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
|
Gu K, Wu A, Liu C, Yu B, He J, Lai X, Chen J, Luo Y, Yan H, Zheng P, Luo J, Pu J, Wang Q, Wang H, Chen D. Absence of gut microbiota alleviates iron overload-induced colitis by modulating ferroptosis in mice. J Adv Res 2024:S2090-1232(24)00608-8. [PMID: 39710300 DOI: 10.1016/j.jare.2024.12.030] [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: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
INTRODUCTION Iron overload disrupts gut microbiota and induces ferroptosis, contributing to colitis. However, whether gut microbiota directly drives iron overload-induced colitis and its underlying mechanism remain unclear. OBJECTIVES The study aimed to explore whether gut microbiota can directly regulate iron overload-induced colitis and its underling mechanism. METHODS Male C57BL/6N mice were fed with ferrous sulfate to establish an iron overload model. Antibiotics and DSS were used to create germ-free and colitis models, respectively. RESULTS Results showed that iron overload caused disruption of systemic iron homeostasis via activating pro-inflammation response, which caused induction of ferroptosis and eventually resulted in colitis in mice. Notably, iron overload inhibited System Xc- and activated the nuclear factor E2-related factor 2/heme oxygenase-1 pathway, driving ferroptosis and colitis progression. Similar results were observed in mouse colon epithelial cells, which were treated with high doses ferric ammonium citrate. Additionally, iron overload exacerbated DSS-induced colitis by activating the ferroptosis and increasing harmful bacteria (e.g., Mucispirillum) abundance. Interestingly, eliminating gut microbiota attenuated iron overload-induced colitis, without affecting systemic inflammation through inhibiting ferroptosis of mice. Depletion of the gut microbiota partially mitigated the exacerbating effect of iron overload on DSS-induced colitis through inhibiting ferroptosis of mice. CONCLUSION Iron overload activates ferroptosis in colonic cells, increases the relative abundance of harmful bacteria, and exacerbates DSS-induced colitis in mice. Iron overload exacerbates DSS-induced ferroptosis and colitis in a microbiota-dependent manner. Targeting gut microbiota may offer new strategies for managing iron overload-induced colitis.
Collapse
Affiliation(s)
- Ke Gu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Aimin Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Chen Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China; Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu 611130, China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Lai
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junzhou Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Junning Pu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Quyuan Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Huifen Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
3
|
Lu W, Song T, Zang Z, Li J, Zhang Y, Lu J. Relaxometry network based on MRI R 2⁎ mapping revealing brain iron accumulation patterns in Parkinson's disease. Neuroimage 2024; 303:120943. [PMID: 39571643 DOI: 10.1016/j.neuroimage.2024.120943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/12/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Excessive iron accumulation in the brain has been implicated in Parkinson's disease (PD). However, the patterns and probable sequences of iron accumulation across the PD brain remain largely unknown. This study aimed to explore the sequence of iron accumulation across the PD brain using R2* mapping and a relaxometry covariance network (RCN) approach. METHODS R2* quantification maps were obtained from PD patients (n = 34) and healthy controls (n = 25). RCN was configured on R2* maps to identify covariance differences in iron levels between the two groups. Regions with excessive iron accumulation and large covariance changes in PD patients compared to controls were defined as propagators of iron. In the PD group, causal RCN analysis was performed on the R2* maps sequenced according to disease duration to investigate the dynamics of iron accumulations from the propagators. The associations between individual connections of the RCN and clinical information were analyzed in PD patients. RESULTS The left substantia nigra pars reticulata (SNpr), left substantia nigra pars compacta (SNpc), and lobule VII of the vermis (VER7) were identified as primary regions for iron accumulation and propagation (propagator). As the disease duration increased, iron accumulation in these three propagators demonstrated positive causal effects on the bilateral pallidum, bilateral gyrus rectus, right middle frontal gyrus, and medial and anterior orbitofrontal cortex (OFC). Furthermore, individual connections of VER7 with the left gyrus rectus and anterior OFC were positively associated with disease duration. CONCLUSIONS Our results indicate that the aberrant iron accumulation in PD involves several regions, mainly starts from the SN and cerebellum and extends to the pallidum and cortices. These findings provide preliminary information on sequences of iron accumulation in PD, which may advance our understanding of the disease.
Collapse
Affiliation(s)
- Weizhao Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Xuanwu Hospital, Beijing, 100053, China
| | - Tianbin Song
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Xuanwu Hospital, Beijing, 100053, China
| | - Zhenxiang Zang
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jiping Li
- Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuqing Zhang
- Beijing Institute of Functional Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Xuanwu Hospital, Beijing, 100053, China.
| |
Collapse
|
4
|
Li Y, Yan S, Li J, Qin Y, Li L, Shen N, Xie Y, Liu D, Fang J, Tian T, Zhu W. Regional homogeneity patterns reveal the genetic and neurobiological basis of State-Trait Anxiety. BMC Psychiatry 2024; 24:837. [PMID: 39567951 PMCID: PMC11577826 DOI: 10.1186/s12888-024-06291-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
OBJECTIVE State anxiety and trait anxiety are differentially mapped in brain function. However, the genetic and neurobiological basis of anxiety-related functional changes remain largely unknown. METHODS Participants aged 18-30 from the community underwent resting-state fMRI and were assessed with the State-Trait Anxiety Inventory. Using a general linear regression model, we analyzed the effects of state and trait anxiety, as well as their sum and difference (delta), on regional homogeneity (ReHo) in cortical areas. ReHo patterns denote the spatial distribution of ReHo associated with anxiety scores. We further explored the spatial correlations between ReHo patterns and neuromaps, including gene expression, neurotransmitter receptor density, myelination, and functional connectivity gradients, to elucidate the genetic and molecular substrates of these ReHo patterns. RESULTS Our findings demonstrated robust spatial correlations between whole-brain ReHo patterns for state and trait anxiety, with trait anxiety and the delta value exhibiting stronger network correlations, notably in the dorsal attention, salience, visual, and sensorimotor networks. Genes highly correlated with ReHo patterns exhibited unique spatiotemporal expression patterns, involvement in oxidative stress, metabolism, and response to stimuli, and were expressed in specific cell types. Furthermore, ReHo patterns significantly correlated with neuromaps of neurotransmitter receptor density, myelination, and functional connectivity gradients. CONCLUSIONS The ReHo patterns associated with anxiety may be driven by genetic and neurobiological traits. Our findings contribute to a deeper understanding of the pathogenesis of anxiety from a genetic and molecular perspective.
Collapse
Affiliation(s)
- Yuanhao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Su Yan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Jia Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Yuanyuan Qin
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Li Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Nanxi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Yan Xie
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Dong Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Jicheng Fang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China
| | - Tian Tian
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China.
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, P.R. China.
| |
Collapse
|
5
|
Wu J, Wu J, Chen T, Cai J, Ren R. Protein aggregation and its affecting mechanisms in neurodegenerative diseases. Neurochem Int 2024; 180:105880. [PMID: 39396709 DOI: 10.1016/j.neuint.2024.105880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Protein aggregation serves as a critical pathological marker in a spectrum of neurodegenerative diseases (NDs), including the formation of amyloid β (Aβ) and Tau neurofibrillary tangles in Alzheimer's disease, as well as α-Synuclein (α-Syn) aggregates in Parkinson's disease, Parkinson's disease-related dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). A significant proportion of patients with amyotrophic lateral sclerosis (ALS) exhibit TDP-43 aggregates. Moreover, a confluence of brain protein pathologies, such as Aβ, Tau, α-Syn, and TDP-43, has been identified in individual NDs cases, highlighting the intricate interplay among these proteins that is garnering heightened scrutiny. Importantly, protein aggregation is modulated by an array of factors, with burgeoning evidence suggesting that it frequently results from perturbations in protein homeostasis, influenced by the cellular membrane milieu, metal ion concentrations, post-translational modifications, and genetic mutations. This review delves into the pathological underpinnings of protein aggregation across various NDs and elucidates the intercommunication among disparate proteins within the same disease context. Additionally, we examine the pathogenic mechanisms by which diverse factors impinge upon protein aggregation, offering fresh perspectives for the future therapeutic intervention of NDs.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jianan Wu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jing Cai
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
6
|
Hervouin A, Bézy-Wendling J, Noury F. How to accurately quantify brain magnetic susceptibility in the context of Parkinson's disease: Validation on phantoms and healthy volunteers at 1.5 and 3 T. NMR IN BIOMEDICINE 2024; 37:e5182. [PMID: 38993048 DOI: 10.1002/nbm.5182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 07/13/2024]
Abstract
Currently, brain iron content represents a new neuromarker for understanding the physiopathological mechanisms leading to Parkinson's disease (PD). In vivo quantification of biological iron is possible by reconstructing magnetic susceptibility maps obtained using quantitative susceptibility mapping (QSM). Applying QSM is challenging, as up to now, no standardization of acquisition protocols and phase image processing has emerged from referenced studies. Our objectives were to compare the accuracy and the sensitivity of 10 QSM pipelines built from algorithms from the literature, applied on phantoms data and on brain data. Two phantoms, with known magnetic susceptibility ranges, were created from several solutions of gadolinium chelate. Twenty healthy volunteers from two age groups were included. Phantoms and brain data were acquired at 1.5 and 3 T, respectively. Susceptibility-weighted images were obtained using a 3D multigradient-recalled-echo sequence. For brain data, 3D anatomical T1- and T2-weighted images were also acquired to segment the deep gray nuclei of interest. Concerning in vitro data, the linear dependence of magnetic susceptibility versus gadolinium concentration and deviations from the theoretically expected values were calculated. For brain data, the accuracy and sensitivity of the QSM pipelines were evaluated in comparison with results from the literature and regarding the expected magnetic susceptibility increase with age, respectively. A nonparametric Mann-Whitney U-test was used to compare the magnetic susceptibility quantification in deep gray nuclei between the two age groups. Our methodology enabled quantifying magnetic susceptibility in human brain and the results were consistent with those from the literature. Statistically significant differences were obtained between the two age groups in all cerebral regions of interest. Our results show the importance of optimizing QSM pipelines according to the application and the targeted magnetic susceptibility range, to achieve accurate quantification. We were able to define the optimal QSM pipeline for future applications on patients with PD.
Collapse
Affiliation(s)
| | | | - Fanny Noury
- Univ Rennes, Inserm, LTSI-UMR 1099, Rennes, France
| |
Collapse
|
7
|
Yao J, Li Z, Zhou Z, Bao A, Wang Z, Wei H, He H. Distinct regional vulnerability to Aβ and iron accumulation in post mortem AD brains. Alzheimers Dement 2024; 20:6984-6997. [PMID: 39175425 PMCID: PMC11485316 DOI: 10.1002/alz.14188] [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: 05/08/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION The paramagnetic iron, diamagnetic amyloid beta (Aβ) plaques and their interaction are crucial in Alzheimer's disease (AD) pathogenesis, complicating non-invasive magnetic resonance imaging for prodromal AD detection. METHODS We used a state-of-the-art sub-voxel quantitative susceptibility mapping method to simultaneously measure Aβ and iron levels in post mortem human brains, validated by histology. Further transcriptomic analysis using Allen Human Brain Atlas elucidated the underlying biological processes. RESULTS Regional increased paramagnetic and diamagnetic susceptibility were observed in medial prefrontal, medial parietal, and para-hippocampal cortices associated with iron deposition (R = 0.836, p = 0.003) and Aβ accumulation (R = 0.853, p = 0.002) in AD brains. Higher levels of gene expression relating to cell cycle, post-translational protein modifications, and cellular response to stress were observed. DISCUSSION These findings provide quantitative insights into the variable vulnerability of cortical regions to higher levels of Aβ aggregation, iron overload, and subsequent neurodegeneration, indicating changes preceding clinical symptoms. HIGHLIGHTS The vulnerability of distinct brain regions to amyloid beta (Aβ) and iron accumulation varies. Histological validation was performed on stained sections of ex-vivo human brains. Regional variations in susceptibility were linked to gene expression profiles. Iron and Aβ levels in ex-vivo brains were simultaneously quantified.
Collapse
Affiliation(s)
- Junye Yao
- Center for Brain Imaging Science and TechnologyZhejiang UniversityHangzhouChina
- College of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Zhenghao Li
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Zihan Zhou
- Center for Brain Imaging Science and TechnologyZhejiang UniversityHangzhouChina
- Stanford University Graduate School of EducationDepartment of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Aimin Bao
- National Human Brain Bank for Health and DiseaseSchool of Brain Science and Brain MedicineZhejiang UniversityHangzhouChina
| | - Zheng Wang
- School of Psychological and Cognitive SciencesBeijing Key Laboratory of Behavior and Mental HealthIDG/McGovern Institute for Brain ResearchPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijingChina
- School of Biomedical EngineeringHainan UniversityHaikouChina
| | - Hongjiang Wei
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Hongjian He
- Center for Brain Imaging Science and TechnologyZhejiang UniversityHangzhouChina
- School of PhysicsZhejiang UniversityHangzhouChina
- State Key Laboratory of Brain‐Machine IntelligenceZhejiang UniversityHangzhouChina
| |
Collapse
|
8
|
Gogna T, Housden BE, Houldsworth A. Exploring the Role of Reactive Oxygen Species in the Pathogenesis and Pathophysiology of Alzheimer's and Parkinson's Disease and the Efficacy of Antioxidant Treatment. Antioxidants (Basel) 2024; 13:1138. [PMID: 39334797 PMCID: PMC11429442 DOI: 10.3390/antiox13091138] [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: 07/24/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's (AD) and Parkinson's Disease (PD) are life-altering diseases that are characterised by progressive memory loss and motor dysfunction. The prevalence of AD and PD is predicted to continuously increase. Symptoms of AD and PD are primarily mediated by progressive neuron death and dysfunction in the hippocampus and substantia nigra. Central features that drive neurodegeneration are caspase activation, DNA fragmentation, lipid peroxidation, protein carbonylation, amyloid-β, and/or α-synuclein formation. Reactive oxygen species (ROS) increase these central features. Currently, there are limited therapeutic options targeting these mechanisms. Antioxidants reduce ROS levels by the induction of antioxidant proteins and direct neutralisation of ROS. This review aims to assess the effectiveness of antioxidants in reducing ROS and neurodegeneration. Antioxidants enhance major endogenous defences against ROS including superoxide dismutase, catalase, and glutathione. Direct neutralisation of ROS by antioxidants protects against ROS-induced cytotoxicity. The combination of Indirect and direct protective mechanisms prevents ROS-induced α-synuclein and/or amyloid-β formation. Antioxidants ameliorate ROS-mediated oxidative stress and subsequent deleterious downstream effects that promote apoptosis. As a result, downstream harmful events including neuron death, dysfunction, and protein aggregation are decreased. The protective effects of antioxidants in human models have yet to directly replicate the success seen in cell and animal models. However, the lack of diversity in antioxidants for clinical trials prevents a definitive answer if antioxidants are protective. Taken together, antioxidant treatment is a promising avenue in neurodegenerative disease therapy and subsequent clinical trials are needed to provide a definitive answer on the protective effects of antioxidants. No current treatment strategies have significant impact in treating advanced AD and PD, but new mimetics of endogenous mitochondrial antioxidant enzymes (Avasopasem Manganese, GC4419 AVA) may be a promising innovative option for decelerating neurodegenerative progress in the future at the mitochondrial level of OS.
Collapse
Affiliation(s)
- Talin Gogna
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| | - Benjamin E Housden
- Living Systems Institute, Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Annwyne Houldsworth
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| |
Collapse
|
9
|
Jiang J, Yang L, Xie Q, Liu X, Jiang J, Zhang J, Zhang S, Zheng H, Li W, Cai X, Liu S, Li R. Synthetic vectors for activating the driving axis of ferroptosis. Nat Commun 2024; 15:7923. [PMID: 39256387 PMCID: PMC11387475 DOI: 10.1038/s41467-024-52312-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/30/2024] [Indexed: 09/12/2024] Open
Abstract
Ferroptosis is a promising strategy for cancer therapy, with numerous inhibitors of its braking axes under investigation as potential drugs. However, few studies have explored the potential of activating the driving axes to induce ferroptosis. Herein, phosphatidylcholine peroxide decorating liposomes (LIPPCPO) are synthesized to induce ferroptosis by targeting divalent metal transporter 1 (DMT1). LIPPCPO is found to boost lysosomal Fe2+ efflux by inducing cysteinylation of lysosomal DMT1, resulting in glutathione peroxidase 4 (GPX4) suppression, glutathione depletion and ferroptosis in breast cancer cells and xenografts. Importantly, LIPPCPO induced ferroptotic cell death is independent of acquired resistance to radiation, chemotherapy, or targeted agents in 11 cancer cell lines. Furthermore, a strong synergistic ferroptosis effect is observed between LIPPCPO and an FDA-approved drug, artesunate, as well as X rays. The formula of LIPPCPO encapsulating artesunate significantly inhibits tumor growth and metastasis and improves the survival rate of breast cancer-bearing female mice. These findings provide a distinct strategy for inducing ferroptosis and highlight the potential of LIPPCPO as a vector to synergize the therapeutic effects of conventional ferroptosis inducers.
Collapse
Affiliation(s)
- Jun Jiang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
- Department of Hepatobiliary and Pancreatic Surgery the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lili Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qianqian Xie
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Xi Liu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Jie Jiang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Huizhen Zheng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Wenjie Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Xiaoming Cai
- School of Public Health, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.
| | - Ruibin Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
10
|
Tunoğlu S, Yalçın B, Tunoğlu ENY, Karaaslan Z, Bilgiç B, Hanağası HA, Tüzün E, Küçükali Cİ. Gene Expression Levels Related to Histone Acetylation are Altered in Parkinson Disease Patients. Noro Psikiyatr Ars 2024; 67:271-274. [PMID: 39258138 PMCID: PMC11382565 DOI: 10.29399/npa.28700] [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: 01/19/2024] [Accepted: 03/02/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Parkinson's Disease (PD) is a neurodegenerative disorder distinguished from other neurodegenerative disorders by the loss of dopaminergic neurons in the substantia nigra region of the brain, and is the most common neurodegenerative disorder, along with Alzheimer's Disease. PD is characterized by the presence of Lewy bodies when evaluated pathologically. Recent studies showed that the incidence of PH development as a result of genetic mutations alone is very low among all PD cases, and that environmental effects contribute significantly to the disease progression. The molecular mechanisms of diseases are associated with the maintenance of gene and protein expressions as a result of epigenetic regulations. The role of these regulations in the development and pathogenesis of neurodegenerative diseases is still not clearly understood. Methods In our study, we examined the expression levels of H3C1, H3C12, HDAC4, HDAC5, ANKRD11, ANKRD12, ITM2B and GABBR1, which are genes involved in epigenetic processes in patients with idiopathic PD. Seventy five patients diagnosed with idiopathic PD and 50 healthy controls were included in the study. Peripheral Blood Mononuclear Cell (PBMC) was obtained from whole blood taken from the patient and control groups, and then total RNA was isolated from PBMC. Results According to the comparison of the patient and control groups, the expression of H3C1, H3C12, ITM2B was high, and the expression of ANKRD11, HDAC4, HDAC5 and GABBR1 was low (p<0.05). Conclusion As conclusion, we propose that histone regulation is one of the epigenetic mechanisms related to the presence of PD.
Collapse
Affiliation(s)
- Servet Tunoğlu
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Çapa, Istanbul, Turkey
| | - Beyzanur Yalçın
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ezgi Nurdan Yenilmez Tunoğlu
- Division of Medical Techniques and Services, Vocational School of Health Services, Demiroglu Science University, Istanbul, Turkey
| | - Zerrin Karaaslan
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Başar Bilgiç
- Istanbul University, Istanbul Faculty of Medicine, Department of Neurology, Behavioral Neurology and Movement Disorders Unit, Istanbul, Turkey
| | - Haşmet Ayhan Hanağası
- Istanbul University, Istanbul Faculty of Medicine, Department of Neurology, Behavioral Neurology and Movement Disorders Unit, Istanbul, Turkey
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Cem İsmail Küçükali
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
11
|
Qiu R, Cai Y, Su Y, Fan K, Sun Z, Zhang Y. Emerging insights into Lipocalin-2: Unraveling its role in Parkinson's Disease. Biomed Pharmacother 2024; 177:116947. [PMID: 38901198 DOI: 10.1016/j.biopha.2024.116947] [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: 03/03/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Parkinson's disease (PD) ranks as the second most prevalent neurodegenerative disorder globally, marked by a complex pathogenesis. Lipocalin-2 (LCN2) emerges as a crucial factor during the progression of PD. Belonging to the lipocalin family, LCN2 is integral to several biological functions, including glial cell activation, iron homeostasis regulation, immune response, inflammatory reactions, and oxidative stress mitigation. Substantial research has highlighted marked increases in LCN2 expression within the substantia nigra (SN), cerebrospinal fluid (CSF), and blood of individuals with PD. This review focuses on the pathological roles of LCN2 in neuroinflammation, aging, neuronal damage, and iron dysregulation in PD. It aims to explore the underlying mechanisms of LCN2 in the disease and potential therapeutic targets that could inform future treatment strategies.
Collapse
Affiliation(s)
- Ruqing Qiu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yunjia Cai
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yana Su
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Kangli Fan
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
12
|
Yan S, Lu J, Duan B, Zhu H, Liu D, Li L, Qin Y, Li Y, Zhu W. Quantitative susceptibility mapping of multiple system atrophy and Parkinson's disease correlates with neurotransmitter reference maps. Neurobiol Dis 2024; 198:106549. [PMID: 38830476 DOI: 10.1016/j.nbd.2024.106549] [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: 05/02/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Multiple system atrophy (MSA) and Parkinson's disease (PD) are neurodegenerative disorders characterized by α-synuclein pathology, disrupted iron homeostasis and impaired neurochemical transmission. Considering the critical role of iron in neurotransmitter synthesis and transport, our study aims to identify distinct patterns of whole-brain iron accumulation in MSA and PD, and to elucidate the corresponding neurochemical substrates. METHODS A total of 122 PD patients, 58 MSA patients and 78 age-, sex-matched health controls underwent multi-echo gradient echo sequences and neurological evaluations. We conducted voxel-wise and regional analyses using quantitative susceptibility mapping to explore MSA or PD-specific alterations in cortical and subcortical iron concentrations. Spatial correlation approaches were employed to examine the topographical alignment of cortical iron accumulation patterns with normative atlases of neurotransmitter receptor and transporter densities. Furthermore, we assessed the associations between the colocalization strength of neurochemical systems and disease severity. RESULTS MSA patients exhibited increased susceptibility in the striatal, midbrain, cerebellar nuclei, as well as the frontal, temporal, occipital lobes, and anterior cingulate gyrus. In contrast, PD patients displayed elevated iron levels in the left inferior occipital gyrus, precentral gyrus, and substantia nigra. The excessive iron accumulation in MSA or PD correlated with the spatial distribution of cholinergic, noradrenaline, glutamate, serotonin, cannabinoids, and opioid neurotransmitters, and the degree of this alignment was related to motor deficits. CONCLUSIONS Our findings provide evidence of the interaction between iron accumulation and non-dopamine neurotransmitters in the pathogenesis of MSA and PD, which inspires research on potential targets for pharmacotherapy.
Collapse
Affiliation(s)
- Su Yan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Lu
- Department of CT & MRI, The First Affiliated Hospital, College of Medicine, Shihezi University, 107 North Second Road, Shihezi, China
| | - Bingfang Duan
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongquan Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Qin
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanhao Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
13
|
Soni P, Ammal Kaidery N, Sharma SM, Gazaryan I, Nikulin SV, Hushpulian DM, Thomas B. A critical appraisal of ferroptosis in Alzheimer's and Parkinson's disease: new insights into emerging mechanisms and therapeutic targets. Front Pharmacol 2024; 15:1390798. [PMID: 39040474 PMCID: PMC11260649 DOI: 10.3389/fphar.2024.1390798] [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: 02/24/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Neurodegenerative diseases represent a pressing global health challenge, and the identification of novel mechanisms underlying their pathogenesis is of utmost importance. Ferroptosis, a non-apoptotic form of regulated cell death characterized by iron-dependent lipid peroxidation, has emerged as a pivotal player in the pathogenesis of neurodegenerative diseases. This review delves into the discovery of ferroptosis, the critical players involved, and their intricate role in the underlying mechanisms of neurodegeneration, with an emphasis on Alzheimer's and Parkinson's diseases. We critically appraise unsolved mechanistic links involved in the initiation and propagation of ferroptosis, such as a signaling cascade resulting in the de-repression of lipoxygenase translation and the role played by mitochondrial voltage-dependent anionic channels in iron homeostasis. Particular attention is given to the dual role of heme oxygenase in ferroptosis, which may be linked to the non-specific activity of P450 reductase in the endoplasmic reticulum. Despite the limited knowledge of ferroptosis initiation and progression in neurodegeneration, Nrf2/Bach1 target genes have emerged as crucial defenders in anti-ferroptotic pathways. The activation of Nrf2 and the inhibition of Bach1 can counteract ferroptosis and present a promising avenue for future therapeutic interventions targeting ferroptosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Priyanka Soni
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC, United States
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| | - Navneet Ammal Kaidery
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC, United States
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| | - Sudarshana M. Sharma
- Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Irina Gazaryan
- Department of Chemical Enzymology, School of Chemistry, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Chemistry and Physical Sciences, Dyson College of Arts and Sciences, Pace University, Pleasantville, NY, United States
| | - Sergey V. Nikulin
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia
| | - Dmitry M. Hushpulian
- Faculty of Biology and Biotechnologies, Higher School of Economics, Moscow, Russia
- A.N.Bach Institute of Biochemistry, Federal Research Center “Fundamentals of Biotechnology” of the Russian Academy of Sciences, Moscow, Russia
| | - Bobby Thomas
- Darby Children’s Research Institute, Medical University of South Carolina, Charleston, SC, United States
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
- Department of Drug Discovery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
14
|
Chen K, Wei X, Zhang W, Wang R, Wang Y, Yang L. Bone morphogenetic protein 4 derived from the cerebrospinal fluid in patients with postherpetic neuralgia induces allodynia via the crosstalk between microglia and astrocyte. Brain Behav Immun 2024; 119:836-850. [PMID: 38735405 DOI: 10.1016/j.bbi.2024.05.007] [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: 12/29/2023] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION During postherpetic neuralgia (PHN), the cerebral spinal fluid (CSF) possesses the capability to trigger glial activation and inflammation, yet the specific changes in its composition remain unclear. Recent findings from our research indicate elevations of central bone morphogenetic protein 4 (BMP4) during neuropathic pain (NP), serving as an independent modulator of glial cells. Herein, the aim of the present study is to test the CSF-BMP4 expressions and its role in the glial modulation in the process of PHN. METHODS CSF samples were collected from both PHN patients and non-painful individuals (Control) to assess BMP4 and its antagonist Noggin levels. Besides, intrathecal administration of both CSF types was conducted in normal rats to evaluate the impact on pain behavior, glial activity, and inflammation.; Additionally, both Noggin and STAT3 antagonist-Stattic were employed to treat the PHN-CSF or exogenous BMP4 challenged cultured astrocytes to explore downstream signals. Finally, microglial depletion was performed prior to the PHN-CSF intervention so as to elucidate the microglia-astrocyte crosstalk. RESULTS BMP4 levels were significantly higher in PHN-CSF compared to Control-CSF (P < 0.001), with a positive correlation with pain duration (P < 0.05, r = 0.502). Comparing with the Control-CSF producing moderate paw withdrawal threshold (PWT) decline and microglial activation, PHN-CSF further exacerbated allodynia and triggered both microglial and astrocytic activation (P < 0.05). Moreover, PHN-CSF rather than Control-CSF evoked microglial proliferation and pro-inflammatory transformation, reinforced iron storage, and activated astrocytes possibly through both SMAD159 and STAT3 signaling, which were all mitigated by the Noggin application (P < 0.05). Next, both Noggin and Stattic effectively attenuated BMP4-induced GFAP and IL-6 upregulation, as well as SMAD159 and STAT3 phosphorylation in the cultured astrocytes (P < 0.05). Finally, microglial depletion diminished PHN-CSF induced astrogliosis, inflammation and endogenous BMP4 expression (P < 0.05). CONCLUSION Our study highlights the role of CSF-BMP4 elevation in glial activation and allodynia during PHN, suggesting a potential therapeutic avenue for future exploration.
Collapse
Affiliation(s)
- Kai Chen
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China
| | - Xiaojin Wei
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China
| | - Wenjuan Zhang
- Department of the Laboratory, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruixuan Wang
- Bourns Engineering, The University of California, Riverside, CA 92521, USA
| | - Yaping Wang
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China.
| | - Lin Yang
- Department of Anesthesiology, the Second Xiangya Hospital, Central South University, Changsha, China; Department of Pain Management, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China; Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, Hunan Province, China; Clinical Research Center for Pain Medicine in Hunan Province, Changsha, Hunan Province, China.
| |
Collapse
|
15
|
Liu N, Wu WL, Wan XR, Wang J, Huang JN, Jiang YY, Sheng YC, Wu JC, Liang ZQ, Qin ZH, Wang Y. Regulation of FSP1 myristoylation by NADPH: A novel mechanism for ferroptosis inhibition. Redox Biol 2024; 73:103176. [PMID: 38705094 PMCID: PMC11074979 DOI: 10.1016/j.redox.2024.103176] [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: 04/16/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
Excitotoxicity is a prevalent pathological event in neurodegenerative diseases. The involvement of ferroptosis in the pathogenesis of excitotoxicity remains elusive. Transcriptome analysis has revealed that cytoplasmic reduced nicotinamide adenine dinucleotide phosphate (NADPH) levels are associated with susceptibility to ferroptosis-inducing compounds. Here we show that exogenous NADPH, besides being reductant, interacts with N-myristoyltransferase 2 (NMT2) and upregulates the N-myristoylated ferroptosis suppressor protein 1 (FSP1). NADPH increases membrane-localized FSP1 and strengthens resistance to ferroptosis. Arg-291 of NMT2 is critical for the NADPH-NMT2-FSP1 axis-mediated suppression of ferroptosis. This study suggests that NMT2 plays a pivotal role by bridging NADPH levels and neuronal susceptibility to ferroptosis. We propose a mechanism by which the NADPH regulates N-myristoylation, which has important implications for ferroptosis and disease treatment.
Collapse
Affiliation(s)
- Na Liu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Wei-Long Wu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Xiao-Rui Wan
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jing Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jia-Ni Huang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yi-Yue Jiang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yi-Chao Sheng
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Jun-Chao Wu
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Zhong-Qin Liang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Zheng-Hong Qin
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Yan Wang
- Department of Pharmacology College of Pharmaceutical Sciences, Suzhou Key Laboratory of Aging and Nervous Diseases, and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
16
|
Chen H, Wang X, Chang Z, Zhang J, Xie D. Evidence for genetic causality between iron homeostasis and Parkinson's disease: A two-sample Mendelian randomization study. J Trace Elem Med Biol 2024; 84:127430. [PMID: 38484633 DOI: 10.1016/j.jtemb.2024.127430] [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: 02/01/2024] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 05/27/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a degenerative disease of the central nervous system, and its specific etiology is still unclear. At present, it is believed that the main pathological basis is the reduction of dopamine concentration in the brain striatum. Although many previous studies have believed that iron as an important nutrient element participates in the occurrence and development of PD, whether there is a causal correlation between total iron binding capacity(TIBC), transferring saturation(TSAT), ferritin and serum iron in iron homeostasis indicators and PD, there has been a lack of effective genetic evidence. METHODS We used Mendelian randomization (MR) as an analytical method to effectively evaluate the genetic association between exposure and outcome, based on the largest genome-wide association study (GWAS) data to date. By using randomly assigned genetic instrumental variables (SNPs, Single Nucleotide Polymorphisms) that are not affected by any causal relationship, we effectively evaluated the causal relationship between iron homeostasis indicators and PD while controlling for confounding factors. RESULTS By coordinated analysis of 86 SNPs associated with iron homeostasis markers and 12,858,066 SNPs associated with PD, a total of 56 SNPs were finally screened for genome-wide significance of iron homeostasis associated with PD. The results of inverse variance weighting(IVW) analysis suggested that iron( β = - 0.524; 95%cl=-0.046 to -0.002; P=0.032) was considered to have a genetic causal relationship with PD. Cochran's Q, Egger intercept and MR-PRESSO global tests did not detect the existence of heterogeneity and pleiotropy (P>0.05). Mr Steiger directionality test further confirmed our estimation of the potential causal direction of iron and PD (P=0.001). In addition, TIBC (β=-0.142; 95%Cl=-0.197-0.481; P=0.414), TSAT (β=-0.316; 95%Cl=-0.861-0.229; P=0.255), and ferritin (β=-0.387; 95%Cl=-1.179-0.405; P=0.338) did not have genetic causal relationships with PD, and the results were not heterogeneous and pleiotropic (P>0.05). In addition, TIBC (β=-0.142; 95%Cl=-0.197-0.481; P=0.414), TSAT (β=-0.316; 95%Cl=-0.861-0.229; P=0.255), and ferritin (β=-0.101; 95%Cl=--0.987 to -0.405; P=0.823) did not have genetic causal relationships with PD, and the results were not heterogeneous and pleiotropic (P>0.05). TIBC (P=0.008), TSAT (P=0.000) and ferritin (P=0.013) were all consistent with the estimation of MR Steiger directivity test. CONCLUSION Our study found that among the four iron homeostasis markers, there was a genetic causal association between serum iron and PD, and the serum iron level was negatively correlated with the risk of PD. In addition, TIBC, TSAT, ferritin had no genetic causal relationship with PD.
Collapse
Affiliation(s)
- Hong Chen
- Anhui University of Chinese Medicine, Hefei 230038, China
| | - Xie Wang
- Anhui University of Chinese Medicine, Hefei 230038, China
| | - Ze Chang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100089, China
| | - Juan Zhang
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China.
| | - Daojun Xie
- Department of Neurology, the First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei 230031, China
| |
Collapse
|
17
|
Yu X, Wang S, Ji Z, Meng J, Mou Y, Wu X, Yang X, Xiong P, Li M, Guo Y. Ferroptosis: An important mechanism of disease mediated by the gut-liver-brain axis. Life Sci 2024; 347:122650. [PMID: 38631669 DOI: 10.1016/j.lfs.2024.122650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/27/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024]
Abstract
AIMS As a unique iron-dependent non-apoptotic cell death, Ferroptosis is involved in the pathogenesis and development of many human diseases and has become a research hotspot in recent years. However, the regulatory role of ferroptosis in the gut-liver-brain axis has not been elucidated. This paper summarizes the regulatory role of ferroptosis and provides theoretical basis for related research. MATERIALS AND METHODS We searched PubMed, CNKI and Wed of Science databases on ferroptosis mediated gut-liver-brain axis diseases, summarized the regulatory role of ferroptosis on organ axis, and explained the adverse effects of related regulatory effects on various diseases. KEY FINDINGS According to our summary, the main way in which ferroptosis mediates the gut-liver-brain axis is oxidative stress, and the key cross-talk of ferroptosis affecting signaling pathway network is Nrf2/HO-1. However, there were no specific marker between different organ axes mediate by ferroptosis. SIGNIFICANCE Our study illustrates the main ways and key cross-talk of ferroptosis mediating the gut-liver-brain axis, providing a basis for future research.
Collapse
Affiliation(s)
- Xinxin Yu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Shihao Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Zhongjie Ji
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Jiaqi Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Yunying Mou
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Xinyi Wu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Xu Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Panyang Xiong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China
| | - Mingxia Li
- Nursing School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, Shandong, China
| | - Yinghui Guo
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, Shandong, China.
| |
Collapse
|
18
|
Dong X, Liu B, Huang W, Chen H, Zhang Y, Yao Z, Shmuel A, Yang A, Dai Z, Ma G, Shu N. Disrupted cerebellar structural connectome in spinocerebellar ataxia type 3 and its association with transcriptional profiles. Cereb Cortex 2024; 34:bhae238. [PMID: 38850215 DOI: 10.1093/cercor/bhae238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is primarily characterized by progressive cerebellar degeneration, including gray matter atrophy and disrupted anatomical and functional connectivity. The alterations of cerebellar white matter structural network in SCA3 and the underlying neurobiological mechanism remain unknown. Using a cohort of 20 patients with SCA3 and 20 healthy controls, we constructed cerebellar structural networks from diffusion MRI and investigated alterations of topological organization. Then, we mapped the alterations with transcriptome data from the Allen Human Brain Atlas to identify possible biological mechanisms for regional selective vulnerability to white matter damage. Compared with healthy controls, SCA3 patients exhibited reduced global and nodal efficiency, along with a widespread decrease in edge strength, particularly affecting edges connected to hub regions. The strength of inter-module connections was lower in SCA3 group and negatively correlated with the Scale for the Assessment and Rating of Ataxia score, International Cooperative Ataxia Rating Scale score, and cytosine-adenine-guanine repeat number. Moreover, the transcriptome-connectome association study identified the expression of genes involved in synapse-related and metabolic biological processes. These findings suggest a mechanism of white matter vulnerability and a potential image biomarker for the disease severity, providing insights into neurodegeneration and pathogenesis in this disease.
Collapse
Affiliation(s)
- Xinyi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- BABRI Centre, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
| | - Bing Liu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jing-wu Road, Jinan, Shandong Province, 250021, China
| | - Weijie Huang
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- BABRI Centre, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- Department of Systems Science, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
| | - Haojie Chen
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- BABRI Centre, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
| | - Yunhao Zhang
- State Key Laboratory of Multimodal Artificial Intelligence Systems, Institute of Automation, Chinese Academy of Sciences, 95 Zhongguancun East Road, Haidian District, Beijing 100190, China
| | - Zeshan Yao
- Institute of Biomedical Engineering, Jingjinji National Center of Technology Innovation, Building 9, No. 6 Dongsheng Science Park North Street, Haidian District, Beijing 100094, China
| | - Amir Shmuel
- McConnell Brain Imaging Centre, Montreal Neurological Institute, 3801 University, Room NW261, Montreal, QC, Canada H3A 2B4
- Departments of Neurology and Neurosurgery, Physiology, and Biomedical Engineering, 3801 University, Room NW261, Montreal, QC, Canada H3A 2B4
| | - Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing 100029, China
| | - Zhengjia Dai
- Department of Psychology, Sun Yat-sen University, 132 Outer Ring East Road, Panyu District, Guangzhou, Guangdong Province, 510275, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, No. 2 East Yinghua Road, Chaoyang District, Beijing 100029, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- BABRI Centre, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, 19 Xiejiekouwai Street, Haidian District, Beijing 100875, China
| |
Collapse
|
19
|
Morys F, Tremblay C, Rahayel S, Hansen JY, Dai A, Misic B, Dagher A. Neural correlates of obesity across the lifespan. Commun Biol 2024; 7:656. [PMID: 38806652 PMCID: PMC11133431 DOI: 10.1038/s42003-024-06361-9] [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: 12/04/2023] [Accepted: 05/20/2024] [Indexed: 05/30/2024] Open
Abstract
Associations between brain and obesity are bidirectional: changes in brain structure and function underpin over-eating, while chronic adiposity leads to brain atrophy. Investigating brain-obesity interactions across the lifespan can help better understand these relationships. This study explores the interaction between obesity and cortical morphometry in children, young adults, adults, and older adults. We also investigate the genetic, neurochemical, and cognitive correlates of the brain-obesity associations. Our findings reveal a pattern of lower cortical thickness in fronto-temporal brain regions associated with obesity across all age cohorts and varying age-dependent patterns in the remaining brain regions. In adults and older adults, obesity correlates with neurochemical changes and expression of inflammatory and mitochondrial genes. In children and older adults, adiposity is associated with modifications in brain regions involved in emotional and attentional processes. Thus, obesity might originate from cognitive changes during early adolescence, leading to neurodegeneration in later life through mitochondrial and inflammatory mechanisms.
Collapse
Affiliation(s)
- Filip Morys
- Montreal Neurological Institute, McGill University, H3A 2B4, Montreal, QC, Canada.
| | - Christina Tremblay
- Montreal Neurological Institute, McGill University, H3A 2B4, Montreal, QC, Canada
| | - Shady Rahayel
- Department of Medicine and Medical Specialties, University of Montreal, Montreal, QC, Canada
- Center for Advanced Research in Sleep Medicine, Hopital du Sacre-Coeur de Montreal, Montreal, QC, Canada
| | - Justine Y Hansen
- Montreal Neurological Institute, McGill University, H3A 2B4, Montreal, QC, Canada
| | - Alyssa Dai
- Montreal Neurological Institute, McGill University, H3A 2B4, Montreal, QC, Canada
| | - Bratislav Misic
- Montreal Neurological Institute, McGill University, H3A 2B4, Montreal, QC, Canada
| | - Alain Dagher
- Montreal Neurological Institute, McGill University, H3A 2B4, Montreal, QC, Canada
| |
Collapse
|
20
|
Maniscalchi A, Benzi Juncos ON, Conde MA, Funk MI, Fermento ME, Facchinetti MM, Curino AC, Uranga RM, Alza NP, Salvador GA. New insights on neurodegeneration triggered by iron accumulation: Intersections with neutral lipid metabolism, ferroptosis, and motor impairment. Redox Biol 2024; 71:103074. [PMID: 38367511 PMCID: PMC10879836 DOI: 10.1016/j.redox.2024.103074] [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: 10/30/2023] [Revised: 12/28/2023] [Accepted: 02/03/2024] [Indexed: 02/19/2024] Open
Abstract
Brain iron accumulation constitutes a pathognomonic indicator in several neurodegenerative disorders. Metal accumulation associated with dopaminergic neuronal death has been documented in Parkinson's disease. Through the use of in vivo and in vitro models, we demonstrated that lipid dysregulation manifests as a neuronal and glial response during iron overload. In this study, we show that cholesterol content and triacylglycerol (TAG) hydrolysis were strongly elevated in mice midbrain. Lipid cacostasis was concomitant with the loss of dopaminergic neurons, astrogliosis and elevated expression of α-synuclein. Exacerbated lipid peroxidation and markers of ferroptosis were evident in the midbrain from mice challenged with iron overload. An imbalance in the activity of lipolytic and acylation enzymes was identified, favoring neutral lipid hydrolysis, and consequently reducing TAG and cholesteryl ester levels. Notably, these observed alterations were accompanied by motor impairment in iron-treated mice. In addition, neuronal and glial cultures along with their secretomes were used to gain further insight into the mechanism underlying TAG hydrolysis and cholesterol accumulation as cellular responses to iron accumulation. We demonstrated that TAG hydrolysis in neurons is triggered by astrocyte secretomes. Moreover, we found that the ferroptosis inhibitor, ferrostatin-1, effectively prevents cholesterol accumulation both in neurons and astrocytes. Taken together, these results indicate that lipid disturbances occur in iron-overloaded mice as a consequence of iron-induced oxidative stress and depend on neuron-glia crosstalk. Our findings suggest that developing therapies aimed at restoring lipid homeostasis may lead to specific treatment for neurodegeneration associated with ferroptosis and brain iron accumulation.
Collapse
Affiliation(s)
- Athina Maniscalchi
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina
| | - Oriana N Benzi Juncos
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Melisa A Conde
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Melania I Funk
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina
| | - María E Fermento
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - María M Facchinetti
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Alejandro C Curino
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Romina M Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina
| | - Natalia P Alza
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Química - UNS, Bahía Blanca, Argentina
| | - Gabriela A Salvador
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Camino La Carrindanga Km7 B8000FWB, Bahía Blanca, Argentina; Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Bahía Blanca, Argentina.
| |
Collapse
|
21
|
Tyczyńska M, Gędek M, Brachet A, Stręk W, Flieger J, Teresiński G, Baj J. Trace Elements in Alzheimer's Disease and Dementia: The Current State of Knowledge. J Clin Med 2024; 13:2381. [PMID: 38673657 PMCID: PMC11050856 DOI: 10.3390/jcm13082381] [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: 03/20/2024] [Revised: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Changes in trace element concentrations are being wildly considered when it comes to neurodegenerative disorders, such as Alzheimer's disease and Parkinson's disease. This study aims to present the role that trace elements play in the central nervous system. Moreover, we reviewed the mechanisms involved in their neurotoxicity. Low zinc concentrations, as well as high levels of copper, manganese, and iron, activate the signalling pathways of the inflammatory, oxidative and nitrosative stress response. Neurodegeneration occurs due to the association between metals and proteins, which is then followed by aggregate formation, mitochondrial disorder, and, ultimately, cell death. In Alzheimer's disease, low Zn levels suppress the neurotoxicity induced by β-amyloid through the selective precipitation of aggregation intermediates. High concentrations of copper, iron and manganese cause the aggregation of intracellular α-synuclein, which results in synaptic dysfunction and axonal transport disruption. Parkinson's disease is caused by the accumulation of Fe in the midbrain dopaminergic nucleus, and the pathogenesis of multiple sclerosis derives from Zn deficiency, leading to an imbalance between T cell functions. Aluminium disturbs the homeostasis of other metals through a rise in the production of oxygen reactive forms, which then leads to cellular death. Selenium, in association with iron, plays a distinct role in the process of ferroptosis. Outlining the influence that metals have on oxidoreduction processes is crucial to recognising the pathophysiology of neurodegenerative diseases and may provide possible new methods for both their avoidance and therapy.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (M.T.); (W.S.)
| | - Marta Gędek
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (M.G.); (A.B.); (G.T.)
| | - Adam Brachet
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (M.G.); (A.B.); (G.T.)
| | - Wojciech Stręk
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (M.T.); (W.S.)
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland;
| | - Grzegorz Teresiński
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland; (M.G.); (A.B.); (G.T.)
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (M.T.); (W.S.)
| |
Collapse
|
22
|
Song N, Mei S, Wang X, Hu G, Lu M. Focusing on mitochondria in the brain: from biology to therapeutics. Transl Neurodegener 2024; 13:23. [PMID: 38632601 PMCID: PMC11022390 DOI: 10.1186/s40035-024-00409-w] [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: 12/10/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Mitochondria have multiple functions such as supplying energy, regulating the redox status, and producing proteins encoded by an independent genome. They are closely related to the physiology and pathology of many organs and tissues, among which the brain is particularly prominent. The brain demands 20% of the resting metabolic rate and holds highly active mitochondrial activities. Considerable research shows that mitochondria are closely related to brain function, while mitochondrial defects induce or exacerbate pathology in the brain. In this review, we provide comprehensive research advances of mitochondrial biology involved in brain functions, as well as the mitochondria-dependent cellular events in brain physiology and pathology. Furthermore, various perspectives are explored to better identify the mitochondrial roles in neurological diseases and the neurophenotypes of mitochondrial diseases. Finally, mitochondrial therapies are discussed. Mitochondrial-targeting therapeutics are showing great potentials in the treatment of brain diseases.
Collapse
Affiliation(s)
- Nanshan Song
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shuyuan Mei
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiangxu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
- Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
23
|
Chen H, Yang A, Huang W, Du L, Liu B, Lv K, Luan J, Hu P, Shmuel A, Shu N, Ma G. Associations of quantitative susceptibility mapping with cortical atrophy and brain connectome in Alzheimer's disease: A multi-parametric study. Neuroimage 2024; 290:120555. [PMID: 38447683 DOI: 10.1016/j.neuroimage.2024.120555] [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: 11/11/2023] [Revised: 01/07/2024] [Accepted: 02/24/2024] [Indexed: 03/08/2024] Open
Abstract
Aberrant susceptibility due to iron level abnormality and brain network disconnections are observed in Alzheimer's disease (AD), with disrupted iron homeostasis hypothesized to be linked to AD pathology and neuronal loss. However, whether associations exist between abnormal quantitative susceptibility mapping (QSM), brain atrophy, and altered brain connectome in AD remains unclear. Based on multi-parametric brain imaging data from 30 AD patients and 26 healthy controls enrolled at the China-Japan Friendship Hospital, we investigated the abnormality of the QSM signal and volumetric measure across 246 brain regions in AD patients. The structural and functional connectomes were constructed based on diffusion MRI tractography and functional connectivity, respectively. The network topology was quantified using graph theory analyses. We identified seven brain regions with both reduced cortical thickness and abnormal QSM (p < 0.05) in AD, including the right superior frontal gyrus, left superior temporal gyrus, right fusiform gyrus, left superior parietal lobule, right superior parietal lobule, left inferior parietal lobule, and left precuneus. Correlations between cortical thickness and network topology computed across patients in the AD group resulted in statistically significant correlations in five of these regions, with higher correlations in functional compared to structural topology. We computed the correlation between network topological metrics, QSM value and cortical thickness across regions at both individual and group-averaged levels, resulting in a measure we call spatial correlations. We found a decrease in the spatial correlation of QSM and the global efficiency of the structural network in AD patients at the individual level. These findings may provide insights into the complex relationships among QSM, brain atrophy, and brain connectome in AD.
Collapse
Affiliation(s)
- Haojie Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Weijie Huang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Lei Du
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bing Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kuan Lv
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jixin Luan
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pianpian Hu
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Amir Shmuel
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Departments of Neurology and Neurosurgery, Physiology, and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China.
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Lee S, Kovacs GG. The Irony of Iron: The Element with Diverse Influence on Neurodegenerative Diseases. Int J Mol Sci 2024; 25:4269. [PMID: 38673855 PMCID: PMC11049980 DOI: 10.3390/ijms25084269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Iron accumulation in the brain is a common feature of many neurodegenerative diseases. Its involvement spans across the main proteinopathies involving tau, amyloid-beta, alpha-synuclein, and TDP-43. Accumulating evidence supports the contribution of iron in disease pathologies, but the delineation of its pathogenic role is yet challenged by the complex involvement of iron in multiple neurotoxicity mechanisms and evidence supporting a reciprocal influence between accumulation of iron and protein pathology. Here, we review the major proteinopathy-specific observations supporting four distinct hypotheses: (1) iron deposition is a consequence of protein pathology; (2) iron promotes protein pathology; (3) iron protects from or hinders protein pathology; and (4) deposition of iron and protein pathology contribute parallelly to pathogenesis. Iron is an essential element for physiological brain function, requiring a fine balance of its levels. Understanding of disease-related iron accumulation at a more intricate and systemic level is critical for advancements in iron chelation therapies.
Collapse
Affiliation(s)
- Seojin Lee
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada;
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gabor G. Kovacs
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 0S8, Canada;
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Edmond J. Safra Program in Parkinson’s Disease, Rossy Program for PSP Research and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
25
|
Varga Z, Keller J, Robinson SD, Serranova T, Nepozitek J, Zogala D, Trnka J, Ruzicka E, Sonka K, Dusek P. Whole brain pattern of iron accumulation in REM sleep behavior disorder. Hum Brain Mapp 2024; 45:e26675. [PMID: 38590155 PMCID: PMC11002348 DOI: 10.1002/hbm.26675] [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: 10/17/2023] [Revised: 03/10/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Isolated REM sleep behavior disorder (iRBD) is an early stage of synucleinopathy with most patients progressing to Parkinson's disease (PD) or related conditions. Quantitative susceptibility mapping (QSM) in PD has identified pathological iron accumulation in the substantia nigra (SN) and variably also in basal ganglia and cortex. Analyzing whole-brain QSM across iRBD, PD, and healthy controls (HC) may help to ascertain the extent of neurodegeneration in prodromal synucleinopathy. 70 de novo PD patients, 70 iRBD patients, and 60 HCs underwent 3 T MRI. T1 and susceptibility-weighted images were acquired and processed to space standardized QSM. Voxel-based analyses of grey matter magnetic susceptibility differences comparing all groups were performed on the whole brain and upper brainstem levels with the statistical threshold set at family-wise error-corrected p-values <.05. Whole-brain analysis showed increased susceptibility in the bilateral fronto-parietal cortex of iRBD patients compared to both PD and HC. This was not associated with cortical thinning according to the cortical thickness analysis. Compared to iRBD, PD patients had increased susceptibility in the left amygdala and hippocampal region. Upper brainstem analysis revealed increased susceptibility within the bilateral SN for both PD and iRBD compared to HC; changes were located predominantly in nigrosome 1 in the former and nigrosome 2 in the latter group. In the iRBD group, abnormal dopamine transporter SPECT was associated with increased susceptibility in nigrosome 1. iRBD patients display greater fronto-parietal cortex involvement than incidental early-stage PD cohort indicating more widespread subclinical neuropathology. Dopaminergic degeneration in the substantia nigra is paralleled by susceptibility increase, mainly in nigrosome 1.
Collapse
Affiliation(s)
- Zsoka Varga
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - Jiri Keller
- Radiodiagnostic DepartmentNa Homolce HospitalPragueCzech Republic
| | - Simon Daniel Robinson
- Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaAustria
| | - Tereza Serranova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - Jiri Nepozitek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - David Zogala
- Department of Nuclear Medicine, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - Jiri Trnka
- Department of Nuclear Medicine, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - Evzen Ruzicka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| | - Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
- Department of Radiology, First Faculty of MedicineCharles University and General University Hospital in PragueCzech Republic
| |
Collapse
|
26
|
Guan X, Lancione M, Ayton S, Dusek P, Langkammer C, Zhang M. Neuroimaging of Parkinson's disease by quantitative susceptibility mapping. Neuroimage 2024; 289:120547. [PMID: 38373677 DOI: 10.1016/j.neuroimage.2024.120547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/21/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease, and apart from a few rare genetic causes, its pathogenesis remains largely unclear. Recent scientific interest has been captured by the involvement of iron biochemistry and the disruption of iron homeostasis, particularly within the brain regions specifically affected in PD. The advent of Quantitative Susceptibility Mapping (QSM) has enabled non-invasive quantification of brain iron in vivo by MRI, which has contributed to the understanding of iron-associated pathogenesis and has the potential for the development of iron-based biomarkers in PD. This review elucidates the biochemical underpinnings of brain iron accumulation, details advancements in iron-sensitive MRI technologies, and discusses the role of QSM as a biomarker of iron deposition in PD. Despite considerable progress, several challenges impede its clinical application after a decade of QSM studies. The initiation of multi-site research is warranted for developing robust, interpretable, and disease-specific biomarkers for monitoring PD disease progression.
Collapse
Affiliation(s)
- Xiaojun Guan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Joint Laboratory of Clinical Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou 31009, China
| | - Marta Lancione
- Laboratory of Medical Physics and Magnetic Resonance, IRCCS Stella Maris Foundation, Pisa, Italy
| | - Scott Ayton
- Florey Institute, The University of Melbourne, Australia
| | - Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia; Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Auenbruggerplatz 22, Prague 8036, Czechia
| | | | - Minming Zhang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Joint Laboratory of Clinical Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou 31009, China.
| |
Collapse
|
27
|
Thomas GE, Hannaway N, Zarkali A, Shmueli K, Weil RS. Longitudinal Associations of Magnetic Susceptibility with Clinical Severity in Parkinson's Disease. Mov Disord 2024; 39:546-559. [PMID: 38173297 PMCID: PMC11141787 DOI: 10.1002/mds.29702] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/29/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Dementia is common in Parkinson's disease (PD), but there is wide variation in its timing. A critical gap in PD research is the lack of quantifiable markers of progression, and methods to identify early stages of dementia. Atrophy-based magnetic resonance imaging (MRI) has limited sensitivity in detecting or tracking changes relating to PD dementia, but quantitative susceptibility mapping (QSM), sensitive to brain tissue iron, shows potential for these purposes. OBJECTIVE The objective of the paper is to study, for the first time, the longitudinal relationship between cognition and QSM in PD in detail. METHODS We present a longitudinal study of clinical severity in PD using QSM, including 59 PD patients (without dementia at study onset), and 22 controls over 3 years. RESULTS In PD, increased baseline susceptibility in the right temporal cortex, nucleus basalis of Meynert, and putamen was associated with greater cognitive severity after 3 years; and increased baseline susceptibility in basal ganglia, substantia nigra, red nucleus, insular cortex, and dentate nucleus was associated with greater motor severity after 3 years. Increased follow-up susceptibility in these regions was associated with increased follow-up cognitive and motor severity, with further involvement of hippocampus relating to cognitive severity. However, there were no consistent increases in susceptibility over 3 years. CONCLUSIONS Our study suggests that QSM may predict changes in cognitive severity many months prior to overt cognitive involvement in PD. However, we did not find robust longitudinal changes in QSM over the course of the study. Additional tissue metrics may be required together with QSM for it to monitor progression in clinical practice and therapeutic trials. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Naomi Hannaway
- Dementia Research CentreUCL Institute of NeurologyLondonUK
| | | | - Karin Shmueli
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Rimona S. Weil
- Dementia Research CentreUCL Institute of NeurologyLondonUK
- Wellcome Centre for Human NeuroimagingUniversity College LondonLondonUK
- Movement Disorders ConsortiumUniversity College LondonLondonUK
| |
Collapse
|
28
|
Wang Y, Xiao Y, Xing Y, Yu M, Wang X, Ren J, Liu W, Zhong Y. Morphometric similarity differences in drug-naive Parkinson's disease correlate with transcriptomic signatures. CNS Neurosci Ther 2024; 30:e14680. [PMID: 38529533 PMCID: PMC10964038 DOI: 10.1111/cns.14680] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Differences in cortical morphology have been reported in individuals with Parkinson's disease (PD). However, the pathophysiological mechanism of transcriptomic vulnerability in local brain regions remains unclear. OBJECTIVE This study aimed to characterize the morphometric changes of brain regions in early drug-naive PD patients and uncover the brain-wide gene expression correlates. METHODS The morphometric similarity (MS) network analysis was used to quantify the interregional structural similarity from multiple magnetic resonance imaging anatomical indices measured in each brain region of 170 early drug-naive PD patients and 123 controls. Then, we applied partial least squares regression to determine the relationship between regional changes in MS and spatial transcriptional signatures from the Allen Human Brain Atlas dataset, and identified the specific genes related to MS differences in PD. We further investigated the biological processes by which the PD-related genes were enriched and the cellular characterization of these genes. RESULTS Our results showed that MS was mainly decreased in cingulate, frontal, and temporal cortical areas and increased in parietal and occipital cortical areas in early drug-naive PD patients. In addition, genes whose expression patterns were associated with regional MS changes in PD were involved in astrocytes, excitatory, and inhibitory neurons and were functionally enriched in neuron-specific biological processes related to trans-synaptic signaling and nervous system development. CONCLUSIONS These findings advance our understanding of the microscale genetic and cellular mechanisms driving macroscale morphological abnormalities in early drug-naive PD patients and provide potential targets for future therapeutic trials.
Collapse
Affiliation(s)
- Yajie Wang
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
- Department of NeurologyThe First People's Hospital of YanchengYanchengChina
| | - Yiwen Xiao
- School of PsychologyNanjing Normal UniversityNanjingChina
| | - Yi Xing
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Miao Yu
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Xiao Wang
- Department of RadiologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Jingru Ren
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Weiguo Liu
- Department of NeurologyThe Affiliated Brain Hospital of Nanjing Medical UniversityNanjingChina
| | - Yuan Zhong
- School of PsychologyNanjing Normal UniversityNanjingChina
| |
Collapse
|
29
|
Zeng W, Cai J, Zhang L, Peng Q. Iron Deposition in Parkinson's Disease: A Mini-Review. Cell Mol Neurobiol 2024; 44:26. [PMID: 38393383 PMCID: PMC10891198 DOI: 10.1007/s10571-024-01459-4] [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: 09/21/2023] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Iron deposition is crucial pathological changes observed in patients with Parkinson's disease (PD). Recently, scientists have actively explored therapeutic approaches targeting iron deposition in PD. However, several clinical studies have failed to yield consistent results. In this review, we provide an overview of iron deposition in PD, from both basic research and clinical perspectives. PD patients exhibit abnormalities in various iron metabolism-related proteins, leading to disruptions in iron distribution, transport, storage, and circulation, ultimately resulting in iron deposition. Excess iron can induce oxidative stress and iron-related cell death, and exacerbate mitochondrial dysfunction, contributing to the progression of PD pathology. Magnetic resonance imaging studies have indicated that the characteristics of iron deposition in the brains of PD patients vary. Iron deposition correlates with the clinical symptoms of PD, and patients with different disease courses and clinical presentations display distinct patterns of iron deposition. These iron deposition patterns may contribute to PD diagnosis. Iron deposition is a promising target for PD treatment. However, further research is required to elucidate the underlying mechanisms and their impacts on PD.
Collapse
Affiliation(s)
- Weiqi Zeng
- Department of Neurology, The First People's Hospital of Foshan, Foshan, China
| | - Jin Cai
- Department of Cardiology, The Second Hospital of Zhangzhou, Zhangzhou, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qiwei Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
30
|
Domínguez D JF, Stewart A, Burmester A, Akhlaghi H, O'Brien K, Bollmann S, Caeyenberghs K. Improving quantitative susceptibility mapping for the identification of traumatic brain injury neurodegeneration at the individual level. Z Med Phys 2024:S0939-3889(24)00001-1. [PMID: 38336583 DOI: 10.1016/j.zemedi.2024.01.001] [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: 06/02/2023] [Revised: 12/19/2023] [Accepted: 01/07/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Emerging evidence suggests that traumatic brain injury (TBI) is a major risk factor for developing neurodegenerative disease later in life. Quantitative susceptibility mapping (QSM) has been used by an increasing number of studies in investigations of pathophysiological changes in TBI. However, generating artefact-free quantitative susceptibility maps in brains with large focal lesions, as in the case of moderate-to-severe TBI (ms-TBI), is particularly challenging. To address this issue, we utilized a novel two-pass masking technique and reconstruction procedure (two-pass QSM) to generate quantitative susceptibility maps (QSMxT; Stewart et al., 2022, Magn Reson Med.) in combination with the recently developed virtual brain grafting (VBG) procedure for brain repair (Radwan et al., 2021, NeuroImage) to improve automated delineation of brain areas. We used QSMxT and VBG to generate personalised QSM profiles of individual patients with reference to a sample of healthy controls. METHODS Chronic ms-TBI patients (N = 8) and healthy controls (N = 12) underwent (multi-echo) GRE, and anatomical MRI (MPRAGE) on a 3T Siemens PRISMA scanner. We reconstructed the magnetic susceptibility maps using two-pass QSM from QSMxT. We then extracted values of magnetic susceptibility in grey matter (GM) regions (following brain repair via VBG) across the whole brain and determined if they deviate from a reference healthy control group [Z-score < -3.43 or > 3.43, relative to the control mean], with the aim of obtaining personalised QSM profiles. RESULTS Using two-pass QSM, we achieved susceptibility maps with a substantial increase in quality and reduction in artefacts irrespective of the presence of large focal lesions, compared to single-pass QSM. In addition, VBG minimised the loss of GM regions and exclusion of patients due to failures in the region delineation step. Our findings revealed deviations in magnetic susceptibility measures from the HC group that differed across individual TBI patients. These changes included both increases and decreases in magnetic susceptibility values in multiple GM regions across the brain. CONCLUSIONS We illustrate how to obtain magnetic susceptibility values at the individual level and to build personalised QSM profiles in ms-TBI patients. Our approach opens the door for QSM investigations in more severely injured patients. Such profiles are also critical to overcome the inherent heterogeneity of clinical populations, such as ms-TBI, and to characterize the underlying mechanisms of neurodegeneration at the individual level more precisely. Moreover, this new personalised QSM profiling could in the future assist clinicians in assessing recovery and formulating a neuroscience-guided integrative rehabilitation program tailored to individual TBI patients.
Collapse
Affiliation(s)
- Juan F Domínguez D
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia.
| | - Ashley Stewart
- School of Information Technology and Electrical Engineering, Faculty of Engineering, Architecture, and Information Technology, The University of Queensland, Brisbane, Australia
| | - Alex Burmester
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia
| | - Hamed Akhlaghi
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia; Department of Emergency Medicine, St. Vincent's Hospital, Melbourne, Australia
| | - Kieran O'Brien
- Siemens Healthcare Pty Ltd, Brisbane, Queensland, Australia
| | - Steffen Bollmann
- School of Information Technology and Electrical Engineering, Faculty of Engineering, Architecture, and Information Technology, The University of Queensland, Brisbane, Australia; Centre for Advanced Imaging, The University of Queensland, Brisbane, Australia
| | - Karen Caeyenberghs
- Cognitive Neuroscience Unit, School of Psychology, Deakin University, Geelong, Australia
| |
Collapse
|
31
|
Wu C, Wu H, Zhou C, Guan X, Guo T, Wu J, Chen J, Wen J, Qin J, Tan S, Duanmu X, Yuan W, Zheng Q, Zhang B, Xu X, Zhang M. Neurovascular coupling alteration in drug-naïve Parkinson's disease: The underlying molecular mechanisms and levodopa's restoration effects. Neurobiol Dis 2024; 191:106406. [PMID: 38199273 DOI: 10.1016/j.nbd.2024.106406] [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: 11/12/2023] [Revised: 12/25/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) patients exhibit an imbalance between neuronal activity and perfusion, referred to as abnormal neurovascular coupling (NVC). Nevertheless, the underlying molecular mechanism and how levodopa, the standard treatment in PD, regulates NVC is largely unknown. MATERIAL AND METHODS A total of 52 drug-naïve PD patients and 49 normal controls (NCs) were enrolled. NVC was characterized in vivo by relating cerebral blood flow (CBF) and amplitude of low-frequency fluctuations (ALFF). Motor assessments and MRI scanning were conducted on drug-naïve patients before and after levodopa therapy (OFF/ON state). Regional NVC differences between patients and NCs were identified, followed by an assessment of the associated receptors/transporters. The influence of levodopa on NVC, CBF, and ALFF within these abnormal regions was analyzed. RESULTS Compared to NCs, OFF-state patients showed NVC dysfunction in significantly lower NVC in left precentral, postcentral, superior parietal cortex, and precuneus, along with higher NVC in left anterior cingulate cortex, right olfactory cortex, thalamus, caudate, and putamen (P-value <0.0006). The distribution of NVC differences correlated with the density of dopaminergic, serotonin, MU-opioid, and cholinergic receptors/transporters. Additionally, levodopa ameliorated abnormal NVC in most of these regions, where there were primarily ALFF changes with limited CBF modifications. CONCLUSION Patients exhibited NVC dysfunction primarily in the striato-thalamo-cortical circuit and motor control regions, which could be driven by dopaminergic and nondopaminergic systems, and levodopa therapy mainly restored abnormal NVC by modulating neuronal activity.
Collapse
Affiliation(s)
- Chenqing Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoting Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Zhou
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Guo
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Wu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingwen Chen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Wen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmei Qin
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sijia Tan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojie Duanmu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijin Yuan
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianshi Zheng
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Baorong Zhang
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Xu
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
32
|
Wang J, Zhao J, Zhao K, Wu S, Chen X, Hu W. The Role of Calcium and Iron Homeostasis in Parkinson's Disease. Brain Sci 2024; 14:88. [PMID: 38248303 PMCID: PMC10813814 DOI: 10.3390/brainsci14010088] [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: 12/11/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Calcium and iron are essential elements that regulate many important processes of eukaryotic cells. Failure to maintain homeostasis of calcium and iron causes cell dysfunction or even death. PD (Parkinson's disease) is the second most common neurological disorder in humans, for which there are currently no viable treatment options or effective strategies to cure and delay progression. Pathological hallmarks of PD, such as dopaminergic neuronal death and intracellular α-synuclein deposition, are closely involved in perturbations of iron and calcium homeostasis and accumulation. Here, we summarize the mechanisms by which Ca2+ signaling influences or promotes PD progression and the main mechanisms involved in ferroptosis in Parkinson's disease. Understanding the mechanisms by which calcium and iron imbalances contribute to the progression of this disease is critical to developing effective treatments to combat this devastating neurological disorder.
Collapse
Affiliation(s)
- Ji Wang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China;
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; (J.Z.); (K.Z.); (S.W.)
| | - Jindong Zhao
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; (J.Z.); (K.Z.); (S.W.)
| | - Kunying Zhao
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; (J.Z.); (K.Z.); (S.W.)
| | - Shangpeng Wu
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; (J.Z.); (K.Z.); (S.W.)
| | - Xinglong Chen
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, China;
| | - Weiyan Hu
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China; (J.Z.); (K.Z.); (S.W.)
| |
Collapse
|
33
|
Fang YY, Teng MJ, Peng JC, Zheng XW, Mo YQ, Ho TT, Lin JJ, Luo JJ, Aschner M, Jiang YM. Combined exposure to manganese and iron decreases oxidative stress-induced nerve damage by increasing Nrf2/HO-1/NQO1 expression. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115853. [PMID: 38128313 DOI: 10.1016/j.ecoenv.2023.115853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Manganese (Mn) and iron (Fe) are essential trace elements for humans, yet excessive exposure to Mn or Fe can accumulate in the central nervous system (CNS) and cause neurotoxicity. The purpose of this study was to investigate the effects of Mn and Fe exposure, alone or in combination, on inducing oxidative stress-induced neurological damage in rat cortical and SH-SY5Y cells, and to determine whether combined exposure to these metals increases their individual toxicity. METHODS SH-SY5Y cells and male Sprague-Dawley rats were used to observe the effects of oxidative stress-induced neurological damage induced by exposure to manganese and iron alone or in combination. To detect the expression of anti-oxidative stress-related proteins, Nrf2, HO-1, and NQO1, and the apoptosis-related proteins, Bcl2 and Bax, and the neurological damage-related protein, α-syn. To detect reactive oxygen species generation and apoptosis. To detect the expression of the rat cortical protein Nrf2. To detect the production of proinflammatory cytokines. RESULTS We demonstrate that juvenile developmental exposure to Mn and Fe and their combination impairs cognitive performance in rats by inducing oxidative stress causing neurodegeneration in the cortex. Mn, Fe, and their combined exposure increased the expression of ROS, Bcl2, Bax, and α-syn, activated the inflammatory factors IL-6 and IL-12, inhibited the activities of SOD and GSH, and induced oxidative stress-induced neurodegeneration both in rats and SH-SY5Y cells. Combined Mn-Fe exposure attenuated the oxidative stress induced by Mn and Fe exposure alone by increasing the expression of antioxidant factors Nrf2, HO-1, and NQO1. CONCLUSION In both in vivo and in vitro studies, manganese and iron alone or in combination induced oxidative stress, leading to neuronal damage. In contrast, combined exposure to manganese and iron mitigated the oxidative stress induced by exposure to manganese and iron alone by increasing the expression of antioxidant factors. Therefore, studies to elucidate the main causes of toxicity and establish the molecular mechanisms of toxicity should help to develop more effective therapeutic modalities in the future.
Collapse
Affiliation(s)
- Yuan-Yuan Fang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Meng-Jun Teng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Jian-Chao Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Xiao-Wei Zheng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Ya-Qi Mo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Thanh-Tung Ho
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Jun-Jie Lin
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Jing-Jing Luo
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China
| | - Michael Aschner
- Department of Molecular Pharmacology at Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Yue-Ming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning 530021, China; Key Laboratory of Prevention and Control of Highly Prevalent Diseases in Guangxi Colleges and Universities, Medical University of Guangxi, Nanning 530021, China.
| |
Collapse
|
34
|
Lin S, Qi R, Lin X, Chen S, Zhang L, Qiu Y. Association Between MRI-Assessed Patterns of Connectome Gradient and Gene-Expression Profiles in Two Independent Patient Cohorts With Hepatitis B Virus-Related Cirrhosis. J Magn Reson Imaging 2023; 58:1863-1874. [PMID: 37022091 DOI: 10.1002/jmri.28732] [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: 01/09/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Patients with hepatitis B virus-related cirrhosis (HBV-RC) exhibit progressive neurologic dysfunction from primary sensorimotor to high-order cognition, as their disease advances. However, the exact neurobiologic mechanisms and the potential association with gene-expression profiles are not fully understood. PURPOSE To explore the hierarchical disorganization in the large-scale functional connectomes in HBV-RC patients and to investigate its potential underlying molecular basis. STUDY TYPE Prospective. POPULATION Fifty HBV-RC patients and 40 controls (Cohort 1) and 30 HBV-RC patients and 38 controls (Cohort 2). FIELD STRENGTH/SEQUENCE Gradient-echo echo-planar and fast field echo sequences at 3.0 T (Cohort 1) and 1.5 T (Cohort 2). ASSESSMENT Data were processed with Dpabi and the BrainSpace package. Gradient scores were evaluated from global to voxel level. Cognitive measurement and patients grouping were based on psychometric hepatic encephalopathy scores. The whole-brain microarray-based gene-expression data were obtained from the AIBS website. STATISTICAL TESTS One-way ANOVA, chi-square test, two-sample t-test, Kruskal-Wallis test, Spearman's correlation coefficient (r), the gaussian random field correction, false discovery rate (FDR) correction and the Bonferroni correction. Significance level: P < 0.05. RESULTS HBV-RC patients exhibited a robust and replicable connectome gradient dysfunction, which was significantly associated with the gene-expression profiles in both cohorts (r = 0.52 and r = 0.56, respectively). The most correlated genes were enriched in γ-aminobutyric acid (GABA) and GABA-related receptor genes (FDR q value <0.05). Moreover, the connectome gradient dysfunction at network level observed in HBV-RC patients correlated with their poor cognitive performance (Cohort 2: visual network, r = -0.56; subcortical network, r = 0.66; frontoparietal network, r = 0.51). DATA CONCLUSION HBV-RC patients had hierarchical disorganization in the large-scale functional connectomes, which may underly their cognitive impairment. In addition, we showed the potential molecular mechanism of the connectome gradient dysfunction, which suggested the importance of GABA and GABA-related receptor genes. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Shiwei Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Rongfeng Qi
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiaoshan Lin
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shengli Chen
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yingwei Qiu
- Department of Radiology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
35
|
Zang Z, Zhang X, Song T, Li J, Nie B, Mei S, Hu Z, Zhang Y, Lu J. Association between gene expression and functional-metabolic architecture in Parkinson's disease. Hum Brain Mapp 2023; 44:5387-5401. [PMID: 37605831 PMCID: PMC10543112 DOI: 10.1002/hbm.26443] [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: 04/20/2023] [Revised: 06/02/2023] [Accepted: 07/23/2023] [Indexed: 08/23/2023] Open
Abstract
Gene expression plays a critical role in the pathogenesis of Parkinson's disease (PD). How gene expression profiles are correlated with functional-metabolic architecture remains obscure. We enrolled 34 PD patients and 25 age-and-sex-matched healthy controls for simultaneous 18 F-FDG-PET/functional MRI scanning during resting state. We investigated the functional gradients and the ratio of standard uptake value. Principal component analysis was used to further combine the functional gradients and glucose metabolism into functional-metabolic architecture. Using partial least squares (PLS) regression, we introduced the transcriptomic data from the Allen Institute of Brain Sciences to identify gene expression patterns underlying the affected functional-metabolic architecture in PD. Between-group comparisons revealed significantly higher gradient variation in the visual, somatomotor, dorsal attention, frontoparietal, default mode, and subcortical network (pFDR < .048) in PD. Increased FDG-uptake was found in the somatomotor and ventral attention network while decreased FDG-uptake was found in the visual network (pFDR < .008). Spatial correlation analysis showed consistently affected patterns of functional gradients and metabolism (p = 2.47 × 10-8 ). PLS analysis and gene ontological analyses further revealed that genes were mainly enriched for metabolic, catabolic, cellular response to ions, and regulation of DNA transcription and RNA biosynthesis. In conclusion, our study provided genetic pathological mechanism to explain imaging-defined brain functional-metabolic architecture of PD.
Collapse
Affiliation(s)
- Zhenxiang Zang
- Department of Radiology and Nuclear Medicine, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain InformaticsBeijingChina
| | - Xiaolong Zhang
- Department of Physiology, College of Basic Medical SciencesArmy Medical UniversityChongqingChina
| | - Tianbin Song
- Department of Radiology and Nuclear Medicine, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain InformaticsBeijingChina
| | - Jiping Li
- Beijing Institute of Functional NeurosurgeryXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Binbin Nie
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy PhysicsChinese Academy of SciencesBeijingChina
| | - Shanshan Mei
- Department of NeurologyXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Zhi'an Hu
- Department of Physiology, College of Basic Medical SciencesArmy Medical UniversityChongqingChina
| | - Yuqing Zhang
- Beijing Institute of Functional NeurosurgeryXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain InformaticsBeijingChina
| |
Collapse
|
36
|
Mateo D, Marquès M, Torrente M. Metals linked with the most prevalent primary neurodegenerative dementias in the elderly: A narrative review. ENVIRONMENTAL RESEARCH 2023; 236:116722. [PMID: 37487923 DOI: 10.1016/j.envres.2023.116722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
The ageing population has been steadily increasing worldwide, leading to a higher risk of cognitive decline and dementia. Environmental toxicants, particularly metals, have been identified as modifiable risk factors for cognitive impairment. Continuous exposure to metals occurs mainly through dietary sources, with older adults being particularly vulnerable. However, imbalances in the gut microbiota, known as dysbiosis, have also been associated with dementia. A literature review was conducted to explore the potential role of metals in the development of cognitive decline and the most prevalent primary neurodegenerative dementias, as well as their interaction with the gut microbiota. High levels of iron (Fe) and copper (Cu) are associated with mild cognitive impairment (MCI) and Alzheimer's disease (AD), while low selenium (Se) levels are linked to poor cognitive status. Parkinson's disease dementia (PDD) is associated with elevated levels of iron (Fe), manganese (Mn), and zinc (Zn), but the role of copper (Cu) remains unclear. The relationship between metals and Lewy body dementia (LBD) requires further investigation. High aluminium (Al) exposure is associated with frontotemporal dementia (FTD), and elevated selenium (Se) levels may be linked to its onset. Challenges in comparing studies arise from the heterogeneity of metal analysis matrices and analytical techniques, as well as the limitations of small study cohorts. More research is needed to understand the influence of metals on cognition through the gut microbiota (GMB) and its potential relevance in the development of these diseases.
Collapse
Affiliation(s)
- David Mateo
- Laboratory of Toxicology and Environmental Health, School of Medicine, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Catalonia, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Universitat Rovira i Virgili, Spain
| | - Montse Marquès
- Laboratory of Toxicology and Environmental Health, School of Medicine, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Catalonia, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Universitat Rovira i Virgili, Spain
| | - Margarita Torrente
- Laboratory of Toxicology and Environmental Health, School of Medicine, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Catalonia, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Universitat Rovira i Virgili, Spain; Department of Psychology, CRAMC (Research Center for Behaviour Assessment), Faculty of Education Sciences and Psychology, Universitat Rovira i Virgili, Crta. de Valls s/n, 43007, Tarragona, Catalonia, Spain; Institute Lerin Neurocognitive, Alzheimer and other Neurocognitive Disorders Association, Av. D'Antoni Planas i Marca, 13, 43205, Reus, Catalonia, Spain.
| |
Collapse
|
37
|
Dutta D, Kanca O, Byeon SK, Marcogliese PC, Zuo Z, Shridharan RV, Park JH, Lin G, Ge M, Heimer G, Kohler JN, Wheeler MT, Kaipparettu BA, Pandey A, Bellen HJ. A defect in mitochondrial fatty acid synthesis impairs iron metabolism and causes elevated ceramide levels. Nat Metab 2023; 5:1595-1614. [PMID: 37653044 PMCID: PMC11151872 DOI: 10.1038/s42255-023-00873-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 07/21/2023] [Indexed: 09/02/2023]
Abstract
In most eukaryotic cells, fatty acid synthesis (FAS) occurs in the cytoplasm and in mitochondria. However, the relative contribution of mitochondrial FAS (mtFAS) to the cellular lipidome is not well defined. Here we show that loss of function of Drosophila mitochondrial enoyl coenzyme A reductase (Mecr), which is the enzyme required for the last step of mtFAS, causes lethality, while neuronal loss of Mecr leads to progressive neurodegeneration. We observe a defect in Fe-S cluster biogenesis and increased iron levels in flies lacking mecr, leading to elevated ceramide levels. Reducing the levels of either iron or ceramide suppresses the neurodegenerative phenotypes, indicating an interplay between ceramide and iron metabolism. Mutations in human MECR cause pediatric-onset neurodegeneration, and we show that human-derived fibroblasts display similar elevated ceramide levels and impaired iron homeostasis. In summary, this study identifies a role of mecr/MECR in ceramide and iron metabolism, providing a mechanistic link between mtFAS and neurodegeneration.
Collapse
Affiliation(s)
- Debdeep Dutta
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Paul C Marcogliese
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Rishi V Shridharan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Guang Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Ming Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Gali Heimer
- Pediatric Neurology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jennefer N Kohler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew T Wheeler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benny A Kaipparettu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Manipal Academy of Higher Education, Manipal, India
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
38
|
Kanaan AS, Yu D, Metere R, Schäfer A, Schlumm T, Bilgic B, Anwander A, Mathews CA, Scharf JM, Müller-Vahl K, Möller HE. Convergent imaging-transcriptomic evidence for disturbed iron homeostasis in Gilles de la Tourette syndrome. Neurobiol Dis 2023; 185:106252. [PMID: 37536382 DOI: 10.1016/j.nbd.2023.106252] [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: 05/10/2023] [Revised: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023] Open
Abstract
Gilles de la Tourette syndrome (GTS) is a neuropsychiatric movement disorder with reported abnormalities in various neurotransmitter systems. Considering the integral role of iron in neurotransmitter synthesis and transport, it is hypothesized that iron exhibits a role in GTS pathophysiology. As a surrogate measure of brain iron, quantitative susceptibility mapping (QSM) was performed in 28 patients with GTS and 26 matched controls. Significant susceptibility reductions in the patients, consistent with reduced local iron content, were obtained in subcortical regions known to be implicated in GTS. Regression analysis revealed a significant negative association of tic scores and striatal susceptibility. To interrogate genetic mechanisms that may drive these reductions, spatially specific relationships between susceptibility and gene-expression patterns from the Allen Human Brain Atlas were assessed. Correlations in the striatum were enriched for excitatory, inhibitory, and modulatory neurochemical signaling mechanisms in the motor regions, mitochondrial processes driving ATP production and iron‑sulfur cluster biogenesis in the executive subdivision, and phosphorylation-related mechanisms affecting receptor expression and long-term potentiation in the limbic subdivision. This link between susceptibility reductions and normative transcriptional profiles suggests that disruptions in iron regulatory mechanisms are involved in GTS pathophysiology and may lead to pervasive abnormalities in mechanisms regulated by iron-containing enzymes.
Collapse
Affiliation(s)
- Ahmad Seif Kanaan
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.
| | - Dongmei Yu
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Riccardo Metere
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Andreas Schäfer
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Siemens Healthcare GmbH, Diagnostic Imaging, Magnetic Resonance, Research and Development, Erlangen, Germany
| | - Torsten Schlumm
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Berkin Bilgic
- Harvard Medical School, Boston, MA, USA; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA; Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alfred Anwander
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Carol A Mathews
- Department of Psychiatry, Center for OCD, Anxiety, and Related Disorders, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jeremiah M Scharf
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Kirsten Müller-Vahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Harald E Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
39
|
Benarroch E. What Is the Role of Ferroptosis in Neurodegeneration? Neurology 2023; 101:312-319. [PMID: 37580137 PMCID: PMC10437014 DOI: 10.1212/wnl.0000000000207730] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 08/16/2023] Open
|
40
|
Rahayel S, Tremblay C, Vo A, Misic B, Lehéricy S, Arnulf I, Vidailhet M, Corvol JC, Gagnon JF, Postuma RB, Montplaisir J, Lewis S, Matar E, Ehgoetz Martens K, Borghammer P, Knudsen K, Hansen AK, Monchi O, Gan-Or Z, Dagher A. Mitochondrial function-associated genes underlie cortical atrophy in prodromal synucleinopathies. Brain 2023; 146:3301-3318. [PMID: 36826230 PMCID: PMC10393413 DOI: 10.1093/brain/awad044] [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: 09/20/2022] [Revised: 01/12/2023] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
Isolated rapid eye movement sleep behaviour disorder (iRBD) is a sleep disorder characterized by the loss of rapid eye movement sleep muscle atonia and the appearance of abnormal movements and vocalizations during rapid eye movement sleep. It is a strong marker of incipient synucleinopathy such as dementia with Lewy bodies and Parkinson's disease. Patients with iRBD already show brain changes that are reminiscent of manifest synucleinopathies including brain atrophy. However, the mechanisms underlying the development of this atrophy remain poorly understood. In this study, we performed cutting-edge imaging transcriptomics and comprehensive spatial mapping analyses in a multicentric cohort of 171 polysomnography-confirmed iRBD patients [67.7 ± 6.6 (49-87) years; 83% men] and 238 healthy controls [66.6 ± 7.9 (41-88) years; 77% men] with T1-weighted MRI to investigate the gene expression and connectivity patterns associated with changes in cortical thickness and surface area in iRBD. Partial least squares regression was performed to identify the gene expression patterns underlying cortical changes in iRBD. Gene set enrichment analysis and virtual histology were then done to assess the biological processes, cellular components, human disease gene terms, and cell types enriched in these gene expression patterns. We then used structural and functional neighbourhood analyses to assess whether the atrophy patterns in iRBD were constrained by the brain's structural and functional connectome. Moreover, we used comprehensive spatial mapping analyses to assess the specific neurotransmitter systems, functional networks, cytoarchitectonic classes, and cognitive brain systems associated with cortical changes in iRBD. All comparisons were tested against null models that preserved spatial autocorrelation between brain regions and compared to Alzheimer's disease to assess the specificity of findings to synucleinopathies. We found that genes involved in mitochondrial function and macroautophagy were the strongest contributors to the cortical thinning occurring in iRBD. Moreover, we demonstrated that cortical thinning was constrained by the brain's structural and functional connectome and that it mapped onto specific networks involved in motor and planning functions. In contrast with cortical thickness, changes in cortical surface area were related to distinct genes, namely genes involved in the inflammatory response, and to different spatial mapping patterns. The gene expression and connectivity patterns associated with iRBD were all distinct from those observed in Alzheimer's disease. In summary, this study demonstrates that the development of brain atrophy in synucleinopathies is constrained by specific genes and networks.
Collapse
Affiliation(s)
- Shady Rahayel
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal H3A 2B4, Canada
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal H4J 1C5, Canada
| | - Christina Tremblay
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal H3A 2B4, Canada
| | - Andrew Vo
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal H3A 2B4, Canada
| | - Bratislav Misic
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal H3A 2B4, Canada
| | - Stéphane Lehéricy
- Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, Sorbonne Université, Paris 75013, France
| | - Isabelle Arnulf
- Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, Sorbonne Université, Paris 75013, France
| | - Marie Vidailhet
- Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, Sorbonne Université, Paris 75013, France
| | - Jean-Christophe Corvol
- Institut du Cerveau–Paris Brain Institute–ICM, INSERM, CNRS, Sorbonne Université, Paris 75013, France
| | - Jean-François Gagnon
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal H4J 1C5, Canada
- Department of Psychology, University of Quebec in Montreal, Montreal H2X 3P2, Canada
- Research Centre, Institut universitaire de gériatrie de Montréal, Montreal H3W 1W5, Canada
| | - Ronald B Postuma
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal H4J 1C5, Canada
- Department of Neurology, Montreal General Hospital, Montreal H3G 1A4, Canada
| | - Jacques Montplaisir
- Centre for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Montreal H4J 1C5, Canada
- Department of Psychiatry, University of Montreal, Montreal H3T 1J4, Canada
| | - Simon Lewis
- ForeFront Parkinson’s Disease Research Clinic, Brain and Mind Centre, University of Sydney, Camperdown NSW 2050, Australia
| | - Elie Matar
- ForeFront Parkinson’s Disease Research Clinic, Brain and Mind Centre, University of Sydney, Camperdown NSW 2050, Australia
| | - Kaylena Ehgoetz Martens
- ForeFront Parkinson’s Disease Research Clinic, Brain and Mind Centre, University of Sydney, Camperdown NSW 2050, Australia
- Department of Kinesiology, University of Waterloo, Waterloo N2L 3G1, Canada
| | - Per Borghammer
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Karoline Knudsen
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Allan K Hansen
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Oury Monchi
- Research Centre, Institut universitaire de gériatrie de Montréal, Montreal H3W 1W5, Canada
- Department of Radiology, Radio-Oncology, and Nuclear Medicine, University of Montreal, Montreal H3T 1A4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal H3A 1A1, Canada
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal H3A 1A1, Canada
- Department of Human Genetics, McGill University, Montreal H3A 0C7, Canada
| | - Alain Dagher
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montreal H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal H3A 1A1, Canada
| | | |
Collapse
|
41
|
Kanaan AS, Yu D, Metere R, Schäfer A, Schlumm T, Bilgic B, Anwander A, Mathews CA, Scharf JM, Müller-Vahl K, Möller HE. Convergent imaging-transcriptomic evidence for disturbed iron homeostasis in Gilles de la Tourette syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.15.23289978. [PMID: 37292704 PMCID: PMC10246056 DOI: 10.1101/2023.05.15.23289978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gilles de la Tourette syndrome (GTS) is a neuropsychiatric movement disorder with reported abnormalities in various neurotransmitter systems. Considering the integral role of iron in neurotransmitter synthesis and transport, it is hypothesized that iron exhibits a role in GTS pathophysiology. As a surrogate measure of brain iron, quantitative susceptibility mapping (QSM) was performed in 28 patients with GTS and 26 matched controls. Significant susceptibility reductions in the patient cohort, consistent with reduced local iron content, were obtained in subcortical regions known to be implicated in GTS. Regression analysis revealed a significant negative association of tic scores and striatal susceptibility. To interrogate genetic mechanisms that may drive these reductions, spatially specific relationships between susceptibility and gene-expression patterns extracted from the Allen Human Brain Atlas were assessed. Correlations in the striatum were enriched for excitatory, inhibitory, and modulatory neurochemical signaling mechanisms in the motor regions, mitochondrial processes driving ATP production and iron-sulfur cluster biogenesis in the executive subdivision, and phosphorylation-related mechanisms that affect receptor expression and long-term potentiation. This link between susceptibility reductions and normative transcriptional profiles suggests that disruptions in iron regulatory mechanisms are involved in GTS pathophysiology and may lead to pervasive abnormalities in mechanisms regulated by iron-containing enzymes.
Collapse
Affiliation(s)
- Ahmad Seif Kanaan
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Dongmei Yu
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Riccardo Metere
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Andreas Schäfer
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Siemens Healthcare GmbH, Diagnostic Imaging, Magnetic Resonance, Research and Development, Erlangen, Germany
| | - Torsten Schlumm
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Berkin Bilgic
- Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alfred Anwander
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Carol A. Mathews
- Department of Psychiatry, Center for OCD, Anxiety, and Related Disorders, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jeremiah M. Scharf
- Center for Human Genetics Research, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kirsten Müller-Vahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Harald E. Möller
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
42
|
Ketone Body β-Hydroxybutyric Acid Ameliorates Dopaminergic Neuron Injury Through Modulating Zinc Finger Protein 36/Acyl-CoA Synthetase Long-Chain Family Member Four Signaling Axis-Mediated Ferroptosis. Neuroscience 2023; 509:157-172. [PMID: 36435477 DOI: 10.1016/j.neuroscience.2022.11.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/13/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022]
Abstract
β-hydroxybutyrate (BHB) is one of main component of ketone body, which plays an important protective role in various tissues and organs. Whereas, its exact regulatory roles and mechanisms in Parkinson's disease (PD) have not been full elucidated. In this study, SN4741 cells and C57BL/6 mice were treated with 1-methyl-4-phenylpyridinium ion (MPP+)/1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to establish the PD model in vitro and in vivo. Cell viability and damage to dopaminergic neurons were measured by cell counting kit 8, Calcein-AM/PI staining, terminal dexynucleotidyl transferase (TdT)-mediated dUTP nick end labeling and hematoxylin & eosin staining. Corresponding assay kits and BODIPY 581/591 C11 probe evaluated oxidative stress and intracellular iron levels. Western blot examined the ferroptosis-related proteins. MPTP/MPP+-treatment reduced cell viability but triggered oxidative stress and ferroptosis in SNA4741 cells and brain tissues of mice. However, these effects were dramatically reversed by BHB and Fer-1 treatment. Mechanistically, Zinc finger protein 36 (ZFP36) was a target of BHB, and its depletion could reverse the anti-oxidative stress and anti-ferroptosis roles of BHB. Moreover, ZFP36 could directly bound to acyl-CoA synthetase long-chain family member 4 (ACSL4) mRNA to decay its expression, thus negatively modulating ACSL4-mediated oxidative stress and ferroptosis. Summary, BHB alleviated oxidative stress and ferroptosis of dopaminergic neurons in PD via modulating ZFP36/ACSL4 axis, which provided some new understanding for PD prevention and treatment.
Collapse
|
43
|
Milotta G, Corbin N, Lambert C, Lutti A, Mohammadi S, Callaghan MF. Mitigating the impact of flip angle and orientation dependence in single compartment R2* estimates via 2-pool modeling. Magn Reson Med 2023; 89:128-143. [PMID: 36161672 PMCID: PMC9827921 DOI: 10.1002/mrm.29428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/08/2022] [Accepted: 08/08/2022] [Indexed: 01/12/2023]
Abstract
PURPOSE The effective transverse relaxation rate (R 2 * $$ {\mathrm{R}}_2^{\ast } $$ ) is influenced by biological features that make it a useful means of probing brain microstructure. However, confounding factors such as dependence on flip angle (α) and fiber orientation with respect to the main field (θ $$ \uptheta $$ ) complicate interpretation. The α- andθ $$ \uptheta $$ -dependence stem from the existence of multiple sub-voxel micro-environments (e.g., myelin and non-myelin water compartments). Ordinarily, it is challenging to quantify these sub-compartments; therefore, neuroscientific studies commonly make the simplifying assumption of a mono-exponential decay obtaining a singleR 2 * $$ {\mathrm{R}}_2^{\ast } $$ estimate per voxel. In this work, we investigated how the multi-compartment nature of tissue microstructure affects single compartmentR 2 * $$ {\mathrm{R}}_2^{\ast } $$ estimates. METHODS We used 2-pool (myelin and non-myelin water) simulations to characterize the bias in single compartmentR 2 * $$ {\mathrm{R}}_2^{\ast } $$ estimates. Based on our numeric observations, we introduced a linear model that partitionsR 2 * $$ {\mathrm{R}}_2^{\ast } $$ into α-dependent and α-independent components and validated this in vivo at 7T. We investigated the dependence of both components on the sub-compartment properties and assessed their robustness, orientation dependence, and reproducibility empirically. RESULTS R 2 * $$ {\mathrm{R}}_2^{\ast } $$ increased with myelin water fraction and residency time leading to a linear dependence on α. We observed excellent agreement between our numeric and empirical results. Furthermore, the α-independent component of the proposed linear model was robust to the choice of α and reduced dependence on fiber orientation, although it suffered from marginally higher noise sensitivity. CONCLUSION We have demonstrated and validated a simple approach that mitigates flip angle and orientation biases in single-compartmentR 2 * $$ {\mathrm{R}}_2^{\ast } $$ estimates.
Collapse
Affiliation(s)
- Giorgia Milotta
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of NeurologyUniversity College London
LondonUnited Kingdom
| | - Nadège Corbin
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of NeurologyUniversity College London
LondonUnited Kingdom
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536CNRS/University BordeauxBordeauxFrance
| | - Christian Lambert
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of NeurologyUniversity College London
LondonUnited Kingdom
| | - Antoine Lutti
- Laboratory for Research in Neuroimaging, Department for Clinical NeuroscienceLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Siawoosh Mohammadi
- Department of Systems NeurosciencesUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Department of NeurophysicsMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Martina F. Callaghan
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of NeurologyUniversity College London
LondonUnited Kingdom
| |
Collapse
|
44
|
Association between Beta Oscillations from Subthalamic Nucleus and Quantitative Susceptibility Mapping in Deep Gray Matter Structures in Parkinson's Disease. Brain Sci 2023; 13:brainsci13010081. [PMID: 36672062 PMCID: PMC9857066 DOI: 10.3390/brainsci13010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/15/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
This study aimed to investigate the association between beta oscillations and brain iron deposition. Beta oscillations were filtered from the microelectrode recordings of local field potentials (LFP) in the subthalamic nucleus (STN), and the ratio of the power spectral density of beta oscillations (PSDXb) to that of the LFP signals was calculated. Iron deposition in the deep gray matter (DGM) structures was indirectly assessed using quantitative susceptibility mapping (QSM). The Unified Parkinson's Disease Rating Scale (UPDRS), part III, was used to assess the severity of symptoms. Spearman correlation coefficients were applied to assess the associations of PSDXb with QSM values in the DGM structures and the severity of symptoms. PSDXb showed a significant positive correlation with the average QSM values in DGM structures, including caudate and substantia nigra (SN) (p = 0.008 and 0.044). Similarly, the PSDXb showed significant negative correlations with the severity of symptoms, including axial symptoms and the gait in the medicine-off state (p = 0.006 for both). The abnormal iron metabolism in the SN and striatum pathways may be one of the underlying mechanisms for the occurrence of abnormal beta oscillations in the STN, and beta oscillations may serve as important pathophysiological biomarkers of PD.
Collapse
|
45
|
Quantitative Susceptibility Mapping: Translating an Investigative Research Tool into High Volume Clinical Diagnostic Imaging. Diagnostics (Basel) 2022; 12:diagnostics12122962. [PMID: 36552969 PMCID: PMC9776933 DOI: 10.3390/diagnostics12122962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Quantitative susceptibility mapping (QSM) is an MRI-based technique for iron quantification of targeted tissue. QSM provides information relevant to clinicians in a broad range of diagnostic contexts, including sickle cell disease, inflammatory/demyelinating processes, and neoplasms. However, major MRI vendors do not offer QSM post-processing in a form ready for general use. This work describes a vendor-agnostic approach for scaling QSM analysis from a research technique to a routine diagnostic test. We provide the details needed to seamlessly integrate hardware, software, and clinical systems to provide QSM processing for a busy clinical radiology workflow. This approach can be generalized to other advanced MRI acquisitions and analyses with proven diagnostic utility, yet without crucial MR vendor support.
Collapse
|
46
|
McColgan P, Gregory S, Zeun P, Zarkali A, Johnson EB, Parker C, Fayer K, Lowe J, Nair A, Estevez-Fraga C, Papoutsi M, Zhang H, Scahill RI, Tabrizi SJ, Rees G. Neurofilament light-associated connectivity in young-adult Huntington's disease is related to neuronal genes. Brain 2022; 145:3953-3967. [PMID: 35758263 PMCID: PMC9679168 DOI: 10.1093/brain/awac227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
Upregulation of functional network connectivity in the presence of structural degeneration is seen in the premanifest stages of Huntington's disease (preHD) 10-15 years from clinical diagnosis. However, whether widespread network connectivity changes are seen in gene carriers much further from onset has yet to be explored. We characterized functional network connectivity throughout the brain and related it to a measure of disease pathology burden (CSF neurofilament light, NfL) and measures of structural connectivity in asymptomatic gene carriers, on average 24 years from onset. We related these measurements to estimates of cortical and subcortical gene expression. We found no overall differences in functional (or structural) connectivity anywhere in the brain comparing control and preHD participants. However, increased functional connectivity, particularly between posterior cortical areas, correlated with increasing CSF NfL level in preHD participants. Using the Allen Human Brain Atlas and expression-weighted cell-type enrichment analysis, we demonstrated that this functional connectivity upregulation occurred in cortical regions associated with regional expression of genes specific to neuronal cells. This relationship was validated using single-nucleus RNAseq data from post-mortem Huntington's disease and control brains showing enrichment of neuronal-specific genes that are differentially expressed in Huntington's disease. Functional brain networks in asymptomatic preHD gene carriers very far from disease onset show evidence of upregulated connectivity correlating with increased disease burden. These changes occur among brain areas that show regional expression of genes specific to neuronal GABAergic and glutamatergic cells.
Collapse
Affiliation(s)
- Peter McColgan
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sarah Gregory
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Paul Zeun
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Angeliki Zarkali
- Dementia Research Centre, University College London, London WC1N 3AR, UK
| | - Eileanoir B Johnson
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Christopher Parker
- Department of Computer Science and Centre for Medical Image Computing, University College London, London WC1V 6LJ, UK
| | - Kate Fayer
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Jessica Lowe
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Akshay Nair
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Carlos Estevez-Fraga
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marina Papoutsi
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Hui Zhang
- Dementia Research Centre, University College London, London WC1N 3AR, UK
| | - Rachael I Scahill
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Sarah J Tabrizi
- Huntington’s Disease Centre, Department of Neurodegenerative disease, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- Dementia Research Centre, University College London, London WC1N 3AR, UK
| | - Geraint Rees
- University College London Institute of Cognitive Neuroscience, University College London, London WC1N 3AZ, UK
| |
Collapse
|
47
|
Vellingiri B, Suriyanarayanan A, Selvaraj P, Abraham KS, Pasha MY, Winster H, Gopalakrishnan AV, G S, Reddy JK, Ayyadurai N, Kumar N, Giridharan B, P S, Rao KRSS, Nachimuthu SK, Narayanasamy A, Mahalaxmi I, Venkatesan D. Role of heavy metals (copper (Cu), arsenic (As), cadmium (Cd), iron (Fe) and lithium (Li)) induced neurotoxicity. CHEMOSPHERE 2022; 301:134625. [PMID: 35439490 DOI: 10.1016/j.chemosphere.2022.134625] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/30/2022] [Accepted: 04/12/2022] [Indexed: 05/15/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative condition characterized by the dopamine (DA) neuronal loss in the substantia nigra. PD impairs motor controls symptoms such as tremor, rigidity, bradykinesia and postural imbalance gradually along with non-motor problems such as olfactory dysfunction, constipation, sleeping disorder. Though surplus of factors and mechanisms have been recognized, the precise PD etiopathogenesis is not yet implied. Reports suggest that various environmental factors play a crucial role in the causality of the PD cases. Epidemiological studies have reported that heavy metals has a role in causing defects in substantia nigra region of brain in PD. Though the reason is unknown, exposure to heavy metals is reported to be an underlying factor in PD development. Metals are classified as either essential or non-essential, and they have a role in physiological processes such protein modification, electron transport, oxygen transport, redox reactions, and cell adhesion. Excessive metal levels cause oxidative stress, protein misfolding, mitochondrial malfunction, autophagy dysregulation, and apoptosis, among other things. In this review, we check out the link between heavy metals like copper (Cu), arsenic (As), cadmium (Cd), iron (Fe), and lithium (Li) in neurodegeneration, and how it impacts the pathological conditions of PD. In conclusion, increase or decrease in heavy metals involve in regulation of neuronal functions that have an impact on neurodegeneration process. Through this review, we suggest that more research is needed in this stream to bring more novel approaches for either disease modelling or therapeutics.
Collapse
Affiliation(s)
- Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Atchaya Suriyanarayanan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Priyanka Selvaraj
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Kripa Susan Abraham
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Md Younus Pasha
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Harysh Winster
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India; Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632 014, India
| | - Singaravelu G
- Department of Education, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | | | - Niraikulam Ayyadurai
- CSIR-Central Leather Research Institute, Adyar, Chennai, 600 020, Tamil Nadu, India
| | - Nandha Kumar
- Department of Zoology, St. Joseph University, 797 115, Dimapur, Nagaland
| | - Bupesh Giridharan
- Department of Forest Science, Nagaland University, Lumami, Zunheboto, Nagaland, India
| | - Sivaprakash P
- Department of Mechanical Engineering, Dr.N.G.P. Institute of Technology, Coimbatore, 641048, Tamil Nadu, India
| | - K R S Sambasiva Rao
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796 004, Mizoram, India
| | - Senthil Kumar Nachimuthu
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796 004, Mizoram, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| | - Iyer Mahalaxmi
- Livestock Farming and Bioresource Technology, Tamil Nadu, India.
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| |
Collapse
|
48
|
Chai C, Wu M, Wang H, Cheng Y, Zhang S, Zhang K, Shen W, Liu Z, Xia S. CAU-Net: A Deep Learning Method for Deep Gray Matter Nuclei Segmentation. Front Neurosci 2022; 16:918623. [PMID: 35720705 PMCID: PMC9204516 DOI: 10.3389/fnins.2022.918623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/03/2022] [Indexed: 12/04/2022] Open
Abstract
The abnormal iron deposition of the deep gray matter nuclei is related to many neurological diseases. With the quantitative susceptibility mapping (QSM) technique, it is possible to quantitatively measure the brain iron content in vivo. To assess the magnetic susceptibility of the deep gray matter nuclei in the QSM, it is mandatory to segment the nuclei of interest first, and many automatic methods have been proposed in the literature. This study proposed a contrast attention U-Net for nuclei segmentation and evaluated its performance on two datasets acquired using different sequences with different parameters from different MRI devices. Experimental results revealed that our proposed method was superior on both datasets over other commonly adopted network structures. The impacts of training and inference strategies were also discussed, which showed that adopting test time augmentation during the inference stage can impose an obvious improvement. At the training stage, our results indicated that sufficient data augmentation, deep supervision, and nonuniform patch sampling contributed significantly to improving the segmentation accuracy, which indicated that appropriate choices of training and inference strategies were at least as important as designing more advanced network structures.
Collapse
Affiliation(s)
- Chao Chai
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Mengran Wu
- College of Electronic Information and Optical Engineering, Nankai University, Tianjin, China
| | - Huiying Wang
- School of Medicine, Nankai University, Tianjin, China
| | - Yue Cheng
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | | | - Kun Zhang
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Wen Shen
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Zhiyang Liu
- College of Electronic Information and Optical Engineering, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Optoelectronic Sensor and Sensing Network Technology, Tianjin, China
- *Correspondence: Zhiyang Liu,
| | - Shuang Xia
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Shuang Xia,
| |
Collapse
|
49
|
Lancione M, Cencini M, Costagli M, Donatelli G, Tosetti M, Giannini G, Zangaglia R, Calandra-Buonaura G, Pacchetti C, Cortelli P, Cosottini M. Diagnostic accuracy of quantitative susceptibility mapping in multiple system atrophy: The impact of echo time and the potential of histogram analysis. Neuroimage Clin 2022; 34:102989. [PMID: 35303599 PMCID: PMC8927993 DOI: 10.1016/j.nicl.2022.102989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/25/2022] [Accepted: 03/10/2022] [Indexed: 11/07/2022]
Abstract
We performed histogram analysis on χ maps at different TEs on MSA patients and HC. We found altered χ distribution in Pu, SN, GP, CN for MSAp and in SN, DN for MSAc. QSM diagnostic accuracy is TE-dependent and is enhanced at short TEs. Short TEs capture rapidly-decaying contributions of high χ sources. Histogram features detect χ spatial heterogeneities improving diagnostic accuracy.
The non-invasive quantification of iron stores via Quantitative Susceptibility Mapping (QSM) could play an important role in the diagnosis and the differential diagnosis of atypical Parkinsonisms. However, the susceptibility (χ) values measured via QSM depend on echo time (TE). This effect relates to the microstructural organization within the voxel, whose composition can be altered by the disease. Moreover, pathological iron deposition in a brain area may not be spatially uniform, and conventional Region of Interest (ROI)-based analysis may fail in detecting alterations. Therefore, in this work we evaluated the impact of echo time on the diagnostic accuracy of QSM on a population of patients with Multiple System Atrophy (MSA) of either Parkinsonian (MSAp) or cerebellar (MSAc) phenotypes. In addition, we tested the potential of histogram analysis to improve QSM classification accuracy. We enrolled 32 patients (19 MSAp and 13 MSAc) and 16 healthy controls, who underwent a 7T MRI session including a gradient-recalled multi-echo sequence for χ mapping. Nine histogram features were extracted from the χ maps computed for each TE in atlas-based ROIs covering deep brain nuclei, and compared among groups. Alterations of susceptibility distribution were found in the Putamen, Substantia Nigra, Globus Pallidus and Caudate Nucleus for MSAp and in the Substantia Nigra and Dentate Nucleus for MSAc. Increased iron deposition was observed in a larger number of ROIs for the two shortest TEs and the standard deviation, the 75th and the 90th percentile were the most informative features yielding excellent diagnostic accuracy with area under the ROC curve > 0.9. In conclusion, short TEs may enhance QSM diagnostic performances, as they can capture variations in rapidly-decaying contributions of high χ sources. The analysis of histogram features allowed to reveal fine heterogeneities in the spatial distribution of susceptibility alteration, otherwise undetected by a simple evaluation of ROI χ mean values.
Collapse
Affiliation(s)
- Marta Lancione
- IRCCS Stella Maris, Pisa, Italy; IMAGO7 Foundation, Pisa, Italy
| | - Matteo Cencini
- IRCCS Stella Maris, Pisa, Italy; IMAGO7 Foundation, Pisa, Italy
| | - Mauro Costagli
- IRCCS Stella Maris, Pisa, Italy; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Sciences (DINOGMI), University of Genova, Genova, Italy.
| | - Graziella Donatelli
- IMAGO7 Foundation, Pisa, Italy; Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Michela Tosetti
- IRCCS Stella Maris, Pisa, Italy; IMAGO7 Foundation, Pisa, Italy
| | - Giulia Giannini
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Roberta Zangaglia
- Parkinson and Movement Disorder Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Giovanna Calandra-Buonaura
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Claudio Pacchetti
- Parkinson and Movement Disorder Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Pietro Cortelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy; IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | |
Collapse
|
50
|
Wang ZL, Yuan L, Li W, Li JY. Ferroptosis in Parkinson’s disease: glia–neuron crosstalk. Trends Mol Med 2022; 28:258-269. [DOI: 10.1016/j.molmed.2022.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/14/2022]
|