1
|
Sadan OR, Avisdris N, Rabinowich A, Link‐Sourani D, Krajden Haratz K, Garel C, Hiersch L, Ben Sira L, Ben Bashat D. Brain Metabolite Differences in Fetuses With Cytomegalovirus Infection: A Magnetic Resonance Spectroscopy Study. J Magn Reson Imaging 2025; 61:1133-1141. [PMID: 38979886 PMCID: PMC11803696 DOI: 10.1002/jmri.29507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) is the most common intrauterine infection and may be associated with unfavorable outcomes. While some CMV-infected fetuses may show gross or subtle brain abnormalities on MRI, their clinical significance may be unclear. Conversely, normal development cannot be guaranteed in CMV-infected fetuses with normal MRI. PURPOSE To assess brain metabolite differences in CMV-infected fetuses using magnetic resonance spectroscopy (MRS). STUDY TYPE Retrospective. SUBJECTS Out of a cohort of 149 cases, 44 with maternal CMV infection, amniocentesis results, and good-quality MRS were included. CMV-infected fetuses with positive polymerase chain reaction (PCR) (N = 35) were divided based on MRI results as follows: typical brain abnormalities (gross findings, N = 8), exclusive white matter hyperintense signal (WMHS) on T2-weighted images (subtle findings, N = 7), and normal MRI (N = 20). Uninfected fetuses (negative PCR) with normal MRI were included as controls (N = 9). FIELD STRENGTH 3 T, T2-weighted half Fourier single-shot turbo spin-echo (HASTE), T2-weighted true fast imaging with steady-state free precession (TrueFISP), T1- and T2*-weighted fast low angle shot (FLASH), and 1H-MRS single-voxel point resolved spectroscopy (PRESS) sequences. ASSESSMENT MRI findings were assessed by three radiologists, and metabolic ratios within the basal ganglia were calculated using LCModel. STATISTICAL TESTS Analysis of covariance test with Bonferroni correction for multiple comparisons was used to compare metabolic ratios between groups while accounting for gestational age. A P-value <0.05 was deemed significant. RESULTS MRS was successfully acquired in 63% of fetuses. Substantial agreement was observed between radiologists (Fleiss' kappa [k] = 0.8). Infected fetuses with gross MRI findings exhibited significantly reduced tNAA/tCr ratios (0.64 ± 0.08) compared with infected fetuses with subtle MRI findings (0.85 ± 0.19), infected fetuses with normal MRI (0.8 ± 0.14) and controls (0.81 ± 0.15). No other significant differences were detected (P ≥ 0.261). CONCLUSION Reduced tNAA/tCr within the apparently normal brain tissue was detected in CMV-infected fetuses with gross brain abnormalities, suggesting extensive brain damage. In CMV-infected fetuses with isolated WMHS, no damage was detected by MRS. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Or R. Sadan
- Sagol Brain InstituteTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- Sagol School of NeuroscienceTel‐Aviv UniversityTel‐AvivIsrael
| | - Netanell Avisdris
- Sagol Brain InstituteTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- School of Computer Science and EngineeringThe Hebrew University of JerusalemJerusalemIsrael
| | - Aviad Rabinowich
- Sagol Brain InstituteTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- Department of RadiologyTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- Faculty of Medical & Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
| | - Daphna Link‐Sourani
- Sagol Brain InstituteTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- Technion Human MRI Research Center, Faculty of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Karina Krajden Haratz
- Faculty of Medical & Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
- Department of Obstetrics and GynecologyLis Hospital for Women, Tel Aviv Sourasky Medical CenterTel‐AvivIsrael
| | - Catherine Garel
- Department of RadiologyTel Aviv Sourasky Medical CenterTel‐AvivIsrael
| | - Liran Hiersch
- Faculty of Medical & Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
- Department of Obstetrics and GynecologyLis Hospital for Women, Tel Aviv Sourasky Medical CenterTel‐AvivIsrael
| | - Liat Ben Sira
- Sagol School of NeuroscienceTel‐Aviv UniversityTel‐AvivIsrael
- Department of RadiologyTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- Faculty of Medical & Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
| | - Dafna Ben Bashat
- Sagol Brain InstituteTel Aviv Sourasky Medical CenterTel‐AvivIsrael
- Sagol School of NeuroscienceTel‐Aviv UniversityTel‐AvivIsrael
- Faculty of Medical & Health SciencesTel‐Aviv UniversityTel‐AvivIsrael
| |
Collapse
|
2
|
Wang Y, Liu H, Ye Y, Fang W, Lin A, Dai X, Ye Q, Chen X, Zhang J. ApoE2 affects insulin signaling in the hippocampus and spatial cognition of aged mice in a sex-dependent manner. Cell Commun Signal 2025; 23:112. [PMID: 40011916 PMCID: PMC11866816 DOI: 10.1186/s12964-025-02093-3] [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: 12/20/2024] [Accepted: 02/08/2025] [Indexed: 02/28/2025] Open
Abstract
Apolipoprotein E (APOE) has garnered significant attention as one of the most influential genetic risk factors for Alzheimer's disease (AD). While the pathogenic role of APOE4 in sporadic AD has been extensively studied, research on the protective effects of the APOE2 genotype and its underlying mechanisms remains limited. Additionally, the existence of sex differences in the protective effects of ApoE2 continues to be a topic of debate. In this study, we utilized humanized ApoE2- and ApoE3- target replacement mice to examine the sex-specific effects of ApoE2 on cognition. Compared with female ApoE3 mice, we found significantly lower spatial cognitive ability and impaired hippocampal synaptic ultrastructure in aged female ApoE2 mice, accompanied by reduced insulin signaling of the hippocampus. Further analyses by target metabolomics and transcriptomic analyses revealed that female ApoE2 mice exhibit an age-related decline in hippocampal inositol levels, and that alterations in inositol levels lower insulin signaling. Importantly, inositol supplementation was found to alleviate peripheral glucose intolerance, enhance insulin signaling, and ultimately improve cognitive function. Interestingly, these differences were not observed between male ApoE2 and ApoE3 mice. The research findings not only provide new insights into the impact of ApoE2 on cognition but also offer a new strategy for cognitive improvement through inositol supplementation in older women.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Hanchen Liu
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yijuan Ye
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Wenting Fang
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Anlan Lin
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoman Dai
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qinyong Ye
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China.
| | - Jing Zhang
- Department of Neurology, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University Union Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
3
|
Sun X, Zhu J, Li R, Peng Y, Gong L. The global research of magnetic resonance imaging in Alzheimer's disease: a bibliometric analysis from 2004 to 2023. Front Neurol 2025; 15:1510522. [PMID: 39882364 PMCID: PMC11774745 DOI: 10.3389/fneur.2024.1510522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Background Alzheimer's disease (AD) is a common neurodegenerative disorder worldwide and the using of magnetic resonance imaging (MRI) in the management of AD is increasing. The present study aims to summarize MRI in AD researches via bibliometric analysis and predict future research hotspots. Methods We searched for records related to MRI studies in AD patients from 2004 to 2023 in the Web of Science Core Collection (WoSCC) database. CiteSpace was applied to analyze institutions, references and keywords. VOSviewer was used for the analysis of countries, authors and journals. Results A total of 13,659 articles were obtained in this study. The number of published articles showed overall exponential growth from 2004 to 2023. The top country and institution were the United States and the University of California System, accounting for 40.30% and 9.88% of the total studies, respectively. Jack CR from the United States was the most productive author. The most productive journal was the Journal of Alzheimers Disease. Keyword burst analysis revealed that "machine learning" and "deep learning" were the keywords that frequently appeared in the past 6 years. Timeline views of the references revealed that "#0 tau pathology" and "#1 deep learning" are currently the latest research focuses. Conclusion This study provides an in-depth overview of publications on MRI studies in AD. The United States is the leading country in this field with a concentration of highly productive researchers and high-level institutions. The current research hotspot is deep learning, which is being applied to develop noninvasive diagnosis and safer treatment of AD.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Jianghua Zhu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Ruowei Li
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Yun Peng
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Lianggeng Gong
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| |
Collapse
|
4
|
Vo DK, Trinh KTL. Emerging Biomarkers in Metabolomics: Advancements in Precision Health and Disease Diagnosis. Int J Mol Sci 2024; 25:13190. [PMID: 39684900 DOI: 10.3390/ijms252313190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Metabolomics has come to the fore as an efficient tool in the search for biomarkers that are critical for precision health approaches and improved diagnostics. This review will outline recent advances in biomarker discovery based on metabolomics, focusing on metabolomics biomarkers reported in cancer, neurodegenerative disorders, cardiovascular diseases, and metabolic health. In cancer, metabolomics provides evidence for unique oncometabolites that are important for early disease detection and monitoring of treatment responses. Metabolite profiling for conditions such as neurodegenerative and mental health disorders can offer early diagnosis and mechanisms into the disease especially in Alzheimer's and Parkinson's diseases. In addition to these, lipid biomarkers and other metabolites relating to cardiovascular and metabolic disorders are promising for patient stratification and personalized treatment. The gut microbiome and environmental exposure also feature among the influential factors in biomarker discovery because they sculpt individual metabolic profiles, impacting overall health. Further, we discuss technological advances in metabolomics, current clinical applications, and the challenges faced by metabolomics biomarker validation toward precision medicine. Finally, this review discusses future opportunities regarding the integration of metabolomics into routine healthcare to enable preventive and personalized approaches.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea
| | - Kieu The Loan Trinh
- BioNano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
5
|
Borghys H, Schwab A, Keppler B. Middle-aged dogs with low and high Aβ CSF concentrations show differences in energy and stress related metabolic profiles in CSF. Heliyon 2024; 10:e39104. [PMID: 39498015 PMCID: PMC11532822 DOI: 10.1016/j.heliyon.2024.e39104] [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: 01/19/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
Background Amyloid beta (Aβ) accumulation in the brain is one of the earliest findings in Alzheimer's disease (AD). The dog is a natural animal model for amyloid processing and early brain amyloid pathology. The goal of this study is to examine which differences in metabolomic profiles in cerebrospinal fluid (CSF) could be detected in dogs with a difference in CSF Aβ concentrations before amyloid accumulation occurs. Method Metabolic profiling was performed on CSF from 4 to 8 year old dogs with different CSF Aβ concentrations. Results Metabolomic profiling of CSF showed differences in brain energy metabolism. More specifically, increases in N-acetylation of amino acids and amino sugars, creatine and pentose metabolism, and a decrease in tricarboxylic acid (TCA) cycle were seen in dogs with a high CSF Aβ concentration. In addition, signs of elevated oxidative stress, higher methionine, lipid and nucleotide metabolism and increased levels of cysteine, myo-inositol and trimethylamine N-oxide were noted in these animals. Conclusions Differences in energy metabolism and stress mediated metabolic changes are seen in the brain of dogs with different CSF Aβ concentrations, before any amyloid deposition occurs. Similar metabolic changes, as in the high Aβ dogs, have been described in AD in humans and/or transgenic AD mice, some of them in very early phases. General significance The differences observed in metabolomic profiles could help in identifying potential biomarkers for an increased risk of developing amyloid pathology in the brain and open the door to the evaluation of preventive treatments for amyloid pathology in humans.
Collapse
Affiliation(s)
- Herman Borghys
- Janssen Research & Development, a division of Janssen Pharmaceutica N.V., Beerse, Belgium
| | | | | |
Collapse
|
6
|
Davies-Jenkins CW, Workman CI, Hupfeld KE, Zöllner HJ, Leoutsakos JM, Kraut MA, Barker PB, Smith GS, Oeltzschner G. Multimodal investigation of neuropathology and neurometabolites in mild cognitive impairment and late-life depression with 11C-PiB beta-amyloid PET and 7T magnetic resonance spectroscopy. Neurobiol Aging 2024; 142:27-40. [PMID: 39111221 PMCID: PMC11916921 DOI: 10.1016/j.neurobiolaging.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 09/02/2024]
Abstract
Positron emission tomography (PET) and magnetic resonance spectroscopy (1H-MRS) are complementary techniques that can be applied to study how proteinopathy and neurometabolism relate to cognitive deficits in preclinical stages of Alzheimer's disease (AD)-mild cognitive impairment (MCI) and late-life depression (LLD). We acquired beta-amyloid (Aβ) PET and 7 T 1H-MRS measures of GABA, glutamate, glutathione, N-acetylaspartate, N-acetylaspartylglutamate, myo-inositol, choline, and lactate in the anterior and posterior cingulate cortices (ACC, PCC) in 13 MCI and 9 LLD patients, and 13 controls. We used linear regression to examine associations between metabolites, Aβ, and cognitive scores, and whether metabolites and Aβ explained cognitive scores better than Aβ alone. In the ACC, higher Aβ was associated with lower GABA in controls but not MCI or LLD patients, but results depended upon MRS data quality control criteria. Greater variance in California Verbal Learning Test scores was better explained by a model that combined ACC glutamate and Aβ deposition than by models that only included one of these variables. These findings identify preliminary associations between Aβ, neurometabolites, and cognition.
Collapse
Affiliation(s)
- Christopher W Davies-Jenkins
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Clifford I Workman
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathleen E Hupfeld
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Helge J Zöllner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Jeannie-Marie Leoutsakos
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael A Kraut
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter B Barker
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Gwenn S Smith
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Nuclear Medicine and Molecular Imaging, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Georg Oeltzschner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
7
|
Kara F, Kantarci K. Understanding Proton Magnetic Resonance Spectroscopy Neurochemical Changes Using Alzheimer's Disease Biofluid, PET, Postmortem Pathology Biomarkers, and APOE Genotype. Int J Mol Sci 2024; 25:10064. [PMID: 39337551 PMCID: PMC11432594 DOI: 10.3390/ijms251810064] [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: 08/23/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
In vivo proton (1H) magnetic resonance spectroscopy (MRS) is a powerful non-invasive method that can measure Alzheimer's disease (AD)-related neuropathological alterations at the molecular level. AD biomarkers include amyloid-beta (Aβ) plaques and hyperphosphorylated tau neurofibrillary tangles. These biomarkers can be detected via postmortem analysis but also in living individuals through positron emission tomography (PET) or biofluid biomarkers of Aβ and tau. This review offers an overview of biochemical abnormalities detected by 1H MRS within the biologically defined AD spectrum. It includes a summary of earlier studies that explored the association of 1H MRS metabolites with biofluid, PET, and postmortem AD biomarkers and examined how apolipoprotein e4 allele carrier status influences brain biochemistry. Studying these associations is crucial for understanding how AD pathology affects brain homeostasis throughout the AD continuum and may eventually facilitate the development of potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Chen AM, Gajdošík M, Ahmed W, Ahn S, Babb JS, Blessing EM, Boutajangout A, de Leon MJ, Debure L, Gaggi N, Gajdošík M, George A, Ghuman M, Glodzik L, Harvey P, Juchem C, Marsh K, Peralta R, Rusinek H, Sheriff S, Vedvyas A, Wisniewski T, Zheng H, Osorio R, Kirov II. Retrospective analysis of Braak stage- and APOE4 allele-dependent associations between MR spectroscopy and markers of tau and neurodegeneration in cognitively unimpaired elderly. Neuroimage 2024; 297:120742. [PMID: 39029606 PMCID: PMC11404707 DOI: 10.1016/j.neuroimage.2024.120742] [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: 03/11/2024] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE The pathological hallmarks of Alzheimer's disease (AD), amyloid, tau, and associated neurodegeneration, are present in the cortical gray matter (GM) years before symptom onset, and at significantly greater levels in carriers of the apolipoprotein E4 (APOE4) allele. Their respective biomarkers, A/T/N, have been found to correlate with aspects of brain biochemistry, measured with magnetic resonance spectroscopy (MRS), indicating a potential for MRS to augment the A/T/N framework for staging and prediction of AD. Unfortunately, the relationships between MRS and A/T/N biomarkers are unclear, largely due to a lack of studies examining them in the context of the spatial and temporal model of T/N progression. Advanced MRS acquisition and post-processing approaches have enabled us to address this knowledge gap and test the hypotheses, that glutamate-plus-glutamine (Glx) and N-acetyl-aspartate (NAA), metabolites reflecting synaptic and neuronal health, respectively, measured from regions on the Braak stage continuum, correlate with: (i) cerebrospinal fluid (CSF) p-tau181 level (T), and (ii) hippocampal volume or cortical thickness of parietal lobe GM (N). We hypothesized that these correlations will be moderated by Braak stage and APOE4 genotype. METHODS We conducted a retrospective imaging study of 34 cognitively unimpaired elderly individuals who received APOE4 genotyping and lumbar puncture from pre-existing prospective studies at the NYU Grossman School of Medicine between October 2014 and January 2019. Subjects returned for their imaging exam between April 2018 and February 2020. Metabolites were measured from the left hippocampus (Braak II) using a single-voxel semi-adiabatic localization by adiabatic selective refocusing sequence; and from the bilateral posterior cingulate cortex (PCC; Braak IV), bilateral precuneus (Braak V), and bilateral precentral gyrus (Braak VI) using a multi-voxel echo-planar spectroscopic imaging sequence. Pearson and Spearman correlations were used to examine the relationships between absolute levels of choline, creatine, myo-inositol, Glx, and NAA and CSF p-tau181, and between these metabolites and hippocampal volume or parietal cortical thicknesses. Covariates included age, sex, years of education, Fazekas score, and months between CSF collection and MRI exam. RESULTS There was a direct correlation between hippocampal Glx and CSF p-tau181 in APOE4 carriers (Pearson's r = 0.76, p = 0.02), but not after adjusting for covariates. In the entire cohort, there was a direct correlation between hippocampal NAA and hippocampal volume (Spearman's r = 0.55, p = 0.001), even after adjusting for age and Fazekas score (Spearman's r = 0.48, p = 0.006). This relationship was observed only in APOE4 carriers (Pearson's r = 0.66, p = 0.017), and was also retained after adjustment (Pearson's r = 0.76, p = 0.008; metabolite-by-carrier interaction p = 0.03). There were no findings in the PCC, nor in the negative control (late Braak stage) regions of the precuneus and precentral gyrus. CONCLUSIONS Our findings are in line with the spatially- and temporally-resolved Braak staging model of pathological severity in which the hippocampus is affected earlier than the PCC. The correlations, between MRS markers of synaptic and neuronal health and, respectively, T and N pathology, were found exclusively within APOE4 carriers, suggesting a connection with AD pathological change, rather than with normal aging. We therefore conclude that MRS has the potential to augment early A/T/N staging, with the hippocampus serving as a more sensitive MRS target compared to the PCC.
Collapse
Affiliation(s)
- Anna M Chen
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Martin Gajdošík
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Wajiha Ahmed
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sinyeob Ahn
- Siemens Medical Solutions USA Inc., Malvern, PA, USA
| | - James S Babb
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Esther M Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Healthy Brain Aging and Sleep Center, NYU Langone Health, New York, NY, USA
| | - Allal Boutajangout
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mony J de Leon
- Retired Director, Center for Brain Health, Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Ludovic Debure
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Naomi Gaggi
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Healthy Brain Aging and Sleep Center, NYU Langone Health, New York, NY, USA
| | - Mia Gajdošík
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ajax George
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mobeena Ghuman
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Lidia Glodzik
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Patrick Harvey
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Christoph Juchem
- Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Karyn Marsh
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Rosemary Peralta
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Henry Rusinek
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alok Vedvyas
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Helena Zheng
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ricardo Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Healthy Brain Aging and Sleep Center, NYU Langone Health, New York, NY, USA.
| | - Ivan I Kirov
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York, NY, USA; Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Noche JA, Vanderlip C, Wright S, Sordo L, Head E, Stark C. Myo-inositol and total NAA in the hippocampus are linked to CSF tau pathology in cognitively normal older adults. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607353. [PMID: 39211099 PMCID: PMC11361118 DOI: 10.1101/2024.08.09.607353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Understanding relationships between in vivo neurometabolic changes and Alzheimer's disease (AD) pathology in the hippocampus, a region vulnerable to early changes in AD, will support early diagnosis. METHODS Two studies using 1 H-MRS examined concentrations of myo-inositol (MI), total creatine (tCr) and total NAA (tNAA) in the hippocampus. The first study compared hippocampal metabolite concentrations in healthy young and older adults and the second study assessed relationships between hippocampal metabolites and cerebrospinal fluid (CSF) measurements of Aβ42, phosphotau 181 (pTau181), and total tau (t-Tau) while adjusting for demographic covariates and spectral characteristics (linewidth, signal- to-noise ratio) in a separate group of older adults ranging from cognitively normal (CN) to AD-dementia. RESULTS Hippocampal MI, but not tCr or tNAA, was increased in cognitively normal older versus young adults. Within the second older adult group, MI and tNAA, but not tCr, were linked to increases in CSF pTau181 and t-Tau, but not Aβ42. DISCUSSION Tau deposition in cognitively normal individuals is associated with biochemical changes related to glial reactivity and neural integrity in the hippocampus.
Collapse
|
10
|
Vints WAJ, Šeikinaitė J, Gökçe E, Kušleikienė S, Šarkinaite M, Valatkeviciene K, Česnaitienė VJ, Verbunt J, Levin O, Masiulis N. Resistance exercise effects on hippocampus subfield volumes and biomarkers of neuroplasticity and neuroinflammation in older adults with low and high risk of mild cognitive impairment: a randomized controlled trial. GeroScience 2024; 46:3971-3991. [PMID: 38478179 PMCID: PMC11226571 DOI: 10.1007/s11357-024-01110-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/25/2024] [Indexed: 07/07/2024] Open
Abstract
Physical exercise is suggested to promote hippocampal neuroplasticity by increasing circulating neurotrophic and anti-inflammatory factors. Our aim was to explore the interplay between the effect of progressive resistance exercise on blood biomarker levels, hippocampal neurometabolite levels and hippocampal volume in older adults with a low compared to a high risk of mild cognitive impairment (MCI). Seventy apparently healthy male/female older adults (aged 60-85 years old) were randomly allocated to a 12 week lower limb progressive resistance or no intervention, stratified for low (< 26/30) or high (≥ 26/30) Montreal Cognitive Assessment (MoCA) score, indicating MCI risk. Outcome measures were blood levels of insulin-like growth factor-1 (IGF-1), interleukin-6 (IL-6) or kynurenine (KYN); hippocampal total and subfield volumes of the cornu ammonis 1 (CA1) and 4 (CA4), subiculum, presubiculum, and dentate gyrus measured with magnetic resonance imaging (MRI); and hippocampus neurometabolites including total N-acetylaspartate (NAA), myo-inositol (mIns), and total creatine (Cr) measured with proton magnetic resonance spectroscopy (1H-MRS). We evaluated the intervention effect, cognitive status effect, their interaction and the bivariate relationship between exercise-induced changes between the outcome measures. Higher kynurenine levels (p = 0.015) and lower subiculum volumes (p = 0.043) were found in older adults with high MCI risk compared to older adults with low MCI risk. Exercise-induced CA1 volume changes were negatively correlated with hippocampal tNAA/mIns level changes (r = -0.605, p = 0.006). This study provides valuable insight in the multifactorial processes related to resistance training in older adults with low or high MCI risk.
Collapse
Affiliation(s)
- Wouter A J Vints
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania.
- Department of Rehabilitation Medicine Research School CAPHRI, Maastricht University, Maastricht, The Netherlands.
- Centre of Expertise in Rehabilitation and Audiology, Adelante Zorggroep, Hoensbroek, The Netherlands.
| | - Julija Šeikinaitė
- Department of Rehabilitation, Physical and Sports Medicine, Institute of Health Science, Vilnius University, Vilnius, Lithuania
| | - Evrim Gökçe
- Sports Rehabilitation Laboratory, Ankara City Hospital, 06800, Ankara, Turkey
| | - Simona Kušleikienė
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
| | - Milda Šarkinaite
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kristina Valatkeviciene
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vida J Česnaitienė
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
| | - Jeanine Verbunt
- Department of Rehabilitation Medicine Research School CAPHRI, Maastricht University, Maastricht, The Netherlands
- Centre of Expertise in Rehabilitation and Audiology, Adelante Zorggroep, Hoensbroek, The Netherlands
| | - Oron Levin
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- Motor Control & Neuroplasticity Research Group, Group Biomedical Sciences, Catholic University Leuven, Heverlee, Belgium
| | - Nerijus Masiulis
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- Department of Rehabilitation, Physical and Sports Medicine, Institute of Health Science, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
11
|
Monnig M, Shah K. Linking alcohol use to Alzheimer's disease: Interactions with aging and APOE along immune pathways. MEDICAL RESEARCH ARCHIVES 2024; 12:10.18103/mra.v12i8.5228. [PMID: 39544182 PMCID: PMC11563488 DOI: 10.18103/mra.v12i8.5228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Although it is known that APOE genotype is the strongest genetic risk factor for late-onset Alzheimer's disease, development is a multifactorial process. Alcohol use is a contributor to the epidemic of Alzheimer's disease and related dementias in the US and globally, yet mechanisms are not fully understood. Carriers of the APOE ε4 allele show elevated risk of dementia in relation to several lifestyle factors, including alcohol use. In this review, we describe how alcohol interacts with APOE genotype and aging with potential implications for Alzheimer's disease promotion. Age-related immune senescence and "inflammaging" (i.e., low-grade inflammation associated with aging) are increasingly recognized as contributors to age-related disease. We focus on three immune pathways that are likely contributors to Alzheimer's disease development, centering on alcohol and APOE genotype interactions, specifically: 1) microbial translocation and immune activation, 2) the senescence associated secretory phenotype, and 3) neuroinflammation. First, microbial translocation, the unphysiological movement of gut products into systemic circulation, elicits a proinflammatory response and increases with aging, with proposed links to Alzheimer's disease. Second, the senescence associated secretory phenotype is a set of intercellular signaling factors, e.g., proinflammatory cytokines and chemokines, growth regulators, and proteases, that drives cellular aging when senescent cells remain metabolically active. The senescence associated secretory phenotype can drive development of aging-diseases such as Alzheimer's disease. Third, neuroinflammation occurs via numerous mechanisms such as microglial activation and is gaining recognition as an etiological factor in the development of Alzheimer's disease. This review focuses on interactions of alcohol with APOE genotype and aging along these three pathways that may promote Alzheimer's disease. Further research on these processes may inform development of strategies to prevent onset and progression of Alzheimer's disease and to delay associated cognitive decline.
Collapse
Affiliation(s)
- Mollie Monnig
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
| | - Krish Shah
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI 02912, USA
| |
Collapse
|
12
|
Dounavi ME, McKiernan E, Langsen M, Gregory S, Muniz-Terrera G, Prats-Sedano MA, Mada MO, Williams GB, Lawlor B, Naci L, Mackay C, Koychev I, Malhotra P, Ritchie K, Ritchie CW, Su L, Waldman AD, O’ Brien JT. Investigating the brain's neurochemical profile at midlife in relation to dementia risk factors. Brain Commun 2024; 6:fcae138. [PMID: 38779354 PMCID: PMC11109818 DOI: 10.1093/braincomms/fcae138] [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: 07/24/2023] [Revised: 01/18/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
Changes in the brain's physiology in Alzheimer's disease are thought to occur early in the disease's trajectory. In this study our aim was to investigate the brain's neurochemical profile in a midlife cohort in relation to risk factors for future dementia using single voxel proton magnetic resonance spectroscopy. Participants in the multi-site PREVENT-Dementia study (age range 40-59 year old) underwent 3T magnetic resonance spectroscopy with the spectroscopy voxel placed in the posterior cingulate/precuneus region. Using LCModel, we quantified the absolute concentrations of myo-inositol, total N-acetylaspartate, total creatine, choline, glutathione and glutamate-glutamine for 406 participants (mean age 51.1; 65.3% female). Underlying partial volume effects were accounted for by applying a correction for the presence of cerebrospinal fluid in the magnetic resonance spectroscopy voxel. We investigated how metabolite concentrations related to apolipoprotein ɛ4 genotype, dementia family history, a risk score (Cardiovascular Risk Factors, Aging and Incidence of Dementia -CAIDE) for future dementia including non-modifiable and potentially-modifiable factors and dietary patterns (adherence to Mediterranean diet). Dementia family history was associated with decreased total N-acetylaspartate and no differences were found between apolipoprotein ɛ4 carriers and non-carriers. A higher Cardiovascular Risk Factors, Aging, and Incidence of Dementia score related to higher myo-inositol, choline, total creatine and glutamate-glutamine, an effect which was mainly driven by older age and a higher body mass index. Greater adherence to the Mediterranean diet was associated with lower choline, myo-inositol and total creatine; these effects did not survive correction for multiple comparisons. The observed associations suggest that at midlife the brain demonstrates subtle neurochemical changes in relation to both inherited and potentially modifiable risk factors for future dementia.
Collapse
Affiliation(s)
- Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK
| | - Elizabeth McKiernan
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK
| | - Michael Langsen
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Sarah Gregory
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Graciela Muniz-Terrera
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh, EH16 4UX, UK
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | | | - Marius Ovidiu Mada
- Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, CB2 7EF, UK
| | - Guy B Williams
- Department of Clinical Neurosciences and Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Brian Lawlor
- Institute of Neuroscience, Trinity College Dublin, University of Dublin, Dublin, D02 PX31, Ireland
| | - Lorina Naci
- Institute of Neuroscience, Trinity College Dublin, University of Dublin, Dublin, D02 PX31, Ireland
| | - Clare Mackay
- Department of Psychiatry, Oxford University, Oxford, OX3 7JX, UK
| | - Ivan Koychev
- Department of Psychiatry, Oxford University, Oxford, OX3 7JX, UK
| | - Paresh Malhotra
- Department of Brain Sciences, Imperial College Healthcare NHS Trust, London, W12 0NN, UK
| | - Karen Ritchie
- INM, Univ Montpellier, INSERM, Montpellier, 34090, France
| | - Craig W Ritchie
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Li Su
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Adam D Waldman
- Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Department of Brain Sciences, Imperial College Healthcare NHS Trust, London, W12 0NN, UK
| | - John T O’ Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SP, UK
| |
Collapse
|
13
|
Sheikh-Bahaei N, Chen M, Pappas I. Magnetic Resonance Spectroscopy (MRS) in Alzheimer's Disease. Methods Mol Biol 2024; 2785:115-142. [PMID: 38427192 DOI: 10.1007/978-1-0716-3774-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
MRS is a noninvasive technique to measure different metabolites in the brain. Changes in the levels of certain metabolites can be used as surrogate markers for Alzheimer's disease. They can potentially be used for diagnosis, prediction of prognosis, or even assessing response to treatment.There are different techniques for MRS acquisitions including STimulated Echo Acquisition Mode (STEAM) and Point Resolved Spectroscopy (PRESS). In terms of localization, single or multi-voxel methods can be used. Based on current data: 1. NAA, marker of neuronal integrity and viability, reduces in AD with longitudinal changes over the time as the disease progresses. There are data claiming that reduction of NAA is associated with tau accumulation, early neurodegenerative processes, and cognitive decline. Therefore, it can be used as a stage biomarker for AD to assess the severity of the disease. With advancement of disease modifying therapies, there is a potential role for NAA in the future to be used as a marker of response to treatment. 2. mI, marker of glial cell proliferation and activation, is associated with AB pathology and has early changes in the course of the disease. The NAA/mI ratio can be predictive of AD development with high specificity and can be utilized in the clinical setting to stratify cases for further evaluation with PET for potential treatments. 3. The changes in the level of other metabolites such as Chol, Glu, Gln, and GABA are controversial because of the lack of standardization of MRS techniques, current technical limitations, and possible region specific changes. 4. Ultrahigh field MRS and more advanced techniques can overcome many of these limitations and enable us to measure more metabolites with higher accuracy. 5. Standardization of MRS techniques, validation of metabolites' changes against PET using PET-guided technique, and longitudinal follow-ups to investigate the temporal changes of the metabolites in relation to other biomarkers and cognition will be crucial to confirm the utility of MRS as a potential noninvasive biomarker for AD.
Collapse
Affiliation(s)
- Nasim Sheikh-Bahaei
- Department of Radiology, Keck School of Medicine of USC, Los Angeles, CA, USA.
| | - Michelle Chen
- Keck School of Medicine of USC, USC, Los Angeles, CA, USA
| | - Ioannis Pappas
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, USC, Los Angeles, CA, USA
| |
Collapse
|
14
|
Rustamzadeh A, Sadigh N, Shabani R, Ahadi R, Vahabi Z, Shabani A, Mohebi N, Khamseh F, Behruzi M, Moradi F. Neurochemical Ameliorating of the Hippocampus in Dyslipidemic Alzheimer Patients Following Silymarin; a Double-Blind Placebo-Controlled Randomized Clinical Trial. Med J Islam Repub Iran 2023; 37:123. [PMID: 38318412 PMCID: PMC10843210 DOI: 10.47176/mjiri.37.123] [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: 08/10/2023] [Indexed: 02/07/2024] Open
Abstract
Background Amyloid-beta (Aβ) production is a normal physiological process, and an imbalance in Aβ production/excretion rate is the basis of the plaque load increase in AD. LRP1 is involved in both central clearance of Aβ from the CNS and transport of Aβ toward peripheral organs. In this study, the effect of silymarin combination compared to rosuvastatin and placebo on neuro-metabolites and serum levels of LRP1 and Aβ1-42 proteins and oxidative stress enzymes and lipid and cognitive tests of Iranian AD patients. Methods In this double-blind placebo-controlled study, thirty-six mild AD patients were divided into groups (n=12) of silymarin 140mg, placebo, and rosuvastatin 10mg. Medications were administered 3 times a day for 6 months. Clinical tests, lipid profile (TG, HDL, TC, and LDL), Aβ1-42, and LRP1 markers were measured at the beginning and end of the intervention. Magnetic resonance spectroscopy (MRS) was used to measure metabolites. Using SPSS software a one-way ANOVA test was used to compare the means of the quantitative variables and Pearson and Spearman's correlations to measure the correlation. GraphPad Prism software was used for drawing graphs. P < 0.05 was considered a significant. Results The levels of LRP1 and Aβ1-42 in the silymarin group were significantly increased compared to the other groups (P < 0.05). NAA/mI in the silymarin group had a significant increase compared to both placebo and rosuvastatin groups (P < 0.05). Right and left hippocampal mI/Cr directly correlated with TG (r = 0.603, P = 0.003 and r = 0.595, P = 0.004, respectively). NAA/Cr of the right and left hippocampus was inversely related to TG (r = -0.511, P = 0.0033, and r = -0.532, P = 0.0021, respectively). NAA/Cr and NAA/mI of bilateral hippocampi directly correlated with HDL (P < 0.05). An inverse correlation was observed between the Aβ1-42 and mI/Cr of the right and left hippocampus (r = -0.661, P = 0.000 and r = -0.638, P = 0.000, respectively). Conclusion Donepezil and silymarin improved lipid profile associated with increased NAA/Cr, and decreased mI/Cr, in AD patients. Biomarker NAA/mI can be clinically significant in examining AD pathology. Measurement of the lipid factors and neurometabolites can be a suitable method for monitoring this disease.
Collapse
Affiliation(s)
- Auob Rustamzadeh
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nader Sadigh
- Department of Emergency Medicine, School of Medicine, Trauma and Injury Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Ahadi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Vahabi
- Department of Geriatric Medicine, Ziaeian Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Shabani
- Saadatabad Medical Imaging Center, Department of Advanced Imaging and Image Processing, Tehran, Iran
| | - Nafiseh Mohebi
- Department of Neurology, Rasool Akram Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khamseh
- Department of Neurology, Faculty of Medicine, Islamic Azad University, Tehran, Iran
| | - Masume Behruzi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
McKiernan E, Su L, O'Brien J. MRS in neurodegenerative dementias, prodromal syndromes and at-risk states: A systematic review of the literature. NMR IN BIOMEDICINE 2023; 36:e4896. [PMID: 36624067 DOI: 10.1002/nbm.4896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND In recent years, MRS has benefited from increased MRI field strengths, new acquisition protocols and new processing techniques. This review aims to determine how this has altered our understanding of MRS neurometabolic markers in neurodegenerative dementias. METHODS Our systematic review of human in vivo MRS literature since 2002 pertains to Alzheimer's disease (AD), dementia with Lewy bodies (DLB), Parkinson's disease dementia, frontotemporal dementia (FTD), prodromal and 'at-risk' states. Studies using field strengths of 3 T or more were included. RESULTS Of 85 studies, AD and/or mild cognitive impairment (MCI) were the most common conditions of interest (58 papers, 68%). Only 14 (16%) studies included other dementia syndromes and 13 (15%) investigated 'at-risk' cohorts. Earlier findings of lower N-acetylaspartate and higher myo-inositol were confirmed. Additionally, lower choline and creatine in AD and MCI were reported, though inconsistently. Previously challenging-to-measure metabolites (glutathione, glutamate and gamma-aminobutyric acid) were reportedly lower in AD, FTD and DLB compared with controls. DISCUSSION Increasing field strength alongside targeted acquisition protocols has revealed additional metabolite changes. Most studies were small and regional metabolite differences between dementia types may not have been captured due to the predominant placement of voxels in the posterior cingulate cortex. The standard of data collection, quality control and analysis is improving due to greater consensus regarding acquisition and processing techniques. Ongoing harmonization of techniques, creation of larger and longitudinal cohorts, and placement of MRS voxels in more diverse regions will strengthen future research.
Collapse
Affiliation(s)
- Elizabeth McKiernan
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Li Su
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - John O'Brien
- Department of Psychiatry, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Calderón-Garcidueñas L, Hernández-Luna J, Aiello-Mora M, Brito-Aguilar R, Evelson PA, Villarreal-Ríos R, Torres-Jardón R, Ayala A, Mukherjee PS. APOE Peripheral and Brain Impact: APOE4 Carriers Accelerate Their Alzheimer Continuum and Have a High Risk of Suicide in PM 2.5 Polluted Cities. Biomolecules 2023; 13:927. [PMID: 37371506 DOI: 10.3390/biom13060927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
This Review emphasizes the impact of APOE4-the most significant genetic risk factor for Alzheimer's disease (AD)-on peripheral and neural effects starting in childhood. We discuss major mechanistic players associated with the APOE alleles' effects in humans to understand their impact from conception through all life stages and the importance of detrimental, synergistic environmental exposures. APOE4 influences AD pathogenesis, and exposure to fine particulate matter (PM2.5), manufactured nanoparticles (NPs), and ultrafine particles (UFPs) associated with combustion and friction processes appear to be major contributors to cerebrovascular dysfunction, neuroinflammation, and oxidative stress. In the context of outdoor and indoor PM pollution burden-as well as Fe, Ti, and Al alloys; Hg, Cu, Ca, Sn, and Si UFPs/NPs-in placenta and fetal brain tissues, urban APOE3 and APOE4 carriers are developing AD biological disease hallmarks (hyperphosphorylated-tau (P-tau) and amyloid beta 42 plaques (Aβ42)). Strikingly, for Metropolitan Mexico City (MMC) young residents ≤ 40 y, APOE4 carriers have 4.92 times higher suicide odds and 23.6 times higher odds of reaching Braak NFT V stage versus APOE4 non-carriers. The National Institute on Aging and Alzheimer's Association (NIA-AA) framework could serve to test the hypothesis that UFPs and NPs are key players for oxidative stress, neuroinflammation, protein aggregation and misfolding, faulty complex protein quality control, and early damage to cell membranes and organelles of neural and vascular cells. Noninvasive biomarkers indicative of the P-tau and Aβ42 abnormal protein deposits are needed across the disease continuum starting in childhood. Among the 21.8 million MMC residents, we have potentially 4 million APOE4 carriers at accelerated AD progression. These APOE4 individuals are prime candidates for early neuroprotective interventional trials. APOE4 is key in the development of AD evolving from childhood in highly polluted urban centers dominated by anthropogenic and industrial sources of pollution. APOE4 subjects are at higher early risk of AD development, and neuroprotection ought to be implemented. Effective reductions of PM2.5, UFP, and NP emissions from all sources are urgently needed. Alzheimer's Disease prevention ought to be at the core of the public health response and physicians-scientist minority research be supported.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- College of Health, The University of Montana, Missoula, MT 59812, USA
- Universidad del Valle de México, Mexico City 14370, Mexico
| | | | - Mario Aiello-Mora
- Otorrinolaryngology Department, Instituto Nacional de Cardiología, Mexico City 14080, Mexico
| | | | - Pablo A Evelson
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113 AAD, Argentina
| | | | - Ricardo Torres-Jardón
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alberto Ayala
- Sacramento Metropolitan Air Quality Management District, Sacramento, CA 95814, USA
- West Virginia University, Morgantown, WV 26506, USA
| | - Partha S Mukherjee
- Interdisciplinary Statistical Research Unit, Indian Statistical Institute, Kolkata 700108, India
| |
Collapse
|
17
|
Cengiz S, Arslan DB, Kicik A, Erdogdu E, Yildirim M, Hatay GH, Tufekcioglu Z, Uluğ AM, Bilgic B, Hanagasi H, Demiralp T, Gurvit H, Ozturk-Isik E. Identification of metabolic correlates of mild cognitive impairment in Parkinson's disease using magnetic resonance spectroscopic imaging and machine learning. MAGMA (NEW YORK, N.Y.) 2022; 35:997-1008. [PMID: 35867235 DOI: 10.1007/s10334-022-01030-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To investigate metabolic changes of mild cognitive impairment in Parkinson's disease (PD-MCI) using proton magnetic resonance spectroscopic imaging (1H-MRSI). METHODS Sixteen healthy controls (HC), 26 cognitively normal Parkinson's disease (PD-CN) patients, and 34 PD-MCI patients were scanned in this prospective study. Neuropsychological tests were performed, and three-dimensional 1H-MRSI was obtained at 3 T. Metabolic parameters and neuropsychological test scores were compared between PD-MCI, PD-CN, and HC. The correlations between neuropsychological test scores and metabolic intensities were also assessed. Supervised machine learning algorithms were applied to classify HC, PD-CN, and PD-MCI groups based on metabolite levels. RESULTS PD-MCI had a lower corrected total N-acetylaspartate over total creatine ratio (tNAA/tCr) in the right precentral gyrus, corresponding to the sensorimotor network (p = 0.01), and a lower tNAA over myoinositol ratio (tNAA/mI) at a part of the default mode network, corresponding to the retrosplenial cortex (p = 0.04) than PD-CN. The HC and PD-MCI patients were classified with an accuracy of 86.4% (sensitivity = 72.7% and specificity = 81.8%) using bagged trees. CONCLUSION 1H-MRSI revealed metabolic changes in the default mode, ventral attention/salience, and sensorimotor networks of PD-MCI patients, which could be summarized mainly as 'posterior cortical metabolic changes' related with cognitive dysfunction.
Collapse
Affiliation(s)
- Sevim Cengiz
- Institute of Biomedical Engineering, Bogazici University, 34684, Istanbul, Turkey
| | - Dilek Betul Arslan
- Institute of Biomedical Engineering, Bogazici University, 34684, Istanbul, Turkey
| | - Ani Kicik
- Neuroimaging Unit, Hulusi Behcet Life Sciences Research Center, Istanbul University, Istanbul, Turkey
- Department of Physiology, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Turkey
| | - Emel Erdogdu
- Neuroimaging Unit, Hulusi Behcet Life Sciences Research Center, Istanbul University, Istanbul, Turkey
- Department of Psychology, Faculty of Economics and Administrative Sciences, Isik University, Istanbul, Turkey
| | - Muhammed Yildirim
- Institute of Biomedical Engineering, Bogazici University, 34684, Istanbul, Turkey
| | - Gokce Hale Hatay
- Institute of Biomedical Engineering, Bogazici University, 34684, Istanbul, Turkey
| | - Zeynep Tufekcioglu
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- Department of Neurology, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| | - Aziz Müfit Uluğ
- Institute of Biomedical Engineering, Bogazici University, 34684, Istanbul, Turkey
- CorTechs Labs, San Diego, CA, USA
| | - Basar Bilgic
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hasmet Hanagasi
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Tamer Demiralp
- Neuroimaging Unit, Hulusi Behcet Life Sciences Research Center, Istanbul University, Istanbul, Turkey
- Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hakan Gurvit
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Esin Ozturk-Isik
- Institute of Biomedical Engineering, Bogazici University, 34684, Istanbul, Turkey.
| |
Collapse
|
18
|
Mechanistic Insights into the Neuroprotective Potential of Sacred Ficus Trees. Nutrients 2022; 14:nu14224731. [PMID: 36432418 PMCID: PMC9695857 DOI: 10.3390/nu14224731] [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: 09/06/2022] [Revised: 10/16/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
Ficus religiosa (Bo tree or sacred fig) and Ficus benghalensis (Indian banyan) are of immense spiritual and therapeutic importance. Various parts of these trees have been investigated for their antioxidant, antimicrobial, anticonvulsant, antidiabetic, anti-inflammatory, analgesic, hepatoprotective, dermoprotective, and nephroprotective properties. Previous reviews of Ficus mostly discussed traditional usages, photochemistry, and pharmacological activities, though comprehensive reviews of the neuroprotective potential of these Ficus species extracts and/or their important phytocompounds are lacking. The interesting phytocompounds from these trees include many bengalenosides, carotenoids, flavonoids (leucopelargonidin-3-O-β-d-glucopyranoside, leucopelargonidin-3-O-α-l-rhamnopyranoside, lupeol, cetyl behenate, and α-amyrin acetate), flavonols (kaempferol, quercetin, myricetin), leucocyanidin, phytosterols (bergapten, bergaptol, lanosterol, β-sitosterol, stigmasterol), terpenes (α-thujene, α-pinene, β-pinene, α-terpinene, limonene, β-ocimene, β-bourbonene, β-caryophyllene, α-trans-bergamotene, α-copaene, aromadendrene, α-humulene, alloaromadendrene, germacrene, γ-cadinene, and δ-cadinene), and diverse polyphenols (tannin, wax, saponin, leucoanthocyanin), contributing significantly to their pharmacological effects, ranging from antimicrobial action to neuroprotection. This review presents extensive mechanistic insights into the neuroprotective potential, especially important phytochemicals from F. religiosa and F. benghalensis. Owing to the complex pathophysiology of neurodegenerative disorders (NDDs), the currently existing drugs merely alleviate the symptoms. Hence, bioactive compounds with potent neuroprotective effects through a multitarget approach would be of great interest in developing pharmacophores for the treatment of NDDs.
Collapse
|
19
|
Spatio-temporal metabolic rewiring in the brain of TgF344-AD rat model of Alzheimer's disease. Sci Rep 2022; 12:16958. [PMID: 36216838 PMCID: PMC9550832 DOI: 10.1038/s41598-022-20962-6] [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: 05/10/2022] [Accepted: 09/21/2022] [Indexed: 12/29/2022] Open
Abstract
Brain damage associated with Alzheimer's disease (AD) occurs even decades before the symptomatic onset, raising the need to investigate its progression from prodromal stages. In this context, animal models that progressively display AD pathological hallmarks (e.g. TgF344-AD) become crucial. Translational technologies, such as magnetic resonance spectroscopy (MRS), enable the longitudinal metabolic characterization of this disease. However, an integrative approach is required to unravel the complex metabolic changes underlying AD progression, from early to advanced stages. TgF344-AD and wild-type (WT) rats were studied in vivo on a 7 Tesla MRI scanner, for longitudinal quantitative assessment of brain metabolic profile changes using MRS. Disease progression was investigated at 4 time points, from 9 to 18 months of age, and in 4 regions: cortex, hippocampus, striatum, and thalamus. Compared to WT, TgF344-AD rats replicated common findings in AD patients, including decreased N-acetylaspartate in the cortex, hippocampus and thalamus, and decreased glutamate in the thalamus and striatum. Different longitudinal evolution of metabolic concentration was observed between TgF344-AD and WT groups. Namely, age-dependent trajectories differed between groups for creatine in the cortex and thalamus and for taurine in cortex, with significant decreases in Tg344-AD animals; whereas myo-inositol in the thalamus and striatum showed greater increase along time in the WT group. Additional analysis revealed divergent intra- and inter-regional metabolic coupling in each group. Thus, in cortex, strong couplings of N-acetylaspartate and creatine with myo-inositol in WT, but with taurine in TgF344-AD rats were observed; whereas in the hippocampus, myo-inositol, taurine and choline compounds levels were highly correlated in WT but not in TgF344-AD animals. Furthermore, specific cortex-hippocampus-striatum metabolic crosstalks were found for taurine levels in the WT group but for myo-inositol levels in the TgF344-AD rats. With a systems biology perspective of metabolic changes in AD pathology, our results shed light into the complex spatio-temporal metabolic rewiring in this disease, reported here for the first time. Age- and tissue-dependent imbalances between myo-inositol, taurine and other metabolites, such as creatine, unveil their role in disease progression, while pointing to the inadequacy of the latter as an internal reference for quantification.
Collapse
|
20
|
Gozdas E, Hinkley L, Fingerhut H, Dacorro L, Gu M, Sacchet MD, Hurd R, Hosseini SMH. 1H-MRS neurometabolites and associations with neurite microstructures and cognitive functions in amnestic mild cognitive impairment. Neuroimage Clin 2022; 36:103159. [PMID: 36063758 PMCID: PMC9450331 DOI: 10.1016/j.nicl.2022.103159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/04/2022] [Accepted: 08/18/2022] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) pathogenesis is associated with alterations in neurometabolites and cortical microstructure. However, our understanding of alterations in neurochemicals in the prefrontal cortex and their relationship with changes in cortical microstructure in AD remains unclear. Here, we studied the levels of neurometabolites in the left dorsolateral prefrontal cortex (DLPFC) in healthy older adults and patients with amnestic Mild Cognitive Impairments (aMCI) using single-voxel proton-magnetic resonance spectroscopy (1H-MRS). N-acetyl aspartate (NAA), glutamate+glutamate (Glx), Myo-inositol (mI), and γ-aminobutyric acid (GABA) brain metabolite levels were quantified relative to total creatine (tCr = Cr + PCr). aMCI had significantly decreased NAA/tCr, Glx/tCr, NAA/mI, and increased mI/tCr levels compared with healthy controls. Further, we leveraged advanced diffusion MRI to extract neurite properties in the left DLPFC and found a significant positive correlation between NAA/tCr, related to neuronal intracellular compartment, and neurite density (ICVF, intracellular volume fraction), and a negative correlation between mI/tCr and neurite orientation (ODI) only in healthy older adults. These data suggest a potential decoupling in the relationship between neurite microstructures and NAA and mI concentrations in DLPFC in the early stage of AD. Together, our results confirm altered DLPFC neurometabolites in prodromal phase of AD and provide unique evidence regarding the imbalance in the association between neurometabolites and neurite microstructure in early stage of AD.
Collapse
Affiliation(s)
- Elveda Gozdas
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA.
| | - Lauren Hinkley
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hannah Fingerhut
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauren Dacorro
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Meng Gu
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Matthew D Sacchet
- Center for Depression, Anxiety, and Stress Research, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Ralph Hurd
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - S M Hadi Hosseini
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
21
|
Lower Posterior Cingulate N-Acetylaspartate to Creatine Level in Early Detection of Biologically Defined Alzheimer's Disease. Brain Sci 2022; 12:brainsci12060722. [PMID: 35741606 PMCID: PMC9220959 DOI: 10.3390/brainsci12060722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/18/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) was recently defined as a biological construct to reflect neuropathologic status, and both abnormal amyloid and tau are required for a diagnosis of AD. We aimed to determine the proton MR spectroscopic (1H-MRS) patterns of the posterior cingulate in biologically defined AD. A total of 68 participants were included in this study, comprising 37 controls, 16 early AD, and 15 late AD, who were classified according to their amyloid and tau status and presence of hippocampal atrophy. Compared with controls, early AD showed lower N-acetylaspartate (NAA)/creatine (Cr) (p = 0.003), whereas late AD showed lower NAA/Cr and higher myoInositol (mI)/Cr (all with p < 0.05). Lower NAA/Cr correlated with a greater global amyloid load (r = −0.47, p < 0.001) and tau load (r = −0.51, p < 0.001) and allowed a discrimination of early AD from controls (p < 0.001). Subgroup analysis showed that NAA/Cr also allowed a differentiation of early AD from controls in the cognitively unimpaired subjects, with an area under the receiver operating characteristics curve, sensitivity, and specificity of 0.96, 100%, and 83.8%, respectively. Lower posterior cingulate NAA levels may help to inform underlying neuropathologic changes in the early stage of AD.
Collapse
|
22
|
Spotorno N, Najac C, Stomrud E, Mattsson-Carlgren N, Palmqvist S, van Westen D, Ronen I, Hansson O. Astrocytic function is associated with both amyloid-β and tau pathology in non-demented APOE ε4 carriers. Brain Commun 2022; 4:fcac135. [PMID: 35702728 PMCID: PMC9185373 DOI: 10.1093/braincomms/fcac135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 02/17/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
A growing body of evidence suggests that astrocytes play a major role in the pathophysiology of Alzheimer’s disease. Given that APOE is primarily expressed in astrocytes, these cells might be an important link between the APOE ε4 allele and development of Alzheimer’s disease pathology. Here, we investigate this hypothesis in vivo by measuring myo-Inositol, a metabolite involved in astrocytic functions, with magnetic resonance spectroscopy. Currently, there are conflicting evidence regarding the relationship between APOE ε4 and myo-Inositol concentration. Furthermore, data supporting a relationship between APOE ε4, myo-Inositol and Alzheimer’s disease pathology (amyloid-beta and tau proteins) in the preclinical stage of Alzheimer’s disease are limited. A previous study revealed differences in myo-Inositol levels between APOE ε4 carriers and noncarriers already in preclinical Alzheimer’s disease participant. However, other reports showed no impact of APOE genotype on the association between myo-Inositol and rate of amyloid-beta accumulation. In the present study we determined the effect of APOE genotype on the association between myo-Inositol and both amyloid-β and tau deposition quantified by PET in 428 cognitively unimpaired elderly and patients with mild cognitive impairment from the Swedish BioFINDER-2 cohort.
APOE genotype impacted the associations between myo-Inositol and amyloid-β pathology as revealed by an interaction effect between APOE genotype and levels of myo-inositol (p < 0.001) such that higher myo-Inositol concentration was related to more amyloid-beta pathology in APOE ε4 carriers only. A similar interaction effect was also found when investigating the effect of APOE on the association between myo-inositol and tau pathology (p < 0.01). Focusing on the APOE ε4 subsample, myo-Inositol partially (17%) mediated the association between amyloid-beta and tau pathology (p < 0.05). Further, in a subgroup of participants with available plasma levels of glial fibrillary acidic protein, a marker of astroglial activation and astrocytosis, we found that glial fibrillary acidic protein correlated with myo-inositol only in APOE e4 carriers (APOE ε4 carriers: p < 0.01; APOE ε4 non carriers: p > 0.8), suggesting that myo-Inosotol might reflect an aspect of the astrocytic involvement in Alzheimer’s pathology which is specific to the impact of APOE ε4. Therefore, we suggest that myo-Inositol is a candidate in vivo marker to study the impact of APOE ε4 on the interplay between astrocytes and the pathophysiology of Alzheimer’s disease.
Collapse
Affiliation(s)
- Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Chloé Najac
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Danielle van Westen
- Image and Function, Skane University Hospital, Lund, Sweden
- Diagnostic Radiology, Institution for Clinical Sciences, Lund University, Lund, Sweden
| | - Itamar Ronen
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
23
|
Kara F, Joers JM, Deelchand DK, Park YW, Przybelski SA, Lesnick TG, Senjem ML, Zeydan B, Knopman DS, Lowe VJ, Vemuri P, Mielke MM, Machulda MM, Jack CR, Petersen RC, Öz G, Kantarci K. 1H MR spectroscopy biomarkers of neuronal and synaptic function are associated with tau deposition in cognitively unimpaired older adults. Neurobiol Aging 2022; 112:16-26. [PMID: 35038671 PMCID: PMC8976711 DOI: 10.1016/j.neurobiolaging.2021.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 12/25/2022]
Abstract
Proton magnetic resonance spectroscopy (1H MRS) may provide information on pathophysiological changes associated with tau deposition in cognitively unimpaired older adults. In this study, the associations of posterior cingulate gyrus tau and amyloid beta (Aβ) deposition on PET with 1H MRS metabolite ratios acquired from bilateral posterior cingulate gyri were investigated in cognitively unimpaired older adults. Participants (n = 40) from the Mayo Clinic Study of Aging underwent single-voxel sLASER 1H MRS from the posterior cingulate gyrus at 3 Tesla, 18F-flortaucipir, and 11C- Pittsburgh Compound B (PiB) PET. An increase in posterior cingulate gyrus tau deposition, but not elevated Aβ, was associated with lower N-acetylaspartate/total creatine (tCr) and glutamate (Glu)/tCr ratios, and sex by tau interaction was observed in association with Glu/tCr. Higher tau levels in cognitively unimpaired older adults are associated with biomarkers of neural and synaptic injury even in the absence of cognitive impairment and these relationships appear to be stronger in women than in men.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - James M Joers
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Dinesh K Deelchand
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Young Woo Park
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Burcu Zeydan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | | | | | - Gülin Öz
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
24
|
Parker AF, Ohlhauser L, Scarapicchia V, Smart CM, Szoeke C, Gawryluk JR. A Systematic Review of Neuroimaging Studies Comparing Individuals with Subjective Cognitive Decline to Healthy Controls. J Alzheimers Dis 2022; 86:1545-1567. [DOI: 10.3233/jad-215249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Individuals with subjective cognitive decline (SCD) are hypothesized to be the earliest along the cognitive continuum between healthy aging and Alzheimer’s disease (AD), although more research is needed on this topic. Given that treatment approaches may be most effective pre-clinically, a primary objective of emerging research is to identify biological markers of SCD using neuroimaging methods. Objective: The current review aimed to comprehensively present the neuroimaging studies on SCD to date. Methods: PubMed and PsycINFO databases were searched for neuroimaging studies of individuals with SCD. Quality assessments were completed using the Appraisal tool for Cross-Sectional Studies. Results: In total, 62 neuroimaging studies investigating differences between participants with SCD and healthy controls were identified. Specifically, the number of studies were as follows: 36 MRI, 6 PET, 8 MRI/PET, 4 EEG, 7 MEG, and 1 SPECT. Across neuroimaging modalities, 48 of the 62 included studies revealed significant differences in brain structure and/or function between groups. Conclusion: Neuroimaging methods can identify differences between healthy controls and individuals with SCD. However, inconsistent results were found within and between neuroimaging modalities. Discrepancies across studies may be best accounted for by methodological differences, notably variable criteria for SCD, and differences in participant characteristics and risk factors for AD. Clinic based recruitment and cross-sectional study design were common and may bias the literature. Future neuroimaging investigations of SCD should consistently incorporate the standardized research criteria for SCD (as recommended by the SCD-Initiative), include more details of their SCD sample and their symptoms, and examine groups longitudinally.
Collapse
Affiliation(s)
- Ashleigh F. Parker
- Department of Psychology, University of Victoria, BC, Canada
- Institute on Aging and Lifelong Health, University of Victoria, BC, Canada
| | - Lisa Ohlhauser
- Department of Psychology, University of Victoria, BC, Canada
- Institute on Aging and Lifelong Health, University of Victoria, BC, Canada
| | - Vanessa Scarapicchia
- Department of Psychology, University of Victoria, BC, Canada
- Institute on Aging and Lifelong Health, University of Victoria, BC, Canada
| | - Colette M. Smart
- Department of Psychology, University of Victoria, BC, Canada
- Institute on Aging and Lifelong Health, University of Victoria, BC, Canada
| | - Cassandra Szoeke
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Jodie R. Gawryluk
- Department of Psychology, University of Victoria, BC, Canada
- Institute on Aging and Lifelong Health, University of Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, BC, Canada
| |
Collapse
|
25
|
Takado Y, Takuwa H, Sampei K, Urushihata T, Takahashi M, Shimojo M, Uchida S, Nitta N, Shibata S, Nagashima K, Ochi Y, Ono M, Maeda J, Tomita Y, Sahara N, Near J, Aoki I, Shibata K, Higuchi M. MRS-measured glutamate versus GABA reflects excitatory versus inhibitory neural activities in awake mice. J Cereb Blood Flow Metab 2022; 42:197-212. [PMID: 34515548 PMCID: PMC8721779 DOI: 10.1177/0271678x211045449] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
To assess if magnetic resonance spectroscopy (MRS)-measured Glutamate (Glu) and GABA reflect excitatory and inhibitory neural activities, respectively, we conducted MRS measurements along with two-photon mesoscopic imaging of calcium signals in excitatory and inhibitory neurons of living, unanesthetized mice. For monitoring stimulus-driven activations of a brain region, MRS signals and mesoscopic neural activities were measured during two consecutive sessions of 15-min prolonged sensory stimulations. In the first session, putative excitatory neuronal activities were increased, while inhibitory neuronal activities remained at the baseline level. In the second half, while excitatory neuronal activities remained elevated, inhibitory neuronal activities were significantly enhanced. We assessed regional neurochemical statuses by measuring MRS signals, which were overall in accordance with the neural activities, and neuronal activities and neurochemical statuses in a mouse model of Dravet syndrome under resting condition. Mesoscopic assessments showed that activities of inhibitory neurons in the cortex were diminished relative to wild-type mice in contrast to spared activities of excitatory neurons. Consistent with these observations, the Dravet model exhibited lower concentrations of GABA than wild-type controls. Collectively, the current investigations demonstrate that MRS-measured Glu and GABA can reflect spontaneous and stimulated activities of neurons producing and releasing these neurotransmitters in an awake condition.
Collapse
Affiliation(s)
- Yuhei Takado
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Yuhei Takado, Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | - Hiroyuki Takuwa
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Hiroyuki Takuwa, Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | - Kazuaki Sampei
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takuya Urushihata
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Manami Takahashi
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masafumi Shimojo
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Shoko Uchida
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Nobuhiro Nitta
- Department of Molecular Imaging and Theranostics, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Sayaka Shibata
- Department of Molecular Imaging and Theranostics, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Keisuke Nagashima
- Kansai Photon Science Institute, National Institutes for Quantum and Radiological Science and Technology, Kyoto, Japan
| | - Yoshihiro Ochi
- Kansai Photon Science Institute, National Institutes for Quantum and Radiological Science and Technology, Kyoto, Japan
| | - Maiko Ono
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Jun Maeda
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yutaka Tomita
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Jamie Near
- Douglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, Canada
| | - Ichio Aoki
- Department of Molecular Imaging and Theranostics, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kazuhisa Shibata
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Laboratory for Human Cognition and Learning, Center for Brain Science, RIKEN, Saitama, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
- Makoto Higuchi, Department of Functional Brain Imaging, Institute of Quantum Medical Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| |
Collapse
|
26
|
Orzyłowska A, Oakden W. Saturation Transfer MRI for Detection of Metabolic and Microstructural Impairments Underlying Neurodegeneration in Alzheimer's Disease. Brain Sci 2021; 12:53. [PMID: 35053797 PMCID: PMC8773856 DOI: 10.3390/brainsci12010053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/25/2021] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common causes of dementia and difficult to study as the pool of subjects is highly heterogeneous. Saturation transfer (ST) magnetic resonance imaging (MRI) methods are quantitative modalities with potential for non-invasive identification and tracking of various aspects of AD pathology. In this review we cover ST-MRI studies in both humans and animal models of AD over the past 20 years. A number of magnetization transfer (MT) studies have shown promising results in human brain. Increased computing power enables more quantitative MT studies, while access to higher magnetic fields improves the specificity of chemical exchange saturation transfer (CEST) techniques. While much work remains to be done, results so far are very encouraging. MT is sensitive to patterns of AD-related pathological changes, improving differential diagnosis, and CEST is sensitive to particular pathological processes which could greatly assist in the development and monitoring of therapeutic treatments of this currently incurable disease.
Collapse
Affiliation(s)
- Anna Orzyłowska
- Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8 (SPSK 4), 20-090 Lublin, Poland
| | - Wendy Oakden
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada;
| |
Collapse
|
27
|
Costigan A, Umla-Runge K, Evans C, Raybould R, Graham K, Lawrence A. Evidence against altered excitatory/inhibitory balance in the posteromedial cortex of young adult APOE E4 carriers: A resting state 1H-MRS study. NEUROIMAGE. REPORTS 2021; 1:100059. [PMID: 36896169 PMCID: PMC9986794 DOI: 10.1016/j.ynirp.2021.100059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/09/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022]
Abstract
A strategy to gain insight into early changes that may predispose people to Alzheimer's disease (AD) is to study the brains of younger cognitively healthy people that are at increased genetic risk of AD. The Apolipoprotein (APOE) E4 allele is the strongest genetic risk factor for AD, and several neuroimaging studies comparing APOE E4 carriers with non-carriers at age ∼20-30 years have detected hyperactivity (or reduced deactivation) in posteromedial cortex (PMC), a key hub of the default network (DN), which has a high susceptibility to early amyloid deposition in AD. Transgenic mouse models suggest such early network activity alterations may result from altered excitatory/inhibitory (E/I) balance, but this is yet to be examined in humans. Here we test the hypothesis that PMC fMRI hyperactivity could be underpinned by altered levels of excitatory (glutamate) and/or inhibitory (GABA) neurotransmitters in this brain region. Forty-seven participants (20 APOE E4 carriers and 27 non-carriers) aged 18-25 years underwent resting-state proton magnetic resonance spectroscopy (1H-MRS), a non-invasive neuroimaging technique to measure glutamate and GABA in vivo. Metabolites were measured in a PMC voxel of interest and in a comparison voxel in the occipital cortex (OCC). There was no difference in either glutamate or GABA between the E4 carriers and non-carriers in either MRS voxel, or in the ratio of glutamate to GABA, a measure of E/I balance. Default Bayesian t-tests revealed evidence in support of this null finding. Our findings suggest that PMC hyperactivity in APOE E4 carriers is unlikely to be associated with, or possibly may precede, alterations in local resting-state PMC neurotransmitters, thus informing our understanding of the spatio-temporal sequence of early network alterations underlying APOE E4 related AD risk.
Collapse
Affiliation(s)
- A.G. Costigan
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - K. Umla-Runge
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - C.J. Evans
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - R. Raybould
- UK Dementia Research Institute, Cardiff, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - K.S. Graham
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - A.D. Lawrence
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| |
Collapse
|
28
|
Ebert T, Heinz DE, Almeida-Corrêa S, Cruz R, Dethloff F, Stark T, Bajaj T, Maurel OM, Ribeiro FM, Calcagnini S, Hafner K, Gassen NC, Turck CW, Boulat B, Czisch M, Wotjak CT. Myo-Inositol Levels in the Dorsal Hippocampus Serve as Glial Prognostic Marker of Mild Cognitive Impairment in Mice. Front Aging Neurosci 2021; 13:731603. [PMID: 34867270 PMCID: PMC8633395 DOI: 10.3389/fnagi.2021.731603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/13/2021] [Indexed: 01/03/2023] Open
Abstract
Dementia is a devastating age-related disorder. Its therapy would largely benefit from the identification of susceptible subjects at early, prodromal stages of the disease. To search for such prognostic markers of cognitive impairment, we studied spatial navigation in male BALBc vs. B6N mice in combination with in vivo magnetic resonance spectroscopy (1H-MRS). BALBc mice consistently showed higher escape latencies than B6N mice, both in the Water Cross Maze (WCM) and the Morris water maze (MWM). These performance deficits coincided with higher levels of myo-inositol (mIns) in the dorsal hippocampus before and after training. Subsequent biochemical analyses of hippocampal specimens by capillary immunodetection and liquid chromatography mass spectrometry-based (LC/MS) metabolomics revealed a higher abundance of glial markers (IBA-1, S100B, and GFAP) as well as distinct alterations in metabolites including a decrease in vitamins (pantothenic acid and nicotinamide), neurotransmitters (acetylcholine), their metabolites (glutamine), and acetyl-L-carnitine. Supplementation of low abundant acetyl-L-carnitine via the drinking water, however, failed to revert the behavioral deficits shown by BALBc mice. Based on our data we suggest (i) BALBc mice as an animal model and (ii) hippocampal mIns levels as a prognostic marker of mild cognitive impairment (MCI), due to (iii) local changes in microglia and astrocyte activity, which may (iv) result in decreased concentrations of promnesic molecules.
Collapse
Affiliation(s)
- Tim Ebert
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Daniel E. Heinz
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
- Max Planck School of Cognition, Leipzig, Germany
| | | | - Renata Cruz
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Frederik Dethloff
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Tibor Stark
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Scientific Core Unit “Neuroimaging”, Max Planck Institute of Psychiatry, Munich, Germany
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Oriana M. Maurel
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Fabiola M. Ribeiro
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Silvio Calcagnini
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Nils C. Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| | - Christoph W. Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Munich, Germany
| | - Benoit Boulat
- Scientific Core Unit “Neuroimaging”, Max Planck Institute of Psychiatry, Munich, Germany
| | - Michael Czisch
- Scientific Core Unit “Neuroimaging”, Max Planck Institute of Psychiatry, Munich, Germany
| | - Carsten T. Wotjak
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, Munich, Germany
- Central Nervous System Diseases Research (CNSDR), Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
29
|
Liu H, Zhang D, Lin H, Zhang Q, Zheng L, Zheng Y, Yin X, Li Z, Liang S, Huang S. Meta-Analysis of Neurochemical Changes Estimated via Magnetic Resonance Spectroscopy in Mild Cognitive Impairment and Alzheimer's Disease. Front Aging Neurosci 2021; 13:738971. [PMID: 34744689 PMCID: PMC8569809 DOI: 10.3389/fnagi.2021.738971] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/26/2021] [Indexed: 11/18/2022] Open
Abstract
The changes of neurochemicals in mild cognitive impairment (MCI) and Alzheimer's disease (AD) patients has been observed via magnetic resonance spectroscopy in several studies. However, whether it exists the consistent pattern of changes of neurochemicals in the encephalic region during the progression of MCI to AD were still not clear. The study performed meta-analysis to investigate the patterns of neurochemical changes in the encephalic region in the progress of AD. We searched the PubMed, Embase, Cochrane Library, and Web of Science databases, and finally included 63 studies comprising 1,086 MCI patients, 1,256 AD patients, and 1,907 healthy controls. It showed that during the progression from MCI to AD, N-acetyl aspartate (NAA) decreased continuously in the posterior cingulate (PC) (SMD: −0.42 [95% CI: −0.62 to −0.21], z = −3.89, P < 0.05), NAA/Cr (creatine) was consistently reduced in PC (SMD: −0.58 [95% CI: −0.86 to −0.30], z = −4.06, P < 0.05) and hippocampus (SMD: −0.65 [95% CI: −1.11 to −0.12], z = −2.44, P < 0.05), while myo-inositol (mI) (SMD: 0.44 [95% CI: 0.26–0.61], z = 4.97, P < 0.05) and mI/Cr (SMD: 0.43 [95% CI: 0.17–0.68], z = 3.30, P < 0.05) were raised in PC. Furthermore, these results were further verified by a sustained decrease in the NAA/mI of PC (SMD: −0.94 [95% CI: −1.24 to −0.65], z = −6.26, P < 0.05). Therefore, the levels of NAA and mI were associated with the cognitive decline and might be used as potentially biomarkers to predict the possible progression from MCI to AD. Systematic Review Registration:https://www.crd.york.ac.uk/PROSPERO/, identifier: CRD42020200308.
Collapse
Affiliation(s)
- Huanhuan Liu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Dandan Zhang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huawei Lin
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Qi Zhang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ling Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuxin Zheng
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiaolong Yin
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zuanfang Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fuzhou, China
| | - Shengxiang Liang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China.,Traditional Chinese Medicine Rehabilitation Research Center of State Administration of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Saie Huang
- Department of Neurological Rehabilitation, Fujian University of Traditional Chinese Medicine Subsidiary Rehabilitation Hospital, Fuzhou, China
| |
Collapse
|
30
|
Yang Z, Wan X, Zhao X, Rong Y, Wu Y, Cao Z, Xie Q, Luo M, Liu Y. Brain neurometabolites differences in individuals with subjective cognitive decline plus: a quantitative single- and multi-voxel proton magnetic resonance spectroscopy study. Quant Imaging Med Surg 2021; 11:4074-4096. [PMID: 34476190 DOI: 10.21037/qims-20-1254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/23/2021] [Indexed: 11/06/2022]
Abstract
Background Subjective cognitive decline plus could be an extremely early phase of Alzheimer's disease; however, changes of N-acetylaspartate, myoinositol, and N-acetylaspartate/myoinositol is still unknown at this stage. This study aimed to explore brain neurometabolic alterations in patients with subjective cognitive decline plus using quantitative single-voxel and multi-voxel 1H-magnetic resonance spectroscopy. Methods A total of 91 participants were enrolled and underwent a GE 3.0-T magnetic resonance imaging, including 33 elderly controls, 27 patients with subjective cognitive decline plus, and 31 patients with amnestic mild cognitive impairment (MCI). Single-voxel and multi-voxel 1H-magnetic resonance spectroscopy were used to investigate the differences in neurometabolite levels among the three groups. Results Compared with elderly controls, patients with subjective cognitive decline plus showed significant decline in N-acetylaspartate and N-acetylaspartate/myoinositol values in multiple regions, and amnestic MCI participants demonstrated more significant decreased N-acetylaspartate and N-acetylaspartate/myoinositol levels in multiple regions. The combined concentrations of N-acetylaspartate with myoinositol showed an excellent discrimination between those with subjective cognitive decline plus and elderly controls as compared to that obtained using N-acetylaspartate/myoinositol ratios with the area under the receiver operating characteristic curve of 0.895 and 0.860, respectively. Likewise, the combined area under the curve for differentiating patients with subjective cognitive decline plus from amnestic MCI was obtained using the combined levels of N-acetylaspartate with myoinositol was 0.892. This was also higher than the combined area under the curve of 0.836 obtained using N-acetylaspartate/myoinositol ratios. Moreover, N-acetylaspartate levels in the left hippocampus and left posterior cingulate cortex (PCC) was positively related to the Auditory Verbal Learning Test delayed recall scores in patients with subjective cognitive decline plus, whereas only the N-acetylaspartate/myoinositol ratio was positively related to this scale scores in the left hippocampus. Conclusions Quantitative single-voxel and multi-voxel 1H-magnetic resonance spectroscopy can provide valuable information to detect alterative brain neurometabolites characteristics in patients with subjective cognitive decline plus. N-acetylaspartate concentrations may be used as one of the earliest neuroimaging markers at this stage, while N-acetylaspartate/myoinositol ratio could be more suitable for monitoring Alzheimer's disease progression.
Collapse
Affiliation(s)
- Zhongxian Yang
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Medical Imaging Center, the Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - Xing Wan
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xinzhu Zhao
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yu Rong
- Department of Neurology, the People's Hospital of Gaozhou City, Maoming, China
| | - Yi Wu
- Department of Neurology, Shantou Central Hospital and Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, China
| | - Zhen Cao
- Medical Imaging Center, the Second Affiliated Hospital, Medical College of Shantou University, Shantou, China
| | - Qiuxia Xie
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Min Luo
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yubao Liu
- Medical Imaging Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China.,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Montal V, Barroeta I, Bejanin A, Pegueroles J, Carmona-Iragui M, Altuna M, Benejam B, Videla L, Fernández S, Padilla C, Aranha MR, Iulita MF, Vidal-Piñeiro D, Alcolea D, Blesa R, Lleó A, Fortea J. Metabolite Signature of Alzheimer's Disease in Adults with Down Syndrome. Ann Neurol 2021; 90:407-416. [PMID: 34309066 DOI: 10.1002/ana.26178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The purpose of this study was to examine the Alzheimer's disease metabolite signature through magnetic resonance spectroscopy in adults with Down syndrome and its relation with Alzheimer's disease biomarkers and cortical thickness. METHODS We included 118 adults with Down syndrome from the Down Alzheimer Barcelona Imaging Initiative and 71 euploid healthy controls from the Sant Pau Initiative on Neurodegeneration cohort. We measured the levels of myo-inositol (a marker of neuroinflammation) and N-acetyl-aspartate (a marker of neuronal integrity) in the precuneus using magnetic resonance spectroscopy. We investigated the changes with age and along the disease continuum (asymptomatic, prodromal Alzheimer's disease, and Alzheimer's disease dementia stages). We assessed the relationship between these metabolites and Aβ42 /Aβ40 ratio, phosphorylated tau-181, neurofilament light (NfL), and YKL-40 cerebrospinal fluid levels as well as amyloid positron emission tomography uptake using Spearman correlations controlling for multiple comparisons. Finally, we computed the relationship between cortical thickness and metabolite levels using Freesurfer. RESULTS Asymptomatic adults with Down syndrome had a 27.5% increase in the levels of myo-inositol, but equal levels of N-acetyl-aspartate compared to euploid healthy controls. With disease progression, myo-inositol levels increased, whereas N-acetyl-aspartate levels decreased in symptomatic stages of the disease. Myo-inositol was associated with amyloid, tau, and neurodegeneration markers, mainly at symptomatic stages of the disease, whereas N-acetyl-aspartate was related to neurodegeneration biomarkers in symptomatic stages. Both metabolites were significantly associated with cortical thinning, mainly in symptomatic participants. INTERPRETATION Magnetic resonance spectroscopy detects Alzheimer's disease related inflammation and neurodegeneration, and could be a good noninvasive disease-stage biomarker in Down syndrome. ANN NEUROL 2021.
Collapse
Affiliation(s)
- Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - María Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Bessy Benejam
- Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Susana Fernández
- Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Concepcion Padilla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Didac Vidal-Piñeiro
- Department of Psychology, Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | | |
Collapse
|
32
|
Piersson AD, Mohamad M, Suppiah S, Rajab NF. Topographical patterns of whole-brain structural alterations in association with genetic risk, cerebrospinal fluid, positron emission tomography biomarkers of Alzheimer’s disease, and neuropsychological measures. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00440-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Foret JT, Dekhtyar M, Cole JH, Gourley DD, Caillaud M, Tanaka H, Haley AP. Network Modeling Sex Differences in Brain Integrity and Metabolic Health. Front Aging Neurosci 2021; 13:691691. [PMID: 34267647 PMCID: PMC8275835 DOI: 10.3389/fnagi.2021.691691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Hypothesis-driven studies have demonstrated that sex moderates many of the relationships between brain health and cardiometabolic disease, which impacts risk for later-life cognitive decline. In the present study, we sought to further our understanding of the associations between multiple markers of brain integrity and cardiovascular risk in a midlife sample of 266 individuals by using network analysis, a technique specifically designed to examine complex associations among multiple systems at once. Separate network models were constructed for male and female participants to investigate sex differences in the biomarkers of interest, selected based on evidence linking them with risk for late-life cognitive decline: all components of metabolic syndrome (obesity, hypertension, dyslipidemia, and hyperglycemia); neuroimaging-derived brain-predicted age minus chronological age; ratio of white matter hyperintensities to whole brain volume; seed-based resting state functional connectivity in the Default Mode Network, and ratios of N-acetyl aspartate, glutamate and myo-inositol to creatine, measured through proton magnetic resonance spectroscopy. Males had a sparse network (87.2% edges = 0) relative to females (69.2% edges = 0), indicating fewer relationships between measures of cardiometabolic risk and brain integrity. The edges in the female network provide meaningful information about potential mechanisms between brain integrity and cardiometabolic health. Additionally, Apolipoprotein ϵ4 (ApoE ϵ4) status and waist circumference emerged as central nodes in the female model. Our study demonstrates that network analysis is a promising technique for examining relationships between risk factors for cognitive decline in a midlife population and that investigating sex differences may help optimize risk prediction and tailor individualized treatments in the future.
Collapse
Affiliation(s)
- Janelle T. Foret
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Maria Dekhtyar
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - James H. Cole
- Department of Computer Science, Centre for Medical Image Computing, University College London, London, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Drew D. Gourley
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Marie Caillaud
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Hirofumi Tanaka
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Andreana P. Haley
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
- Biomedical Imaging Center, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
34
|
Mitolo M, Stanzani-Maserati M, Manners DN, Capellari S, Testa C, Talozzi L, Poda R, Oppi F, Evangelisti S, Gramegna LL, Magarelli S, Pantieri R, Liguori R, Lodi R, Tonon C. The Combination of Metabolic Posterior Cingulate Cortical Abnormalities and Structural Asymmetries Improves the Differential Diagnosis Between Primary Progressive Aphasia and Alzheimer's Disease. J Alzheimers Dis 2021; 82:1467-1473. [PMID: 34151798 DOI: 10.3233/jad-210211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Differential diagnosis between primary progressive aphasia (PPA) and Alzheimer's disease (AD) could be difficult if based on clinical grounds alone. We evaluated the combination of proton MR spectroscopy of posterior cingulate cortex (PCC) and quantitative structural imaging asymmetries to differentiate PPA from AD patients. A greater left-lateralized temporo-parietal atrophy (higher accuracy for the PCC, 81.4%) and metabolic neurodegenerative changes in PCC (accuracy 76.8%) was demonstrated in PPA versus AD. The combined multiparametric approach increased the accuracy to 94%in the differential diagnosis between these two neurodegenerative diseases.
Collapse
Affiliation(s)
- Micaela Mitolo
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy
| | | | - David N Manners
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Claudia Testa
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Lia Talozzi
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Roberto Poda
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Federico Oppi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
| | - Stefania Evangelisti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Laura L Gramegna
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Silvia Magarelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neurologia, Bologna, Italy
| | - Roberta Pantieri
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Neurologia, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Raffaele Lodi
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Caterina Tonon
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, OUC Neurologia, Bologna, Italy.,Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
35
|
Williams ME, Naudé PJW, van der Westhuizen FH. Proteomics and metabolomics of HIV-associated neurocognitive disorders: A systematic review. J Neurochem 2021; 157:429-449. [PMID: 33421125 DOI: 10.1111/jnc.15295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/09/2020] [Accepted: 12/30/2020] [Indexed: 02/01/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) are common features of the effect of human immunodeficiency virus (HIV)-1 within the central nervous system (CNS). The underlying neuropathophysiology of HAND is incompletely known. Furthermore, there are no markers to effectively predict or stratify the risk of HAND. Recent advancements in the fields of proteomics and metabolomics have shown promise in addressing these concerns, however, it is not clear if these approaches may provide new insight into pathways and markers related to HAND. We therefore conducted a systematic review of studies using proteomic and/or metabolomic approaches in the aim of identifying pathways or markers associated with neurocognitive impairment in people living with HIV (PLWH). Thirteen studies were eligible, including 11 proteomic and 2 metabolomic investigations of HIV-positive clinical samples (cerebrospinal fluid (CSF), brain tissue, and serum). Across varying profiling techniques and sample types, the majority of studies found an association of markers with neurocognitive function in PLWH. These included metabolic marker myo-inositol and proteomic markers superoxide dismutase, gelsolin, afamin, sphingomyelin, and ceramide. Certain markers were found to be dysregulated across various sample types. Afamin and gelsolin overlapped in studies of blood and CSF and sphingomyelin and ceramide overlapped in studies of CSF and brain tissue. The association of these markers with neurocognitive functioning may indicate the activity of certain pathways, potentially those related to the underlying neuropathophysiology of HAND.
Collapse
Affiliation(s)
- Monray E Williams
- Human Metabolomics, North-West University, Potchefstroom, South Africa
| | - Petrus J W Naudé
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.,Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
|
36
|
Zeng HM, Han HB, Zhang QF, Bai H. Application of modern neuroimaging technology in the diagnosis and study of Alzheimer's disease. Neural Regen Res 2021; 16:73-79. [PMID: 32788450 PMCID: PMC7818875 DOI: 10.4103/1673-5374.286957] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/14/2020] [Accepted: 03/28/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological abnormalities identified via neuroimaging are common in patients with Alzheimer's disease. However, it is not yet possible to easily detect these abnormalities using head computed tomography in the early stages of the disease. In this review, we evaluated the ways in which modern imaging techniques such as positron emission computed tomography, single photon emission tomography, magnetic resonance spectrum imaging, structural magnetic resonance imaging, magnetic resonance diffusion tensor imaging, magnetic resonance perfusion weighted imaging, magnetic resonance sensitive weighted imaging, and functional magnetic resonance imaging have revealed specific changes not only in brain structure, but also in brain function in Alzheimer's disease patients. The reviewed literature indicated that decreased fluorodeoxyglucose metabolism in the temporal and parietal lobes of Alzheimer's disease patients is frequently observed via positron emission computed tomography. Furthermore, patients with Alzheimer's disease often show a decreased N-acetylaspartic acid/creatine ratio and an increased myoinositol/creatine ratio revealed via magnetic resonance imaging. Atrophy of the entorhinal cortex, hippocampus, and posterior cingulate gyrus can be detected early using structural magnetic resonance imaging. Magnetic resonance sensitive weighted imaging can show small bleeds and abnormal iron metabolism. Task-related functional magnetic resonance imaging can display brain function activity through cerebral blood oxygenation. Resting functional magnetic resonance imaging can display the functional connection between brain neural networks. These are helpful for the differential diagnosis and experimental study of Alzheimer's disease, and are valuable for exploring the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Hong-Mei Zeng
- Department of Neurology, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua-Bo Han
- Department of Radiology, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
| | - Qi-Fang Zhang
- Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, and Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua Bai
- Department of Neurology, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
- Medical Experiment Center, Third Affiliated Hospital of Guizhou Medical University, Duyun, Guizhou Province, China
| |
Collapse
|
37
|
Chen P, Shen Z, Wang Q, Zhang B, Zhuang Z, Lin J, Shen Y, Chen Y, Dai Z, Wu R. Reduced Cerebral Glucose Uptake in an Alzheimer's Rat Model With Glucose-Weighted Chemical Exchange Saturation Transfer Imaging. Front Aging Neurosci 2021; 13:618690. [PMID: 33815088 PMCID: PMC8010663 DOI: 10.3389/fnagi.2021.618690] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 02/24/2021] [Indexed: 02/05/2023] Open
Abstract
A correlation between the abnormal cerebral glucose metabolism and the progression of Alzheimer's disease (AD) has been found in previous studies, suggesting that glucose alterations may be used to predict the histopathological diagnosis in AD. In this study, we investigated the dynamic changes of cerebral glucose uptake in vivo using MR glucose chemical exchange saturation transfer (glucoCEST) imaging in a rat model of AD with an intracerebroventricular (i.c.v) injection of amyloid Aβ-protein (25-35), confirmed by Morris water maze and Nissl staining. In total, 6 rats in the AD group and 6 rats in the control group that were given an injection of sterile normal saline were included. At 28 days after injection, all rats performed a 7.0 T MR exanimation, including glucoCEST, diffusion tensor imaging (DTI) and hippocampus magnetic resonance spectra (MRS), to detect the possible metabolic and structural changes in the rat brain. A significantly elevated brain glucoCEST signal in the brain of AD rats was observed, and a decreased brain glucose uptake was also explored during the progression of glucose infusion compared with those in rats of the control group. In addition, there is a significant positive correlation between glucoCEST enhancement (GCE) and myo-Inosito (Ins) in the AD group and the control group (P < 0.05). A significantly reduced number of neurons in the cortex and hippocampus in AD rats combined with the significantly longer escape and a decreased number of crossings were verified at 28 days after Aβ25-35 injection by Nissl staining and Morris water maze, respectively. Our results indicated that an abnormal brain glucose mechanism in AD rats could be detected by glucoCEST imaging, suggesting a new method to explore the occurrence and progress of diabetes-related AD or dementia.
Collapse
Affiliation(s)
- Peidong Chen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhiwei Shen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- Philips Healthcare, Beijing, China
| | - Qianqian Wang
- Department of Postgraduate, Shantou University Medical College, Shantou, China
| | - Bingna Zhang
- Center for Translational Medicine, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zerui Zhuang
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Jiefen Lin
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yuanyu Shen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yanzhi Chen
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Zhuozhi Dai
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Renhua Wu
- Department of Medical Imaging, The Second Affiliated Hospital, Shantou University Medical College, Shantou, China
- *Correspondence: Renhua Wu,
| |
Collapse
|
38
|
Longitudinal GluCEST MRI Changes and Cerebral Blood Flow in 5xFAD Mice. CONTRAST MEDIA & MOLECULAR IMAGING 2020; 2020:8831936. [PMID: 33304204 PMCID: PMC7714610 DOI: 10.1155/2020/8831936] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 11/17/2022]
Abstract
Many of the focal neurological symptoms associated with Alzheimer's disease (AD) are due to synaptic loss. Glutamate chemical exchange saturation transfer (GluCEST) magnetic resonance imaging (MRI) is a candidate method to assess synaptic dysfunction. We assessed chronological changes in GluCEST in a 5xFAD mouse model of AD, comparing Glucest effects and regional cerebral blood flow (CBF). GluCEST effects and CBF in 5xFAD mice aged 1–15 months and their littermates (WT) were measured. Neurite orientation dispersion and density imaging (NODDI) MRI reflecting dendritic/axonal density was also measured and compared with GluCEST in 7-month-old mice. While regional CBF's decrease began at 7 months, GluCEST-reduction effects preceded hypoperfusion of the temporal cortex and hippocampus. While longitudinal 5xFAD mouse measurements revealed a correlation between the regional GluCEST effects and CBF, a generalized linear mixed model revealed statistically different correlations in cortical and basal brain regions. Further, NODDI-derived neurite density correlated with GluCEST effects in the parietal cortex, but not in the hippocampus, thereby revealing regional differences in pathophysiological mechanisms. Finally, GluCEST's effects correlated with regional synaptophysin. These results demonstrate that GluCEST can reflect subtle synaptic changes and may be a potential imaging method for AD diagnosis as well as serve as a biomarker of AD progression.
Collapse
|
39
|
Early neurotransmission impairment in non-invasive Alzheimer Disease detection. Sci Rep 2020; 10:16396. [PMID: 33009473 PMCID: PMC7532202 DOI: 10.1038/s41598-020-73362-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer Disease (AD) is a pathology suffered by millions of people worldwide and it has a great social and economic impact. Previous studies reported a relationship between alterations in different amino acids and derivatives involved in neurotransmission systems and cognitive impairment. Therefore, in this study the neurotransmission impairment associated to early AD has been evaluated. For this purpose, different amino acids and derivatives were determined in saliva samples from AD patients and healthy subjects, by means of an analytical method based on chromatography coupled to tandem mass spectrometry. Results showed statistically significant differences in salivary levels for the compounds myo-inositol, creatine and acetylcholine; and other compounds (myo-inositol, glutamine, creatine, acetylcholine) showed significant correlations with some cognitive tests scores. Therefore, these compounds were included in a multivariate analysis and the corresponding diagnosis model showed promising indices (AUC 0.806, sensitivity 61%, specificity 92%). In conclusion, some amino acids and derivatives involved in neurotransmission impairment could be potential biomarkers in early and non-invasive AD detection.
Collapse
|
40
|
Hingerl L, Strasser B, Moser P, Hangel G, Motyka S, Heckova E, Gruber S, Trattnig S, Bogner W. Clinical High-Resolution 3D-MR Spectroscopic Imaging of the Human Brain at 7 T. Invest Radiol 2020; 55:239-248. [PMID: 31855587 DOI: 10.1097/rli.0000000000000626] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Available clinical magnetic resonance spectroscopic imaging (MRSI) sequences are hampered by long scan times, low spatial resolution, strong field inhomogeneities, limited volume coverage, and low signal-to-noise ratio. High-resolution, whole-brain mapping of more metabolites than just N-acetylaspartate, choline, and creatine within clinically attractive scan times is urgently needed for clinical applications. The aim is therefore to develop a free induction decay (FID) MRSI sequence with rapid concentric ring trajectory (CRT) encoding for 7 T and demonstrate its clinical feasibility for mapping the whole cerebrum of healthy volunteers and patients. MATERIALS AND METHODS Institutional review board approval and written informed consent were obtained. Time-efficient, 3-dimensional encoding of an ellipsoidal k-space by in-plane CRT and through-plane phase encoding was integrated into an FID-MRSI sequence. To reduce scan times further, repetition times were shortened, and variable temporal interleaves were applied. Measurements with different matrix sizes were performed to validate the CRT encoding in a resolution phantom. One multiple sclerosis patient, 1 glioma patient, and 6 healthy volunteers were prospectively measured. For the healthy volunteers, brain segmentation was performed to quantify median metabolic ratios, Cramér-Rao lower bounds (CRLBs), signal-to-noise ratios, linewidths, and brain coverage among all measured matrix sizes ranging from a 32 × 32 × 31 matrix with 6.9 × 6.9 × 4.2 mm nominal voxel size acquired in ~3 minutes to an 80 × 80 × 47 matrix with 2.7 × 2.7 × 2.7 mm nominal voxel size in ~15 minutes for different brain regions. RESULTS Phantom structures with diameters down to 3 to 4 mm were visible. In vivo MRSI provided high spectral quality (median signal-to-noise ratios, >6.3 and linewidths, <0.082 ppm) and fitting quality. Cramér-Rao lower bounds were ranging from less than 22% for glutamine (highest CRLB in subcortical gray matter) to less than 9.5% for N-acetylaspartate for the 80 × 80 × 47 matrix (highest CRLB in the temporal lobe). This enabled reliable mapping of up to 8 metabolites (N-acetylaspartate, N-acetylaspartyl glutamate, total creatine, glutamine, glutamate, total choline, myo-inositol, glycine) and macromolecules for all resolutions. Coverage of the whole cerebrum allowed visualization of the full extent of diffuse and local multiple sclerosis-related neurochemical changes (eg, up to 100% increased myo-inositol). Three-dimensional brain tumor metabolic maps provided valuable information beyond that of single-slice MRSI, with up to 200% higher choline, up to 100% increased glutamine, and increased glycine in tumor tissue. CONCLUSIONS Seven Tesla FID-MRSI with time-efficient CRT readouts offers clinically attractive acquisition protocols tailored either for speed or for the investigation of small pathologic details and low-abundant metabolites. This can complement clinical MR studies of various brain disorders. Significant metabolic anomalies were demonstrated in a multiple sclerosis and a glioma patient for myo-inositol, glutamine, total choline, glycine, and N-acetylaspartate concentrations.
Collapse
Affiliation(s)
- Lukas Hingerl
- From the High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Bernhard Strasser
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Philipp Moser
- From the High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gilbert Hangel
- From the High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Stanislav Motyka
- From the High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Eva Heckova
- From the High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Stephan Gruber
- From the High Field MR Centre, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
41
|
De Paepe B, Merckx C, Jarošová J, Cannizzaro M, De Bleecker JL. Myo-Inositol Transporter SLC5A3 Associates with Degenerative Changes and Inflammation in Sporadic Inclusion Body Myositis. Biomolecules 2020; 10:biom10040521. [PMID: 32235474 PMCID: PMC7226596 DOI: 10.3390/biom10040521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/20/2020] [Accepted: 03/26/2020] [Indexed: 12/15/2022] Open
Abstract
Myo-inositol exerts many cellular functions, which include osmo-protection, membrane functioning, and secondary messaging. Its Na+/myo-inositol co-transporter SLC5A3 is expressed in muscle tissue and further accumulates in myositis. In this study we focused on the peculiar subgroup of sporadic inclusion body myositis (IBM), in which auto-inflammatory responses and degenerative changes co-exist. A cohort of nine patients was selected with clinically confirmed IBM, in which SLC5A3 protein was immune-localized to the different tissue constituents using immunofluorescence, and expression levels were evaluated using Western blotting. In normal muscle tissue, SLC5A3 expression was restricted to blood vessels and occasional low levels on muscle fiber membranes. In IBM tissues, SLC5A3 staining was markedly increased, with discontinuous staining of the muscle fiber membranes, and accumulation of SLC5A3 near inclusions and on the rims of vacuoles. A subset of muscle-infiltrating auto-aggressive immune cells was SLC5A3 positive, of which most were T-cells and M1 lineage macrophages. We conclude that SLC5A3 is overexpressed in IBM muscle, where it associates with protein aggregation and inflammatory infiltration. Based on our results, functional studies could be initiated to explore the possibilities of therapeutic osmolyte pathway intervention for preventing protein aggregation in muscle cells.
Collapse
|
42
|
Vawter MP, Hamzeh AR, Muradyan E, Civelli O, Abbott GW, Alachkar A. Association of Myoinositol Transporters with Schizophrenia and Bipolar Disorder: Evidence from Human and Animal Studies. MOLECULAR NEUROPSYCHIATRY 2019; 5:200-211. [PMID: 31768373 PMCID: PMC6873027 DOI: 10.1159/000501125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
Evidence from animal and human studies has linked myo-inositol (MI) with the pathophysiology and/or treatment of psychiatric disorders such as schizophrenia and bipolar disorder. However, there is still controversy surrounding the definitive role of MI in these disorders. Given that brain MI is differentially regulated by three transporters - SMIT1, SMIT2 and/or HMIT (encoded by the genes: SLC5A3, SLC5A11, and SLC2A13, respectively) - we used available datasets to describe the distribution in mouse and human brain of the different MI transporters and to examine changes in mRNA expression of these transporters in patients with schizophrenia and bipolar disorder. We found a differential distribution of the mRNA of each of the three MI transporters in both human and mouse brain regions. Interestingly, while individual neurons express SMIT1 and HMIT, non-neuronal cells express SMIT2, thus partially accounting for different uptake levels of MI and concordance to downstream second messenger signaling pathways. We also found that the expression of MI transporters is significantly changed in schizophrenia and bipolar disorder in a diagnostic-, brain region- and subtype-specific manner. We then examined the effects of germline deletion in mice of Slc5a3 on behavioral phenotypes related to schizophrenia and bipolar disorder. This gene deletion produces behavioral deficits that mirror some specific symptoms of schizophrenia and bipolar disorder. Finally, chronic administration of MI was able to reverse particular, but not all, behavioral deficits in Slc5a3 knockout mice; MI itself induced some behavioral deficits. Our data support a strong correlation between the expression of MI transporters and schizophrenia and bipolar disorder, and suggest that brain region-specific aberration of one or more of these transporters determines the partial behavioral phenotypes and/or symptomatic pattern of these disorders.
Collapse
Affiliation(s)
- Marquis P. Vawter
- Department of Psychiatry and Human Behavior, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Abdul Rezzak Hamzeh
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Edgar Muradyan
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Olivier Civelli
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, California, USA
- Department of Pharmaceutical Sciences, School of Medicine, University of California, Irvine, Irvine, California, USA
- Department of Developmental and Cell Biology, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Amal Alachkar
- Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, California, USA
- Department of Pharmaceutical Sciences, School of Medicine, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
43
|
Ma Y, Jun GR, Zhang X, Chung J, Naj AC, Chen Y, Bellenguez C, Hamilton-Nelson K, Martin ER, Kunkle BW, Bis JC, Debette S, DeStefano AL, Fornage M, Nicolas G, van Duijn C, Bennett DA, De Jager PL, Mayeux R, Haines JL, Pericak-Vance MA, Seshadri S, Lambert JC, Schellenberg GD, Lunetta KL, Farrer LA. Analysis of Whole-Exome Sequencing Data for Alzheimer Disease Stratified by APOE Genotype. JAMA Neurol 2019; 76:1099-1108. [PMID: 31180460 PMCID: PMC6563544 DOI: 10.1001/jamaneurol.2019.1456] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 03/22/2019] [Indexed: 12/13/2022]
Abstract
IMPORTANCE Previous genome-wide association studies of common variants identified associations for Alzheimer disease (AD) loci evident only among individuals with particular APOE alleles. OBJECTIVE To identify APOE genotype-dependent associations with infrequent and rare variants using whole-exome sequencing. DESIGN, SETTING, AND PARTICIPANTS The discovery stage included 10 441 non-Hispanic white participants in the Alzheimer Disease Sequencing Project. Replication was sought in 2 independent, whole-exome sequencing data sets (1766 patients with AD, 2906 without AD [controls]) and a chip-based genotype imputation data set (8728 patients with AD, 9808 controls). Bioinformatics and functional analyses were conducted using clinical, cognitive, neuropathologic, whole-exome sequencing, and gene expression data obtained from a longitudinal cohort sample including 402 patients with AD and 647 controls. Data were analyzed between March 2017 and September 2018. MAIN OUTCOMES AND MEASURES Score, Firth, and sequence kernel association tests were used to test the association of AD risk with individual variants and genes in subgroups of APOE ε4 carriers and noncarriers. Results with P ≤ 1 × 10-5 were further evaluated in the replication data sets and combined by meta-analysis. RESULTS Among 3145 patients with AD and 4213 controls lacking ε4 (mean [SD] age, 83.4 [7.6] years; 4363 [59.3.%] women), novel genome-wide significant associations were obtained in the discovery sample with rs536940594 in AC099552 (odds ratio [OR], 88.0; 95% CI, 9.08-852.0; P = 2.22 × 10-7) and rs138412600 in GPAA1 (OR, 1.78; 95% CI, 1.44-2.2; meta-P = 7.81 × 10-8). GPAA1 was also associated with expression in the brain of GPAA1 (β = -0.08; P = .03) and its repressive transcription factor, FOXG1 (β = 0.13; P = .003), and global cognition function (β = -0.53; P = .009). Significant gene-wide associations (threshold P ≤ 6.35 × 10-7) were observed for OR8G5 (P = 4.67 × 10-7), IGHV3-7 (P = 9.75 × 10-16), and SLC24A3 (P = 2.67 × 10-12) in 2377 patients with AD and 706 controls with ε4 (mean [SD] age, 75.2 [9.6] years; 1668 [54.1%] women). CONCLUSIONS AND RELEVANCE The study identified multiple possible novel associations for AD with individual and aggregated rare variants in groups of individuals with and without APOE ε4 alleles that reinforce known and suggest additional pathways leading to AD.
Collapse
Affiliation(s)
- Yiyi Ma
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Center for Translational & Computational Neuroimmunology, Multiple Sclerosis Clinical Care and Research Center, Division of Neuroimmunology, Columbia University Medical Center, New York, New York
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Gyungah R. Jun
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Jaeyoon Chung
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Adam C. Naj
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Yuning Chen
- Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Celine Bellenguez
- Universite de Lille, INSERM UMR1167, Institute Pasteur de Lille, Lille, France
| | - Kara Hamilton-Nelson
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida
| | - Eden R. Martin
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida
| | - Brian W. Kunkle
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle
| | - Stéphanie Debette
- Bordeaux Population Health Research Center, UMR1219, University Bordeaux, Inserm, Bordeaux, France
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
| | - Anita L. DeStefano
- Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Myriam Fornage
- School of Public Health, University of Texas Health Science Center at Houston, Houston
| | - Gaël Nicolas
- UNIROUEN, Inserm U1245, Normandie University, Rouen, France
- Department of Genetics, Rouen University Hospital, Rouen, France
- Normandy Centre for Genomic and Personalized Medicine, Centre National de Référence pour les Malades Alzheimer Jeunes, Rouen, France
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Multiple Sclerosis Clinical Care and Research Center, Division of Neuroimmunology, Columbia University Medical Center, New York, New York
- Department of Neurology, Columbia University Medical Center, New York, New York
- Program in Translational NeuroPsychiatric Genomics, Institute for the Neurosciences, Departments of Neurology and Psychiatry, Brigham and Women's Hospital, Boston, Massachusetts
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts
| | - Richard Mayeux
- Department of Neurology, Columbia University Medical Center, New York, New York
| | - Jonathan L Haines
- Institute for Computational Biology, Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Margaret A. Pericak-Vance
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida
| | - Sudha Seshadri
- Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio
| | | | | | - Kathryn L. Lunetta
- Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics), Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Department of Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Department of Neurology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Department of Ophthalmology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
- Department of Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts
| |
Collapse
|
44
|
Chen Q, Boeve BF, Tosakulwong N, Lesnick T, Brushaber D, Dheel C, Fields J, Forsberg L, Gavrilova R, Gearhart D, Haley D, Gunter JL, Graff‐Radford J, Jones D, Knopman D, Graff‐Radford N, Kraft R, Lapid M, Rademakers R, Wszolek ZK, Rosen H, Boxer AL, Kantarci K. Brain MR Spectroscopy Changes Precede Frontotemporal Lobar Degeneration Phenoconversion in Mapt Mutation Carriers. J Neuroimaging 2019; 29:624-629. [PMID: 31173437 PMCID: PMC6731148 DOI: 10.1111/jon.12642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/16/2019] [Accepted: 05/22/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE The objective of this study was to longitudinally investigate the trajectory of change in 1 H MRS measurements in asymptomatic MAPT mutation carriers who became symptomatic during follow-up, and to determine the time at which the neurochemical alterations accelerated during disease progression. METHODS We identified eight MAPT mutations carriers who transitioned from asymptomatic to symptomatic disease during follow-up. All participants were longitudinally followed with an average of 7.75 years (range 4-11 years) and underwent two or more single voxel 1 H MRS examinations from the posterior cingulate voxel, with a total of 60 examinations. The rate of longitudinal change for each metabolite was estimated using linear mixed models. A flex point model was used to estimate the flex time point of the change in slope. RESULTS The decrease in the NAA/mI ratio accelerated 2.09 years prior to symptom onset, and continued to decline. A similar trajectory was observed in the presumed glial marker mI/Cr ratio accelerating 1.86 years prior to symptom onset. CONCLUSIONS Our findings support the potential use of longitudinal 1 H MRS for monitoring the neurodegenerative progression in MAPT mutation carriers starting from the asymptomatic stage.
Collapse
Affiliation(s)
- Qin Chen
- Department of RadiologyMayo Clinic
- Department of NeurologyMayo Clinic
| | - Bradley F. Boeve
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | | | | | - Danielle Brushaber
- Department of Psychology and PsychiatryMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | - Christina Dheel
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | - Julie Fields
- Department of Clinical Genomic and NeurologyMayo Clinic
| | - Leah Forsberg
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | | | - Debra Gearhart
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | - Dana Haley
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida
| | | | - Jonathan Graff‐Radford
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | | | - David Knopman
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | | | - Ruth Kraft
- Department of Health Sciences ResearchMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| | - Maria Lapid
- Department of Clinical Genomic and NeurologyMayo Clinic
| | - Rosa Rademakers
- Research ServicesMayo ClinicRochesterMinnesota
- Memory and Aging CenterUniversity of California San FranciscoSan Francisco
| | | | - Howie Rosen
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Adam L. Boxer
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
| | - Kejal Kantarci
- Department of RadiologyMayo Clinic
- Research ServicesMayo ClinicRochesterMinnesota
| |
Collapse
|
45
|
Hoffman JD, Yanckello LM, Chlipala G, Hammond TC, McCulloch SD, Parikh I, Sun S, Morganti JM, Green SJ, Lin AL. Dietary inulin alters the gut microbiome, enhances systemic metabolism and reduces neuroinflammation in an APOE4 mouse model. PLoS One 2019; 14:e0221828. [PMID: 31461505 PMCID: PMC6713395 DOI: 10.1371/journal.pone.0221828] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/15/2019] [Indexed: 12/22/2022] Open
Abstract
The apolipoprotein ε4 allele (APOE4) is the strongest genetic risk factor for Alzheimer's disease (AD). APOE4 carriers develop systemic metabolic dysfunction decades before showing AD symptoms. Accumulating evidence shows that the metabolic dysfunction accelerates AD development, including exacerbated amyloid-beta (Aβ) retention, neuroinflammation and cognitive decline. Therefore, preserving metabolic function early on may be critical to reducing the risk for AD. Here, we show that inulin increases beneficial microbiota and decreases harmful microbiota in the feces of young, asymptomatic APOE4 transgenic (E4FAD) mice and enhances metabolism in the cecum, periphery and brain, as demonstrated by increases in the levels of SCFAs, tryptophan-derived metabolites, bile acids, glycolytic metabolites and scyllo-inositol. We show that inulin also reduces inflammatory gene expression in the hippocampus. This knowledge can be utilized to design early precision nutrition intervention strategies that use a prebiotic diet to enhance systemic metabolism and may be useful for reducing AD risk in asymptomatic APOE4 carriers.
Collapse
Affiliation(s)
- Jared D. Hoffman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - Lucille M. Yanckello
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - George Chlipala
- Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Tyler C. Hammond
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, United States of America
| | | | - Ishita Parikh
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, United States of America
| | - Sydney Sun
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Josh M. Morganti
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, United States of America
| | - Stefan J. Green
- Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ai-Ling Lin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Neuroscience, University of Kentucky, Lexington, Kentucky, United States of America
- F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
46
|
Motyka S, Moser P, Hingerl L, Hangel G, Heckova E, Strasser B, Eckstein K, Daniel Robinson S, Poser BA, Gruber S, Trattnig S, Bogner W. The influence of spatial resolution on the spectral quality and quantification accuracy of whole-brain MRSI at 1.5T, 3T, 7T, and 9.4T. Magn Reson Med 2019; 82:551-565. [PMID: 30932248 PMCID: PMC6563461 DOI: 10.1002/mrm.27746] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Inhomogeneities in the static magnetic field (B0 ) deteriorate MRSI data quality by lowering the spectral resolution and SNR. MRSI with low spatial resolution is also prone to lipid bleeding. These problems are increasingly problematic at ultra-high fields. An approach to tackling these challenges independent of B0 -shim hardware is to increase the spatial resolution. Therefore, we investigated the effect of improved spatial resolution on spectral quality and quantification at 4 field strengths. METHODS Whole-brain MRSI data was simulated for 3 spatial resolutions and 4 B0 s based on experimentally acquired MRI data and simulated free induction decay signals of metabolites and lipids. To compare the spectral quality and quantification, we derived SNR normalized to the voxel size (nSNR), linewidth and metabolite concentration ratios, their Cramer-Rao-lower-bounds (CRLBs), and the absolute percentage error (APE) of estimated concentrations compared to the gold standard for the whole-brain and 8 brain regions. RESULTS At 7T, we found up to a 3.4-fold improved nSNR (in the frontal lobe) and a 2.8-fold reduced linewidth (in the temporal lobe) for 1 cm3 versus 0.25 cm3 resolution. This effect was much more pronounced at higher and less homogenous B0 (1.6-fold improved nSNR and 1.8-fold improved linewidth in the parietal lobe at 3T). This had direct implications for quantification: the volume of reliably quantified spectra increased with resolution by 1.2-fold and 1.5-fold (when thresholded by CRLBs or APE, respectively). CONCLUSION MRSI data quality benefits from increased spatial resolution particularly at higher B0 , and leads to more reliable metabolite quantification. In conjunction with the development of better B0 shimming hardware, this will enable robust whole-brain MRSI at ultra-high field.
Collapse
Affiliation(s)
- Stanislav Motyka
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Philipp Moser
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Hingerl
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gilbert Hangel
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Eva Heckova
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Bernhard Strasser
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Radiology, Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Korbinian Eckstein
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Simon Daniel Robinson
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Benedikt A Poser
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands.,Maastricht Brain Imaging Centre, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Stephan Gruber
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Siegfried Trattnig
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Clinical Molecular MR Imaging, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bogner
- High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Ali JI, Smart CM, Gawryluk JR. Subjective Cognitive Decline and APOE ɛ4: A Systematic Review. J Alzheimers Dis 2019; 65:303-320. [PMID: 30040718 DOI: 10.3233/jad-180248] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Individuals with subjective cognitive decline (SCD) report self-perceived declines in cognitive function but perform within normal limits on standardized tests. However, for some, these self-perceived changes may herald eventual decline to Alzheimer's disease (AD). In light of this, the relationship between SCD and APOE ɛ4, a known genetic risk factor for AD, has garnered interest; however, no systematic review of this literature exists. The current review (n = 36 articles) examined the prevalence of APOE ɛ4 in SCD samples relative to healthy and objectively impaired samples, and summarized APOE ɛ4-related risk of conversion from SCD to AD. Univariate ANOVA indicated that APOE ɛ4 frequency was comparable between healthy control and SCD samples, yet significantly higher in objectively impaired samples (i.e., MCI, AD) relative to either of these groups. Narrative review provided mixed evidence linking coincident APOE ɛ4-positive genotype and SCD to structural neuropathology. Though there was little evidence to suggest that APOE ɛ4 predisposes individuals to developing SCD, both APOE ɛ4 and SCD were found to confer individual and multiplicative risk of conversion to objective cognitive impairment. Combined, it is likely that a relationship between APOE ɛ4, SCD, and AD exists, though its exact nature remains undetermined. A clearer understanding of these relationships is hindered by a lack of standardization in SCD classification and a dearth of longitudinal outcome research. Wide-scale adoption of genetic screening for dementia risk in persons with SCD is considered premature at this time. Ethical considerations and clinical implications of genetic testing for dementia risk are discussed.
Collapse
Affiliation(s)
- Jordan I Ali
- Department of Psychology, University of Victoria, Victoria, BC, Canada.,Institute on Aging & Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Colette M Smart
- Department of Psychology, University of Victoria, Victoria, BC, Canada.,Institute on Aging & Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Jodie R Gawryluk
- Department of Psychology, University of Victoria, Victoria, BC, Canada.,Institute on Aging & Lifelong Health, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
48
|
Chen Q, Boeve BF, Tosakulwong N, Lesnick T, Brushaber D, Dheel C, Fields J, Forsberg L, Gavrilova R, Gearhart D, Haley D, Gunter JL, Graff-Radford J, Jones D, Knopman D, Graff-Radford N, Kraft R, Lapid M, Rademakers R, Syrjanen J, Wszolek ZK, Rosen H, Boxer AL, Kantarci K. Frontal lobe 1H MR spectroscopy in asymptomatic and symptomatic MAPT mutation carriers. Neurology 2019; 93:e758-e765. [PMID: 31315971 DOI: 10.1212/wnl.0000000000007961] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 03/26/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To determine the frontal lobe proton magnetic resonance spectroscopy (1H MRS) abnormalities in asymptomatic and symptomatic carriers of microtubule-associated protein tau (MAPT) mutations. METHODS We recruited patients with MAPT mutations from 5 individual families, who underwent single voxel 1H MRS from the medial frontal lobe at 3T (n = 19) from the Longitudinal Evaluation of Familial Frontotemporal Dementia Subjects (LEFFTDS) Study at the Mayo Clinic site. Asymptomatic MAPT mutation carriers (n = 9) had Frontotemporal Lobar Degeneration Clinical Dementia Rating Sum of Boxes (FTLD-CDR SOB) score of zero, and symptomatic MAPT mutation carriers (n = 10) had a median FTLD-CDR SOB score of 5. Noncarriers from healthy first-degree relatives of the patients were recruited as controls (n = 25). The demographic aspects and 1H MRS metabolite ratios were compared by use of the Fisher exact test for sex and linear mixed models to account for within-family correlations. We used Tukey contrasts for pair-wise comparisons. RESULTS Asymptomatic MAPT mutation carriers had lower neuronal marker N-acetylaspartate (NAA)/creatine (Cr) (p = 0.001) and lower NAA/myo-inositol (mI) (p = 0.026) than noncarriers after adjustment for age. Symptomatic MAPT mutation carriers had lower NAA/Cr (p = 0.01) and NAA/mI (p = 0.01) and higher mI/Cr (p = 0.02) compared to noncarriers after adjustment for age. Furthermore, NAA/Cr (p = 0.006) and NAA/mI (p < 0.001) ratios decreased, accompanied by an increase in mI/Cr ratio (p = 0.001), as the ages of carriers approached and passed the age at symptom onset. CONCLUSION Frontal lobe neurochemical alterations measured with 1H MRS precede the symptom onset in MAPT mutation carriers. Frontal lobe 1H MRS is a potential biomarker for early neurodegenerative processes in MAPT mutation carriers.
Collapse
Affiliation(s)
- Qin Chen
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Bradley F Boeve
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Nirubol Tosakulwong
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Timothy Lesnick
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Danielle Brushaber
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Christina Dheel
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Julie Fields
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Leah Forsberg
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Ralitza Gavrilova
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Debra Gearhart
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Dana Haley
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Jeffrey L Gunter
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Jonathan Graff-Radford
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - David Jones
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - David Knopman
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Neill Graff-Radford
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Ruth Kraft
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Maria Lapid
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Rosa Rademakers
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Jeremy Syrjanen
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Zbigniew K Wszolek
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Howie Rosen
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Adam L Boxer
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Kejal Kantarci
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco.
| |
Collapse
|
49
|
Kaufman MJ, Kanayama G, Hudson JI, Pope HG. Supraphysiologic-dose anabolic-androgenic steroid use: A risk factor for dementia? Neurosci Biobehav Rev 2019; 100:180-207. [PMID: 30817935 PMCID: PMC6451684 DOI: 10.1016/j.neubiorev.2019.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 02/06/2023]
Abstract
Supraphysiologic-dose anabolic-androgenic steroid (AAS) use is associated with physiologic, cognitive, and brain abnormalities similar to those found in people at risk for developing Alzheimer's Disease and its related dementias (AD/ADRD), which are associated with high brain β-amyloid (Aβ) and hyperphosphorylated tau (tau-P) protein levels. Supraphysiologic-dose AAS induces androgen abnormalities and excess oxidative stress, which have been linked to increased and decreased expression or activity of proteins that synthesize and eliminate, respectively, Aβ and tau-P. Aβ and tau-P accumulation may begin soon after initiating supraphysiologic-dose AAS use, which typically occurs in the early 20s, and their accumulation may be accelerated by other psychoactive substance use, which is common among non-medical AAS users. Accordingly, the widespread use of supraphysiologic-dose AAS may increase the numbers of people who develop dementia. Early diagnosis and correction of sex-steroid level abnormalities and excess oxidative stress could attenuate risk for developing AD/ADRD in supraphysiologic-dose AAS users, in people with other substance use disorders, and in people with low sex-steroid levels or excess oxidative stress associated with aging.
Collapse
Affiliation(s)
- Marc J Kaufman
- McLean Imaging Center, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| | - Gen Kanayama
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - James I Hudson
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Harrison G Pope
- Biological Psychiatry Laboratory, McLean Hospital, 115 Mill St., Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
50
|
Motor cortex metabolite alterations in amyotrophic lateral sclerosis assessed in vivo using edited and non-edited magnetic resonance spectroscopy. Brain Res 2019; 1718:22-31. [PMID: 31002818 DOI: 10.1016/j.brainres.2019.04.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/09/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022]
Abstract
Previous MRI and proton spectroscopy (1H-MRS) studies have revealed impaired neuronal integrity and altered neurometabolite concentrations in the motor cortex of patients with amyotrophic lateral sclerosis (ALS). Here, we aim to use MRI with conventional and novel MRS sequences to further investigate neurometabolic changes in the motor cortex of ALS patients and their relation to clinical parameters. We utilized the novel HERMES (Hadamard Encoding and Reconstruction of MEGA-Edited Spectroscopy) MRS sequence to simultaneously quantify the inhibitory neurotransmitter GABA and antioxidant glutathione in ALS patients (n = 7) and healthy controls (n = 7). In addition, we have also quantified other MRS observable neurometabolites using a conventional point-resolved MR spectroscopy (PRESS) sequence in ALS patients (n = 20) and healthy controls (n = 20). We observed a trend towards decreasing glutathione concentrations in the motor cortex of ALS patients (p = 0.0842). In addition, we detected a 11% decrease in N-acetylaspartate (NAA) (p = 0.025), a 15% increase in glutamate + glutamine (Glx) (p = 0.0084) and a 21% increase in myo-inositol (mIns) (p = 0.0051) concentrations for ALS patients compared to healthy controls. Furthermore, significant positive correlations were found between GABA-NAA (p = 0.0480; Rρ = 0.7875) and NAA-mIns (p = 0.0448; Rρ = -0.4651) levels among the patients. NAA levels in the bulbar-onset patient group were found to be significantly (p = 0.0097) lower compared to the limb-onset group. A strong correlation (p < 0.0001; Rρ = -0,8801) for mIns and a weak correlation (p = 0.0066; Rρ = -0,6673) for Glx was found for the disease progression, measured by declining of the ALS Functional Rating Scale-Revised criteria (ALSFRS-R). Concentrations of mIns and Glx also correlated with disease severity measured by forced vital capacity (FVC). Results suggest that mean neurometabolite concentrations detected in the motor cortex may indicate clinical and pathological changes in ALS.
Collapse
|