1
|
Kron NS, Fieber LA, Baker L, Campbell C, Schmale MC. Host response to Aplysia Abyssovirus 1 in nervous system and gill. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105211. [PMID: 38885747 PMCID: PMC11378725 DOI: 10.1016/j.dci.2024.105211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
The California sea hare (Aplysia californica) is a model for age associated cognitive decline. Recent researched identified a novel nidovirus, Aplysia Abyssovirus 1, with broad tropism enriched in the Aplysia nervous system. This virus is ubiquitous in wild and maricultured, young and old animals without obvious pathology. Here we re-evaluated gene expression data from several previous studies to investigate differential expression in the nervous system and gill in response to virus and aging as well as the mutational spectrum observed in the viral sequences obtained from these datasets. Viral load and age were highly correlated, indicating persistent infection. Upregulated genes in response to virus were enriched for immune genes and signatures of ER and proteostatic stress, while downregulated genes were enriched for mitochondrial metabolism. Differential expression with respect to age suggested increased iron accumulation and decreased glycolysis, fatty acid metabolism, and proteasome function. Interaction of gene expression trends associated with viral infection and aging suggest that viral infection likely plays a role in aging in the Aplysia nervous system. Mutation analysis of viral RNA identified signatures suggesting ADAR and AID/APOBEC like deaminase act as part of Aplysia anti-viral defense.
Collapse
Affiliation(s)
- Nicholas S Kron
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA.
| | - Lynne A Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| | - Lydia Baker
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| | | | - Michael C Schmale
- Department of Marine Biology and Ecology, Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami, 4600 Rickenbacker Cswy, Miami, FL, 33149, USA
| |
Collapse
|
2
|
Ashraf AA, Aljuhani M, Hubens CJ, Jeandriens J, Parkes HG, Geraki K, Mahmood A, Herlihy AH, So PW. Inflammation subsequent to mild iron excess differentially alters regional brain iron metabolism, oxidation and neuroinflammation status in mice. Front Aging Neurosci 2024; 16:1393351. [PMID: 38836051 PMCID: PMC11148467 DOI: 10.3389/fnagi.2024.1393351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/26/2024] [Indexed: 06/06/2024] Open
Abstract
Iron dyshomeostasis and neuroinflammation, characteristic features of the aged brain, and exacerbated in neurodegenerative disease, may induce oxidative stress-mediated neurodegeneration. In this study, the effects of potential priming with mild systemic iron injections on subsequent lipopolysaccharide (LPS)-induced inflammation in adult C57Bl/6J mice were examined. After cognitive testing, regional brain tissues were dissected for iron (metal) measurements by total reflection X-ray fluorescence and synchrotron radiation X-Ray fluorescence-based elemental mapping; and iron regulatory, ferroptosis-related, and glia-specific protein analysis, and lipid peroxidation by western blotting. Microglial morphology and astrogliosis were assessed by immunohistochemistry. Iron only treatment enhanced cognitive performance on the novel object location task compared with iron priming and subsequent LPS-induced inflammation. LPS-induced inflammation, with or without iron treatment, attenuated hippocampal heme oxygenase-1 and augmented 4-hydroxynonenal levels. Conversely, in the cortex, elevated ferritin light chain and xCT (light chain of System Xc-) were observed in response to LPS-induced inflammation, without and with iron-priming. Increased microglial branch/process lengths and astrocyte immunoreactivity were also increased by combined iron and LPS in both the hippocampus and cortex. Here, we demonstrate iron priming and subsequent LPS-induced inflammation led to iron dyshomeostasis, compromised antioxidant function, increased lipid peroxidation and altered neuroinflammatory state in a brain region-dependent manner.
Collapse
Affiliation(s)
- Azhaar Ahmad Ashraf
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Manal Aljuhani
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Chantal J Hubens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jérôme Jeandriens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
- Department of Human Biology and Toxicology, Faculty of Medicine, University of Mons, Mons, Belgium
| | - Harold G Parkes
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Kalotina Geraki
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom
| | - Ayesha Mahmood
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | | | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Li S, Huang P, Lai F, Zhang T, Guan J, Wan H, He Y. Mechanisms of Ferritinophagy and Ferroptosis in Diseases. Mol Neurobiol 2024; 61:1605-1626. [PMID: 37736794 DOI: 10.1007/s12035-023-03640-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
The discovery of the role of autophagy, particularly the selective form like ferritinophagy, in promoting cells to undergo ferroptosis has inspired us to investigate functional connections between diseases and cell death. Ferroptosis is a novel model of procedural cell death characterized by the accumulation of iron-dependent reactive oxygen species (ROS), mitochondrial dysfunction, and neuroinflammatory response. Based on ferroptosis, the study of ferritinophagy is particularly important. In recent years, extensive research has elucidated the role of ferroptosis and ferritinophagy in neurological diseases and anemia, suggesting their potential as therapeutic targets. Besides, the global emergence and rapid transmission of COVID-19, which is caused by SARS-CoV-2, represents a considerable risk to public health worldwide. The potential involvement of ferroptosis in the pathophysiology of brain injury associated with COVID-19 is still unclear. This review summarizes the pathophysiological changes of ferroptosis and ferritinophagy in neurological diseases, anemia, and COVID-19, and hypothesizes that ferritinophagy may be a potential mechanism of ferroptosis. Advancements in these fields will enhance our comprehension of methods to prevent and address neurological disorders, anemia, and COVID-19.
Collapse
Affiliation(s)
- Siqi Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ping Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Feifan Lai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ting Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiaqi Guan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
4
|
Apostolopoulou EP, Raikos N, Vlemmas I, Michaelidis E, Brellou GD. Metallothionein I/II Expression and Metal Ion Levels in Correlation with Amyloid Beta Deposits in the Aged Feline Brain. Brain Sci 2023; 13:1115. [PMID: 37509045 PMCID: PMC10377600 DOI: 10.3390/brainsci13071115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Brain aging has been correlated with high metallothionein I-II (MT-I/II) expression, iron and zinc dyshomeostasis, and Aβ deposition in humans and experimental animals. In the present study, iron and zinc accumulation, the expression of MT-I/II and Aβ42, and their potential association with aging in the feline brain were assessed. Tissue sections from the temporal and frontal grey (GM) and white (WM) matter, hippocampus, thalamus, striatum, cerebellum, and dentate nucleus were examined histochemically for the presence of age-related histopathological lesions and iron deposits and distribution. We found, using a modified Perl's/DAB method, two types of iron plaques that showed age-dependent accumulation in the temporal GM and WM and the thalamus, along with the age-dependent increment in cerebellar-myelin-associated iron. We also demonstrated an age-dependent increase in MT-I/II immunoreactivity in the feline brain. In cats over 7 years old, Aβ immunoreactivity was detected in vessel walls and neuronal somata; extracellular Aβ deposits were also evident. Interestingly, Aβ-positive astrocytes were also observed in certain cases. ICP-MS analysis of brain content regarding iron and zinc concentrations showed no statistically significant association with age, but a mild increase in iron with age was noticed, while zinc levels were found to be higher in the Mature and Senior groups. Our findings reinforce the suggestion that cats could serve as a dependable natural animal model for brain aging and neurodegeneration; thus, they should be further investigated on the basis of metal ion concentration changes and their effects on aging.
Collapse
Affiliation(s)
- Emmanouela P Apostolopoulou
- Department of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece
| | - Nikolaos Raikos
- Department of Forensic Medicine & Toxicology, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Ioannis Vlemmas
- Department of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece
| | - Efstratios Michaelidis
- Laboratories of the 3rd Army Veterinary Hospital, Chemical Department, 57001 Thessaloniki, Greece
| | - Georgia D Brellou
- Department of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece
| |
Collapse
|
5
|
Xie F, Mao T, Tang J, Zhao L, Guo J, Lin H, Wang D, Zhou G. Evaluation of iron deposition in the motor CSTC loop of a Chinese family with paroxysmal kinesigenic dyskinesia using quantitative susceptibility mapping. Front Neurol 2023; 14:1164600. [PMID: 37483438 PMCID: PMC10358764 DOI: 10.3389/fneur.2023.1164600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Previous studies have revealed structural, functional, and metabolic changes in brain regions inside the cortico-striatal-thalamo-cortical (CSTC) loop in patients with paroxysmal kinesigenic dyskinesia (PKD), whereas no quantitative susceptibility mapping (QSM)-related studies have explored brain iron deposition in these areas. Methods A total of eight familial PKD patients and 10 of their healthy family members (normal controls) were recruited and underwent QSM on a 3T magnetic resonance imaging system. Magnetic susceptibility maps were reconstructed using a multi-scale dipole inversion algorithm. Thereafter, we specifically analyzed changes in local mean susceptibility values in cortical regions and subcortical nuclei inside the motor CSTC loop. Results Compared with normal controls, PKD patients had altered brain iron levels. In the cortical gray matter area involved with the motor CSTC loop, susceptibility values were generally elevated, especially in the bilateral M1 and PMv regions. In the subcortical nuclei regions involved with the motor CSTC loop, susceptibility values were generally lower, especially in the bilateral substantia nigra regions. Conclusion Our results provide new evidence for the neuropathogenesis of PKD and suggest that an imbalance in brain iron levels may play a role in PKD.
Collapse
Affiliation(s)
- Fangfang Xie
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingyi Tang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Linmei Zhao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiuqing Guo
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Huashan Lin
- Department of Pharmaceutical Diagnosis, GE Healthcare, Changsha, China
| | - Dongcui Wang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Gaofeng Zhou
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
6
|
Trigo D, Vitória JJ, da Cruz e Silva OAB. Novel therapeutic strategies targeting mitochondria as a gateway in neurodegeneration. Neural Regen Res 2022; 18:991-995. [PMID: 36254979 PMCID: PMC9827793 DOI: 10.4103/1673-5374.355750] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In recent years, multiple disciplines have focused on mitochondrial biology and contributed to understanding its relevance towards adult-onset neurodegenerative disorders. These are complex dynamic organelles that have a variety of functions in ensuring cellular health and homeostasis. The plethora of mitochondrial functionalities confers them an intrinsic susceptibility to internal and external stressors (such as mutation accumulation or environmental toxins), particularly so in long-lived postmitotic cells such as neurons. Thus, it is reasonable to postulate an involvement of mitochondria in aging-associated neurological disorders, notably neurodegenerative pathologies including Alzheimer's disease and Parkinson's disease. On the other hand, biological effects resulting from neurodegeneration can in turn affect mitochondrial health and function, promoting a feedback loop further contributing to the progression of neuronal dysfunction and cellular death. This review examines state-of-the-art knowledge, focus on current research exploring mitochondrial health as a contributing factor to neuroregeneration, and the development of therapeutic approaches aimed at restoring mitochondrial homeostasis in a pathological setting.
Collapse
Affiliation(s)
- Diogo Trigo
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal,Correspondence to: Diogo Trigo, .
| | - José João Vitória
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Odete A. B. da Cruz e Silva
- Neuroscience and Signalling Laboratory, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
7
|
Tian Y, Tian Y, Yuan Z, Zeng Y, Wang S, Fan X, Yang D, Yang M. Iron Metabolism in Aging and Age-Related Diseases. Int J Mol Sci 2022; 23:3612. [PMID: 35408967 PMCID: PMC8998315 DOI: 10.3390/ijms23073612] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Iron is a trace metal element necessary to maintain life and is also involved in a variety of biological processes. Aging refers to the natural life process in which the physiological functions of the various systems, organs, and tissues decline, affected by genetic and environmental factors. Therefore, it is imperative to investigate the relationship between iron metabolism and aging-related diseases, including neurodegenerative diseases. During aging, the accumulation of nonheme iron destroys the stability of the intracellular environment. The destruction of iron homeostasis can induce cell damage by producing hydroxyl free radicals, leading to mitochondrial dysfunction, brain aging, and even organismal aging. In this review, we have briefly summarized the role of the metabolic process of iron in the body, then discussed recent developments of iron metabolism in aging and age-related neurodegenerative diseases, and finally, explored some iron chelators as treatment strategies for those disorders. Understanding the roles of iron metabolism in aging and neurodegenerative diseases will fill the knowledge gap in the field. This review could provide new insights into the research on iron metabolism and age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yao Tian
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
| | - Yuanliangzi Tian
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
| | - Zhixiao Yuan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
| | - Yutian Zeng
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
| | - Shuai Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
| | - Xiaolan Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Deying Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China; (Y.T.); (Y.T.); (Z.Y.); (Y.Z.); (S.W.); (X.F.); (D.Y.)
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
8
|
Lorio S, Sedlacik J, So PW, Parkes HG, Gunny R, Löbel U, Li YF, Ogunbiyi O, Mistry T, Dixon E, Adler S, Cross JH, Baldeweg T, Jacques TS, Shmueli K, Carmichael DW. Quantitative MRI susceptibility mapping reveals cortical signatures of changes in iron, calcium and zinc in malformations of cortical development in children with drug-resistant epilepsy. Neuroimage 2021; 238:118102. [PMID: 34058334 PMCID: PMC8350142 DOI: 10.1016/j.neuroimage.2021.118102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Malformations of cortical development (MCD), including focal cortical dysplasia (FCD), are the most common cause of drug-resistant focal epilepsy in children. Histopathological lesion characterisation demonstrates abnormal cell types and lamination, alterations in myelin (typically co-localised with iron), and sometimes calcification. Quantitative susceptibility mapping (QSM) is an emerging MRI technique that measures tissue magnetic susceptibility (χ) reflecting it's mineral composition. We used QSM to investigate abnormal tissue composition in a group of children with focal epilepsy with comparison to effective transverse relaxation rate (R2*) and Synchrotron radiation X-ray fluorescence (SRXRF) elemental maps. Our primary hypothesis was that reductions in χ would be found in FCD lesions, resulting from alterations in their iron and calcium content. We also evaluated deep grey matter nuclei for changes in χ with age. METHODS QSM and R2* maps were calculated for 40 paediatric patients with suspected MCD (18 histologically confirmed) and 17 age-matched controls. Patients' sub-groups were defined based on concordant electro-clinical or histopathology data. Quantitative investigation of QSM and R2* was performed within lesions, using a surface-based approach with comparison to homologous regions, and within deep brain regions using a voxel-based approach with regional values modelled with age and epilepsy as covariates. Synchrotron radiation X-ray fluorescence (SRXRF) was performed on brain tissue resected from 4 patients to map changes in iron, calcium and zinc and relate them to MRI parameters. RESULTS Compared to fluid-attenuated inversion recovery (FLAIR) or T1-weighted imaging, QSM improved lesion conspicuity in 5% of patients. In patients with well-localised lesions, quantitative profiling demonstrated decreased χ, but not R2*, across cortical depth with respect to the homologous regions. Contra-lateral homologous regions additionally exhibited increased χ at 2-3 mm cortical depth that was absent in lesions. The iron decrease measured by the SRXRF in FCDIIb lesions was in agreement with myelin reduction observed by Luxol Fast Blue histochemical staining. SRXRF analysis in two FCDIIb tissue samples showed increased zinc and calcium in one patient, and decreased iron in the brain region exhibiting low χ and high R2* in both patients. QSM revealed expected age-related changes in the striatum nuclei, substantia nigra, sub-thalamic and red nucleus. CONCLUSION QSM non-invasively revealed cortical/sub-cortical tissue alterations in MCD lesions and in particular that χ changes in FCDIIb lesions were consistent with reduced iron, co-localised with low myelin and increased calcium and zinc content. These findings suggest that measurements of cortical χ could be used to characterise tissue properties non-invasively in epilepsy lesions.
Collapse
Affiliation(s)
- Sara Lorio
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, UK; Wellcome EPSRC Centre for Medical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Jan Sedlacik
- Biomedical Engineering Department, School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK; Center for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Po-Wah So
- Department of Neuroimaging, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Harold G Parkes
- Department of Neuroimaging, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Roxana Gunny
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ulrike Löbel
- Department of Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Yao-Feng Li
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London and Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; Pathology Department, Tri-Service General Hospital and National Defence Medical Centre, Taipei, Taiwan, ROC
| | - Olumide Ogunbiyi
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London and Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Talisa Mistry
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London and Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Emma Dixon
- MRI Group, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Sophie Adler
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - J Helen Cross
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Torsten Baldeweg
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Thomas S Jacques
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London and Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Karin Shmueli
- MRI Group, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - David W Carmichael
- Developmental Neurosciences, Great Ormond Street Institute of Child Health, University College London, London, UK; Wellcome EPSRC Centre for Medical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
9
|
Han K, Jin X, Guo X, Cao G, Tian S, Song Y, Zuo Y, Yu P, Gao G, Chang YZ. Nrf2 knockout altered brain iron deposition and mitigated age-related motor dysfunction in aging mice. Free Radic Biol Med 2021; 162:592-602. [PMID: 33248265 DOI: 10.1016/j.freeradbiomed.2020.11.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/05/2020] [Accepted: 11/17/2020] [Indexed: 01/09/2023]
Abstract
The transcription factor NF-E2-related factor 2 (Nrf2) is a central regulator of cellular antioxidant and detoxification response. The association between Nrf2 activity and iron-related oxidative stress in neurodegenerative diseases has been studied, and Nrf2 was found to transcriptionally regulate the expression of iron transporters and ferroptosis-related factors. However, the role of Nrf2 in age-related motor dysfunction and its link to iron metabolism dysregulation in brain have not been fully elucidated. In this study, with different ages of Nrf2 knockout (KO) and wild type (WT) mice, we investigated the effects of Nrf2 deficiency on brain oxidative stress, iron metabolism and the motor coordination ability of mice. In contrast to the predicted neuroprotective role of Nrf2 in oxidative stress-related diseases, we found that Nrf2 KO remarkably improved the motor coordination of aged mice, which was associated with the reduced ROS level and decreased apoptosis of dopaminergic neurons in substantia nigra (SN) of 18-month-old Nrf2 KO mice. With high-iron and Parkinson's disease (PD) mouse models, we revealed that Nrf2 KO prevented the deposition of brain iron, particularly in SN and striatum, which may subsequently delay motor dysfunction in aged mice. The regulation of Nrf2 KO on brain iron metabolism was likely mediated by decreasing the ferroportin 1 (FPN1) level on brain microvascular endothelial cells, thus hindering the process of iron entry into the brain. Nrf2 may be a potential therapeutic target in age-related motor dysfunction diseases for its role in regulating brain iron homeostasis.
Collapse
Affiliation(s)
- Kang Han
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Xiaofang Jin
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Xin Guo
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China; Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China
| | - Guoli Cao
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Siyu Tian
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Yiming Song
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Yuanyuan Zuo
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Peng Yu
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China.
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei Province, 050024, China.
| |
Collapse
|
10
|
Ijomone OM, Ifenatuoha CW, Aluko OM, Ijomone OK, Aschner M. The aging brain: impact of heavy metal neurotoxicity. Crit Rev Toxicol 2020; 50:801-814. [PMID: 33210961 DOI: 10.1080/10408444.2020.1838441] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aging process is accompanied by critical changes in cellular and molecular functions, which upset the homeostatic balance in the central nervous system. Accumulation of metals renders the brain susceptible to neurotoxic insults by mechanisms such as mitochondrial dysfunction, neuronal calcium-ion dyshomeostasis, buildup of damaged molecules, compromised DNA repair, reduction in neurogenesis, and impaired energy metabolism. These hallmarks have been identified to be responsible for neuronal injuries, resulting in several neurological disorders. Various studies have shown solid associations between metal accumulation, abnormal protein expressions, and pathogenesis of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and Amyotrophic lateral sclerosis. This review highlights metals (such as manganese, zinc, iron, copper, and nickel) for their accumulation, and consequences in the development of neurological disorders, in relation to the aging brain.
Collapse
Affiliation(s)
- Omamuyovwi M Ijomone
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.,Department of Human Anatomy, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Chibuzor W Ifenatuoha
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Oritoke M Aluko
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.,Department of Physiology, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria
| | - Olayemi K Ijomone
- The Neuro-Lab, School of Health and Health Technology, Federal University of Technology, Akure, Nigeria.,Department of Anatomy, University of Medical Sciences, Ondo, Nigeria
| | - Michael Aschner
- Departments of Molecular Pharmacology, Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
11
|
Kron NS, Schmale MC, Fieber LA. Changes in Metabolism and Proteostasis Drive Aging Phenotype in Aplysia californica Sensory Neurons. Front Aging Neurosci 2020; 12:573764. [PMID: 33101008 PMCID: PMC7522570 DOI: 10.3389/fnagi.2020.573764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/12/2020] [Indexed: 12/29/2022] Open
Abstract
Aging is associated with cognitive declines that originate in impairments of function in the neurons that make up the nervous system. The marine mollusk Aplysia californica (Aplysia) is a premier model for the nervous system uniquely suited to investigation of neuronal aging due to uniquely identifiable neurons and molecular techniques available in this model. This study describes the molecular processes associated with aging in two populations of sensory neurons in Aplysia by applying RNA sequencing technology across the aging process (age 6-12 months). Differentially expressed genes clustered into four to five coherent expression patterns across the aging time series in the two neuron populations. Enrichment analysis of functional annotations in these neuron clusters revealed decreased expression of pathways involved in energy metabolism and neuronal signaling, suggesting that metabolic and signaling pathways are intertwined. Furthermore, increased expression of pathways involved in protein processing and translation suggests that proteostatic stress also occurs in aging. Temporal overlap of enrichment for energy metabolism, proteostasis, and neuronal function suggests that cognitive impairments observed in advanced age result from the ramifications of broad declines in energy metabolism.
Collapse
Affiliation(s)
- Nicholas S Kron
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, United States
| | - Michael C Schmale
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, United States
| | - Lynne A Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami, Miami, FL, United States
| |
Collapse
|
12
|
Iron Aggravates the Depressive Phenotype of Stressed Mice by Compromising the Glymphatic System. Neurosci Bull 2020; 36:1542-1546. [PMID: 32578069 DOI: 10.1007/s12264-020-00539-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
|
13
|
Hernández-Gallardo AK, Missirlis F. Cellular iron sensing and regulation: Nuclear IRP1 extends a classic paradigm. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118705. [PMID: 32199885 DOI: 10.1016/j.bbamcr.2020.118705] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/02/2020] [Accepted: 03/16/2020] [Indexed: 01/26/2023]
Abstract
The classic view is that iron regulatory proteins operate at the post-transcriptional level. Iron Regulatory Protein 1 (IRP1) shifts between an apo-form that binds mRNAs and a holo-form that harbors a [4Fe4S] cluster. The latter form is not considered relevant to iron regulation, but rather thought to act as a non-essential cytosolic aconitase. Recent work in Drosophila, however, shows that holo-IRP1 can also translocate to the nucleus, where it appears to downregulate iron metabolism genes, preparing the cell for a decline in iron uptake. The shifting of IRP1 between states requires a functional mitoNEET pathway that includes a glycogen branching enzyme for the repair or disassembly of IRP1's oxidatively damaged [3Fe4S] cluster. The new findings add to the notion that glucose metabolism is modulated by iron metabolism. Furthermore, we propose that ferritin ferroxidase activity participates in the repair of the IRP1 [3Fe4S] cluster leading to the hypothesis that cytosolic ferritin directly contributes to cellular iron sensing.
Collapse
Affiliation(s)
| | - Fanis Missirlis
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav, CDMX, Mexico.
| |
Collapse
|
14
|
Ashraf A, Jeandriens J, Parkes HG, So PW. Iron dyshomeostasis, lipid peroxidation and perturbed expression of cystine/glutamate antiporter in Alzheimer's disease: Evidence of ferroptosis. Redox Biol 2020; 32:101494. [PMID: 32199332 PMCID: PMC7083890 DOI: 10.1016/j.redox.2020.101494] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Iron dyshomeostasis is implicated in Alzheimer’s disease (AD) alongside β-amyloid and tau pathologies. Despite the recent discovery of ferroptosis, an iron-dependent form cell death, hitherto, in vivo evidence of ferroptosis in AD is lacking. The present study uniquely adopts an integrated multi-disciplinary approach, combining protein (Western blot) and elemental analysis (total reflection X-ray fluorescence) with metabolomics (1H nuclear magnetic resonance spectroscopy) to identify iron dyshomeostasis and ferroptosis, and possible novel interactions with metabolic dysfunction in age-matched male cognitively normal (CN) and AD post-mortem brain tissue (n = 7/group). Statistical analysis was used to compute differences between CN and AD, and to examine associations between proteins, elements and/or metabolites. Iron dyshomeostasis with elevated levels of ferritin, in the absence of increased elemental iron, was observed in AD. Moreover, AD was characterised by enhanced expression of the light-chain subunit of the cystine/glutamate transporter (xCT) and lipid peroxidation, reminiscent of ferroptosis, alongside an augmented excitatory glutamate to inhibitory GABA ratio. Protein, element and metabolite associations also greatly differed between CN and AD suggesting widespread metabolic dysregulation in AD. We demonstrate iron dyshomeostasis, upregulated xCT (impaired glutathione metabolism) and lipid peroxidation in AD, suggesting anti-ferroptotic therapies may be efficacious in AD.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Jérôme Jeandriens
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Department of Human Biology and Toxicology, Faculty of Medicine, University of Mons, Place du Parc 20, Mons, Belgium
| | - Harold G Parkes
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
15
|
Ashraf A, Michaelides C, Walker TA, Ekonomou A, Suessmilch M, Sriskanthanathan A, Abraha S, Parkes A, Parkes HG, Geraki K, So PW. Regional Distributions of Iron, Copper and Zinc and Their Relationships With Glia in a Normal Aging Mouse Model. Front Aging Neurosci 2019; 11:351. [PMID: 31920630 PMCID: PMC6930884 DOI: 10.3389/fnagi.2019.00351] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/02/2019] [Indexed: 01/08/2023] Open
Abstract
Microglia and astrocytes can quench metal toxicity to maintain tissue homeostasis, but with age, increasing glial dystrophy alongside metal dyshomeostasis may predispose the aged brain to acquire neurodegenerative diseases. The aim of the present study was to investigate age-related changes in brain metal deposition along with glial distribution in normal C57Bl/6J mice aged 2-, 6-, 19- and 27-months (n = 4/age). Using synchrotron-based X-ray fluorescence elemental mapping, we demonstrated age-related increases in iron, copper, and zinc in the basal ganglia (p < 0.05). Qualitative assessments revealed age-associated increases in iron, particularly in the basal ganglia and zinc in the white matter tracts, while copper showed overt enrichment in the choroid plexus/ventricles. Immunohistochemical staining showed augmented numbers of microglia and astrocytes, as a function of aging, in the basal ganglia (p < 0.05). Moreover, qualitative analysis of the glial immunostaining at the level of the fimbria and ventral commissure, revealed increments in the number of microglia but decrements in astroglia, in older aged mice. Upon morphological evaluation, aged microglia and astroglia displayed enlarged soma and thickened processes, reminiscent of dystrophy. Since glial cells have major roles in metal metabolism, we performed linear regression analysis and found a positive association between iron (R2 = 0.57, p = 0.0008), copper (R2 = 0.43, p = 0.0057), and zinc (R2 = 0.37, p = 0.0132) with microglia in the basal ganglia. Also, higher levels of iron (R2 = 0.49, p = 0.0025) and zinc (R2 = 0.27, p = 0.040) were correlated to higher astroglia numbers. Aging was accompanied by a dissociation between metal and glial levels, as we found through the formulation of metal to glia ratios, with regions of basal ganglia being differentially affected. For example, iron to astroglia ratio showed age-related increases in the substantia nigra and globus pallidus, while the ratio was decreased in the striatum. Meanwhile, copper and zinc to astroglia ratios showed a similar regional decline. Our findings suggest that inflammation at the choroid plexus, part of the blood-cerebrospinal-fluid barrier, prompts accumulation of, particularly, copper and iron in the ventricles, implying a compromised barrier system. Moreover, age-related glial dystrophy/senescence appears to disrupt metal homeostasis, likely due to induced oxidative stress, and hence increase the risk of neurodegenerative diseases.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Christos Michaelides
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Thomas A Walker
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Antigoni Ekonomou
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Maria Suessmilch
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Achvini Sriskanthanathan
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Semhar Abraha
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Adam Parkes
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Harold G Parkes
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| | - Kalotina Geraki
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, United Kingdom
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
16
|
Zou Z, Shao S, Zou R, Qi J, Chen L, Zhang H, Shen Q, Yang Y, Ma L, Guo R, Li H, Tian H, Li P, Yu M, Wang L, Kong W, Li C, Yu Z, Huang Y, Chen L, Shao Q, Gao X, Chen X, Zhang Z, Yan J, Shao X, Pan R, Xu L, Fang J, Zhao L, Huang Y, Li A, Zhang Y, Huang W, Tian K, Hu M, Xie L, Wu L, Wu Y, Luo Z, Xiao W, Ma S, Wang J, Huang K, He S, Yang F, Zhou S, Jia M, Zhang H, Lu H, Wang X, Tan J. Linking the low-density lipoprotein receptor-binding segment enables the therapeutic 5-YHEDA peptide to cross the blood-brain barrier and scavenge excess iron and radicals in the brain of senescent mice. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:717-731. [PMID: 31921964 PMCID: PMC6944740 DOI: 10.1016/j.trci.2019.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Introduction Iron accumulates in the brain during aging, which catalyzes radical formation, causing neuronal impairment, and is thus considered a pathogenic factor in Alzheimer's disease (AD). To scavenge excess iron-catalyzed radicals and thereby protect the brain and decrease the incidence of AD, we synthesized a soluble pro-iron 5-YHEDA peptide. However, the blood-brain barrier (BBB) blocks large drug molecules from entering the brain and thus strongly reduces their therapeutic effects. However, alternative receptor- or transporter-mediated approaches are possible. Methods A low-density lipoprotein receptor (LDLR)-binding segment of Apolipoprotein B-100 was linked to the 5-YHEDA peptide (bs-5-YHEDA) and intracardially injected into senescent (SN) mice that displayed symptoms of cognitive impairment similar to those of people with AD. Results We successfully delivered 5-YHEDA across the BBB into the brains of the SN mice via vascular epithelium LDLR-mediated endocytosis. The data showed that excess brain iron and radical-induced neuronal necrosis were reduced after the bs-5-YHEDA treatment, together with cognitive amelioration in the SN mouse, and that the senescence-associated ferritin and transferrin increase, anemia and inflammation reversed without kidney or liver injury. Discussion bs-5-YHEDA may be a mild and safe iron remover that can cross the BBB and enter the brain to relieve excessive iron- and radical-induced cognitive disorders.
Collapse
Affiliation(s)
- Zhenyou Zou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China.,Medical School of Taizhou University, Taizhou, ZJ, China.,Biochemistry Department, Purdue University, West Lafayette, USA
| | - Shengxi Shao
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom
| | - Ruyi Zou
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Jini Qi
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Liguan Chen
- Zhejiang Armed Police Corps, Hangzhou, ZJ, China
| | - Hui Zhang
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou, HN, China
| | - Qiqiong Shen
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Yue Yang
- Clinical Laboratory Department, Wenzhou Medical University, ZJ, China
| | - Liman Ma
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Ruzeng Guo
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Hongwen Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Haibo Tian
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Pengxin Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Mingfang Yu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Lu Wang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Wenjuan Kong
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Caiyu Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Zhenhai Yu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Yuping Huang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Li Chen
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Qi Shao
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Xinyan Gao
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Xiaolin Chen
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Zhengbo Zhang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Jianguo Yan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Xiaoyun Shao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Ru Pan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| | - Lu Xu
- Clinical Laboratory of Jingyou Hospital, Xiaoshan, ZJ, China
| | - Jing Fang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Lei Zhao
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Yaohui Huang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Anqi Li
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Yuchong Zhang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Wenkao Huang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Kechun Tian
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Minxin Hu
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Linchao Xie
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Lingbin Wu
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Yu Wu
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Zhen Luo
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Wenxin Xiao
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Shanshan Ma
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Jianan Wang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Kaixin Huang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Siyuan He
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Fan Yang
- Chemistry Engineering Department, Shangrao Normal University, Shangrao, JX, China
| | - Shuni Zhou
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Mo Jia
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Hui Zhang
- Pathology Department, Affiliated Hospital of Taizhou University, ZJ, China
| | - Hongsheng Lu
- Pathology Department, Affiliated Hospital of Taizhou University, ZJ, China
| | - Xinjuan Wang
- Medical School of Taizhou University, Taizhou, ZJ, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX, China
| |
Collapse
|
17
|
Dorsal hippocampal changes in T2 relaxation times are associated with early spatial cognitive deficits in 5XFAD mice. Brain Res Bull 2019; 153:150-161. [PMID: 31422072 DOI: 10.1016/j.brainresbull.2019.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 01/01/2023]
Abstract
T2 relaxation time (T2) alterations may serve as markers for early detection and disease progression monitoring by reflecting brain microstructural integrity in Alzheimer's disease (AD). However, the characteristics of T2 alterations during the early stage of AD remain elusive. We explored T2 alterations and their possible correlations with cognitive function in 5XFAD mice at early ages (1, 2, 3, and 5 months of age). Voxel-based analysis (VBA) and region of interest (ROI) analysis showed a decreased T2 in the hippocampus of 2-, 3-, and 5-month-old 5XFAD mice compared to those of controls. The dorsal hippocampal T2 decreased earlier than the ventral hippocampus T2. A significant correlation was observed between Morris water maze (MWM) test cognitive behavior and the dorsal hippocampus T2 in 5XFAD mice. These results indicated that the microstructural integrity of brain tissues, particularly the hippocampus, was impaired early and the impairment became more extensive and severe during disease progression. Furthermore, the dorsal hippocampus is a crucial component involved in spatial cognition impairment in young 5XFAD mice.
Collapse
|
18
|
Rodríguez-Callejas JDD, Cuervo-Zanatta D, Rosas-Arellano A, Fonta C, Fuchs E, Perez-Cruz C. Loss of ferritin-positive microglia relates to increased iron, RNA oxidation, and dystrophic microglia in the brains of aged male marmosets. Am J Primatol 2019; 81:e22956. [DOI: 10.1002/ajp.22956] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/09/2019] [Accepted: 01/12/2019] [Indexed: 12/26/2022]
Affiliation(s)
| | - Daniel Cuervo-Zanatta
- Department of Pharmacology; Center of Research in Advance Studies; Mexico City Mexico
| | | | - Caroline Fonta
- Brain and Cognition Research Centre (CERCO); CNRS/University of Toulouse; Toulouse France
| | - Eberhard Fuchs
- German Primate Center; Leibniz Institute for Primate Research; Göttingen Germany
| | - Claudia Perez-Cruz
- Department of Pharmacology; Center of Research in Advance Studies; Mexico City Mexico
| |
Collapse
|
19
|
Zou Z, Cai J, Zhong A, Zhou Y, Wang Z, Wu Z, Yang Y, Li X, Cheng X, Tan J, Fan Y, Zhang X, Lu Y, Zhou Y, Yang L, Zhang C, Zhao Q, Fu D, Shen Q, Chen J, Bai S, Wu L, Chen Y, Chen X, Chen J, Zheng H, Wang H, Lou Y, Ding Y, Shen S, Ye Y, Chen L, Lin Y, Huang J, Zou K, Zhang J, Bian B, Huang C, Rong C, Dai L, Xu Y, Cheng L, Chen Y, Luo Y, Zhang S, Li L. Using the synthesized peptide HAYED (5) to protect the brain against iron catalyzed radical attack in a naturally senescence Kunming mouse model. Free Radic Biol Med 2019; 130:458-470. [PMID: 30448512 DOI: 10.1016/j.freeradbiomed.2018.11.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/27/2018] [Accepted: 11/13/2018] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease of the brain. It cannot be cured currently, and those suffering from AD place a great burden on their caregivers and society. AD is characterized by high levels of iron ions in the brain, which catalyze radicals that damage the neurons. Knowing that the Aβ42 peptide precipitates iron by binding iron ions at amino acid residues D1, E3, H11, H13, and H14, we synthesized a 5-repeat (HAYED) sequence peptide. By treating iron-stressed SH-SY5Y cells with it and injecting it into the cerebrospinal fluid (CSF) of naturally senescence Kunming mouse, which displaying AD-similar symptoms such as learning and memory dysfunction, neuron degeneration and high level of iron in brain, we found that HAYED (5) decreased the iron and radical levels in the cell culture medium and in the CSF. Specially, the synthesized peptide prevented cell and brain damage. Furthermore, functional magnetic resonance imaging (fMRI), Morris water maze and passive avoidance tests demonstrated that the peptide ameliorated brain blood-oxygen metabolism and slowed cognitive loss in the experimental senescence mice, and clinical and blood tests showed that HAYED (5) was innoxious to the kidney, the liver and blood and offset the AD-associated inflammation and anemia.
Collapse
Affiliation(s)
- Zhenyou Zou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX 541199, China; Medical School of Taizhou University, Taizhou, ZJ 318000, China; Biochemistry Department, Purdue University, West Lafayette, IN 47906, USA.
| | - Jing Cai
- Genetic Department of Nanjing Medical University, Nanjing, JS 210000, China
| | - Aiguo Zhong
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yong Zhou
- Clinical Laboratory of Affiliated Hospital of Taizhou University, Taizhou, ZJ 318000, China.
| | - Zengxian Wang
- Medical Image Center of Affiliated Hospital of Taizhou University, Taizhou, ZJ 318000, China.
| | - Zhongmin Wu
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yue Yang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Xin Li
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Xiaoying Cheng
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX 541199, China
| | - Yihao Fan
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Xiaotong Zhang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yuxiang Lu
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yaping Zhou
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Liu Yang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | | | - Qiang Zhao
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Derong Fu
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Qiqiong Shen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Jie Chen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Shi Bai
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Lijuan Wu
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yongfeng Chen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Xin Chen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Jiaren Chen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Hongjie Zheng
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Hongdian Wang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yingjie Lou
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yarong Ding
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Shiyi Shen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Ying Ye
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Lifen Chen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yukai Lin
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Jue Huang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Kechun Zou
- Shangli Teacher Training School, Pingxiang, JX 337009, China
| | - Jianxing Zhang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Baohua Bian
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Chengbo Huang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Cuiping Rong
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX 541199, China
| | - Limiao Dai
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yali Xu
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Lin Cheng
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Ye Chen
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Yewen Luo
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Shanshan Zhang
- Medical School of Taizhou University, Taizhou, ZJ 318000, China
| | - Liangxian Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, GX 541199, China
| |
Collapse
|
20
|
da Silva VK, de Freitas BS, Garcia RCL, Monteiro RT, Hallak JE, Zuardi AW, Crippa JAS, Schröder N. Antiapoptotic effects of cannabidiol in an experimental model of cognitive decline induced by brain iron overload. Transl Psychiatry 2018; 8:176. [PMID: 30177808 PMCID: PMC6120904 DOI: 10.1038/s41398-018-0232-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/03/2018] [Accepted: 07/24/2018] [Indexed: 12/26/2022] Open
Abstract
Iron accumulation in the brain has been recognized as a common feature of both normal aging and neurodegenerative diseases. Cognitive dysfunction has been associated to iron excess in brain regions in humans. We have previously described that iron overload leads to severe memory deficits, including spatial, recognition, and emotional memory impairments in adult rats. In the present study we investigated the effects of neonatal iron overload on proteins involved in apoptotic pathways, such as Caspase 8, Caspase 9, Caspase 3, Cytochrome c, APAF1, and PARP in the hippocampus of adult rats, in an attempt to establish a causative role of iron excess on cell death in the nervous system, leading to memory dysfunction. Cannabidiol (CBD), the main non-psychotropic component of Cannabis sativa, was examined as a potential drug to reverse iron-induced effects on the parameters analyzed. Male rats received vehicle or iron carbonyl (30 mg/kg) from the 12th to the 14th postnatal days and were treated with vehicle or CBD (10 mg/kg) for 14 days in adulthood. Iron increased Caspase 9, Cytochrome c, APAF1, Caspase 3 and cleaved PARP, without affecting cleaved Caspase 8 levels. CBD reversed iron-induced effects, recovering apoptotic proteins Caspase 9, APAF1, Caspase 3 and cleaved PARP to the levels found in controls. These results suggest that iron can trigger cell death pathways by inducing intrinsic apoptotic proteins. The reversal of iron-induced effects by CBD indicates that it has neuroprotective potential through its anti-apoptotic action.
Collapse
Affiliation(s)
- Vanessa Kappel da Silva
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Rebeca Carvalho Lacerda Garcia
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Ricardo Tavares Monteiro
- Neurobiology and Developmental Biology Laboratory, Faculty of Biosciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, 90619-900, Brazil
| | - Jaime Eduardo Hallak
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - Antônio Waldo Zuardi
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - José Alexandre S Crippa
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, São Paulo, 14048-900, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil.
- Departamento de Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, 90050-170, Brazil.
| |
Collapse
|
21
|
Abstract
Brain iron is tightly regulated by a multitude of proteins to ensure homeostasis. Iron dyshomeostasis has become a molecular signature associated with aging which is accompanied by progressive decline in cognitive processes. A common theme in neurodegenerative diseases where age is the major risk factor, iron dyshomeostasis coincides with neuroinflammation, abnormal protein aggregation, neurodegeneration, and neurobehavioral deficits. There is a great need to determine the mechanisms governing perturbations in iron metabolism, in particular to distinguish between physiological and pathological aging to generate fruitful therapeutic targets for neurodegenerative diseases. The aim of the present review is to focus on the age-related alterations in brain iron metabolism from a cellular and molecular biology perspective, alongside genetics, and neuroimaging aspects in man and rodent models, with respect to normal aging and neurodegeneration. In particular, the relationship between iron dyshomeostasis and neuroinflammation will be evaluated, as well as the effects of systemic iron overload on the brain. Based on the evidence discussed here, we suggest a synergistic use of iron-chelators and anti-inflammatories as putative anti-brain aging therapies to counteract pathological aging in neurodegenerative diseases.
Collapse
Affiliation(s)
- Azhaar Ashraf
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| | - Maryam Clark
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Po-Wah So
- Institute of Psychiatry, Psychology and Neuroscience, Department of Neuroimaging, King's College London, London, United Kingdom
| |
Collapse
|
22
|
Caldeira C, Cunha C, Vaz AR, Falcão AS, Barateiro A, Seixas E, Fernandes A, Brites D. Key Aging-Associated Alterations in Primary Microglia Response to Beta-Amyloid Stimulation. Front Aging Neurosci 2017; 9:277. [PMID: 28912710 PMCID: PMC5583148 DOI: 10.3389/fnagi.2017.00277] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/03/2017] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by a progressive cognitive decline and believed to be driven by the self-aggregation of amyloid-β (Aβ) peptide into oligomers and fibrils that accumulate as senile plaques. It is widely accepted that microglia-mediated inflammation is a significant contributor to disease pathogenesis; however, different microglia phenotypes were identified along AD progression and excessive Aβ production was shown to dysregulate cell function. As so, the contribution of microglia to AD pathogenesis remains to be elucidated. In this study, we wondered if isolated microglia cultured for 16 days in vitro (DIV) would react differentially from the 2 DIV cells upon treatment with 1000 nM Aβ1-42 for 24 h. No changes in cell viability were observed and morphometric alterations associated to microglia activation, such as volume increase and process shortening, were obvious in 2 DIV microglia, but less evident in 16 DIV cells. These cells showed lower phagocytic, migration and autophagic properties after Aβ treatment than the 2 DIV cultured microglia. Reduced phagocytosis may derive from increased CD33 expression, reduced triggering receptor expressed on myeloid cells 2 (TREM2) and milk fat globule-EGF factor 8 protein (MFG-E8) levels, which were mainly observed in 16 DIV cells. Activation of inflammatory mediators, such as high mobility group box 1 (HMGB1) and pro-inflammatory cytokines, as well as increased expression of Toll-like receptor 2 (TLR2), TLR4 and fractalkine/CX3C chemokine receptor 1 (CX3CR1) cell surface receptors were prominent in 2 DIV microglia, while elevation of matrix metalloproteinase 9 (MMP9) was marked in 16 DIV cells. Increased senescence-associated β-galactosidase (SA-β-gal) and upregulated miR-146a expression that were observed in 16 DIV cells showed to increase by Aβ in 2 DIV microglia. Additionally, Aβ downregulated miR-155 and miR-124, and reduced the CD11b+ subpopulation in 2 DIV microglia, while increased the number of CD86+ cells in 16 DIV microglia. Simultaneous M1 and M2 markers were found after Aβ treatment, but at lower expression in the in vitro aged microglia. Data show key-aging associated responses by microglia when incubated with Aβ, with a loss of reactivity from the 2 DIV to the 16 DIV cells, which course with a reduced phagocytosis, migration and lower expression of inflammatory miRNAs. These findings help to improve our understanding on the heterogeneous responses that microglia can have along the progression of AD disease and imply that therapeutic approaches may differ from early to late stages.
Collapse
Affiliation(s)
- Cláudia Caldeira
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Carolina Cunha
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Ana R Vaz
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Ana S Falcão
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Andreia Barateiro
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Elsa Seixas
- Obesity Laboratory, Instituto Gulbenkian de CiênciaOeiras, Portugal
| | - Adelaide Fernandes
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| | - Dora Brites
- Neuron Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de LisboaLisbon, Portugal
| |
Collapse
|
23
|
Ingram T, Chakrabarti L. Proteomic profiling of mitochondria: what does it tell us about the ageing brain? Aging (Albany NY) 2016; 8:3161-3179. [PMID: 27992860 PMCID: PMC5270661 DOI: 10.18632/aging.101131] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/01/2016] [Indexed: 02/07/2023]
Abstract
Mitochondrial dysfunction is evident in numerous neurodegenerative and age-related disorders. It has also been linked to cellular ageing, however our current understanding of the mitochondrial changes that occur are unclear. Functional studies have made some progress reporting reduced respiration, dynamic structural modifications and loss of membrane potential, though there are conflicts within these findings. Proteomic analyses, together with functional studies, are required in order to profile the mitochondrial changes that occur with age and can contribute to unravelling the complexity of the ageing phenotype. The emergence of improved protein separation techniques, combined with mass spectrometry analyses has allowed the identification of age and cell-type specific mitochondrial changes in energy metabolism, antioxidants, fusion and fission machinery, chaperones, membrane proteins and biosynthesis pathways. Here, we identify and review recent data from the analyses of mitochondria from rodent brains. It is expected that knowledge gained from understanding age-related mitochondrial changes of the brain should lead to improved biomarkers of normal ageing and also age-related disease progression.
Collapse
Affiliation(s)
- Thomas Ingram
- SVMS, Faculty of Medicine, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| | - Lisa Chakrabarti
- SVMS, Faculty of Medicine, University of Nottingham, Sutton Bonington, LE12 5RD, UK
| |
Collapse
|