1
|
Goldman JG, Jagota P, Matar E. Managing cognitive impairment in Parkinson's disease: an update of the literature. Expert Rev Neurother 2025:1-21. [PMID: 39773313 DOI: 10.1080/14737175.2025.2450668] [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: 11/10/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Cognitive impairment in Parkinson's disease (PD) substantially affects patient outcomes, function, and quality of life. PD-related cognitive dysfunction is often heterogeneous in clinical presentation and rates of progression. As cognitive changes occur in many people with PD, it is essential to evaluate cognition, provide education, and implement management strategies for cognitive symptoms. AREAS COVERED This article describes the symptomatology, epidemiology, risk factors, and pathobiology of cognitive impairment in PD. Additionally, the article provides an overview of evidence-based management and other therapeutic and coping strategies for cognitive impairment and dementia in PD. Comment is offered on challenges and opportunities for trials and emerging therapeutics targeting cognitive symptoms or decline. EXPERT OPINION While our understanding of cognitive dysfunction in PD has grown, effective and safe therapeutics are still needed to not only treat cognitive impairment and dementia symptomatically, but also slow down or prevent cognitive decline. Further research is needed to elucidate the pathobiology of PD cognitive impairment, develop validated biomarkers reflecting cognitive change, and ultimately, integrate clinical and biological frameworks. Consensus regarding cognitive evaluations, definitions and criteria of cognitive impairment, evaluating functional abilities in the context of cognitive impairment, and determining optimal outcome measures for clinical trials remain unmet needs.
Collapse
Affiliation(s)
- Jennifer G Goldman
- Enterprises LLC, Chicago, IL
- Adjunct Professor, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Priya Jagota
- Chulalongkorn Centre of Excellence for Parkinson's Disease and Related Disorders, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Elie Matar
- Central Clinical School, Faculty of Medicine and Health, University of Sydney
- Department of Neurology, Royal Prince Alfred Hospital, Camperdown, NSW
| |
Collapse
|
2
|
Bjørklund G, Wallace DR, Hangan T, Butnariu M, Gurgas L, Peana M. Cerebral iron accumulation in multiple sclerosis: Pathophysiology and therapeutic implications. Autoimmun Rev 2025; 24:103741. [PMID: 39756528 DOI: 10.1016/j.autrev.2025.103741] [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: 12/14/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system characterized by demyelination, neuroinflammation, and neurodegeneration. Recent studies highlight the role of cerebral iron (Fe) accumulation in exacerbating MS pathophysiology. Fe, essential for neural function, contributes to oxidative stress and inflammation when dysregulated, particularly in the brain's gray matter and demyelinated lesions. Advanced imaging techniques, including susceptibility-weighted and quantitative susceptibility mapping, have revealed abnormal Fe deposition patterns in MS patients, suggesting its involvement in disease progression. Iron's interaction with immune cells, such as microglia, releases pro-inflammatory cytokines, further amplifying neuroinflammation and neuronal damage. These findings implicate Fe dysregulation as a significant factor in MS progression, contributing to clinical manifestations like cognitive impairment. Therapeutic strategies targeting Fe metabolism, including Fe chelation therapies, show promise in reducing Fe-related damage, instilling optimism about the future of MS treatment. However, challenges such as crossing the blood-brain barrier and maintaining Fe homeostasis remain. Emerging approaches, such as Fe-targeted nanotherapeutics and biologics, offer new possibilities for personalized treatments. However, the journey is far from over. Continued research into the molecular mechanisms of Fe-induced neuroinflammation and oxidative damage is essential. Through this research, we can develop effective interventions that could slow MS progression and improve patient outcomes.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway.
| | - David R Wallace
- Department of Pharmacology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, Timis, Romania; CONEM Romania Biotechnology and Environmental Sciences Group, University of Life Sciences "King Mihai I" from Timisoara, Timis, Romania
| | - Leonard Gurgas
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Italy
| |
Collapse
|
3
|
Salazar Ariza JF, Lux F, Da Cruz-Boisson F, Resende de Azevedo J, Vera R, Tillement O, Montembault A, David L. Chitosan based hydrogel for iron (III) chelation in biological conditions. Carbohydr Polym 2025; 347:122670. [PMID: 39486926 DOI: 10.1016/j.carbpol.2024.122670] [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: 07/12/2024] [Accepted: 08/27/2024] [Indexed: 11/04/2024]
Abstract
In this study, a chitosan derivative with strong iron (III) chelating capabilities was developed by grafting the Deferoxamine (DFO) chelator to achieve a substitution degree of 3.8 ± 0.2 %. Through blending with ungrafted chitosan of low degree of acetylation (DA), a formulation able to form a physical hydrogels was formed in aqueous media, without the requirement of a cross-linking agent. The functionalization of chitosan with DFO led to xerogels exhibiting superior iron (III) chelation capacity and higher swelling when exposed to aqueous solutions, in comparison with to an unmodified chitosan xerogel. Notably, this material extracts iron (III) even against the strong iron chelator deferiprone. Furthermore, the material demonstrates selectivity for iron (III) chelation even in the presence of competing cations like copper (II) and zinc (II).
Collapse
Affiliation(s)
- Juan Felipe Salazar Ariza
- Universite Claude Bernard Lyon 1, CNRS, Institut Lumière-Matière (ILM), UMR 5306, 2 rue Victor Grignard, F-69622 Villeurbanne Cedex, France; Universite Claude Bernard Lyon 1, INSA de Lyon, Universite Jean Monnet, CNRS, Ingénierie des Matériaux Polymères (IMP), UMR 5223, 15 bd Latarjet, F-69622 Villeurbanne, France
| | - François Lux
- Universite Claude Bernard Lyon 1, CNRS, Institut Lumière-Matière (ILM), UMR 5306, 2 rue Victor Grignard, F-69622 Villeurbanne Cedex, France; Institut Universitaire de France (IUF), 75231 Paris, France
| | - Fernande Da Cruz-Boisson
- Universite Claude Bernard Lyon 1, INSA de Lyon, Universite Jean Monnet, CNRS, Ingénierie des Matériaux Polymères (IMP), UMR 5223, 15 bd Latarjet, F-69622 Villeurbanne, France
| | - Jacqueline Resende de Azevedo
- Universite Claude Bernard Lyon 1, CNRS, Laboratoire d'Automatique, de Génie des Procédés et de Génie Pharmaceutique (LAGEPP), UMR5007, 3, rue Victor Grignard, Bâtiment CPE, F-69100 Villeurbanne Cedex, France
| | - Ruben Vera
- Universite Claude Bernard Lyon 1, Centre de Diffractométrie Henri Longchambon, 5 rue de La Doua, F-69100 Villeurbanne, France
| | - Olivier Tillement
- Universite Claude Bernard Lyon 1, CNRS, Institut Lumière-Matière (ILM), UMR 5306, 2 rue Victor Grignard, F-69622 Villeurbanne Cedex, France
| | - Alexandra Montembault
- Universite Claude Bernard Lyon 1, INSA de Lyon, Universite Jean Monnet, CNRS, Ingénierie des Matériaux Polymères (IMP), UMR 5223, 15 bd Latarjet, F-69622 Villeurbanne, France
| | - Laurent David
- Universite Claude Bernard Lyon 1, INSA de Lyon, Universite Jean Monnet, CNRS, Ingénierie des Matériaux Polymères (IMP), UMR 5223, 15 bd Latarjet, F-69622 Villeurbanne, France.
| |
Collapse
|
4
|
Wang N, Liao C, Cao X, Nishimura M, Brackenier YWE, Yurt M, Gao M, Abraham D, Alkan C, Iyer SS, Zhou Z, Jeong H, Kerr A, Haldar JP, Setsompop K. Spherical echo-planar time-resolved imaging (sEPTI) for rapid 3D quantitative T 2 * and susceptibility imaging. Magn Reson Med 2025; 93:121-137. [PMID: 39250435 DOI: 10.1002/mrm.30255] [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: 03/28/2024] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE To develop a 3D spherical EPTI (sEPTI) acquisition and a comprehensive reconstruction pipeline for rapid high-quality whole-brain submillimeterT 2 * $$ {\mathrm{T}}_2^{\ast } $$ and QSM quantification. METHODS For the sEPTI acquisition, spherical k-space coverage is utilized with variable echo-spacing and maximum kx ramp-sampling to improve efficiency and signal incoherency compared to existing EPTI approaches. For reconstruction, an iterative rank-shrinking B0 estimation and odd-even high-order phase correction algorithms were incorporated into the reconstruction to better mitigate artifacts from field imperfections. A physics-informed unrolled network was utilized to boost the SNR, where 1-mm and 0.75-mm isotropic whole-brain imaging were performed in 45 and 90 s at 3 T, respectively. These protocols were validated through simulations, phantom, and in vivo experiments. Ten healthy subjects were recruited to provide sufficient data for the unrolled network. The entire pipeline was validated on additional five healthy subjects where different EPTI sampling approaches were compared. Two additional pediatric patients with epilepsy were recruited to demonstrate the generalizability of the unrolled reconstruction. RESULTS sEPTI achieved 1.4× $$ \times $$ faster imaging with improved image quality and quantitative map precision compared to existing EPTI approaches. The B0 update and the phase correction provide improved reconstruction performance with lower artifacts. The unrolled network boosted the SNR, achieving high-qualityT 2 * $$ {\mathrm{T}}_2^{\ast } $$ and QSM quantification with single average data. High-quality reconstruction was also obtained in the pediatric patients using this network. CONCLUSION sEPTI achieved whole-brain distortion-free multi-echo imaging andT 2 * $$ {\mathrm{T}}_2^{\ast } $$ and QSM quantification at 0.75 mm in 90 s which has the potential to be useful for wide clinical applications.
Collapse
Affiliation(s)
- Nan Wang
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Congyu Liao
- Department of Radiology, Stanford University, Stanford, California, USA
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Xiaozhi Cao
- Department of Radiology, Stanford University, Stanford, California, USA
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Mark Nishimura
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | | | - Mahmut Yurt
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Mengze Gao
- Department of Radiology, Stanford University, Stanford, California, USA
| | - Daniel Abraham
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Cagan Alkan
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| | - Siddharth Srinivasan Iyer
- Department of Radiology, Stanford University, Stanford, California, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, Massachusetts, USA
| | - Zihan Zhou
- Department of Radiology, Stanford University, Stanford, California, USA
| | | | - Adam Kerr
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
- Cognitive and Neurobiological Imaging Center, Stanford University, Stanford, California, USA
| | - Justin P Haldar
- Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, California, USA
| | - Kawin Setsompop
- Department of Radiology, Stanford University, Stanford, California, USA
- Department of Electrical Engineering, Stanford University, Stanford, California, USA
| |
Collapse
|
5
|
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
|
6
|
Bai M, Xiong Z, Zhang Y, Wang Z, Zeng X. Associations between quantitative susceptibility mapping with male obstructive sleep apnea clinical and imaging markers. Sleep Med 2024; 124:154-161. [PMID: 39303362 DOI: 10.1016/j.sleep.2024.09.019] [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: 06/07/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE To quantitatively measure and compare whole-brain iron deposition between OSA patients and a healthy control group, we initially utilized QSM and evaluated its correlation with PSG results and cognitive function. MATERIALS AND METHODS A total of 28 OSA patients and 22 healthy control subjects matched in age, education level, and BMI were enrolled in our study. Each participant underwent scanning with 3D T1 and multi-echo GRE sequences. Additionally, PSG results were collected from OSA patients, and they underwent simple cognitive assessments. Finally, we analyzed the relationship between iron content in different brain regions, PSG results, and cognitive ability. RESULTS In OSA patients, iron content increased in the left temporal-pole-sup and right putamen, while it decreased in the left fusiform gyrus, left middle temporal gyrus, right inferior occipital gyrus, and right superior temporal gyrus. The correlation analysis between brain iron content and PSG results/cognitive scales is as follows: left fusiform gyrus and MMSE (r = -0.416, p = 0.028); right superior temporal gyrus and MMSE (r = 0.422, p = 0.025); left middle temporal gyrus and average oxygen saturation (r = -0.418, p = 0.027); left temporal-pole-sup and REM stage (rs = 0.466, p = 0.012); the right putamen and N1 stage (rs = 0.393. p = 0.039). Moreover, both MoCA (r = 0.598, p = 0.001) and MMSE (r = 0.456, p = 0.015) show a positive correlation with average oxygen saturation. CONCLUSION This study is the first to use QSM technology to show abnormal brain iron levels in OSA. Correlations between brain iron content, PSG, and cognition in OSA may reveal neuropathological mechanisms, aiding OSA diagnosis.
Collapse
Affiliation(s)
- Mingxian Bai
- GuiZhou University Medical College, Guiyang, China; Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zhenliang Xiong
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China; College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Yan Zhang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China; College of Computer Science and Technology, Guizhou University, Guiyang, China
| | - Zhongxin Wang
- Department of Pulmonary and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xianchun Zeng
- GuiZhou University Medical College, Guiyang, China; Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, China.
| |
Collapse
|
7
|
Cai LM, Zeng JY, Huang HW, Tang Y, Li D, Li JQ, Chen HJ. Quantitative susceptibility mapping reveals brain iron accumulation in minimal hepatic encephalopathy: associations with neurocognitive changes. Metab Brain Dis 2024; 40:22. [PMID: 39565400 DOI: 10.1007/s11011-024-01440-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/20/2024] [Indexed: 11/21/2024]
Abstract
Brain iron deposition is correlated with minimal hepatic encephalopathy (MHE). This study aimed to investigate the pattern of altered iron distribution, using quantitative susceptibility mapping (QSM), and to clarify the relationship between iron deposition and neurocognitive changes in MHE. We enrolled 32 cirrhotic patients without MHE (NHE), 21 cirrhotic patients with MHE, and 24 healthy controls, and used the Psychometric Hepatic Encephalopathy Score (PHES) to assess neurocognitive function. All participants underwent magnetic resonance scans with a gradient-echo sequence reconstructing for QSM. We performed voxel-wise and region-of-interest (ROI)-wise analyses to investigate the QSM difference across three groups and to examine the relationship between susceptibility value and PHES. MHE patients exhibited increased susceptibility value in widespread brain areas (family-wise error (FWE)-corrected P < 0.05), which was located mainly in cognition-related regions (such as the prefrontal lobe, precuneus, inferior parietal lobule, insula, thalamus, and superior longitudinal fasciculus), sensorimotor regions (such as the precentral/postcentral gyrus, superior parietal lobule, and posterior corona radiata), visual regions (such as the occipital cortex and posterior thalamic radiation), and auditory regions (such as the temporal lobe). NHE patients also followed a trend of increasing susceptibility in the scattered brain regions, but which did not reach statistical significance (FWE-corrected P > 0.05). We observed negative correlations between cirrhotic patients' PHES and regional susceptibility values (FWE-corrected P < 0.05). Brain iron accumulation (measured using QSM) contributes to cognitive impairments in MHE patients. QSM could provide new insights into the pathogenesis of MHE and facilitate monitoring disease development.
Collapse
Affiliation(s)
- Li-Min Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jing-Yi Zeng
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Hui-Wei Huang
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Ying Tang
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, 200062, China
| | - Dan Li
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Jian-Qi Li
- Shanghai Key Laboratory of Magnetic Resonance, School of Physics and Electronic Science, East China Normal University, Shanghai, 200062, China.
| | - Hua-Jun Chen
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
8
|
Mi X, Li M, Zhang Y, Qu L, Xu A, Xie J, Song N. Intracerebroventricular injection of α-synuclein preformed fibrils do not induce motor and olfactory impairment in C57BL/6 mice. Neuroscience 2024; 559:293-301. [PMID: 39251058 DOI: 10.1016/j.neuroscience.2024.09.014] [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: 03/26/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Alpha-synuclein (αSyn) is believed to play a central role in the pathogenesis of Parkinson's disease (PD). Cerebrospinal fluid (CSF) total αSyn were significantly lower in PD patients, whereas the aggregates were higher, and this phenomenon was further exacerbated with longer disease duration. However, whether CSF αSyn can be the cause and/or a consequence in PD is not fully elucidated. METHOD We administered 2 ng or 200 ng αSyn preformed fibrils (PFFs) by intracerebroventricular injection for consecutive 7 days in C57BL/6 mice. The olfactory function was assessed by the olfactory discrimination test and buried food-seeking test. The locomotor function was assessed by the rotarod test, pole test, open field test and CatWalk gait analysis. Phosphorylated αSyn at serine 129 was detected by the immunohistochemistry staining. Iron levels was determined by Perl's-diaminobenzidine iron staining and synchrotron-based X-ray fluorescence. RESULTS The mice did not exhibit any diffuse synucleinopathy in the brain for up to 30 weeks, although αSyn PFFs induced aggregation in SH-SY5Y cells and in the substantia nigra and striatum of mice with stereotactic injection. No impairment of motor behaviors or olfactory functions were observed, although there was a temporary motor enhancement at 1 week. We then demonstrated iron levels were comparable in certain brain regions, suggesting there was no iron deposition/redistribution occurred. CONCLUSION The intraventricular injection of αSyn PFFs does not induce synucleinopathy or behavioral symptoms. These findings have implications that CSF αSyn aggregates may not necessarily contribute to the onset or progression in PD.
Collapse
Affiliation(s)
- Xiaoqing Mi
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Mengyu Li
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Yaru Zhang
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Le Qu
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Aoyang Xu
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Junxia Xie
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China.
| | - Ning Song
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
9
|
Qian ZM, Li W, Guo Q. Lactoferrin/lactoferrin receptor: Neurodegenerative or neuroprotective in Parkinson's disease? Ageing Res Rev 2024; 101:102474. [PMID: 39197711 DOI: 10.1016/j.arr.2024.102474] [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: 07/11/2024] [Revised: 08/04/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Lactoferrin (Lf) is a multifunctional protein in the transferrin family. It is involved in many physiological functions, including the regulation of iron absorption and immune response. It also has antibacterial, antiviral, anti-inflammatory, anticancer and antioxidant capabilities under pathophysiological conditions. The mammalian lactoferrin receptor (LfR) plays a key role in mediating multiple functions of Lf. Studies have shown that Lf/LfR is abnormally expressed in the brain of Parkinson's disease, and the excessive accumulation of iron in the brain caused by the overexpression of Lf and LfR is considered to be one of the initial causes of the degeneration of dopaminergic neurons in Parkinson's disease. On the other hand, a number of recent studies have reported that Lf/LfR has a significant neuroprotective effect on Parkinson's disease. In other words, it seems paradoxical that Lf/LfR has both neurodegenerative and neuroprotective effects in Parkinson's disease. This article focuses on recent advances in the possible mechanisms of the neurodegenerative and neuroprotective effects of Lf/LfR in Parkinson's disease and discusses why Lf/LfR has a seemingly contradictory role in the development of Parkinson's disease. Based on the evidence obtained so far, we believed that Lf/LfR has a neuroprotective effect on Parkinson's disease, while as to whether the overexpressed Lf/LfR is the cause of the development of Parkinson's disease, the current evidence is insufficient and further investigation needed.
Collapse
Affiliation(s)
- Zhong-Ming Qian
- Shanghai 411 Hospital, China RongTong Medical Healthcare Group Co.Ltd. / 411 Hospital, Shanghai University, Shanghai, China; Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong 226001, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Wei Li
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong 226001, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qian Guo
- Shanghai 411 Hospital, China RongTong Medical Healthcare Group Co.Ltd. / 411 Hospital, Shanghai University, Shanghai, China; Laboratory of Drug Delivery, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, China.
| |
Collapse
|
10
|
Straumann N, Combes BF, Dean Ben XL, Sternke‐Hoffmann R, Gerez JA, Dias I, Chen Z, Watts B, Rostami I, Shi K, Rominger A, Baumann CR, Luo J, Noain D, Nitsch RM, Okamura N, Razansky D, Ni R. Visualizing alpha-synuclein and iron deposition in M83 mouse model of Parkinson's disease in vivo. Brain Pathol 2024; 34:e13288. [PMID: 38982662 PMCID: PMC11483525 DOI: 10.1111/bpa.13288] [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: 12/23/2023] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Abnormal alpha-synuclein (αSyn) and iron accumulation in the brain play an important role in Parkinson's disease (PD). Herein, we aim to visualize αSyn inclusions and iron deposition in the brains of M83 (A53T) mouse models of PD in vivo. The fluorescent pyrimidoindole derivative THK-565 probe was characterized by means of recombinant fibrils and brains from 10- to 11-month-old M83 mice. Concurrent wide-field fluorescence and volumetric multispectral optoacoustic tomography (vMSOT) imaging were subsequently performed in vivo. Structural and susceptibility weighted imaging (SWI) magnetic resonance imaging (MRI) at 9.4 T as well as scanning transmission x-ray microscopy (STXM) were performed to characterize the iron deposits in the perfused brains. Immunofluorescence and Prussian blue staining were further performed on brain slices to validate the detection of αSyn inclusions and iron deposition. THK-565 showed increased fluorescence upon binding to recombinant αSyn fibrils and αSyn inclusions in post-mortem brain slices from patients with PD and M83 mice. Administration of THK-565 in M83 mice showed higher cerebral retention at 20 and 40 min post-intravenous injection by wide-field fluorescence compared to nontransgenic littermate mice, in congruence with the vMSOT findings. SWI/phase images and Prussian blue indicated the accumulation of iron deposits in the brains of M83 mice, presumably in the Fe3+ form, as evinced by the STXM results. In conclusion, we demonstrated in vivo mapping of αSyn by means of noninvasive epifluorescence and vMSOT imaging and validated the results by targeting the THK-565 label and SWI/STXM identification of iron deposits in M83 mouse brains ex vivo.
Collapse
Affiliation(s)
- Nadja Straumann
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Benjamin F. Combes
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Xose Luis Dean Ben
- Institute for Biomedical EngineeringUniversity of Zurich & ETH ZurichZurichSwitzerland
| | | | - Juan A. Gerez
- Laboratory of Physical Chemistry, Department of Chemistry and Applied BiosciencesETH ZurichZurichSwitzerland
| | - Ines Dias
- Neurology DepartmentUniversity Hospital ZurichZurichSwitzerland
| | - Zhenyue Chen
- Institute for Biomedical EngineeringUniversity of Zurich & ETH ZurichZurichSwitzerland
| | - Benjamin Watts
- Photon Science DivisionPaul Scherrer InstituteVilligenSwitzerland
| | - Iman Rostami
- Microscopic Anatomy and Structural BiologyUniversity of BernBernSwitzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, InselspitalBern University Hospital, University of BernBernSwitzerland
| | - Axel Rominger
- Department of Nuclear Medicine, InselspitalBern University Hospital, University of BernBernSwitzerland
| | | | - Jinghui Luo
- Department of Biology and ChemistryPaul Scherrer InstituteVilligenSwitzerland
| | - Daniela Noain
- Neurology DepartmentUniversity Hospital ZurichZurichSwitzerland
| | - Roger M. Nitsch
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of MedicineTohoku Medical and Pharmaceutical UniversitySendaiJapan
| | - Daniel Razansky
- Institute for Biomedical EngineeringUniversity of Zurich & ETH ZurichZurichSwitzerland
| | - Ruiqing Ni
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Institute for Biomedical EngineeringUniversity of Zurich & ETH ZurichZurichSwitzerland
- Department of Nuclear Medicine, InselspitalBern University Hospital, University of BernBernSwitzerland
| |
Collapse
|
11
|
Pirozzi MA, Canna A, Nardo FD, Sansone M, Trojsi F, Cirillo M, Esposito F. Reliability of quantitative magnetic susceptibility imaging metrics for cerebral cortex and major subcortical structures. J Neuroimaging 2024; 34:720-731. [PMID: 39210534 DOI: 10.1111/jon.13234] [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: 06/28/2024] [Revised: 08/02/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Susceptibility estimates derived from quantitative susceptibility mapping (QSM) images for the cerebral cortex and major subcortical structures are variably reported in brain magnetic resonance imaging (MRI) studies, as average of all (μ all ${{{{\mu}}}_{{\mathrm{all}}}}$ ), absolute (μ abs ${{{{\mu}}}_{{\mathrm{abs}}}}$ ), or positive- (μ p ${{{{\mu}}}_{\mathrm{p}}}$ ) and negative-only (μ n ${{{{\mu}}}_{\mathrm{n}}}$ ) susceptibility values using a region of interest (ROI) approach. This pilot study presents a reliability analysis of currently used ROI-QSM metrics and an alternative ROI-based approach to obtain voxel-weighted ROI-QSM metrics (μ wp ${{{{\mu}}}_{{\mathrm{wp}}}}$ andμ wn ${{{{\mu}}}_{{\mathrm{wn}}}}$ ). METHODS Ten healthy subjects underwent repeated (test-retest) 3-dimensional multi-echo gradient-echo (3DMEGE) 3 Tesla MRI measurements. Complex-valued 3DMEGE images were acquired and reconstructed with slice thicknesses of 1 and 2 mm (3DMEGE1, 3DMEGE2) along with 3DT1-weighted isometric (voxel 1 mm3) images for independent registration and ROI segmentation. Agreement, consistency, and reproducibility of ROI-QSM metrics were assessed through Bland-Altman analysis, intraclass correlation coefficient, and interscan and intersubject coefficient of variation (CoV). RESULTS All ROI-QSM metrics exhibited good to excellent consistency and test-retest agreement with no proportional bias. Interscan CoV was higher forμ all ${{{{\mu}}}_{{\mathrm{all}}}}$ in comparison to the other metrics where it was below 15%, in both 3DMEGE1 and 3DMEGE2 datasets. Intersubject CoV forμ all ${{{{\mu}}}_{{\mathrm{all}}}}$ andμ abs ${{{{\mu}}}_{{\mathrm{abs}}}}$ exceeded 50% in all ROIs. CONCLUSIONS Among the evaluated ROI-QSM metrics,μ all ${{{{\mu}}}_{{\mathrm{all}}}}$ andμ abs ${{{{\mu}}}_{{\mathrm{abs}}}}$ estimates were less reliable, whereas separating positive and negative values (usingμ p , μ n , μ wp , μ wn ${{{{\mu}}}_{\mathrm{p}}},\ {{{{\mu}}}_{\mathrm{n}}},\ {{{{\mu}}}_{{\mathrm{wp}}}},\ {{{{\mu}}}_{{\mathrm{wn}}}}$ ) improved the reproducibility within, and the comparability between, subjects, even when reducing the slice thickness. These preliminary findings may offer valuable insights toward standardizing ROI-QSM metrics across different patient cohorts and imaging settings in future clinical MRI studies.
Collapse
Affiliation(s)
- Maria Agnese Pirozzi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonietta Canna
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Federica Di Nardo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Sansone
- Department of Electrical Engineering and Information Technologies, University of Naples "Federico II", Naples, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Cirillo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fabrizio Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
12
|
Patrick K, Cousins E, Spitznagel MB. Associations between cognitive screening performance and motor symptoms in Parkinson's disease:a systematic review and meta-analysis. Dement Neuropsychol 2024; 18:e20230102. [PMID: 39258165 PMCID: PMC11386525 DOI: 10.1590/1980-5764-dn-2023-0102] [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: 03/22/2024] [Accepted: 04/27/2024] [Indexed: 09/12/2024] Open
Abstract
Although the most prominent symptoms of Parkinson's disease (PD) are those impacting movement, cognitive dysfunction is prevalent and often presents early in the disease process. Individuals with cognitive symptoms of PD often complete cognitive screening, making it important to identify factors associated with cognitive screening performance to ensure prompt and accurate detection of cognitive impairments. Objective Despite a body of research examining relationships between motor symptoms and cognitive dysfunction in PD, no prior study has undertaken a systematic review of the magnitude of the relationship between motor symptoms and cognitive screening performance in PD. Methods This study was a systematic review and meta-analysis of the relationship between cognitive screening performance, as assessed by the Montreal Cognitive Assessment (MoCA), and motor symptoms of PD. After the systematic screening, 20 studies were included, and meta-regressions using mixed-effects models were conducted. Results Motor symptoms across included studies were relatively mild, but average MoCA scores were at the established cutoff for risk of dementia in PD. The average disease duration was 5 years. Consistent with hypotheses, more severe motor symptoms were associated with lower MoCA scores (r=-0.22 (95%CI -0.29 to -0.16), p<0.001), indicating worse cognitive functioning. Conclusion The results indicate a significant negative correlation between MoCA performance and motor symptoms of PD. Average MoCA scores captured early disease-stage cognitive impairment when motor symptoms remained relatively mild. Serial screening for cognitive impairment beginning early in the disease course may be of benefit to ensure that cognitive dysfunction is detected as it arises.
Collapse
Affiliation(s)
- Karlee Patrick
- Kent State University, College of Arts and Sciences, Department of Psychological Sciences, Kent, Ohio, USA
| | - Elizabeth Cousins
- Kent State University, College of Arts and Sciences, Department of Psychological Sciences, Kent, Ohio, USA
| | - Mary Beth Spitznagel
- Kent State University, College of Arts and Sciences, Department of Psychological Sciences, Kent, Ohio, USA
| |
Collapse
|
13
|
Spence H, Mengoa-Fleming S, Sneddon AA, McNeil CJ, Waiter GD. Associations between sex, systemic iron and inflammatory status and subcortical brain iron. Eur J Neurosci 2024; 60:5069-5085. [PMID: 39113267 DOI: 10.1111/ejn.16467] [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: 01/12/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
Brain iron increases in several neurodegenerative diseases are associated with disease progression. However, the causes of increased brain iron remain unclear. This study investigates relationships between subcortical iron, systemic iron and inflammatory status. Brain magnetic resonance imaging (MRI) scans and blood plasma samples were collected from cognitively healthy females (n = 176, mean age = 61.4 ± 4.5 years, age range = 28-72 years) and males (n = 152, mean age = 62.0 ± 5.1 years, age range = 32-74 years). Regional brain iron was quantified using quantitative susceptibility mapping. To assess systemic iron, haematocrit, ferritin and soluble transferrin receptor were measured, and total body iron index was calculated. To assess systemic inflammation, C-reactive protein (CRP), neutrophil:lymphocyte ratio (NLR), macrophage colony-stimulating factor 1 (MCSF), interleukin 6 (IL6) and interleukin 1β (IL1β) were measured. We demonstrated that iron levels in the right hippocampus were higher in males compared with females, while iron in the right caudate was higher in females compared with males. There were no significant associations observed between subcortical iron levels and blood markers of iron and inflammatory status indicating that such blood measures are not markers of brain iron. These results suggest that brain iron may be regulated independently of blood iron and so directly targeting global iron change in the treatment of neurodegenerative disease may have differential impacts on blood and brain iron.
Collapse
Affiliation(s)
- Holly Spence
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Stephanie Mengoa-Fleming
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Christopher J McNeil
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Gordon D Waiter
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
14
|
Mohammadi S, Ghaderi S, Mohammadi H, Fatehi F. Simultaneous Increase of Mean Susceptibility and Mean Kurtosis in the Substantia Nigra as an MRI Neuroimaging Biomarker for Early-Stage Parkinson's Disease: A Systematic Review and Meta-Analysis. J Magn Reson Imaging 2024. [PMID: 39210501 DOI: 10.1002/jmri.29569] [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: 06/13/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disorder. Early detection is crucial for treatment and slowing disease progression. HYPOTHESIS Simultaneous alterations in mean susceptibility (MS) from quantitative susceptibility mapping (QSM) and mean kurtosis (MK) from diffusion kurtosis imaging (DKI) can serve as reliable neuroimaging biomarkers for early-stage PD (ESPD) in the basal ganglia nuclei, including the substantia nigra (SN), putamen (PUT), globus pallidus (GP), and caudate nucleus (CN). STUDY TYPE Systematic review and meta-analysis. POPULATION One hundred eleven patients diagnosed with ESPD and 81 healthy controls (HCs) were included from four studies that utilized both QSM and DKI in both subject groups. FIELD STRENGTH/SEQUENCE Three-dimensional multi-echo gradient echo sequence for QSM and spin echo planar imaging sequence for DKI at 3 Tesla. ASSESSMENT A systematic review and meta-analysis using PRISMA guidelines searched PubMed, Web of Science, and Scopus. STATISTICAL TESTS Random-effects model, standardized mean difference (SMD) to compare MS and MK between ESPD patients and HCs, I2 statistic for heterogeneity, Newcastle-Ottawa Scale (NOS) for risk of bias, and Egger's test for publication bias. A P-value <0.05 was considered significant. RESULTS MS values were significantly higher in SN (SMD 0.72, 95% CI 0.31 to 1.12), PUT (SMD 0.68, 95% CI 0.29 to 1.07), and GP (SMD 0.53, 95% CI 0.19 to 0.87) in ESPD patients compared to HCs. CN did not show a significant difference in MS values (P = 0.15). MK values were significantly higher only in SN (SMD = 0.72, 95% CI 0.16 to 1.27). MK values were not significantly different in PUT (P = 1.00), GP (P = 0.97), and CN (P = 0.59). Studies had high quality (NOS 7-8) and no publication bias (P = 0.967). DATA CONCLUSION Simultaneous use of MS and MK may be useful as an early neuroimaging biomarker for ESPD detection and its differentiation from HCs, with significant differences observed in the SN. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Ghaderi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Mohammadi
- Department of Bioimaging, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences (IUMS), Isfahan, Iran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Neurology Department, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
15
|
Yang M, Tang C, Peng F, Luo C, Chen G, Kong R, Peng P. Abdominal multi-organ iron content and the risk of Parkinson's disease: a Mendelian randomization study. Front Aging Neurosci 2024; 16:1416014. [PMID: 39206119 PMCID: PMC11349543 DOI: 10.3389/fnagi.2024.1416014] [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: 04/11/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Background To evaluate the causal relationship between abdominal multi-organ iron content and PD risk using publicly available genome-wide association study (GWAS) data. Methods We conducted MR analysis to assess the effects of iron content in various abdominal organs on PD risk, followed by reverse analysis. Additionally, MVMR analysis evaluated the independent effects of organ-specific iron content on PD. We utilized genetic variation data from the UK Biobank, including liver iron content (n = 32,858), spleen iron content (n = 35,324), and pancreas iron content (n = 25,617), as well as summary-level data for Parkinson's disease from the FinnGen (n = 218,473) and two other large GWAS datasets of European populations (First dataset n = 480,018; Second dataset n = 2,829). The primary MR analysis used the inverse variance-weighted (IVW) method, confirmed by MR-Egger and weighted median methods. Sensitivity analysis was performed to address potential pleiotropy and heterogeneity. Observational cohort results were validated through replication cohort analysis, followed by meta-analysis. Results IVW analysis revealed a causal relationship between increased liver iron content and elevated risk of PD (OR = 1.27; 95% CI: 1.05-1.53; p = 0.015). No significant causal relationship was observed between spleen (OR = 1.00; 95% CI: 0.76-1.32; p = 0.983) and pancreatic (OR = 0.93; 95% CI: 0.72-1.20; p = 0.573) iron content and increased risk of PD. Meta-analysis of GWAS data for PD from three different sources using the random-effects IVW method showed a statistically significant causal relationship between liver iron content and the occurrence of PD (OR = 1.17, 95% CI: 1.01-1.35; p = 0.012). Conclusion This study presents evidence from Mendelian randomization (MR) analysis indicating a significant causal link between increased liver iron content and a higher risk of Parkinson's disease (PD). These findings suggest that interventions targeting body iron metabolism, particularly liver iron levels, may be effective in preventing PD.
Collapse
Affiliation(s)
- Mingrui Yang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Cheng Tang
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fei Peng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chaotian Luo
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Guowei Chen
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rong Kong
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Peng Peng
- Department of Radiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- NHC Key Laboratory of Thalassemia Medicine, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
16
|
Ma ZL, Wang ZL, Zhang FY, Liu HX, Mao LH, Yuan L. Biomarkers of Parkinson's Disease: From Basic Research to Clinical Practice. Aging Dis 2024; 15:1813-1830. [PMID: 37815899 PMCID: PMC11272192 DOI: 10.14336/ad.2023.1005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized pathologically by dopaminergic neuron loss and the formation of Lewy bodies, which are enriched with aggregated α-synuclein (α-syn). PD currently has no cure, but therapeutic strategies are available to alleviate symptoms. Early diagnosis can greatly improve therapeutic interventions, but the clinical diagnosis of PD remains challenging and depends mainly on clinical features and imaging tests. Efficient and specific biomarkers are crucial for the diagnosis, monitoring, and evaluation of PD. Here, we reviewed the biomarkers of PD in different tissues and biofluids, along with the current clinical biochemical detection methods. We found that the sensitivity and specificity of single biomarkers are limited, and selecting appropriate indicators for combined detection can improve the diagnostic accuracy of PD.
Collapse
Affiliation(s)
| | | | - Fei-yue Zhang
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Hong-xun Liu
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Li-hong Mao
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| | - Lin Yuan
- Laboratory of Research in Parkinson’s Disease and Related Disorders, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute, China Medical University, Shenyang, China
| |
Collapse
|
17
|
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
|
18
|
Zarkali A, Thomas GEC, Zetterberg H, Weil RS. Neuroimaging and fluid biomarkers in Parkinson's disease in an era of targeted interventions. Nat Commun 2024; 15:5661. [PMID: 38969680 PMCID: PMC11226684 DOI: 10.1038/s41467-024-49949-9] [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: 07/26/2023] [Accepted: 06/19/2024] [Indexed: 07/07/2024] Open
Abstract
A major challenge in Parkinson's disease is the variability in symptoms and rates of progression, underpinned by heterogeneity of pathological processes. Biomarkers are urgently needed for accurate diagnosis, patient stratification, monitoring disease progression and precise treatment. These were previously lacking, but recently, novel imaging and fluid biomarkers have been developed. Here, we consider new imaging approaches showing sensitivity to brain tissue composition, and examine novel fluid biomarkers showing specificity for pathological processes, including seed amplification assays and extracellular vesicles. We reflect on these biomarkers in the context of new biological staging systems, and on emerging techniques currently in development.
Collapse
Affiliation(s)
- Angeliki Zarkali
- Dementia Research Centre, Institute of Neurology, UCL, London, UK.
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Rimona S Weil
- Dementia Research Centre, Institute of Neurology, UCL, London, UK
- Department of Advanced Neuroimaging, UCL, London, UK
- Movement Disorders Centre, UCL, London, UK
| |
Collapse
|
19
|
Xi J, Huang Y, Bao L. Quantitative susceptibility mapping based basal ganglia segmentation via AGSeg: leveraging active gradient guiding mechanism in deep learning. Quant Imaging Med Surg 2024; 14:4417-4435. [PMID: 39022266 PMCID: PMC11250355 DOI: 10.21037/qims-23-1858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/14/2024] [Indexed: 07/20/2024]
Abstract
Background With better visual contrast and the ability for magnetic susceptibility quantification analysis, quantitative susceptibility mapping (QSM) has emerged as an important magnetic resonance imaging (MRI) method for basal ganglia studies. Precise segmentation of basal ganglia is a prerequisite for quantification analysis of tissue magnetic susceptibility, which is crucial for subsequent disease diagnosis and surgical planning. The conventional method of localizing and segmenting basal ganglia heavily relies on layer-by-layer manual annotation by experts, resulting in a tedious amount of workload. Although several morphology registration and deep learning based methods have been developed to automate segmentation, the voxels around the nuclei boundary remain a challenge to distinguish due to insufficient tissue contrast. This paper proposes AGSeg, an active gradient guidance-based susceptibility and magnitude information complete (MIC) network for real-time and accurate basal ganglia segmentation. Methods Various datasets, including clinical scans and data from healthy volunteers, were collected across multiple centers with different magnetic field strengths (3T/5T/7T), with a total of 210 three-dimensional (3D) susceptibility measurements. Manual segmentations following fixed rules for anatomical borders annotated by experts were used as ground truth labels. The proposed network took QSM maps and Magnitude images as two individual inputs, of which the features are selectively enhanced in the proposed magnitude information complete (MIC) module. AGSeg utilized a dual-branch architecture, with Seg-branch aiming to generate a proper segmentation map and Grad-branch to reconstruct the gradient map of regions of interest (ROIs). With the support of the newly designed active gradient module (AGM) and gradient guiding module (GGM), the Grad-branch provided attention guidance for the Seg-branch, facilitating it to focus on the boundary of target nuclei. Results Ablation studies were conducted to assess the functionality of the proposed modules. Significant performance decrement was observed after ablating relative modules. AGSeg was evaluated against several existing methods on both healthy and clinical data, achieving an average Dice similarity coefficient (DSC) =0.874 and average 95% Hausdorff distance (HD95) =2.009. Comparison experiments indicated that our model had superior performance on basal ganglia segmentation and better generalization ability over existing methods. The AGSeg outperformed all implemented comparison deep learning algorithms with average DSC enhancement ranging from 0.036 to 0.074. Conclusions The current work integrates a deep learning-based method into automated basal ganglia segmentation. The high processing speed and segmentation robustness of AGSeg contribute to the feasibility of future surgery planning and intraoperative navigation. Experiments show that leveraging active gradient guidance mechanisms and magnitude information completion can facilitate the segmentation process. Moreover, this approach also offers a portable solution for other multi-modality medical image segmentation tasks.
Collapse
Affiliation(s)
- Jiaxiu Xi
- Department of Electronic Science, Xiamen University, Xiamen, China
| | - Yuqing Huang
- Department of Electronic Science, Xiamen University, Xiamen, China
| | - Lijun Bao
- Department of Electronic Science, Xiamen University, Xiamen, China
| |
Collapse
|
20
|
Zhang M, Feng R, Li Z, Feng J, Wu Q, Zhang Z, Ma C, Wu J, Yan F, Liu C, Zhang Y, Wei H. A subject-specific unsupervised deep learning method for quantitative susceptibility mapping using implicit neural representation. Med Image Anal 2024; 95:103173. [PMID: 38657424 DOI: 10.1016/j.media.2024.103173] [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: 10/09/2023] [Revised: 03/11/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024]
Abstract
Quantitative susceptibility mapping (QSM) is an MRI-based technique that estimates the underlying tissue magnetic susceptibility based on phase signal. Deep learning (DL)-based methods have shown promise in handling the challenging ill-posed inverse problem for QSM reconstruction. However, they require extensive paired training data that are typically unavailable and suffer from generalization problems. Recent model-incorporated DL approaches also overlook the non-local effect of the tissue phase in applying the source-to-field forward model due to patch-based training constraint, resulting in a discrepancy between the prediction and measurement and subsequently suboptimal QSM reconstruction. This study proposes an unsupervised and subject-specific DL method for QSM reconstruction based on implicit neural representation (INR), referred to as INR-QSM. INR has emerged as a powerful framework for learning a high-quality continuous representation of the signal (image) by exploiting its internal information without training labels. In INR-QSM, the desired susceptibility map is represented as a continuous function of the spatial coordinates, parameterized by a fully-connected neural network. The weights are learned by minimizing a loss function that includes a data fidelity term incorporated by the physical model and regularization terms. Additionally, a novel phase compensation strategy is proposed for the first time to account for the non-local effect of tissue phase in data consistency calculation to make the physical model more accurate. Our experiments show that INR-QSM outperforms traditional established QSM reconstruction methods and the compared unsupervised DL method both qualitatively and quantitatively, and is competitive against supervised DL methods under data perturbations.
Collapse
Affiliation(s)
- Ming Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ruimin Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenghao Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Feng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Wu
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhiyong Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chengxin Ma
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinsong Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Yuyao Zhang
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hongjiang Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China; Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Jin J, Su D, Zhang J, Lam JST, Zhou J, Feng T. Iron deposition in subcortical nuclei of Parkinson's disease: A meta-analysis of quantitative iron-sensitive magnetic resonance imaging studies. Chin Med J (Engl) 2024:00029330-990000000-01086. [PMID: 38809051 DOI: 10.1097/cm9.0000000000003167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Iron deposition plays a crucial role in the pathophysiology of Parkinson's disease (PD), yet the distribution pattern of iron deposition in the subcortical nuclei has been inconsistent across previous studies. We aimed to assess the difference patterns of iron deposition detected by quantitative iron-sensitive magnetic resonance imaging (MRI) between patients with PD and patients with atypical parkinsonian syndromes (APSs), and between patients with PD and healthy controls (HCs). METHODS A systematic literature search was conducted on PubMed, Embase, and Web of Science databases to identify studies investigating the iron content in PD patients using the iron-sensitive MRI techniques (R2* and quantitative susceptibility mapping [QSM]), up until May 1, 2023. The quality assessment of case-control and cohort studies was performed using the Newcastle-Ottawa Scale, whereas diagnostic studies were assessed using the Quality Assessment of Diagnostic Accuracy Studies-2. Standardized mean differences and summary estimates of sensitivity, specificity, and area under the curve (AUC) were calculated for iron content, using a random effects model. We also conducted the subgroup-analysis based on the MRI sequence and meta-regression. RESULTS Seventy-seven studies with 3192 PD, 209 multiple system atrophy (MSA), 174 progressive supranuclear palsy (PSP), and 2447 HCs were included. Elevated iron content in substantia nigra (SN) pars reticulata (P <0.001) and compacta (P <0.001), SN (P <0.001), red nucleus (RN, P <0.001), globus pallidus (P <0.001), putamen (PUT, P = 0.009), and thalamus (P = 0.046) were found in PD patients compared with HCs. PD patients showed lower iron content in PUT (P <0.001), RN (P = 0.003), SN (P = 0.017), and caudate nucleus (P = 0.027) than MSA patients, and lower iron content in RN (P = 0.001), PUT (P <0.001), globus pallidus (P = 0.004), SN (P = 0.015), and caudate nucleus (P = 0.001) than PSP patients. The highest diagnostic accuracy distinguishing PD from HCs was observed in SN (AUC: 0.85), and that distinguishing PD from MSA was found in PUT (AUC: 0.90). In addition, the best diagnostic performance was achieved in the RN for distinguishing PD from PSP (AUC: 0.84). CONCLUSION Quantitative iron-sensitive MRI could quantitatively detect the iron content of subcortical nuclei in PD and APSs, while it may be insufficient to accurately diagnose PD. Future studies are needed to explore the role of multimodal MRI in the diagnosis of PD. REGISTRISION PROSPERO; CRD42022344413.
Collapse
Affiliation(s)
- Jianing Jin
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Dongning Su
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Junjiao Zhang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| | - Joyce S T Lam
- Pacific Parkinson's Research Centre, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Junhong Zhou
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Roslindale, MA 02131, United States
- Harvard Medical School, Boston, MA 02210, United States
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
| |
Collapse
|
22
|
Liu N, Yu W, Sun M, Zhou D, Sun J, Jiang T, Zhang W, Wang M. Research trends and hotspots of ferroptosis in neurodegenerative diseases from 2013 to 2023: A bibliometrics study. Heliyon 2024; 10:e29418. [PMID: 38638970 PMCID: PMC11024616 DOI: 10.1016/j.heliyon.2024.e29418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
Abstract
Background With the aging population, the incidence of neurodegenerative diseases increases yearly, seriously impacting human health. Various journals have published studies on the pathogenesis of ferroptosis in neurodegenerative diseases. However, bibliometric analysis in this field is still lacking. The study aims to visually analyze global research trends in this field over the past decade. Methods The articles and reviews regarding ferroptosis in neurodegenerative diseases were retrieved from the Web of Science on September 1, 2023. Citespace [version 6.2. R4 (64-bit)] and VOSviewer (version 1.6.18) were used to conduct the bibliometric and knowledge-map analysis. Results In total, 370 studies were included in the paper and ranked by their citation frequency. Many articles on ferroptosis in neurodegenerative diseases have been published in the past decade. The country, institution, author, and journal with the highest publications were China, Guangzhou Medical University, Maher, Pamela, and Free Radical Biology And Medicine, respectively. The analysis of keyword co-occurrence indicated that research frontiers were molecular mechanisms of ferroptosis in neurodegenerative diseases, especially a few key pathways that triggered ferroptosis in these diseases, including lipid peroxidation signaling, iron metabolism, and GSH/GPX4 signaling. In addition, ferroptosis inhibitors such as liproxstatins and ferrostatins had protective effects in animal models of neurodegenerative diseases. Therefore, future attention should also be focused on therapeutic drugs that target ferroptosis. Conclusion This study comprehensively analyzed the publications on ferroptosis in neurodegenerative diseases from a bibliometric perspective. Research on this topic is currently expanding at a rapid pace, and the China holds a leading position in this field by its scientific achievements and productivity. Moreover, the research frontiers were molecular mechanisms of ferroptosis in neurodegenerative diseases and developing targeted therapeutic drugs. In summary, our results showed an all-sided overview of the knowledge atlas and a valuable reference for the future research in this field.
Collapse
Affiliation(s)
- Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wuhan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, China
- Department of Anus and Intestine Surgery, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, 523000, China
| | - Mengjiao Sun
- Capital Medical University, Beijing, 100000, China
| | - Dan Zhou
- Department of Neurology, Xi ‘an Ninth Hospital, Xi ‘an, 710000, China
| | - Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Taotao Jiang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Wenjing Zhang
- Department of Neurology, Qinghai Provincial People's Hospital, Qinghai, 810000, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, 730030, China
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Mohammadi S, Ghaderi S. Parkinson's disease and Parkinsonism syndromes: Evaluating iron deposition in the putamen using magnetic susceptibility MRI techniques - A systematic review and literature analysis. Heliyon 2024; 10:e27950. [PMID: 38689949 PMCID: PMC11059419 DOI: 10.1016/j.heliyon.2024.e27950] [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: 12/10/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 05/02/2024] Open
Abstract
Magnetic resonance imaging (MRI) techniques, such as quantitative susceptibility mapping (QSM) and susceptibility-weighted imaging (SWI), can detect iron deposition in the brain. Iron accumulation in the putamen (PUT) can contribute to the pathogenesis of Parkinson's disease (PD) and atypical Parkinsonian disorders. This systematic review aimed to synthesize evidence on iron deposition in the PUT assessed by MRI susceptibility techniques in PD and Parkinsonism syndromes. The PubMed and Scopus databases were searched for relevant studies. Thirty-four studies from January 2007 to October 2023 that used QSM, SWI, or other MRI susceptibility methods to measure putaminal iron in PD, progressive supranuclear palsy (PSP), multiple system atrophy (MSA), and healthy controls (HCs) were included. Most studies have found increased putaminal iron levels in PD patients versus HCs based on higher quantitative susceptibility. Putaminal iron accumulation correlates with worse motor scores and cognitive decline in patients with PD. Evidence regarding differences in susceptibility between PD and atypical Parkinsonism is emerging, with several studies showing greater putaminal iron deposition in PSP and MSA than in PD patients. Alterations in putaminal iron levels help to distinguish these disorders from PD. Increased putaminal iron levels appear to be associated with increased disease severity and progression. Thus, magnetic susceptibility MRI techniques can detect abnormal iron accumulation in the PUT of patients with Parkinsonism. Moreover, quantifying putaminal susceptibility may serve as an MRI biomarker to monitor motor and cognitive changes in PD and aid in the differential diagnosis of Parkinsonian disorders.
Collapse
Affiliation(s)
- Sana Mohammadi
- Department of Medical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sadegh Ghaderi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
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
|
27
|
Chen Y, Wu Z, Li S, Chen Q, Wang L, Qi X, Tian C, Yang M. Mapping the Research of Ferroptosis in Parkinson's Disease from 2013 to 2023: A Scientometric Review. Drug Des Devel Ther 2024; 18:1053-1081. [PMID: 38585257 PMCID: PMC10999190 DOI: 10.2147/dddt.s458026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
Methods Related studies on PD and ferroptosis were searched in Web of Science Core Collection (WOSCC) from inception to 2023. VOSviewer, CiteSpace, RStudio, and Scimago Graphica were employed as bibliometric analysis tools to generate network maps about the collaborations between authors, countries, and institutions and to visualize the co-occurrence and trends of co-cited references and keywords. Results A total of 160 original articles and reviews related to PD and ferroptosis were retrieved, produced by from 958 authors from 162 institutions. Devos David was the most prolific author, with 9 articles. China and the University of Melbourne had leading positions in publication volume with 84 and 12 publications, respectively. Current hot topics focus on excavating potential new targets for treating PD based on ferroptosis by gaining insight into specific molecular mechanisms, including iron metabolism disorders, lipid peroxidation, and imbalanced antioxidant regulation. Clinical studies aimed at treating PD by targeting ferroptosis remain in their preliminary stages. Conclusion A continued increase was shown in the literature within the related field over the past decade. The current study suggested active collaborations among authors, countries, and institutions. Research into the pathogenesis and treatment of PD based on ferroptosis has remained a prominent topic in the field in recent years, indicating that ferroptosis-targeted therapy is a potential approach to halting the progression of PD.
Collapse
Affiliation(s)
- Yingfan Chen
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Zhenhui Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, People’s Republic of China
| | - Shaodan Li
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Qi Chen
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Liang Wang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| | - Xiaorong Qi
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Chujiao Tian
- Medical School of Chinese People’s Liberation Army, Beijing, People’s Republic of China
| | - Minghui Yang
- Department of Traditional Chinese Medicine, the Six Medical Center of Chinese People’s Liberation Army General Hospital, Beijing, People’s Republic of China
| |
Collapse
|
28
|
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
|
29
|
Bissolotti L, Artiles-Sánchez J, Alonso-Pérez JL, Fernández-Carnero J, Abuín-Porras V, Sinatti P, Villafañe JH. Virtual Reality-Based Assessment for Rehabilitation of the Upper Limb in Patients with Parkinson's Disease: A Pilot Cross-Sectional Study. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:555. [PMID: 38674201 PMCID: PMC11051709 DOI: 10.3390/medicina60040555] [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: 02/16/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024]
Abstract
Background and Objectives: This study aimed to examine the responsiveness and concurrent validity of a serious game and its correlation between the use of serious games and upper limbs (UL) performance in Parkinson's Disease (PD) patients. Materials and Methods: Twenty-four consecutive upper limbs (14 males, 8 females, age: 55-83 years) of PD patients were assessed. The clinical assessment included: the Box and Block test (BBT), Nine-Hole Peg test (9HPT), and sub-scores of the Unified Parkinson's Disease Rating-Scale Motor section (UPDRS-M) to assess UL disability. Performance scores obtained in two different tests (Ex. A and Ex. B, respectively, the Trolley test and Mushrooms test) based on leap motion (LM) sensors were used to study the correlations with clinical scores. Results: The subjective fatigue experienced during LM tests was measured by the Borg Rating of Perceived Exertion (RPE, 0-10); the BBT and 9HPT showed the highest correlation coefficients with UPDRS-M scores (ICCs: -0.652 and 0.712, p < 0.05). Exercise A (Trolley test) correlated with UPDRS-M (ICC: 0.31, p < 0.05), but not with the 9HPT and BBT tests (ICCs: -0.447 and 0.390, p < 0.05), while Exercise B (Mushroom test) correlated with UPDRS-M (ICC: -0.40, p < 0.05), as did these last two tests (ICCs: -0.225 and 0.272, p < 0.05). The mean RPE during LM tests was 3.4 ± 3.2. The evaluation of upper limb performance is feasible and does not induce relevant fatigue. Conclusions: The analysis of the ICC supports the use of Test B to evaluate UL disability and performance in PD patients, while Test A is mostly correlated with disability. Specifically designed serious games on LM can serve as a method of impairment in the PD population.
Collapse
Affiliation(s)
- Luciano Bissolotti
- Fondazione Teresa Camplani Casa di Cura Domus Salutis, 25123 Brescia, Italy;
| | - Justo Artiles-Sánchez
- Musculoskeletal Pain and Motor Control Research Group, Faculty of Health Sciences, Universidad Europea de Canarias, C/Inocencio García 1, 38300 Santa Cruz de Tenerife, Spain;
- Department of Physiotherapy, Faculty of Health Sciences, Universidad Europea de Canarias, 38300 Santa Cruz de Tenerife, Spain
| | - José Luís Alonso-Pérez
- Musculoskeletal Pain and Motor Control Research Group, Faculty of Health Sciences, Universidad Europea de Canarias, C/Inocencio García 1, 38300 Santa Cruz de Tenerife, Spain;
- Department of Physiotherapy, Faculty of Health Sciences, Universidad Europea de Canarias, 38300 Santa Cruz de Tenerife, Spain
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain; (V.A.-P.); (P.S.)
- Musculoskeletal Pain and Motor Control Research Group, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain
- Onelife Center, Multidisciplinary Pain Treatment Center, 28925 Alcorcón, Spain
| | - Josué Fernández-Carnero
- Department of Physical Therapy, Occupational Therapy, Rehabilitation and Physical Medicine, Rey Juan Carlos University, 28032 Madrid, Spain;
- Grupo Multidisciplinar de Investigación y Tratamiento del Dolor, Grupo de Excelencia Investigadora URJC-Banco de Santander, 28922 Madrid, Spain
- Motion in Brains Research Group, Institute of Neuroscience and Movement Sciences (INCIMOV), Centro Superior de Estudios Universitarios La Salle, Universidad Autonoma de Madrid, 28049 Madrid, Spain
- La Paz Hospital Institute for Health Research, IdiPAZ, 28029 Madrid, Spain
| | - Vanesa Abuín-Porras
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain; (V.A.-P.); (P.S.)
| | - Pierluigi Sinatti
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain; (V.A.-P.); (P.S.)
| | - Jorge Hugo Villafañe
- Department of Physiotherapy, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain; (V.A.-P.); (P.S.)
- Musculoskeletal Pain and Motor Control Research Group, Faculty of Sport Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Spain
| |
Collapse
|
30
|
Zhang X, Hu Y, Wang B, Yang S. Ferroptosis: Iron-mediated cell death linked to disease pathogenesis. J Biomed Res 2024; 38:1-23. [PMID: 38808552 PMCID: PMC11461536 DOI: 10.7555/jbr.37.20230224] [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: 09/19/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 05/30/2024] Open
Abstract
Ferroptosis is an iron-mediated regulatory cell death pattern characterized by oxidative damage. The molecular regulating mechanisms are related to iron metabolism, lipid peroxidation, and glutathione metabolism. Additionally, some immunological signaling pathways, such as the cyclic GMP-AMP synthase-stimulator ofinterferon genes axis, Janus kinase-signal transducer and activator of transcription 1 axis, and transforming growth factor beta 1-Smad3 axis may also participate in the regulation of ferroptosis. Studies have shown that ferroptosis is closely related to many diseases such as cancer, neurodegenerative diseases, inflammatory diseases, and autoimmune diseases. Considering the pivotal role of ferroptosis-regulating signaling in the pathogenesis of diverse diseases, the development of ferroptosis inducers or inhibitors may have significant clinical potential for the treatment of the aforementioned conditions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingchao Hu
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Shuo Yang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Gusu School, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Alushaj E, Handfield-Jones N, Kuurstra A, Morava A, Menon RS, Owen AM, Sharma M, Khan AR, MacDonald PA. Increased iron in the substantia nigra pars compacta identifies patients with early Parkinson'sdisease: A 3T and 7T MRI study. Neuroimage Clin 2024; 41:103577. [PMID: 38377722 PMCID: PMC10944193 DOI: 10.1016/j.nicl.2024.103577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/19/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024]
Abstract
Degeneration in the substantia nigra (SN) pars compacta (SNc) underlies motor symptoms in Parkinson's disease (PD). Currently, there are no neuroimaging biomarkers that are sufficiently sensitive, specific, reproducible, and accessible for routine diagnosis or staging of PD. Although iron is essential for cellular processes, it also mediates neurodegeneration. MRI can localize and quantify brain iron using magnetic susceptibility, which could potentially provide biomarkers of PD. We measured iron in the SNc, SN pars reticulata (SNr), total SN, and ventral tegmental area (VTA), using quantitative susceptibility mapping (QSM) and R2* relaxometry, in PD patients and age-matched healthy controls (HCs). PD patients, diagnosed within five years of participation and HCs were scanned at 3T (22 PD and 23 HCs) and 7T (17 PD and 21 HCs) MRI. Midbrain nuclei were segmented using a probabilistic subcortical atlas. QSM and R2* values were measured in midbrain subregions. For each measure, groups were contrasted, with Age and Sex as covariates, and receiver operating characteristic (ROC) curve analyses were performed with repeated k-fold cross-validation to test the potential of our measures to classify PD patients and HCs. Statistical differences of area under the curves (AUCs) were compared using the Hanley-MacNeil method (QSM versus R2*; 3T versus 7T MRI). PD patients had higher QSM values in the SNc at both 3T (padj = 0.001) and 7T (padj = 0.01), but not in SNr, total SN, or VTA, at either field strength. No significant group differences were revealed using R2* in any midbrain region at 3T, though increased R2* values in SNc at 7T MRI were marginally significant in PDs compared to HCs (padj = 0.052). ROC curve analyses showed that SNc iron measured with QSM, distinguished early PD patients from HCs at the single-subject level with good diagnostic accuracy, using 3T (mean AUC = 0.83, 95 % CI = 0.82-0.84) and 7T (mean AUC = 0.80, 95 % CI = 0.79-0.81) MRI. Mean AUCs reported here are from averages of tests in the hold-out fold of cross-validated samples. The Hanley-MacNeil method demonstrated that QSM outperforms R2* in discriminating PD patients from HCs at 3T, but not 7T. There were no significant differences between 3T and 7T in diagnostic accuracy of QSM values in SNc. This study highlights the importance of segmenting midbrain subregions, performed here using a standardized atlas, and demonstrates high accuracy of SNc iron measured with QSM at 3T MRI in identifying early PD patients. QSM measures of SNc show potential for inclusion in neuroimaging diagnostic biomarkers of early PD. An MRI diagnostic biomarker of PD would represent a significant clinical advance.
Collapse
Affiliation(s)
- Erind Alushaj
- Department of Neuroscience, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 3K7, Canada; Western Institute for Neuroscience, Western University, London, Ontario N6A 3K7, Canada
| | - Nicholas Handfield-Jones
- Department of Neuroscience, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 3K7, Canada; Western Institute for Neuroscience, Western University, London, Ontario N6A 3K7, Canada
| | - Alan Kuurstra
- Robarts Research Institute, Western University, London, Ontario N6A 3K7, Canada; Department of Medical Biophysics, Western University, London, Ontario N6A 3K7, Canada
| | - Anisa Morava
- School of Kinesiology, Faculty of Health Sciences, Western University, London, Ontario N6A 3K7, Canada
| | - Ravi S Menon
- Robarts Research Institute, Western University, London, Ontario N6A 3K7, Canada; Department of Medical Biophysics, Western University, London, Ontario N6A 3K7, Canada
| | - Adrian M Owen
- Western Institute for Neuroscience, Western University, London, Ontario N6A 3K7, Canada; Department of Physiology and Pharmacology, Western University, London, Ontario N6A 3K7, Canada
| | - Manas Sharma
- Department of Radiology, Western University, London, Ontario N6A 3K7, Canada; Department of Clinical Neurological Sciences, Western University, London, Ontario N6A 3K7, Canada
| | - Ali R Khan
- Robarts Research Institute, Western University, London, Ontario N6A 3K7, Canada; Department of Medical Biophysics, Western University, London, Ontario N6A 3K7, Canada
| | - Penny A MacDonald
- Western Institute for Neuroscience, Western University, London, Ontario N6A 3K7, Canada; Department of Clinical Neurological Sciences, Western University, London, Ontario N6A 3K7, Canada.
| |
Collapse
|
34
|
Wang X, Zhu Z, Sun J, Jia L, Cai L, Chen Q, Yang W, Wang Y, Zhang Y, Guo S, Liu W, Yang Z, Zhao P, Wang Z, Lv H. Changes in iron load in specific brain areas lead to neurodegenerative diseases of the central nervous system. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110903. [PMID: 38036035 DOI: 10.1016/j.pnpbp.2023.110903] [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: 07/30/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
The causes of neurodegenerative diseases remain largely elusive, increasing their personal and societal impacts. To reveal the causal effects of iron load on Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis and multiple sclerosis, we used Mendelian randomisation and brain imaging data from a UK Biobank genome-wide association study of 39,691 brain imaging samples (predominantly of European origin). Using susceptibility-weighted images, which reflect iron load, we analysed genetically significant brain regions. Inverse variance weighting was used as the main estimate, while MR Egger and weighted median were used to detect heterogeneity and pleiotropy. Nine clear associations were obtained. For AD and PD, an increased iron load was causative: the right pallidum for AD and the right caudate, left caudate and right accumbens for PD. However, a reduced iron load was identified in the right and left caudate for multiple sclerosis, the bilateral hippocampus for mixed vascular dementia and the left thalamus and bilateral accumbens for subcortical vascular dementia. Thus, changes in iron load in different brain regions have causal effects on neurodegenerative diseases. Our results are crucial for understanding the pathogenesis and investigating the treatment of these diseases.
Collapse
Affiliation(s)
- Xinghao Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Zaimin Zhu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, People's Republic of China
| | - Jing Sun
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Li Jia
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Linkun Cai
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China; School of Biological Science and Medical Engineering, Beihang University, No.37 XueYuan Road, Beijing 100191, People's Republic of China
| | - Qian Chen
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Wenbo Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Yiling Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Yufan Zhang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Sihui Guo
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Wenjuan Liu
- Department of Radiology, Aerospace Center Hospital, Beijing, People's Republic of China; Peking University Aerospace School of Clinical Medicine, Beijing 100049, People's Republic of China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Pengfei Zhao
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China.
| | - Han Lv
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 YongAn Road, Beijing 100050, People's Republic of China.
| |
Collapse
|
35
|
Li QM, Xu T, Zha XQ, Feng XW, Zhang FY, Luo JP. Buddlejasaponin IVb ameliorates ferroptosis of dopaminergic neuron by suppressing IRP2-mediated iron overload in Parkinson's disease. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117196. [PMID: 37717841 DOI: 10.1016/j.jep.2023.117196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Parkinson's disease (PD) is the second neurodegenerative disease that lacks effective treatments. Buddlejasaponin IVb (BJP-IVb) is the main bioactive component of herbs in genus Clinopodium which display antioxidative, anti-inflammatory and neuroprotective activities. However, the role of BJP-IVb in PD still remains unknown. AIM OF THE STUDY This study aimed to evaluate the effect of BJP-IVb on dopaminergic neurodegeneration in PD and clarified the underlying mechanisms from the aspect of iron overload-mediated ferroptosis. MATERIALS AND METHODS One-methyl-4-phenylpyridinium (MPP+) and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD models were established in this study. Behavioral tests, cell cytotoxicity assay, tyrosine hydroxylase (TH) and Nissl staining were performed to evaluate the antiparkinsonian effect of BJP-IVb. Cellular ultrastructure, iron content and lipid peroxidation were detected to evaluate iron overload-mediated dopaminergic neuron ferroptosis. Iron regulatory protein 2 (IRP2) and iron transport-related proteins were detected by immunofluorescence and Western blot to evaluated iron transport. Finally, plasmid vector-mediated IRP2 overexpression were performed to further clarify the molecular mechanism. RESULTS BJP-IVb alleviated MPP+-induced neurotoxicity in vitro and improved MPTP-induced dopaminergic neuron loss and motor dysfunctions of PD mice, confirming an effect of BJP-IVb against dopaminergic neurodegeneration of PD. Further results revealed that BJP-IVb protected against PD by suppressing iron overload-mediated dopaminergic neuron ferroptosis, as evidenced by the attenuated lipid peroxidation, decreased iron content and changes in cellular ultrastructure. Finally, the decreased iron regulatory protein (IRP2) was confirmed to be responsible for BJP-IVb-mediated ferroptosis suppression by modulating iron transport-related proteins and alleviating iron overload. CONCLUSION BJP-IVb suppressed iron overload-mediated dopaminergic neuron ferroptosis and improved motor dysfunctions in PD, which was achieved by inhibiting IRP2-mediated iron overload. This study provided a potential drug candidate for the treatment of PD.
Collapse
Affiliation(s)
- Qiang-Ming Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China
| | - Tong Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China
| | - Xue-Qiang Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China
| | - Xiao-Wen Feng
- School of Pharmacy, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Feng-Yun Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People's Republic of China.
| | - Jian-Ping Luo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, People's Republic of China.
| |
Collapse
|
36
|
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
|
37
|
Chen L, Xin G, He Y, Tian Q, Kong X, Fu Y, Wang J, Zhang H, Wang L. Study of molecular patterns associated with ferroptosis in Parkinson's disease and its immune signature. PLoS One 2023; 18:e0295699. [PMID: 38127902 PMCID: PMC10734959 DOI: 10.1371/journal.pone.0295699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disease in the world. We downloaded data on Parkinson's disease and Ferroptosis-related genes from the GEO and FerrDb databases. We used WCGAN and Random Forest algorithm to screen out five Parkinson's disease ferroptosis-related hub genes. Two genes were identified for the first time as possibly playing a role in Braak staging progression. Unsupervised clustering analysis based on hub genes yielded ferroptosis isoforms, and immune infiltration analysis indicated that these isoforms are associated with immune cells and may represent different immune patterns. FRHGs scores were obtained to quantify the level of ferroptosis modifications in each individual. In addition, differences in interleukin expression were found between the two ferroptosis subtypes. The biological functions involved in the hub gene are analyzed. The ceRNA regulatory network of hub genes was mapped. The disease classification diagnosis model and risk prediction model were also constructed by applying hub genes based on logistic regression. Multiple external datasets validated the hub gene and classification diagnostic model with some accuracy. This study explored hub genes associated with ferroptosis in Parkinson's disease and their molecular patterns and immune signatures to provide new ideas for finding new targets for intervention and predictive biomarkers.
Collapse
Affiliation(s)
- Lixia Chen
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Guanghao Xin
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Yijie He
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Qinghua Tian
- Department of Neurology, The 962 Hospital of the Chinese People’s Liberation Army Joint Logistic Support Force, City Harbin, Province Heilongjiang, China
| | - Xiaotong Kong
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Yanchi Fu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Huixue Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, City Harbin, Province Heilongjiang, China
| |
Collapse
|
38
|
Yan Y, Wang Z, Wei W, Yang Z, Guo L, Wang Z, Wei X. Correlation of brain iron deposition and freezing of gait in Parkinson's disease: a cross-sectional study. Quant Imaging Med Surg 2023; 13:7961-7972. [PMID: 38106290 PMCID: PMC10721991 DOI: 10.21037/qims-23-267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/07/2023] [Indexed: 12/19/2023]
Abstract
Background Quantitative susceptibility mapping (QSM) is a novel imaging method for detecting iron content in the brain. The study aimed determine whether the iron deposition in the brains of people with Parkinson's disease (PD) is correlated with freezing of gait (FOG). Methods We retrospectively collected the data of 24 patients with PD from the Movement Disorders Program and 36 healthy controls (HCs) from January 2021 to December 2021. Clinical assessments included mental intelligence scales, Parkinson rating scales, motor-related scales, and clinical gait assessments. All exercise scales and gait assessments were performed in the "ON" and "OFF" states. Magnetic resonance imaging (MRI) data were collected using 3-dimensional fast low-angle shot sequences. We chose the bilateral red nucleus, substantia nigra, thalamus, putamen, caudate nucleus, and globus pallidus as regions of interest for QSM analysis. Results The iron deposition in the substantia nigra of the PD group was significantly higher than that of the HC group (P<0.01). In the PD group, the iron deposition in the substantia nigra of patients with FOG was significantly higher than that in patients without FOG (P=0.04). The iron deposition in the substantia nigra was positively correlated with the New Freezing of Gait Questionnaire (P=0.03). The scores for depression and anxiety of the PD group were significantly higher than those of the HC group, while the Berg balance scale score was significantly lower (P<0.01). Conclusions The iron deposition in the substantia nigra of patients with PD is increased compared with that of controls and is associated with FOG. QSM can be used to detect brain iron deposition in patients with PD, which would help to explore the mechanism of abnormal neurobiological activity in FOG.
Collapse
Affiliation(s)
- Ying Yan
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Wei
- Division of Science and Technology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lingfei Guo
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xuan Wei
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
39
|
Zhou Z, Zhang M, Fang Q, Huang J. Relationship between Parkinson's disease and cardio-cerebrovascular diseases: a Mendelian randomized study. Sci Rep 2023; 13:20428. [PMID: 37993489 PMCID: PMC10665329 DOI: 10.1038/s41598-023-47708-2] [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: 08/13/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023] Open
Abstract
Parkinson's disease (PD) and cardio-cerebrovascular diseases are related, according to earlier studies, but these studies have some controversy. Our aim was to assess the impact of PD on cardiocerebrovascular diseases using a Mendelian randomization (MR) method. The data for PD were single nucleotide polymorphisms (SNPs) from a publicly available genome-wide association study (GWAS) dataset containing data on 482,730 individuals. And the outcome SNPs data is were derived from five different GWAS datasets. The basic method for MR analysis was the inverse variance weighted (IVW) approach. We use the weighted median method and the MR-Egger method to supplement the MR analysis conclusion. Finally, We used Cochran's Q test to test heterogeneity, MR-PRESSO method and leave-one-out analysis method to perform sensitivity analysis. We used ratio ratios (OR) to assess the strength of the association between exposure and outcome, and 95% confidence intervals (CI) to show the reliability of the results. Our findings imply that PD is linked to a higher occurrence of coronary artery disease (CAD) (OR = 1.055, 95% CI 1.020-1.091, P = 0.001), stroke (OR = 1.039, 95% CI 1.007-1.072, P = 0.014). IVW analyses for stroke's subgroups of ischemic stroke (IS) and 95% CI 1.007-1.072, P = 0.014). IVW analyses for stroke's subgroups of ischemic stroke (IS) and cardioembolic stroke (CES) also yielded positive results, respectively (OR = 1.043, 95% CI 1.008-1.079, P = 0.013), (OR = 1.076, 95% CI 1.008-1.149, P = 0.026). There is no evidence of a relationship between PD and other cardio-cerebrovascular diseases. Additionally, sensitivity analysis revealed reliable outcomes. Our MR study analysis that PD is related with an elevated risk of CAD, stroke, IS, and CES.
Collapse
Affiliation(s)
- Zhongzheng Zhou
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muzi Zhang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghua Fang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
40
|
Hu H, Zhou J, Fang W, Chen HH, Jiang WH, Pu XY, Xu XQ, Gu WH, Wu FY. Increased brain iron in patients with thyroid-associated ophthalmopathy: a whole-brain analysis. Front Endocrinol (Lausanne) 2023; 14:1268279. [PMID: 38034014 PMCID: PMC10687634 DOI: 10.3389/fendo.2023.1268279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Background To investigate the whole-brain iron deposition alternations in patients with thyroid-associated ophthalmopathy (TAO) using quantitative susceptibility mapping (QSM). Methods Forty-eight patients with TAO and 33 healthy controls (HCs) were enrolled. All participants underwent brain magnetic resonance imaging scans and clinical scale assessments. QSM values were calculated and compared between TAO and HCs groups using a voxel-based analysis. A support vector machine (SVM) analysis was performed to evaluate the performance of QSM values in differentiating patients with TAO from HCs. Results Compared with HCs, patients with TAO showed significantly increased QSM values in the bilateral caudate nucleus (CN), left thalamus (TH), left cuneus, left precuneus, right insula and right middle frontal gyrus. In TAO group, QSM values in left TH were positively correlated with Hamilton Depression Rating Scale (HDRS) scores (r = 0.414, p = 0.005). The QSM values in right CN were negatively correlated with Montreal Cognitive Assessment (MoCA) scores (r = -0.342, p = 0.021). Besides that, a nearly negative correlation was found between QSM values in left CN and MoCA scores (r = -0.286, p = 0.057). The SVM model showed a good performance in distinguishing patients with TAO from the HCs (area under the curve, 0.958; average accuracy, 90.1%). Conclusion Patients with TAO had significantly increased iron deposition in brain regions corresponding to known visual, emotional and cognitive deficits. QSM values could serve as potential neuroimaging markers of TAO.
Collapse
Affiliation(s)
- Hao Hu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiang Zhou
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Fang
- Department of Radiology, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, Taicang, China
| | - Huan-Huan Chen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen-Hao Jiang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiong-Ying Pu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Quan Xu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen-Hao Gu
- Department of Radiology, Taicang Affiliated Hospital of Soochow University, The First People’s Hospital of Taicang, Taicang, China
| | - Fei-Yun Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
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
|
42
|
Chen L, Ma Y, Ma X, Liu L, Jv X, Li A, Shen Q, Jia W, Qu L, Shi L, Xie J. TFEB regulates cellular labile iron and prevents ferroptosis in a TfR1-dependent manner. Free Radic Biol Med 2023; 208:445-457. [PMID: 37683766 DOI: 10.1016/j.freeradbiomed.2023.09.004] [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: 05/09/2023] [Revised: 08/07/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Autophagy is a major clearance pathway for misfolded α-synuclein which promotes ferroptosis through NCOA4-mediated ferritin degradation. The regulation of these two processes to achieve improved neuroprotection in Parkinson's disease (PD) must be elucidated. Transcription factor EB (TFEB) is a master regulator of both autophagy and lysosome biogenesis, and lysosomes are important cellular iron storage organelles; however, the role of TFEB in ferroptosis and iron metabolism remains unclear. In this study, TFEB overexpression promoted the clearance of misfolded α-synuclein and prevented ferroptosis and iron overload. TFEB overexpression up-regulated transferrin receptor 1 (TfR1) synthesis and increased the localization of TfR1 in the lysosome, facilitating lysosomal iron import and transient lysosomal iron storage. TFEB overexpression increased the levels of cellular iron-safe storage proteins (both ferritin light and heavy chains). These functions in iron metabolism maintain the cellular labile iron at a low level and electrical activity, even under iron overload conditions. Notably, lower levels of cellular labile iron and the upregulation of ferritin light and heavy chains were reversed after TfR1 knockdown in cells overexpressing TFEB, indicating that TFEB regulates cellular labile iron and suppresses ferroptosis in a TfR1 dependent manner. Taken together, this evidence of the regulation of iron metabolism enriches our understanding of the function of TFEB. In addition, TFEB overexpression protects against ferroptosis and iron overload and provides a new direction and perspective for autophagy regulation in PD.
Collapse
Affiliation(s)
- Leilei Chen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China.
| | - Yue Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Xizhen Ma
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Lin Liu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Xianhui Jv
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Ang Li
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Qingqing Shen
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Wenting Jia
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Le Qu
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China
| | - Limin Shi
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China.
| | - Junxia Xie
- Institute of Brain Science and Disease, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Qingdao University, Qingdao, Shandong, China; Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
43
|
Kiersnowski OC, Winston GP, Caciagli L, Biondetti E, Elbadri M, Buck S, Duncan JS, Thornton JS, Shmueli K, Vos SB. Quantitative susceptibility mapping identifies hippocampal and other subcortical grey matter tissue composition changes in temporal lobe epilepsy. Hum Brain Mapp 2023; 44:5047-5064. [PMID: 37493334 PMCID: PMC10502681 DOI: 10.1002/hbm.26432] [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: 01/31/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/27/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is associated with widespread brain alterations. Using quantitative susceptibility mapping (QSM) alongside transverse relaxation rate (R 2 * ), we investigated regional brain susceptibility changes in 36 patients with left-sided (LTLE) or right-sided TLE (RTLE) secondary to hippocampal sclerosis, and 27 healthy controls (HC). We compared three susceptibility calculation methods to ensure image quality. Correlations of susceptibility andR 2 * with age of epilepsy onset, frequency of focal-to-bilateral tonic-clonic seizures (FBTCS), and neuropsychological test scores were examined. Weak-harmonic QSM (WH-QSM) successfully reduced noise and removed residual background field artefacts. Significant susceptibility increases were identified in the left putamen in the RTLE group compared to the LTLE group, the right putamen and right thalamus in the RTLE group compared to HC, and a significant susceptibility decrease in the left hippocampus in LTLE versus HC. LTLE patients who underwent epilepsy surgery showed significantly lower left-versus-right hippocampal susceptibility. SignificantR 2 * changes were found between TLE and HC groups in the amygdala, putamen, thalamus, and in the hippocampus. Specifically, decreased R2 * was found in the left and right hippocampus in LTLE and RTLE, respectively, compared to HC. Susceptibility andR 2 * were significantly correlated with cognitive test scores in the hippocampus, globus pallidus, and thalamus. FBTCS frequency correlated positively with ipsilateral thalamic and contralateral putamen susceptibility and withR 2 * in bilateral globi pallidi. Age of onset was correlated with susceptibility in the hippocampus and putamen, and withR 2 * in the caudate. Susceptibility andR 2 * changes observed in TLE groups suggest selective loss of low-myelinated neurons alongside iron redistribution in the hippocampi, predominantly ipsilaterally, indicating QSM's sensitivity to local pathology. Increased susceptibility andR 2 * in the thalamus and putamen suggest increased iron content and reflect disease severity.
Collapse
Affiliation(s)
- Oliver C. Kiersnowski
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Gavin P. Winston
- Department of Clinical and Experimental EpilepsyUniversity College LondonLondonUK
- Department of Medicine, Division of NeurologyQueen's UniversityKingstonCanada
| | - Lorenzo Caciagli
- Department of Clinical and Experimental EpilepsyUniversity College LondonLondonUK
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emma Biondetti
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
- Department of Neuroscience, Imaging and Clinical SciencesInstitute for Advanced Biomedical Technologies, “D'Annunzio” University of Chieti‐PescaraChietiItaly
| | - Maha Elbadri
- Department of NeurologyQueen Elizabeth HospitalBirminghamUK
| | - Sarah Buck
- Department of Clinical and Experimental EpilepsyUniversity College LondonLondonUK
| | - John S. Duncan
- Department of Clinical and Experimental EpilepsyUniversity College LondonLondonUK
| | - John S. Thornton
- Neuroradiological Academic UnitUCL Queen Square Institute of Neurology, University College LondonLondonUK
| | - Karin Shmueli
- Department of Medical Physics and Biomedical EngineeringUniversity College LondonLondonUK
| | - Sjoerd B. Vos
- Neuroradiological Academic UnitUCL Queen Square Institute of Neurology, University College LondonLondonUK
- Centre for Microscopy, Characterisation, and AnalysisThe University of Western AustraliaNedlandsAustralia
- Centre for Medical Image Computing, Computer Science departmentUniversity College LondonLondonUK
| |
Collapse
|
44
|
Zeng S, Ma H, Xie D, Huang Y, Wang M, Zeng W, Zhu N, Ma Z, Yang Z, Chu J, Zhao J. Quantitative susceptibility mapping evaluation of glioma. Eur Radiol 2023; 33:6636-6647. [PMID: 37095360 DOI: 10.1007/s00330-023-09647-4] [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: 04/29/2022] [Revised: 12/28/2022] [Accepted: 02/24/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVES To comprehensively evaluate the glioma using quantitative susceptibility mapping (QSM). MATERIALS AND METHODS Forty-two patients (18 women; mean age, 45 years) with pathologically confirmed gliomas were retrospectively included. All the patients underwent conventional and advanced MRI examinations (QSM, DWI, MRS, etc.). Five patients underwent paired QSM (pre- and post-enhancement). Four Visually Accessible Rembrandt Image (VASARI) features and intratumoural susceptibility signal (ITSS) were observed. Three ROIs each were manually drawn separately in the tumour parenchyma with relatively high and low magnetic susceptibility. The association between the tumour's magnetic susceptibility and other MRI parameters was also analysed. RESULTS Morphologically, gliomas with heterogeneous ITSS were more similar to high-grade gliomas (p = 0.006, AUC: 0.72, sensitivity: 70%, and specificity: 73%). Heterogeneous ITSS was significantly associated with tumour haemorrhage, necrosis, diffusion restriction, and avid enhancement but did not change between pre- and post-enhanced QSM. Quantitatively, tumour parenchyma magnetic susceptibility had limited value in grading gliomas and identifying IDH mutation status, whereas the relatively low magnetic susceptibility of the tumour parenchyma helped identify oligodendrogliomas in IDH mutated gliomas (AUC = 0.78) with high specificity (100%). The relatively high tumour magnetic susceptibility significantly increased after enhancement (p = 0.039). Additionally, we found that the magnetic susceptibility of the tumour parenchyma was significantly correlated with ADC (r = 0.61) and Cho/NAA (r = 0.40). CONCLUSIONS QSM is a promising candidate for the comprehensive evaluation of gliomas, except for IDH mutation status. The magnetic susceptibility of tumour parenchyma may be affected by tumour cell proliferation. KEY POINTS • Morphologically, gliomas with a heterogeneous intratumoural susceptibility signal (ITSS) are more similar to high-grade gliomas (p = 0.006; AUC, 0.72; sensitivity, 70%; and specificity, 73%). Heterogeneous ITSS was significantly associated with tumour haemorrhage, necrosis, diffusion restriction, and avid enhancement but did not change between pre- and post-enhanced QSM. • Tumour parenchyma's relatively low magnetic susceptibility helped identify oligodendroglioma with high specificity. • Tumour parenchyma magnetic susceptibility was significantly correlated with ADC (r = 0.61) and Cho/NAA (r = 0.40).
Collapse
Affiliation(s)
- Shanmei Zeng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Hui Ma
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Dingxiang Xie
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Yingqian Huang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Mengzhu Wang
- Department of MR Scientific Marketing, Siemens Healthineers, Guangzhou, Guangdong, People's Republic of China
| | - Wenting Zeng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Nengjin Zhu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Zuliwei Ma
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Zhiyun Yang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China
| | - Jianping Chu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China.
| | - Jing Zhao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58 Zhongshan Road 2, Guangdong, 510080, Guangzhou, People's Republic of China.
| |
Collapse
|
45
|
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
|
46
|
Li X, Wang X, Guo L, Wu K, Wang L, Rao L, Liu X, Kang C, Jiang B, Li Q, Li H, He F, Lu C. Association between lipocalin-2 and mild cognitive impairment or dementia: A systematic review and meta-analysis of population-based evidence. Ageing Res Rev 2023; 89:101984. [PMID: 37330019 DOI: 10.1016/j.arr.2023.101984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND The associations between lipocalin-2 (LCN2) with mild cognitive impairment (MCI) and dementia have gained growing interest. However, population-based studies have yielded inconsistent findings. Therefore, we conducted this essential systematic review and meta-analysis to analyze and summarize the existing population-based evidence. METHODS PubMed, EMBASE, and Web of Science were systematically searched until Mar 18, 2022. Meta-analysis was performed to generate the standard mean difference (SMD) of peripheral blood and cerebrospinal fluid (CSF) LCN2. A qualitative review was performed to summarize the evidence from postmortem brain tissue studies. RESULTS In peripheral blood, the overall pooled results showed no significant difference in LCN2 across Alzheimer's disease (AD), MCI and control groups. Further subgroup analysis revealed higher serum LCN2 levels in AD compared to controls (SMD =1.28 [0.44;2.13], p = 0.003), while the difference remained insignificant in plasma (SMD =0.04 [-0.82;0.90], p = 0.931). Besides, peripheral blood LCN2 were higher in AD when age difference between AD and controls ≥ 4 years (SMD =1.21 [0.37;2.06], p = 0.005). In CSF, no differences were found in LCN2 across groups of AD, MCI and controls. However, CSF LCN2 was higher in vascular dementia (VaD) compared to controls (SMD =1.02 [0.17;1.87], p = 0.018), as well as compared to AD (SMD =1.19 [0.58;1.80], p < 0.001). Qualitative analysis supported that LCN2 was increased in the brain tissue of AD-related areas, especially in astrocytes and microglia; while LCN2 increased in infarct-related brain areas and over-expressed in astrocytes and macrophages in mixed dementia (MD). CONCLUSION The difference in peripheral blood LCN2 between AD and controls may be affected by the type of biofluid and age. No differences were found in CSF LCN2 across AD, MCI and controls groups. In contrast, CSF LCN2 was elevated in VaD patients. Moreover, LCN2 was increased in AD-related brain areas and cells in AD, while in infarcts-related brain areas and cells in MD.
Collapse
Affiliation(s)
- Xiuwen Li
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou 510080, Guangdong, People's Republic of China
| | - Xiaojie Wang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Lan Guo
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou 510080, Guangdong, People's Republic of China
| | - Keying Wu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou 510080, Guangdong, People's Republic of China
| | - Li Wang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Lu Rao
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Xinjian Liu
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Chenyao Kang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Bin Jiang
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Qian Li
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Huling Li
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China
| | - Fenfen He
- Department of Neurology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen 518067, Guangdong, People's Republic of China.
| | - Ciyong Lu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, People's Republic of China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou 510080, Guangdong, People's Republic of China.
| |
Collapse
|
47
|
Jia X, Li Y, Ying Y, Jia X, Tang W, Bian Y, Zhang J, Wang DJJ, Cheng X, Yang Q. Effect of corticosubcortical iron deposition on dysfunction in CADASIL is mediated by white matter microstructural damage. Neuroimage Clin 2023; 39:103485. [PMID: 37542975 PMCID: PMC10407949 DOI: 10.1016/j.nicl.2023.103485] [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: 03/30/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Iron dysregulation may attenuate cognitive performance in patients with CADASIL. However, the underlying pathophysiological mechanisms remain incompletely understood. Whether white matter microstructural changes mediate these processes is largely unclear. In the present study, 30 cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) patients were confirmed via genetic analysis and 30 sex- and age-matched healthy controls underwent multimodal MRI examinations and neuropsychological assessments. Quantitative susceptibility mapping and peak width of skeletonized mean diffusivity (PSMD) were analyzed. Mediation effect analysis was performed to explore the interrelationship between iron deposition, white matter microstructural changes and cognitive deficits in CADASIL. Cognitive deterioration was most affected in memory and executive function, followed by attention and working memory in CADASIL. Excessive iron in the temporal-precuneus pathway and deep gray matter specific to CADASIL were identified. Mediation analysis further revealed that PSMD mediated the relationship between iron concentration and cognitive profile in CADASIL. The present findings provide a new perspective on iron deposition in the corticosubcortical circuit and its contribution to disease-related selective cognitive decline, in which iron concentration may affect cognition by white matter microstructural changes in CADASIL.
Collapse
Affiliation(s)
- Xiuqin Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China
| | - Yingying Li
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yunqing Ying
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xuejia Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Weijun Tang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yueyan Bian
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiajia Zhang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, United States
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China.
| |
Collapse
|
48
|
Xie F, Mao T, Tang J, Zhao L, Guo J, Lin H, Wang D, Zhou G. Evaluation of iron deposition in the motor CSTC loop of a Chinese family with paroxysmal kinesigenic dyskinesia using quantitative susceptibility mapping. Front Neurol 2023; 14:1164600. [PMID: 37483438 PMCID: PMC10358764 DOI: 10.3389/fneur.2023.1164600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/12/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Previous studies have revealed structural, functional, and metabolic changes in brain regions inside the cortico-striatal-thalamo-cortical (CSTC) loop in patients with paroxysmal kinesigenic dyskinesia (PKD), whereas no quantitative susceptibility mapping (QSM)-related studies have explored brain iron deposition in these areas. Methods A total of eight familial PKD patients and 10 of their healthy family members (normal controls) were recruited and underwent QSM on a 3T magnetic resonance imaging system. Magnetic susceptibility maps were reconstructed using a multi-scale dipole inversion algorithm. Thereafter, we specifically analyzed changes in local mean susceptibility values in cortical regions and subcortical nuclei inside the motor CSTC loop. Results Compared with normal controls, PKD patients had altered brain iron levels. In the cortical gray matter area involved with the motor CSTC loop, susceptibility values were generally elevated, especially in the bilateral M1 and PMv regions. In the subcortical nuclei regions involved with the motor CSTC loop, susceptibility values were generally lower, especially in the bilateral substantia nigra regions. Conclusion Our results provide new evidence for the neuropathogenesis of PKD and suggest that an imbalance in brain iron levels may play a role in PKD.
Collapse
Affiliation(s)
- Fangfang Xie
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingyi Tang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Linmei Zhao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jiuqing Guo
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Huashan Lin
- Department of Pharmaceutical Diagnosis, GE Healthcare, Changsha, China
| | - Dongcui Wang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Gaofeng Zhou
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
49
|
Straumann N, Combes BF, Dean Ben XL, Sternke-Hoffmann R, Gerez JA, Dias I, Chen Z, Watts B, Rostami I, Shi K, Rominger A, Baumann CR, Luo J, Noain D, Nitsch RM, Okamura N, Razansky D, Ni R. Visualizing alpha-synuclein and iron deposition in M83 mouse model of Parkinson's disease in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546962. [PMID: 37425954 PMCID: PMC10327184 DOI: 10.1101/2023.06.28.546962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background Abnormal alpha-synuclein and iron accumulation in the brain play an important role in Parkinson's disease (PD). Herein, we aim at visualizing alpha-synuclein inclusions and iron deposition in the brains of M83 (A53T) mouse models of PD in vivo. Methods Fluorescently labelled pyrimidoindole-derivative THK-565 was characterized by using recombinant fibrils and brains from 10-11 months old M83 mice, which subsequently underwent in vivo concurrent wide-field fluorescence and volumetric multispectral optoacoustic tomography (vMSOT) imaging. The in vivo results were verified against structural and susceptibility weighted imaging (SWI) magnetic resonance imaging (MRI) at 9.4 Tesla and scanning transmission X-ray microscopy (STXM) of perfused brains. Brain slice immunofluorescence and Prussian blue staining were further performed to validate the detection of alpha-synuclein inclusions and iron deposition in the brain, respectively. Results THK-565 showed increased fluorescence upon binding to recombinant alpha-synuclein fibrils and alpha-synuclein inclusions in post-mortem brain slices from patients with Parkinson's disease and M83 mice. i.v. administration of THK-565 in M83 mice showed higher cerebral retention at 20 and 40 minutes post-injection by wide-field fluorescence compared to non-transgenic littermate mice, in congruence with the vMSOT findings. SWI/phase images and Prussian blue indicated the accumulation of iron deposits in the brains of M83 mice, presumably in the Fe3+ form, as evinced by the STXM results. Conclusion We demonstrated in vivo mapping of alpha-synuclein by means of non-invasive epifluorescence and vMSOT imaging assisted with a targeted THK-565 label and SWI/STXM identification of iron deposits in M83 mouse brains ex vivo.
Collapse
Affiliation(s)
- Nadja Straumann
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Benjamin F. Combes
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Xose Luis Dean Ben
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | | | - Juan A. Gerez
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Zurich, Switzerland
| | - Ines Dias
- Neurology Department, University Hospital Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Benjamin Watts
- Photon Science Division, Paul Scherrer Institute, Villigen, Switzerland
| | - Iman Rostami
- Microscopic Anatomy and Structural Biology, University of Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Jinghui Luo
- Department of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland
| | - Daniela Noain
- Neurology Department, University Hospital Zurich, Zurich, Switzerland
| | - Roger M. Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Daniel Razansky
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| |
Collapse
|
50
|
Snyder J, Wu Z. Origins of nervous tissue susceptibility to ferroptosis. CELL INSIGHT 2023; 2:100091. [PMID: 37398634 PMCID: PMC10308196 DOI: 10.1016/j.cellin.2023.100091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/03/2023] [Accepted: 03/11/2023] [Indexed: 07/04/2023]
Abstract
Ferroptosis is a newly defined form of programmed cell death. It possesses unique processes of cell demise, cytopathological changes, and independent signal regulation pathways. Ferroptosis is considered to be deeply involved in the development of many diseases, including cancer, cardiovascular diseases, and neurodegeneration. Intriguingly, why cells in certain tissues and organs (such as the central nervous system, CNS) are more sensitive to changes in ferroptosis remains a question that has not been carefully discussed. In this Holmesian review, we discuss lipid composition as a potential but often overlooked determining factor in ferroptosis sensitivity and the role of polyunsaturated fatty acids (PUFAs) in the pathogenesis of several common human neurodegenerative diseases. In subsequent studies of ferroptosis, lipid composition needs to be given special attention, as it may significantly affect the susceptibility of the cell model used (or the tissue studied).
Collapse
Affiliation(s)
- Jessica Snyder
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Zhihao Wu
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| |
Collapse
|