfn2 was associated with the expression of active caspase-3 following Mn exposure. Importantly, overexpression of Mfn2 apparently attenuated Mn-induced neuronal apoptosis. Notably, treatment with caspase-3 inhibitor Ac-DEVD-CH could not rescue Mn-induced downregulation of Mfn2, suggesting that Mn-induced mfn2 occurs prior to neuronal apoptosis. Taken together, these results indicated that down-regulated expression of Mfn2 might contribute to the pathological processes underlying Mn neurotoxicity.
Collapse
Affiliation(s)
- Xinhang Liu
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Jianbin Yang
- Department of Public Health, The Second People's Hospital of Nantong, Nantong, Jiangsu Province, People's Republic of China
| | - Chunhua Lu
- Department of Occupational Health and Occupational Diseases, Nantong Center for Disease Control and Prevention, Nantong, Jiangsu Province, People's Republic of China
| | - Shengyang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Xiaoke Nie
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Jingling Han
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Lifeng Yin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| | - Junkang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, Jiangsu Province, People's Republic of China.
| |
Collapse
|
35
|
Bichell TJV, Wegrzynowicz M, Tipps KG, Bradley EM, Uhouse MA, Bryan M, Horning K, Fisher N, Dudek K, Halbesma T, Umashanker P, Stubbs AD, Holt HK, Kwakye GF, Tidball AM, Colbran RJ, Aschner M, Neely MD, Di Pardo A, Maglione V, Osmand A, Bowman AB. Reduced bioavailable manganese causes striatal urea cycle pathology in Huntington's disease mouse model. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1596-1604. [PMID: 28213125 DOI: 10.1016/j.bbadis.2017.02.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 01/23/2017] [Accepted: 02/13/2017] [Indexed: 01/12/2023]
Abstract
Huntington's disease (HD) is caused by a mutation in the huntingtin gene (HTT), resulting in profound striatal neurodegeneration through an unknown mechanism. Perturbations in the urea cycle have been reported in HD models and in HD patient blood and brain. In neurons, arginase is a central urea cycle enzyme, and the metal manganese (Mn) is an essential cofactor. Deficient biological responses to Mn, and reduced Mn accumulation have been observed in HD striatal mouse and cell models. Here we report in vivo and ex vivo evidence of a urea cycle metabolic phenotype in a prodromal HD mouse model. Further, either in vivo or in vitro Mn supplementation reverses the urea-cycle pathology by restoring arginase activity. We show that Arginase 2 (ARG2) is the arginase enzyme present in these mouse brain models, with ARG2 protein levels directly increased by Mn exposure. ARG2 protein is not reduced in the prodromal stage, though enzyme activity is reduced, indicating that altered Mn bioavailability as a cofactor leads to the deficient enzymatic activity. These data support a hypothesis that mutant HTT leads to a selective deficiency of neuronal Mn at an early disease stage, contributing to HD striatal urea-cycle pathophysiology through an effect on arginase activity.
Collapse
Affiliation(s)
- Terry Jo V Bichell
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA; Vanderbilt Brain Institute, VU
| | - Michal Wegrzynowicz
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - K Grace Tipps
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Emma M Bradley
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Michael A Uhouse
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Miles Bryan
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA; Vanderbilt Brain Institute, VU
| | - Kyle Horning
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA; Vanderbilt Brain Institute, VU
| | - Nicole Fisher
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Karrie Dudek
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Timothy Halbesma
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Preethi Umashanker
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Andrew D Stubbs
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Hunter K Holt
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Gunnar F Kwakye
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | - Andrew M Tidball
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA
| | | | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - M Diana Neely
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA; Vanderbilt Brain Institute, VU
| | - Alba Di Pardo
- Centre for Neurogenetics and Rare Diseases, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Vittorio Maglione
- Centre for Neurogenetics and Rare Diseases, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Alexander Osmand
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| | - Aaron B Bowman
- Department of Pediatrics, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Neurology, Vanderbilt University (VU), Vanderbilt University Medical Center (VUMC), Nashville, TN, USA; Department of Biochemistry, Vanderbilt University (VU), Nashville, TN, USA; Vanderbilt Brain Institute, VU.
| |
Collapse
|
36
|
Abstract
Copper is an essential trace metal that is required for several important biological processes, however, an excess of copper can be toxic to cells. Therefore, systemic and cellular copper homeostasis is tightly regulated, but dysregulation of copper homeostasis may occur in disease states, resulting either in copper deficiency or copper overload and toxicity. This chapter will give an overview on the biological roles of copper and of the mechanisms involved in copper uptake, storage, and distribution. In addition, we will describe potential mechanisms of the cellular toxicity of copper and copper oxide nanoparticles. Finally, we will summarize the current knowledge on the connection of copper toxicity with neurodegenerative diseases.
Collapse
Affiliation(s)
- Felix Bulcke
- Center for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany
- Center for Environmental Research and Sustainable Technology, Bremen, Germany
| | - Ralf Dringen
- Center for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany
- Center for Environmental Research and Sustainable Technology, Bremen, Germany
| | - Ivo Florin Scheiber
- Center for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany.
- Center for Environmental Research and Sustainable Technology, Bremen, Germany.
| |
Collapse
|
37
|
|
38
|
Dusek P, Bahn E, Litwin T, Jabłonka-Salach K, Łuciuk A, Huelnhagen T, Madai VI, Dieringer MA, Bulska E, Knauth M, Niendorf T, Sobesky J, Paul F, Schneider SA, Czlonkowska A, Brück W, Wegner C, Wuerfel J. Brain iron accumulation in Wilson disease: apost mortem7 Tesla MRI - histopathological study. Neuropathol Appl Neurobiol 2016; 43:514-532. [DOI: 10.1111/nan.12341] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/18/2016] [Accepted: 08/20/2016] [Indexed: 12/12/2022]
Affiliation(s)
- P. Dusek
- Institute of Neuroradiology; University Medical Center Göttingen; Göttingen Germany
- Department of Neurology and Center of Clinical Neuroscience; 1 Faculty of Medicine and General University Hospital in Prague; Charles University in Prague; Praha Czech Republic
| | - E. Bahn
- Institute of Neuropathology; University Medical Center Göttingen; Göttingen Germany
| | - T. Litwin
- 2 Department of Neurology; Institute Psychiatry and Neurology; Warsaw Poland
| | - K. Jabłonka-Salach
- Faculty of Chemistry; Biological and Chemical Research Centre; University of Warsaw; Warsaw Poland
| | - A. Łuciuk
- Faculty of Chemistry; Biological and Chemical Research Centre; University of Warsaw; Warsaw Poland
| | - T. Huelnhagen
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
| | - V. I. Madai
- Department of Neurology and Center for Stroke Research Berlin (CSB); Charité-Universitätsmedizin; Berlin Germany
| | - M. A. Dieringer
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
- Experimental and Clinical Research Center (ECRC); Charité-Universitätsmedizin and Max Delbrück Center for Molecular Medicine (MDC); Berlin Germany
| | - E. Bulska
- Faculty of Chemistry; Biological and Chemical Research Centre; University of Warsaw; Warsaw Poland
| | - M. Knauth
- Institute of Neuroradiology; University Medical Center Göttingen; Göttingen Germany
| | - T. Niendorf
- Berlin Ultrahigh Field Facility (B.U.F.F.); Max-Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
- Experimental and Clinical Research Center (ECRC); Charité-Universitätsmedizin and Max Delbrück Center for Molecular Medicine (MDC); Berlin Germany
| | - J. Sobesky
- Department of Neurology and Center for Stroke Research Berlin (CSB); Charité-Universitätsmedizin; Berlin Germany
- Experimental and Clinical Research Center (ECRC); Charité-Universitätsmedizin and Max Delbrück Center for Molecular Medicine (MDC); Berlin Germany
| | - F. Paul
- Experimental and Clinical Research Center (ECRC); Charité-Universitätsmedizin and Max Delbrück Center for Molecular Medicine (MDC); Berlin Germany
- NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center; Department of Neurology; Charité-Universitätsmedizin; Berlin Germany
| | - S. A. Schneider
- Neurology Department; University of Kiel; Kiel Germany
- Department of Neurology; Ludwig-Maximilians-University; Munich Germany
| | - A. Czlonkowska
- 2 Department of Neurology; Institute Psychiatry and Neurology; Warsaw Poland
- Department of Experimental and Clinical Pharmacology; Medical University; Warsaw Poland
| | - W. Brück
- Institute of Neuropathology; University Medical Center Göttingen; Göttingen Germany
| | - C. Wegner
- Institute of Neuropathology; University Medical Center Göttingen; Göttingen Germany
| | - J. Wuerfel
- Institute of Neuroradiology; University Medical Center Göttingen; Göttingen Germany
- NeuroCure Clinical Research Center and Clinical and Experimental Multiple Sclerosis Research Center; Department of Neurology; Charité-Universitätsmedizin; Berlin Germany
- Medical Imaging Analysis Center AG; Basel Switzerland
| |
Collapse
|
39
|
Langford-Smith A, Tilakaratna V, Lythgoe PR, Clark SJ, Bishop PN, Day AJ. Age and Smoking Related Changes in Metal Ion Levels in Human Lens: Implications for Cataract Formation. PLoS One 2016; 11:e0147576. [PMID: 26794210 PMCID: PMC4721641 DOI: 10.1371/journal.pone.0147576] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022] Open
Abstract
Age-related cataract formation is the primary cause of blindness worldwide and although treatable by surgical removal of the lens the majority of sufferers have neither the finances nor access to the medical facilities required. Therefore, a better understanding of the pathogenesis of cataract may identify new therapeutic targets to prevent or slow its progression. Cataract incidence is strongly correlated with age and cigarette smoking, factors that are often associated with accumulation of metal ions in other tissues. Therefore this study evaluated the age-related changes in 14 metal ions in 32 post mortem human lenses without known cataract from donors of 11 to 82 years of age by inductively coupled plasma mass spectrometry; smoking-related changes in 10 smokers verses 14 non-smokers were also analysed. A significant age-related increase in selenium and decrease in copper ions was observed for the first time in the lens tissue, where cadmium ion levels were also increased as has been seen previously. Aluminium and vanadium ions were found to be increased in smokers compared to non-smokers (an analysis that has only been carried out before in lenses with cataract). These changes in metal ions, i.e. that occur as a consequence of normal ageing and of smoking, could contribute to cataract formation via induction of oxidative stress pathways, modulation of extracellular matrix structure/function and cellular toxicity. Thus, this study has identified novel changes in metal ions in human lens that could potentially drive the pathology of cataract formation.
Collapse
Affiliation(s)
- Alex Langford-Smith
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Viranga Tilakaratna
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Paul R Lythgoe
- School of Earth, Atmospheric and Environmental Sciences and Williamson Research Centre for Molecular Environmental Science, University of Manchester, Manchester, United Kingdom
| | - Simon J Clark
- Centre for Ophthalmology and Vision Sciences, Institute of Human Development, University of Manchester, Manchester, United Kingdom.,Centre for Advanced Discovery and Experimental Therapeutics, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Paul N Bishop
- Centre for Ophthalmology and Vision Sciences, Institute of Human Development, University of Manchester, Manchester, United Kingdom.,Centre for Advanced Discovery and Experimental Therapeutics, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom.,Manchester Royal Eye Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
40
|
Xu G, Su R, Li B, Lv J, Sun W, Hu B, Li X, Gu J, Yu X. Trace Element Concentrations in Human Tissues of Death Cases Associated With Secondary Infection and MOF After Severe Trauma. Biol Trace Elem Res 2015; 168:335-9. [PMID: 26043914 DOI: 10.1007/s12011-015-0378-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/20/2015] [Indexed: 02/05/2023]
Abstract
Proper trace element level is crucial for the organs in maintaining normal physiological functions. Multiple organ failure (MOF) might be added to critically ill patients due to a lack of trace elements. Alterations of trace element levels in brain, heart, liver, and kidney after severe trauma, however, have been little studied so far. In this study, tissue samples of the frontal cortex of the brain, interventricular septum of the heart, right lobe of the liver, and upper pole of the kidney were obtained from forensic autopsies, of which 120 cases died during the 5th to 15th day of hospitalization, whereas the trauma death group and 43 cases immediately died due to severe craniocerebral trauma as the control group. Copper (Cu), iron (Fe), zinc (Zn), and selenium (Se) were quantified by inductively coupled plasma atomic emission spectrophotometry (ICP-AES). Cu, Fe, Zn, and Se concentrations in the brain, heart, liver, and kidney in the trauma group decreased dramatically (p<0.05) compared to the control group. The incidence of secondary infection and multiple organ failure (MOF) in the trauma death group were 78.33 and 29.17%, respectively. The concentrations of all elements exhibited a significant correlation with secondary infection and MOF (p<0.01). Our data suggest that low concentrations of Cu, Fe, Zn, and Se in pivotal organs may contribute to the incidence of secondary infection and MOF after severe trauma, which to some extent results in death.
Collapse
Affiliation(s)
- Guangtao Xu
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China.
- Department of Forensic and Pathology, Jiaxing University Medical College, 314001, Jiaxing, Zhejiang Province, People's Republic of China.
- Department of Forensic Pathology, Jiaxing Zhiyuan Forensic Science Institute, 314001, Jiaxing, Zhejiang Province, People's Republic of China.
| | - Ruibing Su
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China
| | - Bo Li
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China
| | - Junyao Lv
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China
| | - Weiqi Sun
- Department of Preventive Medicine, School of Public Health, Beihua University, 132013, Jilin, Jilin Province, People's Republic of China
| | - Bo Hu
- Department of Pathology, Jiaxing Hospital of Traditional Chinese Medicine, 314001, Jiaxing, Zhejiang Province, People's Republic of China
| | - Xianxian Li
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China
| | - Jiang Gu
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China
| | - Xiaojun Yu
- National Key Disciplines, Department of Forensic and Pathology, Shantou University Medical College, 515041, Shantou, Guangdong Province, People's Republic of China.
| |
Collapse
|
41
|
Domenech M, Ruiz S, Moscoso M, García E. In vitro biofilm development of Streptococcus pneumoniae and formation of choline-binding protein-DNA complexes. ENVIRONMENTAL MICROBIOLOGY REPORTS 2015; 7:715-727. [PMID: 25950767 DOI: 10.1111/1758-2229.12295] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/08/2015] [Indexed: 06/04/2023]
Abstract
Extracellular deoxyribonucleic acid (eDNA) is an essential component of bacterial biofilm matrices, and is required in their formation and maintenance. Extracellular DNA binds to exopolysaccharides or extracellular proteins, affording biofilms greater structural integrity. Recently, we reported evidence of intercellular eDNA-LytC complexes in pneumococcal biofilms. The LytC lysozyme is a member of the choline-binding family of proteins (CBPs) located on the pneumococcal surface. The present work shows that other CBPs, i.e. LytA, LytB, Pce, PspC and CbpF, which have a pI between 5 and 6, can bind DNA in vitro. This process requires the presence of divalent cations other than Mg(2+). This DNA binding capacity of CBPs appears to be independent of their enzymatic activity and, at least in the case of LytA, does not require the choline-binding domain characteristic of CBPs. Positively charged, surface-exposed, 25 amino acid-long peptides derived from the catalytic domain of LytB, were also found capable of DNA binding through electrostatic interactions. Confocal laser scanning microcopy revealed the existence of cell-associated LytB-eDNA complexes in Streptococcus pneumoniae biofilms. These and other findings suggest that these surface-located proteins of S. pneumoniae could play roles of varying importance in the colonization and/or invasion of human host where different environmental conditions exist.
Collapse
Affiliation(s)
- Mirian Domenech
- Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, 28040, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Susana Ruiz
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Miriam Moscoso
- Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, 28040, Spain
| | - Ernesto García
- Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, 28040, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
42
|
Fu S, Jiang W, Zheng W. Age-dependent increase of brain copper levels and expressions of copper regulatory proteins in the subventricular zone and choroid plexus. Front Mol Neurosci 2015; 8:22. [PMID: 26106293 PMCID: PMC4458609 DOI: 10.3389/fnmol.2015.00022] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/25/2015] [Indexed: 12/14/2022] Open
Abstract
Our recent data suggest a high accumulation of copper (Cu) in the subventricular zone (SVZ) along the wall of brain ventricles. Anatomically, SVZ is in direct contact with cerebrospinal fluid (CSF), which is secreted by a neighboring tissue choroid plexus (CP). Changes in Cu regulatory gene expressions in the SVZ and CP as the function of aging may determine Cu levels in the CSF and SVZ. This study was designed to investigate the associations between age, Cu levels, and Cu regulatory genes in SVZ and plexus. The SVZ and CP were dissected from brains of 3-week, 10-week, or 9-month old male rats. Analyses by atomic absorption spectroscopy revealed that the SVZ of adult and old animals contained the highest Cu level compared with other tested brain regions. Significantly positive correlations between age and Cu levels in SVZ and plexus were observed; the SVZ Cu level of old animals was 7.5- and 5.8-fold higher than those of young and adult rats (p < 0.01), respectively. Quantitation by qPCR of the transcriptional expressions of Cu regulatory proteins showed that the SVZ expressed the highest level of Cu storage protein metallothioneins (MTs), while the CP expressed the high level of Cu transporter protein Ctr1. Noticeably, Cu levels in the SVZ were positively associated with type B slow proliferating cell marker Gfap (p < 0.05), but inversely associated with type A proliferating neuroblast marker Dcx (p < 0.05) and type C transit amplifying progenitor marker Nestin (p < 0.01). Dmt1 had significant positive correlations with age and Cu levels in the plexus (p < 0.01). These findings suggest that Cu levels in all tested brain regions are increased as the function of age. The SVZ shows a different expression pattern of Cu-regulatory genes from the CP. The age-related increase of MTs and decrease of Ctr1 may contribute to the high Cu level in this neurogenesis active brain region.
Collapse
Affiliation(s)
- Sherleen Fu
- School of Health Sciences, Purdue University West Lafayette, IN, USA
| | - Wendy Jiang
- School of Health Sciences, Purdue University West Lafayette, IN, USA
| | - Wei Zheng
- School of Health Sciences, Purdue University West Lafayette, IN, USA
| |
Collapse
|
43
|
Abstract
The understanding of manganese (Mn) biology, in particular its cellular regulation and role in neurological disease, is an area of expanding interest. Mn is an essential micronutrient that is required for the activity of a diverse set of enzymatic proteins (e.g., arginase and glutamine synthase). Although necessary for life, Mn is toxic in excess. Thus, maintaining appropriate levels of intracellular Mn is critical. Unlike other essential metals, cell-level homeostatic mechanisms of Mn have not been identified. In this review, we discuss common forms of Mn exposure, absorption, and transport via regulated uptake/exchange at the gut and blood-brain barrier and via biliary excretion. We present the current understanding of cellular uptake and efflux as well as subcellular storage and transport of Mn. In addition, we highlight the Mn-dependent and Mn-responsive pathways implicated in the growing evidence of its role in Parkinson's disease and Huntington's disease. We conclude with suggestions for future focuses of Mn health-related research.
Collapse
Affiliation(s)
- Kyle J Horning
- Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232; , ,
| | | | | | | | | |
Collapse
|
44
|
Soria M, González-Haro C, Ansón M, López-Colón JL, Escanero JF. Plasma levels of trace elements and exercise induced stress hormones in well-trained athletes. J Trace Elem Med Biol 2015; 31:113-9. [PMID: 26004901 DOI: 10.1016/j.jtemb.2015.04.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/27/2015] [Accepted: 04/13/2015] [Indexed: 01/04/2023]
Abstract
This study analyzed the variation and relationship of several trace elements, metabolic substrates and stress hormones activated by exercise during incremental exercise. Seventeen well-trained endurance athletes performed a cycle ergometer test: after a warm-up of 10 min at 2.0 W kg(-1), the workload was increased by 0.5 W kg(-1) every 10 min until exhaustion. Prior diet, activity patterns, and levels of exercise training were controlled, and tests timed to minimize variations due to the circadian rhythm. Oxygen uptake, blood lactate concentration, plasma ions (Zn, Se, Mn and Co), serum glucose, non-esterified fatty acids (NEFAs) and several hormones were measured at rest, at the end of each stage and 3, 5 and 7 min post-exercise. Urine specific gravity was measured before and after the test, and participants drank water ad libitum. Significant differences were found in plasma Zn and Se levels as a function of exercise intensity. Zn was significantly correlated with epinephrine, norepinephrine and cortisol (r = 0.884, P < 0.01; r = 0.871, P < 0.01; and r = 0.808, P = 0.05); and Se showed significant positive correlations whit epinephrine and cortisol (r = 0.743, P < 0.05; and r = 0.776, P < 0.05). Neither Zn nor Se levels were associated with insulin or glucagon, and neither Mn nor Co levels were associated with any of the hormones or substrate metabolites studied. Further, while Zn levels were found to be associated only with lactate, plasma Se was significantly correlated with lactate and glucose (respectively for Zn: r = 0.891, P < 0.01; and for Se: r = 0.743, P < 0.05; r = 0.831, P < 0.05). In conclusion, our data suggest that there is a positive correlation between the increases in plasma Zn or Se and stress hormones variations induced by exercise along different submaximal intensities in well-hydrated well-trained endurance athletes.
Collapse
Affiliation(s)
- Marisol Soria
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Zaragoza, C/Domingo Miral, s/n, 50009 Zaragoza, Spain.
| | - Carlos González-Haro
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Zaragoza, C/Domingo Miral, s/n, 50009 Zaragoza, Spain.
| | - Miguel Ansón
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Zaragoza, C/Domingo Miral, s/n, 50009 Zaragoza, Spain
| | - José L López-Colón
- Toxicology Service, Hospital Central de la Defensa, Glorieta del Ejército, s/n, 28047 Madrid, Spain.
| | - Jesús F Escanero
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Zaragoza, C/Domingo Miral, s/n, 50009 Zaragoza, Spain.
| |
Collapse
|