51
|
Frantellizzi V, Pani A, Ricci M, Locuratolo N, Fattapposta F, De Vincentis G. Neuroimaging in Vascular Cognitive Impairment and Dementia: A Systematic Review. J Alzheimers Dis 2021; 73:1279-1294. [PMID: 31929166 DOI: 10.3233/jad-191046] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cerebrovascular diseases are well established causes of cognitive impairment. Different etiologic entities, such as vascular dementia (VaD), vascular cognitive impairment, subcortical (ischemic) VaD, and vascular cognitive disorder, are included in the umbrella definition of vascular cognitive impairment and dementia (VCID). Because of the variability of VCID clinical presentation, there is no agreement on criteria defining the neuropathological threshold of this disorder. In fact, VCID is characterized by cerebral hemodynamic alteration which ranges from decreased cerebral blood flow to small vessels disease and involves a multifactorial process that leads to demyelination and gliosis, including blood-brain barrier disruption, hypoxia, and hypoperfusion, oxidative stress, neuroinflammation and alteration on neurovascular unit coupling, cerebral microbleeds, or superficial siderosis. Numerous criteria for the definition of VaD have been described: the National Institute of Neurological Disorders and Stroke Association Internationale pour Recherche'-et-l'Enseignement en Neurosciences criteria, the State of California Alzheimer's Disease Diagnostic and Treatment Centers criteria, DSM-V criteria, the Diagnostic Criteria for Vascular Cognitive Disorders (a VASCOG Statement), and Vascular Impairment of Cognition Classification Consensus Study. Neuroimaging is fundamental for definition and diagnosis of VCID and should be used to assess the extent, location, and type of vascular lesions. MRI is the most sensible technique, especially if used according to standardized protocols, even if CT plays an important role in several conditions. Functional neuroimaging, in particular functional MRI and PET, may facilitate differential diagnosis among different forms of dementia. This systematic review aims to explore the state of the art and future perspective of non-invasive diagnostics of VCID.
Collapse
Affiliation(s)
| | - Arianna Pani
- Clinical Pharmacology and Toxicology, University of Milan "Statale", Italy
| | - Maria Ricci
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | | | | | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
52
|
Sarikaya I, Kamel WA, Ateyah KK, Essa NB, AlTailji S, Sarikaya A. Visual versus semiquantitative analysis of 18F- fluorodeoxyglucose-positron emission tomography brain images in patients with dementia. World J Nucl Med 2021; 20:82-89. [PMID: 33850493 PMCID: PMC8034786 DOI: 10.4103/wjnm.wjnm_53_18] [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: 05/15/2018] [Revised: 05/31/2018] [Accepted: 07/19/2018] [Indexed: 11/04/2022] Open
Abstract
Various studies have reported to the superiority of semiquantitative (SQ) analysis over visual analysis in detecting metabolic changes in the brain. In this study, we aimed to determine the limitations of SQ analysis programs and the current status of 18F- fluorodeoxyglucose (FDG)-positron emission tomography (PET) scan in dementia. 18F- FDG-PET/computed tomography (CT) brain images of 39 patients with a history of dementia were analyzed both visually and semiquantitatively. Using the visually markedly abnormal 18F- FDG-PET images as standard, we wanted to test the accuracy of two commercially available SQ analysis programs. SQ analysis results were classified as matching, partially matching and nonmatching with visually markedly abnormal studies. On visual analysis, 18F- FDG-PET showed marked regional hypometabolism in 19 patients, mild abnormalities in 8 and was normal in 12 patients. SQ analysis-1 results matched with visual analysis in 8 patients (42.1%) and partially matched in 11. SQ analysis-2 findings matched with visual analysis in 11 patients (57.8%) and partially matched in 7 and did not match in 1. Marked regional hypometabolism was either on the left side of the brain or was more significant on the left than the right in 63% of patients. Preservation of metabolism in sensorimotor cortex was seen in various dementia subtypes. Reviewing images in color scale and maximum intensity projection (MIP) image was helpful in demonstrating and displaying regional abnormalities, respectively. SQ analysis provides less accurate results as compared to visual analysis by experts. Due to suboptimal image registration and selection of brain areas, SQ analysis provides inaccurate results, particularly in small areas and areas in close proximity. Image registration and selection of areas with SQ programs should be checked carefully before reporting the results.
Collapse
Affiliation(s)
- Ismet Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Kuwait University, Kuwait University, Kuwait
| | - Walaa A Kamel
- Department of Neurology, Faculty of Medicine, Beni-Suef University, Egypt.,Department of Nuclear Medicine, Ibn Sina Hospital, Kuwait
| | | | - Nooraessa Bin Essa
- Department of Nuclear Medicine, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| | | | - Ali Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
53
|
Borja AJ, Hancin EC, Zhang V, Koa B, Bhattaru A, Rojulpote C, Detchou DK, Aly M, Kaghazchi F, Gerke O, Patil S, Gonuguntla K, Werner TJ, Revheim ME, Høilund-Carlsen PF, Alavi A. Global brain glucose uptake on 18F-FDG-PET/CT is influenced by chronic cardiovascular risk. Nucl Med Commun 2021; 42:444-450. [PMID: 33323870 DOI: 10.1097/mnm.0000000000001349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE The goal of this study was to assess global cerebral glucose uptake in subjects with known cardiovascular risk factors by employing a quantitative 18F-fluorodeoxyglucose-PET/computed tomography (FDG-PET/CT) technique. We hypothesized that at-risk subjects would demonstrate decreased global brain glucose uptake compared to healthy controls. METHODS We compared 35 healthy male controls and 14 male subjects at increased risk for cardiovascular disease (CVD) as assessed by the systematic coronary risk evaluation (SCORE) tool. All subjects were grouped into two age-matched cohorts: younger (<50 years) and older (≥50 years). The global standardized uptake value mean (Avg SUVmean) was measured by mapping regions of interest of the entire brain across the supratentorial structures and cerebellum. Wilcoxon's rank-sum test was used to assess the differences in Avg SUVmean between controls and at-risk subjects. RESULTS Younger subjects demonstrated higher brain Avg SUVmean than older subjects. In addition, in both age strata, the 10-year risk for fatal CVD according to the SCORE tool was significantly greater in the at-risk groups than in healthy controls (younger: P = 0.0304; older: P = 0.0436). In the younger cohort, at-risk subjects demonstrated significantly lower brain Avg SUVmean than healthy controls (P = 0.0355). In the older cohort, at-risk subjects similarly had lower Avg SUVmean than controls (P = 0.0343). CONCLUSIONS Global brain glucose uptake appears to be influenced by chronic cardiovascular risk factors. Therefore, FDG-PET/CT may play a role in determining the importance of CVD on brain function and has potential for monitoring the efficacy of various therapeutic interventions.
Collapse
Affiliation(s)
- Austin J Borja
- Department of Radiology, Hospital of the University of Pennsylvania
- Perelman School of Medicine, University of Pennsylvania
| | - Emily C Hancin
- Department of Radiology, Hospital of the University of Pennsylvania
- Lewis Katz School of Medicine, Temple University
| | - Vincent Zhang
- Department of Radiology, Hospital of the University of Pennsylvania
| | - Benjamin Koa
- Department of Radiology, Hospital of the University of Pennsylvania
- Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Abhijit Bhattaru
- Department of Radiology, Hospital of the University of Pennsylvania
| | | | - Donald K Detchou
- Department of Radiology, Hospital of the University of Pennsylvania
- Perelman School of Medicine, University of Pennsylvania
| | - Mahmoud Aly
- Department of Radiology, Hospital of the University of Pennsylvania
| | | | - Oke Gerke
- Department of Nuclear Medicine, Odense University Hospital
- Department of Clinical Research, Research Unit of Clinical Physiology and Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Shivaraj Patil
- Department of Radiology, Hospital of the University of Pennsylvania
- Department of Medicine, University of Connecticut, Hartford, Connecticut, USA
| | - Karthik Gonuguntla
- Department of Radiology, Hospital of the University of Pennsylvania
- Department of Medicine, University of Connecticut, Hartford, Connecticut, USA
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania
| | - Mona-Elisabeth Revheim
- Division of Radiology and Nuclear Medicine, Oslo University Hospital
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital
- Department of Clinical Research, Research Unit of Clinical Physiology and Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania
| |
Collapse
|
54
|
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and accounts for approximately 50% to 80% of all cases of dementia. The diagnosis of probable AD is based on clinical criteria and overlapping clinical features pose a challenge to accurate diagnosis. However, neuroimaging has been included as a biomarker in various published criteria for the diagnosis of probable AD, in the absence of a confirmatory diagnostic test during life. Advances in neuroimaging techniques and their inclusion in diagnostic and research criteria for the diagnosis of AD includes the use of positron emission tomography (PET) imaging as a biomarker in various therapeutic and prognostic studies in AD. The development and application of a range of PET tracers will allow more detailed assessment of people with AD and will improve diagnostic specificity and targeted therapy of AD. The aim of this review is to summarize current evidence on PET imaging using the non-specific tracer [18F]fluorodeoxyglucose and specific tracers that target amyloid and tau pathology in people with AD.
Collapse
Affiliation(s)
- Shailendra Mohan Tripathi
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| |
Collapse
|
55
|
Kumar P, Liu C, Hsu JW, Chacko S, Minard C, Jahoor F, Sekhar RV. Glycine and N-acetylcysteine (GlyNAC) supplementation in older adults improves glutathione deficiency, oxidative stress, mitochondrial dysfunction, inflammation, insulin resistance, endothelial dysfunction, genotoxicity, muscle strength, and cognition: Results of a pilot clinical trial. Clin Transl Med 2021; 11:e372. [PMID: 33783984 PMCID: PMC8002905 DOI: 10.1002/ctm2.372] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/07/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Oxidative stress (OxS) and mitochondrial dysfunction are implicated as causative factors for aging. Older adults (OAs) have an increased prevalence of elevated OxS, impaired mitochondrial fuel-oxidation (MFO), elevated inflammation, endothelial dysfunction, insulin resistance, cognitive decline, muscle weakness, and sarcopenia, but contributing mechanisms are unknown, and interventions are limited/lacking. We previously reported that inducing deficiency of the antioxidant tripeptide glutathione (GSH) in young mice results in mitochondrial dysfunction, and that supplementing GlyNAC (combination of glycine and N-acetylcysteine [NAC]) in aged mice improves naturally-occurring GSH deficiency, mitochondrial impairment, OxS, and insulin resistance. This pilot trial in OA was conducted to test the effect of GlyNAC supplementation and withdrawal on intracellular GSH concentrations, OxS, MFO, inflammation, endothelial function, genotoxicity, muscle and glucose metabolism, body composition, strength, and cognition. METHODS A 36-week open-label clinical trial was conducted in eight OAs and eight young adults (YAs). After all the participants underwent an initial (pre-supplementation) study, the YAs were released from the study. OAs were studied again after GlyNAC supplementation for 24 weeks, and GlyNAC withdrawal for 12 weeks. Measurements included red-blood cell (RBC) GSH, MFO; plasma biomarkers of OxS, inflammation, endothelial function, glucose, and insulin; gait-speed, grip-strength, 6-min walk test; cognitive tests; genomic-damage; glucose-production and muscle-protein breakdown rates; and body-composition. RESULTS GlyNAC supplementation for 24 weeks in OA corrected RBC-GSH deficiency, OxS, and mitochondrial dysfunction; and improved inflammation, endothelial dysfunction, insulin-resistance, genomic-damage, cognition, strength, gait-speed, and exercise capacity; and lowered body-fat and waist-circumference. However, benefits declined after stopping GlyNAC supplementation for 12 weeks. CONCLUSIONS GlyNAC supplementation for 24-weeks in OA was well tolerated and lowered OxS, corrected intracellular GSH deficiency and mitochondrial dysfunction, decreased inflammation, insulin-resistance and endothelial dysfunction, and genomic-damage, and improved strength, gait-speed, cognition, and body composition. Supplementing GlyNAC in aging humans could be a simple and viable method to promote health and warrants additional investigation.
Collapse
Affiliation(s)
- Premranjan Kumar
- Translational Metabolism Unit, Division of Endocrinology, Diabetes and MetabolismDepartment of Medicine, Baylor College of MedicineHoustonTexas77030USA
| | - Chun Liu
- Translational Metabolism Unit, Division of Endocrinology, Diabetes and MetabolismDepartment of Medicine, Baylor College of MedicineHoustonTexas77030USA
| | - Jean W. Hsu
- USDA/ARS Children's Nutritional Research CenterHoustonTexasUSA
| | - Shaji Chacko
- USDA/ARS Children's Nutritional Research CenterHoustonTexasUSA
| | - Charles Minard
- Institute of Clinical and Translational Research, Baylor College of MedicineHoustonTexas
| | - Farook Jahoor
- USDA/ARS Children's Nutritional Research CenterHoustonTexasUSA
| | - Rajagopal V. Sekhar
- Translational Metabolism Unit, Division of Endocrinology, Diabetes and MetabolismDepartment of Medicine, Baylor College of MedicineHoustonTexas77030USA
| |
Collapse
|
56
|
He L, Loika Y, Park Y, Bennett DA, Kellis M, Kulminski AM. Exome-wide age-of-onset analysis reveals exonic variants in ERN1 and SPPL2C associated with Alzheimer's disease. Transl Psychiatry 2021; 11:146. [PMID: 33637690 PMCID: PMC7910483 DOI: 10.1038/s41398-021-01263-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/07/2021] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Despite recent discoveries in genome-wide association studies (GWAS) of genomic variants associated with Alzheimer's disease (AD), its underlying biological mechanisms are still elusive. The discovery of novel AD-associated genetic variants, particularly in coding regions and from APOE ε4 non-carriers, is critical for understanding the pathology of AD. In this study, we carried out an exome-wide association analysis of age-of-onset of AD with ~20,000 subjects and placed more emphasis on APOE ε4 non-carriers. Using Cox mixed-effects models, we find that age-of-onset shows a stronger genetic signal than AD case-control status, capturing many known variants with stronger significance, and also revealing new variants. We identified two novel variants, rs56201815, a rare synonymous variant in ERN1, and rs12373123, a common missense variant in SPPL2C in the MAPT region in APOE ε4 non-carriers. Besides, a rare missense variant rs144292455 in TACR3 showed the consistent direction of effect sizes across all studies with a suggestive significant level. In an attempt to unravel their regulatory and biological functions, we found that the minor allele of rs56201815 was associated with lower average FDG uptake across five brain regions in ADNI. Our eQTL analyses based on 6198 gene expression samples from ROSMAP and GTEx revealed that the minor allele of rs56201815 was potentially associated with elevated expression of ERN1, a key gene triggering unfolded protein response (UPR), in multiple brain regions, including the posterior cingulate cortex and nucleus accumbens. Our cell-type-specific eQTL analysis using ~80,000 single nuclei in the prefrontal cortex revealed that the protective minor allele of rs12373123 significantly increased the expression of GRN in microglia, and was associated with MAPT expression in astrocytes. These findings provide novel evidence supporting the hypothesis of the potential involvement of the UPR to ER stress in the pathological pathway of AD, and also give more insights into underlying regulatory mechanisms behind the pleiotropic effects of rs12373123 in multiple degenerative diseases including AD and Parkinson's disease.
Collapse
Affiliation(s)
- Liang He
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA.
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA
| | - Yongjin Park
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA.
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
57
|
Pontifex MG, Malik MMAH, Connell E, Müller M, Vauzour D. Citrus Polyphenols in Brain Health and Disease: Current Perspectives. Front Neurosci 2021; 15:640648. [PMID: 33679318 PMCID: PMC7933480 DOI: 10.3389/fnins.2021.640648] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/25/2021] [Indexed: 12/17/2022] Open
Abstract
In addition to essential micronutrients such as vitamin C, citrus fruits represent a considerably rich source of non-essential bioactive compounds, in particular flavanones which form a sub-set of the flavonoid group. Preclinical studies have demonstrated the neuroprotective potential of citrus flavonoids and have highlighted both the well-established (anti-inflammatory and anti-oxidative properties), and newly emerging (influence upon blood-brain barrier function/integrity) mechanistic actions by which these neurological effects are mediated. Encouragingly, results from human studies, although limited in number, appear to support this preclinical basis, with improvements in cognitive performance and disease risk observed across healthy and disease states. Therefore, citrus fruits - both as whole fruit and 100% juices - should be encouraged within the diet for their potential neurological benefit. In addition, there should be further exploration of citrus polyphenols to establish therapeutic efficacy, particularly in the context of well-designed human interventions.
Collapse
Affiliation(s)
- Matthew G Pontifex
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Mohammad M A H Malik
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Emily Connell
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - Michael Müller
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| | - David Vauzour
- Norwich Medical School, Biomedical Research Centre, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
58
|
Cortes-Canteli M, Gispert JD, Salvadó G, Toribio-Fernandez R, Tristão-Pereira C, Falcon C, Oliva B, Mendiguren J, Fernandez-Friera L, Sanz J, Garcia-Ruiz JM, Fernandez-Ortiz A, Sanchez-Gonzalez J, Ibanez B, Molinuevo JL, Fuster V. Subclinical Atherosclerosis and Brain Metabolism in Middle-Aged Individuals: The PESA Study. J Am Coll Cardiol 2021; 77:888-898. [PMID: 33602472 DOI: 10.1016/j.jacc.2020.12.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND Atherosclerosis has been linked to cognitive decline in late life; however, the impact of cardiovascular risk factors (CVRFs) and subclinical atherosclerosis on brain metabolism at earlier stages remains unexplored. OBJECTIVES This study sought to determine the association between brain metabolism, subclinical atherosclerosis, and CVRFs in middle-aged asymptomatic individuals. METHODS This study included 547 asymptomatic middle-aged participants (50 ± 4 years, 82% men) from the PESA (Progression of Early Subclinical Atherosclerosis) study with evidence of subclinical atherosclerosis. Participants underwent 18F-fluorodeoxyglucose (FDG)-positron emission tomography. Global brain FDG uptake and voxel-wise analyses were used to evaluate the associations of cerebral metabolism with CVRFs and atherosclerotic plaque burden in carotids and femorals assessed by 3-dimensional vascular ultrasound. RESULTS Global FDG uptake showed an inverse correlation with 30-year Framingham Risk Score (FRS) (β = -0.15, p < 0.001). This association was mainly driven by the presence of hypertension (d = 0.36, p < 0.001). Carotid plaque burden was inversely associated with global brain FDG uptake (β = -0.16, p < 0.001), even after adjusting for 30-year FRS. Voxel-wise approaches revealed that the brain areas most strongly affected by hypometabolism in association with 30-year FRS, hypertension, and carotid plaque burden were parietotemporal regions (angular, supramarginal, and inferior/middle temporal gyri) and the cingulate gyrus. CONCLUSIONS In asymptomatic middle-aged individuals, cardiovascular risk is associated with brain hypometabolism, with hypertension being the modifiable CVRF showing the strongest association. Subclinical carotid plaque burden is also linked to reduced brain metabolism independently of CVRFs. Cerebral areas showing hypometabolism include those known to be affected in dementia. These data reinforce the need to control CVRFs early in life in order to potentially reduce the brain's midlife vulnerability to future cognitive dysfunction.
Collapse
Affiliation(s)
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Gemma Salvadó
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | | | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Belen Oliva
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Leticia Fernandez-Friera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; HM Hospitales-Centro Integral de Enfermedades Cardiovasculares, Universidad San Pablo-CEU, Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain
| | - Javier Sanz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jose M Garcia-Ruiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Hospital Universitario Central de Oviedo, Asturias, Spain
| | - Antonio Fernandez-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos, Universidad Complutense, Madrid, Spain
| | | | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; CIBER de enfermedades Cardiovasculares, Madrid, Spain; Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Madrid, Spain.
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
59
|
Contino S, Suelves N, Vrancx C, Vadukul DM, Payen VL, Stanga S, Bertrand L, Kienlen-Campard P. Presenilin-Deficient Neurons and Astrocytes Display Normal Mitochondrial Phenotypes. Front Neurosci 2021; 14:586108. [PMID: 33551720 PMCID: PMC7862347 DOI: 10.3389/fnins.2020.586108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/14/2020] [Indexed: 01/13/2023] Open
Abstract
Presenilin 1 (PS1) and Presenilin 2 (PS2) are predominantly known as the catalytic subunits of the γ-secretase complex that generates the amyloid-β (Aβ) peptide, the major constituent of the senile plaques found in the brain of Alzheimer's disease (AD) patients. Apart from their role in γ-secretase activity, a growing number of cellular functions have been recently attributed to PSs. Notably, PSs were found to be enriched in mitochondria-associated membranes (MAMs) where mitochondria and endoplasmic reticulum (ER) interact. PS2 was more specifically reported to regulate calcium shuttling between these two organelles by controlling the formation of functional MAMs. We have previously demonstrated in mouse embryonic fibroblasts (MEF) an altered mitochondrial morphology along with reduced mitochondrial respiration and increased glycolysis in PS2-deficient cells (PS2KO). This phenotype was restored by the stable re-expression of human PS2. Still, all these results were obtained in immortalized cells, and one bottom-line question is to know whether these observations hold true in central nervous system (CNS) cells. To that end, we carried out primary cultures of PS1 knockdown (KD), PS2KO, and PS1KD/PS2KO (PSdKO) neurons and astrocytes. They were obtained from the same litter by crossing PS2 heterozygous; PS1 floxed (PS2+/-; PS1flox/flox) animals. Genetic downregulation of PS1 was achieved by lentiviral expression of the Cre recombinase in primary cultures. Strikingly, we did not observe any mitochondrial phenotype in PS1KD, PS2KO, or PSdKO primary cultures in basal conditions. Mitochondrial respiration and membrane potential were similar in all models, as were the glycolytic flux and NAD+/NADH ratio. Likewise, mitochondrial morphology and content was unaltered by PS expression. We further investigated the differences between results we obtained here in primary nerve cells and those previously reported in MEF cell lines by analyzing PS2KO primary fibroblasts. We found no mitochondrial dysfunction in this model, in line with observations in PS2KO primary neurons and astrocytes. Together, our results indicate that the mitochondrial phenotype observed in immortalized PS2-deficient cell lines cannot be extrapolated to primary neurons, astrocytes, and even to primary fibroblasts. The PS-dependent mitochondrial phenotype reported so far might therefore be the consequence of a cell immortalization process and should be critically reconsidered regarding its relevance to AD.
Collapse
Affiliation(s)
- Sabrina Contino
- Alzheimer Research Group, Molecular and Cellular Division (CEMO), Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Nuria Suelves
- Alzheimer Research Group, Molecular and Cellular Division (CEMO), Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Céline Vrancx
- Alzheimer Research Group, Molecular and Cellular Division (CEMO), Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Devkee M. Vadukul
- Alzheimer Research Group, Molecular and Cellular Division (CEMO), Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Valery L. Payen
- Laboratory of Advanced Drug Delivery and Biomaterial (ADDB), Louvain Drug Research Institute (LDRI), Université Catholique de Louvain, Brussels, Belgium
| | - Serena Stanga
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience, University of Torino, Torino, Italy
| | - Luc Bertrand
- Pole of Cardiovascular Research, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Pascal Kienlen-Campard
- Alzheimer Research Group, Molecular and Cellular Division (CEMO), Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
60
|
Schaeffer MJ, Chan L, Barber PA. The neuroimaging of neurodegenerative and vascular disease in the secondary prevention of cognitive decline. Neural Regen Res 2021; 16:1490-1499. [PMID: 33433462 PMCID: PMC8323688 DOI: 10.4103/1673-5374.303011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Structural brain changes indicative of dementia occur up to 20 years before the onset of clinical symptoms. Efforts to modify the disease process after the onset of cognitive symptoms have been unsuccessful in recent years. Thus, future trials must begin during the preclinical phases of the disease before symptom onset. Age related cognitive decline is often the result of two coexisting brain pathologies: Alzheimer’s disease (amyloid, tau, and neurodegeneration) and vascular disease. This review article highlights some of the common neuroimaging techniques used to visualize the accumulation of neurodegenerative and vascular pathologies during the preclinical stages of dementia such as structural magnetic resonance imaging, positron emission tomography, and white matter hyperintensities. We also describe some emerging neuroimaging techniques such as arterial spin labeling, diffusion tensor imaging, and quantitative susceptibility mapping. Recent literature suggests that structural imaging may be the most sensitive and cost-effective marker to detect cognitive decline, while molecular positron emission tomography is primarily useful for detecting disease specific pathology later in the disease process. Currently, the presence of vascular disease on magnetic resonance imaging provides a potential target for optimizing vascular risk reduction strategies, and the presence of vascular disease may be useful when combined with molecular and metabolic markers of neurodegeneration for identifying the risk of cognitive impairment.
Collapse
Affiliation(s)
- Morgan J Schaeffer
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Leona Chan
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Philip A Barber
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
61
|
Medhi B, Soni H, Goyal M, Sarma P, Singh H, Modi M, Sharma A, Mohanty M, Vishnu V, Kumar A, Mittal B. Evaluation of plasma amyloid peptides Aβ 1-40 and Aβ 1-42 as diagnostic biomarker of alzheimer's disease, its association with different grades of clinical severity and 18f-fluorodeoxyglucose positron emission tomography Z score in the Indian population: A case-control study. Indian J Nucl Med 2021; 36:391-397. [PMID: 35125757 PMCID: PMC8771055 DOI: 10.4103/ijnm.ijnm_50_21] [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: 04/06/2021] [Revised: 07/01/2021] [Accepted: 08/06/2021] [Indexed: 11/04/2022] Open
Abstract
Background: We estimated plasma amyloid-peptides levels (Aβ1-42 and Aβ1-40) as diagnostic biomarker of Alzheimer's disease (AD) and evaluated its association with clinical severity and 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) Z score of the different brain regions in the Indian population. Patients and Methods: A case-control study was conducted. Diagnostic and statistical manual-IV, Dubois, and NIA-AA criteria were used for the diagnosis of AD. The plasma Aβ1-42 and Aβ1-40 concentration and 18F-FDG PET Z score were estimated for different brain regions. Results: Forty-seven cognitive impairment patients (AD = 29, mild cognitive impairment = 18) and 33 age-matched controls were enrolled. Plasma Aβ1-42 level was significantly higher in the AD group compared to controls (P = 0.046) and a cut-off >5.7 ng/mL has a specificity of 96.9%, sensitivity of 27.6%, positive predictive value 88.9%, and negative predictive value 60.4% for differentiating AD patients from controls. Significant correlation was seen between Aβ1-40/Aβ1-42 ratio and 18F-FDG PET Z score in the bilateral-parietal, temporal, frontal-association area, and posterior-cingulate areas. Conclusion: As a diagnostic biomarker of AD, plasma Aβ1-42 level showed good specificity but low sensitivity in the Indian population.
Collapse
|
62
|
Dave A, Hansen N, Downey R, Johnson C. FDG-PET Imaging of Dementia and Neurodegenerative Disease. Semin Ultrasound CT MR 2020; 41:562-571. [DOI: 10.1053/j.sult.2020.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
63
|
Faustman DL. Benefits of BCG-induced metabolic switch from oxidative phosphorylation to aerobic glycolysis in autoimmune and nervous system diseases. J Intern Med 2020; 288:641-650. [PMID: 32107806 DOI: 10.1111/joim.13050] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/10/2020] [Accepted: 01/24/2020] [Indexed: 12/19/2022]
Abstract
The most commonly used vaccine worldwide, bacillus Calmette-Guerin (BCG), appears to have the ability to restore blood sugar control in humans with early-onset but long-duration type 1 diabetes when a repeat vaccination strategy is used. This is a process that may be driven by a metabolic switch from overactive oxidative phosphorylation to accelerated aerobic glycolysis and a reset of the immune system. BCG is a live, attenuated strain of Mycobacteria bovis, a cousin of M. tuberculosis. Humans and Mycobacteria, which are found in the environment and in warm-blooded hosts, share a long coevolutionary history. In recent times, humans have had fewer exposures to these and other microorganisms that historically helped shape the immune response. By 're-introducing' an attenuated form of Mycobacteria via BCG vaccination, humans might benefit from an immunological perspective, a concept supported by a growing body of data in autoimmunity and robust data on the nonspecific immune effects of BCG related to protection from diverse infections and early mortality. New findings of immune and metabolic defects in type 1 diabetes that can be corrected with repeat BCG vaccination suggest that this therapeutic strategy may be applicable in other diseases with inadequate aerobic glycolysis, including Parkinson's disease, dementia, depression and other disorders affecting the nervous system.
Collapse
Affiliation(s)
- D L Faustman
- From the, Department of Medicine, Immunobiology Laboratories, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
64
|
von Morze C, Engelbach JA, Blazey T, Quirk JD, Reed GD, Ippolito JE, Garbow JR. Comparison of hyperpolarized 13 C and non-hyperpolarized deuterium MRI approaches for imaging cerebral glucose metabolism at 4.7 T. Magn Reson Med 2020; 85:1795-1804. [PMID: 33247884 DOI: 10.1002/mrm.28612] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/12/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE The purpose of this study was to directly compare two isotopic metabolic imaging approaches, hyperpolarized (HP) 13 C MRI and deuterium metabolic imaging (DMI), for imaging specific closely related segments of cerebral glucose metabolism at 4.7 T. METHODS Comparative HP-13 C and DMI neuroimaging experiments were conducted consecutively in normal rats during the same scanning session. Localized conversions of [1-13 C]pyruvate and [6,6-2 H2 ]glucose to their respective downstream metabolic products were measured by spectroscopic imaging, using an identical 2D-CSI sequence with parameters optimized for the respective experiments. To facilitate direct comparison, a pair of substantially equivalent 2.5-cm double-tuned X/1 H RF surface coils was developed. For improved results, multidimensional low-rank reconstruction was applied to denoise the raw DMI data. RESULTS Localized conversion of HP [1-13 C]pyruvate to [1-13 C]lactate, and [6,6-2 H2 ]glucose to [3,3-2 H2 ]lactate and Glx-d (glutamate and glutamine), was detected in rat brain by spectroscopic imaging at 4.7 T. The SNR and spatial resolution of HP-13 C MRI was superior to DMI but limited to a short time window, whereas the lengthy DMI acquisition yielded maps of not only lactate, but also Glx production, albeit with relatively poor spectral discrimination between metabolites at this field strength. Across the individual rats, there was an apparent inverse correlation between cerebral production of HP [1-13 C]lactate and Glx-d, along with a trend toward increased [3,3-2 H2 ]lactate. CONCLUSION The HP-13 C MRI and DMI methods are both feasible at 4.7 T and have significant potential for metabolic imaging of specific segments of glucose metabolism.
Collapse
Affiliation(s)
- Cornelius von Morze
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, USA
| | - John A Engelbach
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, USA
| | - Tyler Blazey
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, USA
| | - James D Quirk
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, USA
| | | | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, USA
| | - Joel R Garbow
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
65
|
Du L, Liu F, Liu K, Yao X, Risacher SL, Han J, Saykin AJ, Shen L. Associating Multi-Modal Brain Imaging Phenotypes and Genetic Risk Factors via a Dirty Multi-Task Learning Method. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:3416-3428. [PMID: 32746095 PMCID: PMC7705646 DOI: 10.1109/tmi.2020.2995510] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Brain imaging genetics becomes more and more important in brain science, which integrates genetic variations and brain structures or functions to study the genetic basis of brain disorders. The multi-modal imaging data collected by different technologies, measuring the same brain distinctly, might carry complementary information. Unfortunately, we do not know the extent to which the phenotypic variance is shared among multiple imaging modalities, which further might trace back to the complex genetic mechanism. In this paper, we propose a novel dirty multi-task sparse canonical correlation analysis (SCCA) to study imaging genetic problems with multi-modal brain imaging quantitative traits (QTs) involved. The proposed method takes advantages of the multi-task learning and parameter decomposition. It can not only identify the shared imaging QTs and genetic loci across multiple modalities, but also identify the modality-specific imaging QTs and genetic loci, exhibiting a flexible capability of identifying complex multi-SNP-multi-QT associations. Using the state-of-the-art multi-view SCCA and multi-task SCCA, the proposed method shows better or comparable canonical correlation coefficients and canonical weights on both synthetic and real neuroimaging genetic data. In addition, the identified modality-consistent biomarkers, as well as the modality-specific biomarkers, provide meaningful and interesting information, demonstrating the dirty multi-task SCCA could be a powerful alternative method in multi-modal brain imaging genetics.
Collapse
Affiliation(s)
- Lei Du
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Fang Liu
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Kefei Liu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Xiaohui Yao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Junwei Han
- School of Automation, Northwestern Polytechnical University, Xi’an 710072, China
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
66
|
Valotassiou V, Angelidis G, Psimadas D, Tsougos I, Georgoulias P. In the era of FDG PET, is it time for brain perfusion SPECT to gain a place in Alzheimer's disease imaging biomarkers? Eur J Nucl Med Mol Imaging 2020; 48:969-971. [PMID: 33078261 DOI: 10.1007/s00259-020-05077-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 11/24/2022]
Affiliation(s)
- Varvara Valotassiou
- Department of Nuclear Medicine, University Hospital of Larissa, Thessaly, Greece.
| | - George Angelidis
- Department of Nuclear Medicine, University Hospital of Larissa, Thessaly, Greece
| | - Dimitrios Psimadas
- Department of Nuclear Medicine, University Hospital of Larissa, Thessaly, Greece
| | - Ioannis Tsougos
- Medical Physics Department, Medical School, University of Thessaly, Thessaly, Greece
| | | |
Collapse
|
67
|
Ganesh A, Genesh P, Adil MM, Varma M, Smith EE. Practice Current: How do you manage mild cognitive impairment? Neurol Clin Pract 2020; 10:362-370. [PMID: 32983617 DOI: 10.1212/cpj.0000000000000890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/08/2020] [Indexed: 11/15/2022]
Abstract
Mild cognitive impairment (MCI) is characterized by evidence of cognitive impairment with minimal disruption of instrumental activities of daily living and carries a substantial risk of progression of dementia. Whereas current guidelines support a relatively minimalistic workup to identify reversible or structural causes, the field has witnessed the rapid development of various sophisticated imaging, biomarker, and genetic investigations in the past few years. The role of these investigations in routine practice is uncertain. Similarly, although there are no approved treatments for MCI, neurologists may experience uncertainty about using cholinesterase inhibitors or other medications or supplements that have been studied in MCI with limited success, particularly when patients or families are keen to try pharmacologic options. Given these uncertainties, and the paucity of high-quality data in the literature, we sought expert opinion from around the globe on how to investigate and treat patients with MCI. Similar questions were posed to the rest of our readership in an online survey, the preliminary results of which are also presented.
Collapse
Affiliation(s)
- Aravind Ganesh
- Department of Clinical Neurosciences (AG), University of Calgary; Learning and Support Services (PG), Alzheimer Society of Calgary, Canada; Johns Hopkins University School of Medicine (MMA), Bethesda, MD; Advanced Health Analytics (AHA Health Ltd) (MV), Calgary, and Katthy Taylor Chair in Vascular Dementia (EES), Department of Clinical Neurosciences, University of Calgary, Canada
| | - Padmaja Genesh
- Department of Clinical Neurosciences (AG), University of Calgary; Learning and Support Services (PG), Alzheimer Society of Calgary, Canada; Johns Hopkins University School of Medicine (MMA), Bethesda, MD; Advanced Health Analytics (AHA Health Ltd) (MV), Calgary, and Katthy Taylor Chair in Vascular Dementia (EES), Department of Clinical Neurosciences, University of Calgary, Canada
| | - Malik M Adil
- Department of Clinical Neurosciences (AG), University of Calgary; Learning and Support Services (PG), Alzheimer Society of Calgary, Canada; Johns Hopkins University School of Medicine (MMA), Bethesda, MD; Advanced Health Analytics (AHA Health Ltd) (MV), Calgary, and Katthy Taylor Chair in Vascular Dementia (EES), Department of Clinical Neurosciences, University of Calgary, Canada
| | - Malavika Varma
- Department of Clinical Neurosciences (AG), University of Calgary; Learning and Support Services (PG), Alzheimer Society of Calgary, Canada; Johns Hopkins University School of Medicine (MMA), Bethesda, MD; Advanced Health Analytics (AHA Health Ltd) (MV), Calgary, and Katthy Taylor Chair in Vascular Dementia (EES), Department of Clinical Neurosciences, University of Calgary, Canada
| | - Eric E Smith
- Department of Clinical Neurosciences (AG), University of Calgary; Learning and Support Services (PG), Alzheimer Society of Calgary, Canada; Johns Hopkins University School of Medicine (MMA), Bethesda, MD; Advanced Health Analytics (AHA Health Ltd) (MV), Calgary, and Katthy Taylor Chair in Vascular Dementia (EES), Department of Clinical Neurosciences, University of Calgary, Canada
| |
Collapse
|
68
|
Abstract
FTLD (Frontotemporal lobar degeneration) is a clinically and pathologically heterogeneous group of degenerative disorders, characterized by predominantly asymmetric degeneration of frontal and temporal lobes with selective neuronal loss and gliosis. The disease presents with variable degrees of impairment in behavior, language, executive control, and motor symptoms with progressive loss of cognition. On the basis of presenting clinical symptoms, FTLD is further divided into behavioral variant, nonfluent/agrammatic variant primary progressive aphasia (PPA), semantic variant PPA, logopenic variant PPA, progressive supranuclear palsy, and corticobasal ganglionic degeneration. Here we illustrate the utility of FDG PET with statistical parametric analysis for evaluation of these patients.
Collapse
|
69
|
Glutamate-glutamine homeostasis is perturbed in neurons and astrocytes derived from patient iPSC models of frontotemporal dementia. Mol Brain 2020; 13:125. [PMID: 32928252 PMCID: PMC7491073 DOI: 10.1186/s13041-020-00658-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 08/21/2020] [Indexed: 02/08/2023] Open
Abstract
Frontotemporal dementia (FTD) is amongst the most prevalent early onset dementias and even though it is clinically, pathologically and genetically heterogeneous, a crucial involvement of metabolic perturbations in FTD pathology is being recognized. However, changes in metabolism at the cellular level, implicated in FTD and in neurodegeneration in general, are still poorly understood. Here we generate induced human pluripotent stem cells (hiPSCs) from patients carrying mutations in CHMP2B (FTD3) and isogenic controls generated via CRISPR/Cas9 gene editing with subsequent neuronal and glial differentiation and characterization. FTD3 neurons show a dysregulation of glutamate-glutamine related metabolic pathways mapped by 13C-labelling coupled to mass spectrometry. FTD3 astrocytes show increased uptake of glutamate whilst glutamate metabolism is largely maintained. Using quantitative proteomics and live-cell metabolic analyses, we elucidate molecular determinants and functional alterations of neuronal and glial energy metabolism in FTD3. Importantly, correction of the mutations rescues such pathological phenotypes. Notably, these findings implicate dysregulation of key enzymes crucial for glutamate-glutamine homeostasis in FTD3 pathogenesis which may underlie vulnerability to neurodegeneration. Neurons derived from human induced pluripotent stem cells (hiPSCs) of patients carrying mutations in CHMP2B (FTD3) display major metabolic alterations compared to CRISPR/Cas9 generated isogenic controls. Using quantitative proteomics, 13C-labelling coupled to mass spectrometry metabolic mapping and seahorse analyses, molecular determinants and functional alterations of neuronal and astrocytic energy metabolism in FTD3 were characterized. Our findings implicate dysregulation of glutamate-glutamine homeostasis in FTD3 pathogenesis. In addition, FTD3 neurons recapitulate glucose hypometabolism observed in FTD patient brains. The impaired mitochondria function found here is concordant with disturbed TCA cycle activity and decreased glycolysis in FTD3 neurons. FTD3 neuronal glutamine hypermetabolism is associated with up-regulation of PAG expression and, possibly, ROS production. Distinct compartments of glutamate metabolism can be suggested for the FTD3 neurons. Endogenous glutamate generated from glutamine via PAG may enter the TCA cycle via AAT (left side of neuron) while exogenous glutamate taken up from the extracellular space may be incorporated into the TCA cycle via GDH (right side of the neuron) FTD3 astrocytic glutamate uptake is upregulated whilst glutamate metabolism is largely maintained. Finally, pharmacological reversal of glutamate hypometabolism manifesting from decreased GDH expression should be explored as a novel therapeutic intervention for treating FTD3.
Collapse
|
70
|
Lesman-Segev OH, Edwards L, Rabinovici GD. Chronic Traumatic Encephalopathy: A Comparison with Alzheimer's Disease and Frontotemporal Dementia. Semin Neurol 2020; 40:394-410. [PMID: 32820492 DOI: 10.1055/s-0040-1715134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The clinical diagnosis of chronic traumatic encephalopathy (CTE) is challenging due to heterogeneous clinical presentations and overlap with other neurodegenerative dementias. Depending on the clinical presentation, the differential diagnosis of CTE includes Alzheimer's disease (AD), behavioral variant frontotemporal dementia (bvFTD), Parkinson's disease, amyotrophic lateral sclerosis, primary mood disorders, posttraumatic stress disorder, and psychotic disorders. The aim of this article is to compare the clinical aspects, genetics, fluid biomarkers, imaging, treatment, and pathology of CTE to those of AD and bvFTD. A detailed clinical evaluation, neurocognitive assessment, and structural brain imaging can inform the differential diagnosis, while molecular biomarkers can help exclude underlying AD pathology. Prospective studies that include clinicopathological correlations are needed to establish tools that can more accurately determine the cause of neuropsychiatric decline in patients at risk for CTE.
Collapse
Affiliation(s)
- Orit H Lesman-Segev
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - Lauren Edwards
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, California.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California.,Weill Neuroscience Institute, University of California San Francisco, San Francisco, California
| |
Collapse
|
71
|
Cistaro A, Quartuccio N, Piccardo A, Fania P, Spunton M, Liava A, Danesino C, Albani G, Guala A. 18F-FDG PET Identifies Altered Brain Metabolism in Patients with Cri du Chat Syndrome. J Nucl Med 2020; 61:1195-1199. [PMID: 31836684 DOI: 10.2967/jnumed.119.236893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023] Open
Abstract
Cri du chat syndrome (CDCS) is a rare genetic disease that is caused by a deletion in the short arm of chromosome 5 (5p) and has a variable clinical spectrum. To our knowledge, no study in the literature has ever applied 18F-FDG PET/CT to investigate alterations in brain glucose metabolism in these subjects. The aims of this study were to detect differences in brain 18F-FDG metabolism in CDCS patients with different clinical presentations and identify possible brain metabolic phenotypes of this syndrome. Methods: Six patients (5 male and 1 female; age range, 10-27 y) with CDCS were assessed for the presence of cognitive and behavioral symptoms using a battery of neuropsychologic tests and then classified as having either a severe or a mild phenotype. The patients then underwent brain 18F-FDG PET/CT. The PET/CT findings were compared with an age- and sex-matched control group using statistical parametric mapping (SPM). Whether there was an association between different clinical phenotypes and 18F-FDG PET/CT findings was investigated. Results: Four patients had the severe phenotype, and 2 patients demonstrated the mild phenotype. SPM single-subject analysis, and a group analysis in comparison with the control cohort, revealed significant hypometabolism in the left temporal lobe (Brodmann areas [BAs] 20, 36, and 38), in the right frontal subcallosal gyrus (BA 34) and caudate body, and in the cerebellar tonsils (P < 0.001). Hypermetabolism (P = 0.001) was revealed in the right superior and precentral frontal gyrus (BA 6) in the patient group, compared with the control cohort. In SPM single-subject analysis, the hypermetabolic areas were detected only in patients with the severe phenotype. Conclusion: This study revealed different patterns of brain glucose metabolism in patients with the severe and mild phenotypes, compared with control subjects. In particular, abnormal hypermetabolism in the brain, as evaluated by18F-FDG PET/CT, seems to correlate with the severe CDCS phenotype.
Collapse
Affiliation(s)
- Angelina Cistaro
- Nuclear Medicine Department, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
- AIMN Pediatric Study Group, Milan, Italy
- Scientific Committee of ABC Associazione Nazionale Bambini Cri du Chat, San Casciano in Val di Pesa (Firenze), Italy
| | - Natale Quartuccio
- AIMN Pediatric Study Group, Milan, Italy
- Nuclear Medicine Unit, ARNAS Ospedali Civico, Di Cristina e Benfratelli, Palermo, Italy
| | - Arnoldo Piccardo
- Nuclear Medicine Department, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
- AIMN Pediatric Study Group, Milan, Italy
| | | | - Marianna Spunton
- Scientific Committee of ABC Associazione Nazionale Bambini Cri du Chat, San Casciano in Val di Pesa (Firenze), Italy
- Paediatric Unit, Castelli Hospital, Verbania, Italy
| | - Alexandra Liava
- Scientific Committee of ABC Associazione Nazionale Bambini Cri du Chat, San Casciano in Val di Pesa (Firenze), Italy
- Child Neuropsychiatric Unit, Castelli Hospital, Verbania, Italy
| | - Cesare Danesino
- Scientific Committee of ABC Associazione Nazionale Bambini Cri du Chat, San Casciano in Val di Pesa (Firenze), Italy
- Department of Molecular Medicine, IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy; and
| | - Giovanni Albani
- Scientific Committee of ABC Associazione Nazionale Bambini Cri du Chat, San Casciano in Val di Pesa (Firenze), Italy
- Department of Neurology, Istituto Auxologico Italiano, IRCCS, Piancavallo-Verbania, Italy
| | - Andrea Guala
- Scientific Committee of ABC Associazione Nazionale Bambini Cri du Chat, San Casciano in Val di Pesa (Firenze), Italy
- Paediatric Unit, Castelli Hospital, Verbania, Italy
| |
Collapse
|
72
|
Gramkow MH, Gjerum L, Koikkalainen J, Lötjönen J, Law I, Hasselbalch SG, Waldemar G, Frederiksen KS. Prognostic value of complementary biomarkers of neurodegeneration in a mixed memory clinic cohort. PeerJ 2020; 8:e9498. [PMID: 32714664 PMCID: PMC7354835 DOI: 10.7717/peerj.9498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/17/2020] [Indexed: 11/20/2022] Open
Abstract
Background Biomarkers of neurodegeneration, e.g. MRI brain atrophy and [18F]FDG-PET hypometabolism, are often evaluated in patients suspected of neurodegenerative disease. Objective Our primary objective was to investigate prognostic properties of atrophy and hypometabolism. Methods From March 2015-June 2016, 149 patients referred to a university hospital memory clinic were included. The primary outcome was progression/stable disease course as assessed by a clinician at 12 months follow-up. Intracohort defined z-scores of baseline MRI automatic quantified volume and [18F]FDG-PET standardized uptake value ratios were calculated for all unilaterally defined brain lobes and dichotomized as pronounced atrophy (+A)/ pronounced hypometabolism (+H) at z-score <0. A logistic regression model with progression status as the outcome was carried out with number of lobes with the patterns +A/-H, -A/+H, +A/+H respectively as predictors. The model was mutually adjusted along with adjustment for age and sex. A sensitivity analysis with a z-score dichotomization at −0.1 and −0.5 and dichotomization regarding number of lobes affected at one and three lobes was done. Results Median follow-up time was 420 days [IQR: 387-461 days] and 50 patients progressed. Patients with two or more lobes affected by the pattern +A/+H compared to patients with 0–1 lobes affected had a statistically significant increased risk of progression (odds ratio, 95 % confidence interval: 4.33, 1.90–9.86) in a multivariable model. The model was partially robust to the applied sensitivity analysis. Conclusion Combined atrophy and hypometabolism as assessed by MRI and [18F]FDG-PET in patients under suspicion of neurodegenerative disease predicts progression over 1 year.
Collapse
Affiliation(s)
- Mathias Holsey Gramkow
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Le Gjerum
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Steen Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
73
|
A critical review of radiotracers in the positron emission tomography imaging of traumatic brain injury: FDG, tau, and amyloid imaging in mild traumatic brain injury and chronic traumatic encephalopathy. Eur J Nucl Med Mol Imaging 2020; 48:623-641. [DOI: 10.1007/s00259-020-04926-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
|
74
|
Gatto EM, Rojas GJ, Nemirovsky SI, Da Prat G, Persi G, Cesarini M, Etcheverry JL, Rojas NG, Parisi V, Cordoba M, Sevlever G, Allegri RF, Turjanski AG. A novel mutation in PSEN1 (p.Arg41Ser) in an Argentinian woman with early onset Parkinsonism. Parkinsonism Relat Disord 2020; 77:21-25. [PMID: 32590294 DOI: 10.1016/j.parkreldis.2020.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/02/2020] [Accepted: 06/05/2020] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Mutations in presenilin-1 (PSEN1) account for the majority of cases of familial autosomal dominant early-onset Alzheimer's disease (AD) as well as in sporadic forms. Atypical presentations are reported including extrapyramidal signs. In the last years, a pleiotropic effect of some PSEN1 variants has been reported in Parkinson's disease (PD). OBJECTIVE to report a new PSEN1 mutation characterized by early-onset Parkinsonism (EOPD) without dementia or classical AD biomarkers phenotype. PATIENT AND METHODS An Argentinian 46 years old woman was diagnosed with EOPD at 35 years old with no family history of neurodegenerative disorders. Her medical history included iron deficiency and anemia since childhood. A brain MRI showed moderate frontal atrophy. 18FDG-PET and PiB-PET as well as CSF biomarkers were inconclusive for AD. Two neuropsychological examinations were compatible with a mild non amnestic cognitive impairment. Whole blood DNA was extracted and whole exome sequencing and analysis was performed. RESULTS AND CONCLUSION A heterozygous novel missense PSEN1 mutation (position 14:73637540, A > T, pArg41Ser) was identified as a likely causative mutation in this patient. To the best of our knowledge, this case is the first PSEN1 mutation with a l-dopa responsive Parkinsonism lacking distinctive classical AD biomarkers. This case opens a new window to explore the pathophysiological link among PSEN1 and EOPDs and contributes to increase the phenotypes of PSEN1 variants.
Collapse
Affiliation(s)
- Emilia M Gatto
- Departamento de Neurología. Sanatorio de la Trinidad Mitre, Bartolomé Mitre, 2553, Buenos Aires, Argentina; Instituto de Neurociencias Buenos Aires, INEBA, Guardia Vieja, 4435, Buenos Aires, Argentina.
| | - Galeno J Rojas
- Unidad de Neurociencias Cognitivas del Sanatorio de la Trinidad Mitre, Bartolomé Mitre, 2553, Buenos Aires, Argentina
| | - Sergio I Nemirovsky
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET Universidad de Buenos Aires, Av. Int. Cantilo, Buenos Aires, C1428EGA, Argentina
| | - Gustavo Da Prat
- Instituto de Neurociencias Buenos Aires, INEBA, Guardia Vieja, 4435, Buenos Aires, Argentina
| | - Gabriel Persi
- Departamento de Neurología. Sanatorio de la Trinidad Mitre, Bartolomé Mitre, 2553, Buenos Aires, Argentina
| | - Martin Cesarini
- Instituto de Neurociencias Buenos Aires, INEBA, Guardia Vieja, 4435, Buenos Aires, Argentina
| | - Jose L Etcheverry
- Instituto de Neurociencias Buenos Aires, INEBA, Guardia Vieja, 4435, Buenos Aires, Argentina
| | - Natalia Gonzalez Rojas
- Instituto de Neurociencias Buenos Aires, INEBA, Guardia Vieja, 4435, Buenos Aires, Argentina
| | - Virginia Parisi
- Departamento de Neurología. Sanatorio de la Trinidad Mitre, Bartolomé Mitre, 2553, Buenos Aires, Argentina
| | - Marta Cordoba
- Instituto de Neurociencias Buenos Aires, INEBA, Guardia Vieja, 4435, Buenos Aires, Argentina
| | - Gustavo Sevlever
- Laboratorio de Enfermedades Neurodegenerativas (LEN) Departamento de Neuropatología y Biología Molecular, Instituto de Investigaciones Neurológicas FLENI, Montañeses, 2325, Buenos Aires, Argentina
| | - Ricardo F Allegri
- Centro de Memoria y Envejecimiento, Instituto de Investigaciones Neurológicas FLENI, Montañeses, 2325, Buenos Aires, Argentina; Departamento de Neurociencias, Universidad de la Costa (CUC), Colombia
| | - Adrian G Turjanski
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, UBA, IQUIBICEN-CONICET, Buenos Aires, Argentina; Bitgenia, Ciudad Autónoma de Buenos Aires, Argentina, Av. Int. Cantilo, Buenos Aires C1428EGA, Argentina
| |
Collapse
|
75
|
Mapstone M, Gross TJ, Macciardi F, Cheema AK, Petersen M, Head E, Handen BL, Klunk WE, Christian BT, Silverman W, Lott IT, Schupf N, for the Alzheimer's Biomarkers Consortium–Down Syndrome (ABC‐DS) Investigators. Metabolic correlates of prevalent mild cognitive impairment and Alzheimer's disease in adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12028. [PMID: 32258359 PMCID: PMC7131985 DOI: 10.1002/dad2.12028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/14/2020] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Disruption of metabolic function is a recognized feature of late onset Alzheimer's disease (LOAD). We sought to determine whether similar metabolic pathways are implicated in adults with Down syndrome (DS) who have increased risk for Alzheimer's disease (AD). METHODS We examined peripheral blood from 292 participants with DS who completed baseline assessments in the Alzheimer's Biomarkers Consortium-Down Syndrome (ABC-DS) using untargeted mass spectrometry (MS). Our sample included 38 individuals who met consensus criteria for AD (DS-AD), 43 who met criteria for mild cognitive impairment (DS-MCI), and 211 who were cognitively unaffected and stable (CS). RESULTS We measured relative abundance of 8,805 features using MS and 180 putative metabolites were differentially expressed (DE) among the groups at false discovery rate-corrected q< 0.05. From the DE features, a nine-feature classifier model classified the CS and DS-AD groups with receiver operating characteristic area under the curve (ROC AUC) of 0.86 and a two-feature model classified the DS-MCI and DS-AD groups with ROC AUC of 0.88. Metabolite set enrichment analysis across the three groups suggested alterations in fatty acid and carbohydrate metabolism. DISCUSSION Our results reveal metabolic alterations in DS-AD that are similar to those seen in LOAD. The pattern of results in this cross-sectional DS cohort suggests a dynamic time course of metabolic dysregulation which evolves with clinical progression from non-demented, to MCI, to AD. Metabolomic markers may be useful for staging progression of DS-AD.
Collapse
Affiliation(s)
- Mark Mapstone
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Thomas J Gross
- Department of NeurologyUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Fabio Macciardi
- Department of Psychiatry and Human BehaviorUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Amrita K Cheema
- Departments of Biochemistry and Molecular & Cellular BiologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Melissa Petersen
- Institute for Translational ResearchUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Elizabeth Head
- Department of Pathology and Laboratory MedicineUniversity of California‐IrvineIrvineCaliforniaUSA
| | - Benjamin L Handen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - William E Klunk
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Bradley T Christian
- Departments of Medical Physics and PsychiatryWaisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Wayne Silverman
- Department of PediatricsUniversity of California‐ IrvineIrvineCaliforniaUSA
| | - Ira T Lott
- Department of PediatricsUniversity of California‐ IrvineIrvineCaliforniaUSA
| | - Nicole Schupf
- Taub Institute for Research in Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of EpidemiologyJoseph P. Mailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
- Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
76
|
Alzheimer's Disease Diagnosis Using Misfolding Proteins in Blood. Dement Neurocogn Disord 2020; 19:1-18. [PMID: 32174051 PMCID: PMC7105719 DOI: 10.12779/dnd.2020.19.1.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/02/2019] [Accepted: 12/09/2019] [Indexed: 01/06/2023] Open
Abstract
Alzheimer's disease (AD) is pathologically characterized by a long progressive phase of neuronal changes, including accumulation of extracellular amyloid-β (Aβ) and intracellular neurofibrillary tangles, before the onset of observable symptoms. Many efforts have been made to develop a blood-based diagnostic method for AD by incorporating Aβ and tau as plasma biomarkers. As blood tests have the advantages of being highly accessible and low cost, clinical implementation of AD blood tests would provide preventative screening to presymptomatic individuals, facilitating early identification of AD patients and, thus, treatment development in clinical research. However, the low concentration of AD biomarkers in the plasma has posed difficulties for accurate detection, hindering the development of a reliable blood test. In this review, we introduce three AD blood test technologies emerging in South Korea, which have distinctive methods of heightening detection sensitivity of specific plasma biomarkers. We discuss in detail the multimer detection system, the self-standard analysis of Aβ biomarkers quantified by interdigitated microelectrodes, and a biomarker ratio analysis comprising Aβ and tau.
Collapse
|
77
|
Effects of Fever on 18F-FDG Distribution In Vivo: a Preliminary Study. Mol Imaging Biol 2020; 22:1116-1123. [PMID: 32100227 DOI: 10.1007/s11307-020-01486-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE Elevated body temperature might change glucose metabolism in human organs. The purpose of this study is to explore 18F-FDG distribution in febrile patients on the day of 18F-FDG PET/CT scanning and compare it with patients with a normal temperature. PROCEDURES 18F-FDG PET/CT was performed on 69 febrile patients and 82 patients with a normal temperature. Patient sociodemographic data, blood glucose levels before PET/CT, body temperature on the day of the exam, and laboratory test results were collected. Maximal standard uptake values (SUVmax) in the brain, mediastinal blood pool, liver, spleen, and the bone marrow were compared. RESULTS Compared with the controls, SUVmax of the febrile patients was significantly lower in the brain, mediastinal blood pool, and the liver (p < 0.01), and higher in the spleen and bone marrow (p < 0.01). In the febrile group, SUVmax was not significantly different between the FDG burden and non-FDG burden patients (p > 0.05). Body temperature was found negatively correlated with SUVmax in the brain (r = - 0.646), mediastinal blood pool (r = - 0.530), and the liver (r = - 0.384), and positively correlated with the SUVmax in the spleen (r = 0.592) and bone marrow (r = 0.651). Multivariate linear regression established body temperature on the day of PET/CT as an independent affecting factor (p < 0.01) for the SUVmax in the brain, mediastinal blood pool, liver, spleen, and bone marrow. The SUV in the brain, liver, and mediastinal blood pool remained different (p < 0.05) after corrected with the SUVmax in the blood pool or liver. CONCLUSIONS Fever influences 18F-FDG distribution in multiple human tissues and organs. Altered 18F-FDG distribution in vivo might affect results of disease lesion detection and tumor therapy response assessment. Correction with blood pool or liver SUV fails to cancel the effects of fever. The day of fever should be avoided for PET/CT scan, especially in assessing tumor therapy response.
Collapse
|
78
|
Goud NS, Joshi RK, Bharath RD, Kumar P. Fluorine-18: A radionuclide with diverse range of radiochemistry and synthesis strategies for target based PET diagnosis. Eur J Med Chem 2020; 187:111979. [DOI: 10.1016/j.ejmech.2019.111979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/25/2022]
|
79
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
80
|
Lee I, Na HR, Byun BH, Lim I, Kim BI, Choi CW, Ko IO, Lee KC, Kim KM, Park SY, Kim YK, Lee JY, Bu SH, Kim JH, Kil HS, Park C, Chi DY, Ha JH, Lim SM. Clinical Usefulness of 18F-FC119S Positron-Emission Tomography as an Auxiliary Diagnostic Method for Dementia: An Open-Label, Single-Dose, Evaluator-Blind Clinical Trial. J Clin Neurol 2020; 16:131-139. [PMID: 31942769 PMCID: PMC6974833 DOI: 10.3988/jcn.2020.16.1.131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND PURPOSE The aim of this study was to determine the diagnostic performance and safety of a new ¹⁸F-labeled amyloid tracer, ¹⁸F-FC119S. METHODS This study prospectively recruited 105 participants, comprising 53 with Alzheimer's disease (AD) patients, 16 patients with dementia other than AD (non-AD), and 36 healthy controls (HCs). In the first screening visit, the Seoul Neuropsychological Screening Battery cognitive function test was given to the dementia group, while HC subjects completed the Korean version of the Mini Mental State Examination. Individuals underwent ¹⁸F-FC119S PET, ¹⁸F-fluorodeoxyglucose (FDG) PET, and brain MRI. The diagnostic performance of ¹⁸F-FC119S PET for AD was compared to a historical control (comprising previously reported and currently used amyloid-beta PET agents), ¹⁸F-FDG PET, and MRI. The standardized uptake value (SUV) ratio (ratio of the cerebral cortical SUV to the cerebellar SUV) was measured for each PET data set to provide semiquantitative analysis. All adverse effects during the clinical trial periods were monitored. RESULTS Visual assessments of the ¹⁸F-FC119S PET data revealed a sensitivity of 92% and a specificity of 84% in detecting AD. ¹⁸F-FC119S PET demonstrated equivalent or better diagnostic performance for AD detection than the historical control, ¹⁸F-FDG PET (sensitivity of 80.0% and specificity of 76.0%), and MRI (sensitivity of 98.0% and specificity of 50.0%). The SUV ratios differed significantly between AD patients and the other groups, at 1.44±0.17 (mean±SD) for AD, 1.24±0.09 for non-AD, and 1.21±0.08 for HC. No clinically significant adverse effects occurred during the trial periods. CONCLUSIONS ¹⁸F-FC119S PET provides high sensitivity and specificity in detecting AD and therefore may be considered a useful diagnostic tool for AD.
Collapse
Affiliation(s)
- Inki Lee
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hae Ri Na
- Department of Neurology, Bobath Memorial Hospital, Seongnam, Korea
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Ilhan Lim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Byung Il Kim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Chang Woon Choi
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - In Ok Ko
- Division of Applied RI, Research Institute of Radiological & Medical Sciences, Korea Institutes of Radiological & Medical Sciences, Seoul, Korea
| | - Kyo Chul Lee
- Division of Applied RI, Research Institute of Radiological & Medical Sciences, Korea Institutes of Radiological & Medical Sciences, Seoul, Korea
| | - Kyeong Min Kim
- Division of Applied RI, Research Institute of Radiological & Medical Sciences, Korea Institutes of Radiological & Medical Sciences, Seoul, Korea
| | - Su Yeon Park
- Department of Neurology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Yu Keong Kim
- Department of Nuclear Medicine, Seoul National University College of Medicine & SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Jun Young Lee
- Department of Psychiatry and Behavioural Science, College of Medicine, Seoul National University, Boramae Hospital, Seoul, Korea
| | - Seon Hee Bu
- Department of Neurology, Seoul Bukbu Hospital, Seoul, Korea
| | - Jung Hwa Kim
- Department of Neurology, Seoul Bukbu Hospital, Seoul, Korea
| | - Hee Seup Kil
- Research Institute of Labelling, FutureChem Co., Ltd, Seoul, Korea
| | - Chansoo Park
- Research Institute of Labelling, FutureChem Co., Ltd, Seoul, Korea
| | - Dae Yoon Chi
- Research Institute of Labelling, FutureChem Co., Ltd, Seoul, Korea.,Department of Chemistry, Sogang University, Seoul, Korea
| | - Jeong Ho Ha
- Department of Neurology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.
| | - Sang Moo Lim
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea.
| |
Collapse
|
81
|
Suantio AM, Huang HL, Kwok CSN, Teo DCH, Nguyen MH. FDG-PET in suspected dementia with Lewy bodies: a case report. BMC Geriatr 2019; 19:150. [PMID: 31138141 PMCID: PMC6540462 DOI: 10.1186/s12877-019-1166-3] [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: 10/18/2018] [Accepted: 05/21/2019] [Indexed: 11/20/2022] Open
Abstract
Background Dementia with Lewy bodies (DLB) is still underdiagnosed or mistaken for other types of neurodegenerative diseases. Biomarkers such as 18-Fluorodeoxyglucose Positron Emission Tomography (FDG-PET) can be helpful. Case presentation A 72-year-old gentleman presented with postural hypotension, hallucination, Parkinsonism and recurrent falls. He also had rapidly progressing cognitive impairment. CT and MRI brain showed atrophy of the frontal lobes with preservation of the hippocampi. FDG-PET was suggestive of DLB. He was subsequently treated with Rivastigmine, with significant improvement of his symptoms. Conclusion This case highlights the challenges in diagnosis of an elderly patient with DLB, the use of neuro-imaging as a diagnostic biomarker, the avoidance of the use of antipsychotic and the response to pharmacological treatment with Rivastigmine after a probable diagnosis of DLB.
Collapse
|
82
|
Ferrari BL, Neto GDCC, Nucci MP, Mamani JB, Lacerda SS, Felício AC, Amaro E, Gamarra LF. The accuracy of hippocampal volumetry and glucose metabolism for the diagnosis of patients with suspected Alzheimer's disease, using automatic quantitative clinical tools. Medicine (Baltimore) 2019; 98:e17824. [PMID: 31702636 PMCID: PMC6855664 DOI: 10.1097/md.0000000000017824] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The hippocampus is one of the earliest sites involved in the pathology of Alzheimer's disease (AD). Therefore, we specifically investigated the sensitivity and specificity of hippocampal volume and glucose metabolism in patients being evaluated for AD, using automated quantitative tools (NeuroQuant - magnetic resonance imaging [MRI] and Scenium - positron emission tomography [PET]) and clinical evaluation.This retrospective study included adult patients over the age of 45 years with suspected AD, who had undergone fluorodeoxyglucose positron emission tomography-computed tomography (FDG-PET-CT) and MRI. FDG-PET-CT images were analyzed both qualitatively and quantitatively. In quantitative volumetric MRI analysis, the percentage of the total intracranial volume of each brain region, as well as the total hippocampal volume, were considered in comparison to an age-adjusted percentile. The remaining brain regions were compared between groups according to the final diagnosis.Thirty-eight patients were included in this study. After a mean follow-up period of 23 ± 11 months, the final diagnosis for 16 patients was AD or high-risk mild cognitive impairment (MCI). Out of the 16 patients, 8 patients were women, and the average age of all patients was 69.38 ± 10.98 years. Among the remaining 22 patients enrolled in the study, 14 were women, and the average age was 67.50 ± 11.60 years; a diagnosis of AD was initially excluded, but the patients may have low-risk MCI. Qualitative FDG-PET-CT analysis showed greater accuracy (0.87), sensitivity (0.76), and negative predictive value (0.77), when compared to quantitative PET analysis, hippocampal MRI volumetry, and specificity. The positive predictive value of FDG-PET-CT was similar to the MRI value.The performance of FDG-PET-CT qualitative analysis was significantly more effective compared to MRI volumetry. At least in part, this observation could corroborate the sequential hypothesis of AD pathophysiology, which posits that functional changes (synaptic dysfunction) precede structural changes (atrophy).
Collapse
Affiliation(s)
| | | | - Mariana Penteado Nucci
- LIM44, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
83
|
Oka S, Ogata M, Takano M, Minamimoto R, Hotta M, Tajima T, Nagata N, Tsukada K, Teruya K, Kikuchi Y, Gatanaga H. Non-AIDS-defining malignancies in Japanese hemophiliacs with HIV-1 infection. Glob Health Med 2019; 1:49-54. [PMID: 33330754 DOI: 10.35772/ghm.2019.01015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 11/08/2022]
Abstract
Along improvement of prognosis of HIV-1-infected patients due to successful anti-retroviral therapy, main causes of death in the patients have been changing from AIDS to non-AIDS defining malignancies (NADM) recently. However, little is known about the prevalence and incidence of NADM in patients, and especially in HIV-1-infected hemophiliacs. We prospectively conducted NADM screening with FDG-PET, chest CT, upper gastrointestinal endoscopy, tumor markers, and stool occult blood in hemophiliacs with a mean age of 48.9 years. Screening was done twice from December 2016 through March 2019; the first screening was used to calculate prevalence in 69 patients and the second was used to calculate incidence in 56 patients. The first screening revealed 4 cases of malignancies; three were cases of thyroid cancer and one was a case of a neuroendocrine tumor in the pancreas; prevalence was 5.8% (95% CI: 0.2-11.4%). During a mean follow-up of 1.2 years with 68.2 person-years (PYs), cancer was diagnosed in 2 cases (pancreatic and liver cancer) during the second screening. Incidence was 2.99/100 PY. It can be speculated that there might be around 40 cases of undiagnosed NADM currently and 20 cases of new NADM annually in this population, because 718 HIV-1-infected hemophiliacs are surviving in Japan according to the 2018 Nationwide Survey on Coagulation Disorders. Screening for NADM in HIV-1-infected hemophiliacs at other hospitals is strongly recommended.
Collapse
Affiliation(s)
- Shinichi Oka
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mikiko Ogata
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Misao Takano
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ryogo Minamimoto
- Department of Nuclear Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masatoshi Hotta
- Department of Nuclear Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tsuyoshi Tajima
- Department of Diagnostic Radiology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Naoyoshi Nagata
- Department of Gastroenterology, National Center for Global Health and Medicine, Tokyo, Japan.,Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Kunihisa Tsukada
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsuji Teruya
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yoshimi Kikuchi
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroyuki Gatanaga
- AIDS Clinical Center, National Center for Global Health and Medicine, Tokyo, Japan
| | | |
Collapse
|
84
|
Abstract
Importance Worldwide, 47 million people live with dementia and, by 2050, the number is expected to increase to 131 million. Observations Dementia is an acquired loss of cognition in multiple cognitive domains sufficiently severe to affect social or occupational function. In the United States, Alzheimer disease, one cause of dementia, affects 5.8 million people. Dementia is commonly associated with more than 1 neuropathology, usually Alzheimer disease with cerebrovascular pathology. Diagnosing dementia requires a history evaluating for cognitive decline and impairment in daily activities, with corroboration from a close friend or family member, in addition to a thorough mental status examination by a clinician to delineate impairments in memory, language, attention, visuospatial cognition such as spatial orientation, executive function, and mood. Brief cognitive impairment screening questionnaires can assist in initiating and organizing the cognitive assessment. However, if the assessment is inconclusive (eg, symptoms present, but normal examination findings), neuropsychological testing can help determine whether dementia is present. Physical examination may help identify the etiology of dementia. For example, focal neurologic abnormalities suggest stroke. Brain neuroimaging may demonstrate structural changes including, but not limited to, focal atrophy, infarcts, and tumor, that may not be identified on physical examination. Additional evaluation with cerebrospinal fluid assays or genetic testing may be considered in atypical dementia cases, such as age of onset younger than 65 years, rapid symptom onset, and/or impairment in multiple cognitive domains but not episodic memory. For treatment, patients may benefit from nonpharmacologic approaches, including cognitively engaging activities such as reading, physical exercise such as walking, and socialization such as family gatherings. Pharmacologic approaches can provide modest symptomatic relief. For Alzheimer disease, this includes an acetylcholinesterase inhibitor such as donepezil for mild to severe dementia, and memantine (used alone or as an add-on therapy) for moderate to severe dementia. Rivastigmine can be used to treat symptomatic Parkinson disease dementia. Conclusions and Relevance Alzheimer disease currently affects 5.8 million persons in the United States and is a common cause of dementia, which is usually accompanied by other neuropathology, often cerebrovascular disease such as brain infarcts. Causes of dementia can be diagnosed by medical history, cognitive and physical examination, laboratory testing, and brain imaging. Management should include both nonpharmacologic and pharmacologic approaches, although efficacy of available treatments remains limited.
Collapse
Affiliation(s)
- Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL
- Dept of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Raj C. Shah
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL
- Dept of Family Medicine, Rush University Medical Center, Chicago, IL
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL
- Dept of Neurological Sciences, Rush University Medical Center, Chicago, IL
| |
Collapse
|
85
|
Abdul Aziz SA, Jia Ling L, Ahmad Saad FF, Nordin AJ, Ibrahim N, Nuruddin A, Tunan E, Rosalina, Saripan MI, Suppiah S. Voxel-wise analysis of 18F-fluorodeoxyglucose metabolism in correlation with variations in the presentation of Alzheimer’s disease: a clinician’s guide. MEDICAL JOURNAL OF INDONESIA 2019. [DOI: 10.13181/mji.v28i3.2770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND Diagnostic imaging can be applied in the management of Alzheimer’s disease as it provides structural and functional information to exclude possible secondary causes and offers additional information, especially in atypical cases of Alzheimer’s disease. The utility of positron emission tomography/computed tomography (PET/CT) can help in the noninvasive diagnosis of Alzheimer’s disease by voxel-wise quantification of cerebral 18F-fluorodeoxyglucose (FDG) metabolism.
METHODS This prospective study was conducted among 10 subjects with Alzheimer’s disease and 10 healthy control subjects who underwent neuropsychological testing and 18F-FDG PET/CT scans. Images of the brain were postprocessed using voxel-wise analysis and segmented into 20 regions of interest. The standardized uptake value (SUV)max/SUVmean/standard deviation of SUVmean results were analyzed accordingly and correlated with the subjects’ Montreal cognitive assessment (MoCA) results that were adjusted for age and education level.
RESULTS Hypometabolism at the right parietal lobe significantly correlated with increasing age and lower MoCA scores. Global hypometabolism was observed in subjects who had advanced Alzheimer’s disease but preserved primary somatosensory cortices (S1) region metabolism. Predominance of frontal lobe hypometabolism was a feature of subjects with Alzheimer’s disease having associated depressive symptoms.
CONCLUSIONS 18F-FDG PET/CT voxel-wise analysis can be used for quantitative assessment and can assist clinicians in the diagnosis of Alzheimer’s disease and other variations of the disease spectrum.
Collapse
|
86
|
Dumba M, Khan S, Patel N, Perry L, Malhotra P, Perry R, Nijran K, Barwick T, Wallitt K, Win Z. Clinical 18F-FDG and amyloid brain positron emission tomography/CT in the investigation of cognitive impairment: where are we now? Br J Radiol 2019; 92:20181027. [PMID: 30982329 PMCID: PMC6732924 DOI: 10.1259/bjr.20181027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/07/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The number of people living with dementia is increasing, but as yet there remains no cure or disease-modifying treatment. This review aims to help readers understand the role of 18F-FDG PET/CT imaging in the investigation of cognitive impairment and how the advent of amyloid PET/CT imaging may hold the key to radically changing management of the most common form of dementia - Alzheimer's disease. The indications for 18F-FDG PET/CT and amyloid PET/CT imaging in cognitive impairment are outlined. Additionally, the mechanisms of action, technique, patient preparation and acquisition parameters for both are detailed. We conclude by providing a framework for interpreting 18F-FDG PET/CT and amyloid PET/CT imaging in the more common conditions that lead to cognitive impairment conditions with tips on avoiding pitfalls in interpretation.
Collapse
Affiliation(s)
- Maureen Dumba
- Department of Neuroradiology, Imperial College Healthcare NHS Trust, London UK
| | - Sairah Khan
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
| | - Neva Patel
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
- Radiological Sciences Unit, Imperial College Healthcare NHS Trust, London UK
| | - Laura Perry
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
- Radiological Sciences Unit, Imperial College Healthcare NHS Trust, London UK
| | - Paresh Malhotra
- Division of Brain Sciences, Imperial College, London UK
- Department of Neurology, Imperial College Healthcare NHS Trust, London UK
| | - Richard Perry
- Division of Brain Sciences, Imperial College, London UK
- Department of Neurology, Imperial College Healthcare NHS Trust, London UK
| | - Kuldip Nijran
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
- Radiological Sciences Unit, Imperial College Healthcare NHS Trust, London UK
| | - Tara Barwick
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
| | - Kathryn Wallitt
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
| | - Zarni Win
- Department of Nuclear Medicine, Imperial College Healthcare NHS Trust, London UK
| |
Collapse
|
87
|
Gross TJ, Doran E, Cheema AK, Head E, Lott IT, Mapstone M. Plasma metabolites related to cellular energy metabolism are altered in adults with Down syndrome and Alzheimer's disease. Dev Neurobiol 2019; 79:622-638. [PMID: 31419370 DOI: 10.1002/dneu.22716] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/02/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
Down syndrome (DS) is a well-known neurodevelopmental disorder most commonly caused by trisomy of chromosome 21. Because individuals with DS almost universally develop heavy amyloid burden and Alzheimer's disease (AD), biomarker discovery in this population may be extremely fruitful. Moreover, any AD biomarker in DS that does not directly involve amyloid pathology may be of high value for understanding broader mechanisms of AD generalizable to the neurotypical population. In this retrospective biomarker discovery study, we examined banked peripheral plasma samples from 78 individuals with DS who met clinical criteria for AD at the time of the blood draw (DS-AD) and 68 individuals with DS who did not (DS-NAD). We measured the relative abundance of approximately 5,000 putative features in the plasma using untargeted mass spectrometry (MS). We found significantly higher levels of a peak putatively annotated as lactic acid in the DS-AD group (q = .014), a finding confirmed using targeted MS (q = .011). Because lactate is the terminal product of glycolysis and subsequent lactic acid fermentation, we performed additional targeted MS focusing on central carbon metabolism which revealed significantly increased levels of pyruvic (q = .03) and methyladipic (q = .03) acids in addition to significantly lower levels of uridine (q = .007) in the DS-AD group. These data suggest that AD in DS is accompanied by a shift from aerobic respiration toward the less efficient fermentative metabolism and that bioenergetically derived metabolites observable in peripheral blood may be useful for detecting this shift.
Collapse
Affiliation(s)
- Thomas J Gross
- Department of Neurology, The University of California, Irvine, Irvine, California
| | - Eric Doran
- Department of Pediatrics, The University of California, Irvine, Irvine, California
| | - Amrita K Cheema
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, The University of California, Irvine, Irvine, California
| | - Ira T Lott
- Department of Pediatrics, The University of California, Irvine, Irvine, California
| | - Mark Mapstone
- Department of Neurology, The University of California, Irvine, Irvine, California
| |
Collapse
|
88
|
Eguchi A, Kimura N, Aso Y, Yabuuchi K, Ishibashi M, Hori D, Sasaki Y, Nakamichi A, Uesugi S, Jikumaru M, Sumi K, Shimomura T, Matsubara E. Relationship Between the Japanese Version of the Montreal Cognitive Assessment and PET Imaging in Subjects with Mild Cognitive Impairment. Curr Alzheimer Res 2019; 16:852-860. [PMID: 31385770 DOI: 10.2174/1567205016666190805155230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/28/2019] [Accepted: 07/23/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND The Montreal Cognitive Assessment (MoCA) test has high sensitivity and specificity for detecting mild cognitive impairment or early dementia. How the MoCA score relates to findings of positron emission tomography imaging, however, remains unclear. OBJECTIVE This prospective study examined the relationship between the Japanese version of the MoCA (MoCA-J) test and brain amyloid deposition or cerebral glucose metabolism among subjects with mild cognitive impairment. METHODS A total of 125 subjects with mild cognitive impairment underwent the MoCA-J test, and amyloid- and 18F-fluorodeoxyglucose- positron emission tomography. Linear correlation analysis and multiple linear regression analysis were conducted to investigate the relationship between the MoCA-J score and demographic characteristics, amyloid deposition, and cerebral glucose metabolism. Moreover, Statistical Parametric Mapping 8 was used for a voxel-wise regression analysis of the MoCA-J score and cerebral glucose metabolism. RESULTS The MoCA-J score significantly correlated with age, years of education, and the Mini-Mental State Examination score. After adjusting for age, sex, and education, the MoCA-J score significantly correlated negatively with amyloid retention (β= -0.174, p= 0.031) and positively with cerebral glucose metabolism (β= 0.183, p= 0.044). Statistical Parametric Mapping showed that Japanese version of MoCA score correlated with glucose metabolism in the bilateral frontal and parietal lobes, and the left precuneus. CONCLUSION The total MoCA-J score correlated with amyloid deposition and frontal and parietal glucose metabolism in subjects with mild cognitive impairment. Our findings support the usefulness of the MoCA-J test for screening subjects at high risk for Alzheimer's disease.
Collapse
Affiliation(s)
- Atsuko Eguchi
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Noriyuki Kimura
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Yasuhiro Aso
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Kenichi Yabuuchi
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Masato Ishibashi
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Daiji Hori
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Yuuki Sasaki
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Atsuhito Nakamichi
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Souhei Uesugi
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Mika Jikumaru
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Kaori Sumi
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| | - Tsuyoshi Shimomura
- Department of Neurosurgery, Oita University, Faculty of Medicine, Yufu, Oita 879-5593, Japan
| | - Etsuro Matsubara
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan
| |
Collapse
|
89
|
Altered Brain Glucose Metabolism Assessed by 18F-FDG PET Imaging Is Associated with the Cognitive Impairment of CADASIL. Neuroscience 2019; 417:35-44. [PMID: 31394195 DOI: 10.1016/j.neuroscience.2019.07.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/26/2022]
Abstract
Recurrent stroke and cognitive impairment are the primary features of patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). The cognitive deficits in these patients are known to be correlated with structural brain changes, such as white matter lesions and lacunae, and resting-state functional connectivity in brain networks. However, the associations between changes in brain glucose metabolism based on 18F-2-fluoro-2-deoxy-D-glucose (18F-FDG) positron emission tomography (PET) imaging and cognitive scores in CADASIL patients remain unclear. In the present study, 24 CADASIL patients and 24 matched healthy controls underwent 18F-FDG PET imaging. Brain glucose metabolism was measured in all subjects and Pearson's correlation analyses were performed to evaluate relationships between abnormal glucose metabolism in various brain areas and cognitive scores. Compared to controls, CADASIL patients exhibited significantly lower metabolism in the right cerebellar posterior lobe, left cerebellar anterior lobe, bilateral thalamus and left limbic lobe. Additionally, hypermetabolism was observed in the left precentral and postcentral gyri. Importantly, glucose metabolism in the left limbic lobe was positively associated with cognitive scores on the Mini-Mental State Examination (MMSE). Furthermore, glucose metabolism in the left precentral gyri was negatively correlated with cognitive scores on the Montreal Cognitive Assessment (MoCA). The present findings provide strong support for the presence of altered brain glucose metabolism in CADASIL patients as well as the associations between abnormal metabolism and cognitive scales in this population. The present findings suggest that patterns of brain glucose metabolism may become useful markers of cognitive impairment in CADASIL patients.
Collapse
|
90
|
Mukku SSR, Sivakumar PT, Nagaraj C, Mangalore S, Harbishettar V, Varghese M. Clinical utility of 18F-FDG-PET/MRI brain in dementia: Preliminary experience from a geriatric clinic in South India. Asian J Psychiatr 2019; 44:99-105. [PMID: 31336358 DOI: 10.1016/j.ajp.2019.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND 18F-FDG-PET is a potential sensitive biomarker indicating neuronal damage. 18F-FDG-PET has proven to be useful in subtyping dementia. Utility of simultaneous 18F-FDG-PET and MRI-brain was investigated in the evaluation of dementia in this facility. METHOD All case notes of patients who underwent 18 F-FDG-PET/MRI brain attending the Geriatric Clinic for 18 month period between January 2017 and June 2018 were retrospectively reviewed. Their socio-demographic details, MRI-brain finding, 18F- FDG-PET findings and comorbid illnesses were studied. RESULTS A total of 21 patients underwent 18F-FDG-PET/MRI brain during study period. The mean age was 61.23, SD-8.6 years (range: 36-75 years). Among them 5 (23.8%) had Mild Cognitive Impairment (MCI) and 16 (76.2%) had dementia. Majority of patients had early onset cognitive decline (76.2%). Based on the pattern of hypometabolism, the MCI group had one patient each indicative of AD, Semantic-Frontotemporal dementia (Semantic-FTD), mixed Alzheimer's dementia (AD + FTD) and two patients had patterns suggestive of Behaviour Variant of FTD (Bv-FTD). In Dementia group the pattern of hypometabolism was indicative of Bv-FTD in seven, AD in four, Posterior Cortical Atrophy (PCA) in one, Semantic-FTD in one, Mixed AD-Diffuse Lewy Body Dementia (DLBD) in one and no specific pattern in two patients. MRI and 18 F-FDG-PET brain had concordance in 9 (56.26%) patients. DISCUSSION 18F-FDG-PET/MRI helped in overall clinical diagnosis and management in 19 (90.5%) patients especially with early onset dementia. In MCI group it indicated underlying aetiology and in dementia group it helped in subtyping. CONCLUSION The study supports the role of 18F-FDG-PET/MRI as an emerging diagnostic tool to assist in dementia evaluation in India.
Collapse
Affiliation(s)
- Shiva Shanker Reddy Mukku
- Geriatric Clinic & Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India.
| | - Palanimuthu Thangaraju Sivakumar
- Geriatric Clinic & Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India.
| | - Chandana Nagaraj
- Department of Neuroimaging and interventional Radiology, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India.
| | - Sandhya Mangalore
- Department of Neuroimaging and interventional Radiology, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India.
| | - Vijaykumar Harbishettar
- Geriatric Clinic & Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India.
| | - Mathew Varghese
- Geriatric Clinic & Services, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bangalore, 560029, India.
| |
Collapse
|
91
|
Hrubešová K, Fousková M, Habartová L, Fišar Z, Jirák R, Raboch J, Setnička V. Search for biomarkers of Alzheimer's disease: Recent insights, current challenges and future prospects. Clin Biochem 2019; 72:39-51. [PMID: 30953619 DOI: 10.1016/j.clinbiochem.2019.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
Due to the trend of prolonged lifespan leading to higher incidence of age-related diseases, the demand for reliable biomarkers of dementia rises. In this review, we present novel biomarkers of high potential, especially those found in blood, urine or saliva, which could lead to a more comfortable patient experience and better time- and cost-effectivity, compared to the currently used diagnostic methods. We focus on biomarkers that might allow for the detection of Alzheimer's disease before its clinical manifestations. Such biomarkers might be helpful for better understanding the etiology of the disease and identifying its risk factors. Moreover, it could be a base for developing new treatment or at least help to prolong the presymptomatic stage in patients suffering from Alzheimer's disease. As potential candidates, we present, for instance, neurofilament light in both cerebrospinal fluid and blood plasma or amyloid β in plasma. Above all, we provide an overview of different approaches to the diagnostics, analyzing patient's biofluids as a whole using molecular spectroscopy. Infrared and Raman spectroscopy and especially chiroptical methods provide information not only on the chemical composition, but also on molecular structure. Therefore, these techniques are promising for the diagnostics of Alzheimer's disease, as the accumulation of amyloid β in abnormal conformation is one of the hallmarks of this disease.
Collapse
Affiliation(s)
- Kateřina Hrubešová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Markéta Fousková
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Ke Karlovu 11, 120 00 Prague 2, Czech Republic
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
92
|
Viglianti BL, Wale DJ, Ma T, Johnson TD, Bohnen NI, Wong KK, Ky C, Frey KA, Townsend DM, Rubello D, Gross MD. Effects of plasma glucose levels on regional cerebral 18F-fluorodeoxyglucose uptake: Implications for dementia evaluation with brain PET imaging. Biomed Pharmacother 2019; 112:108628. [PMID: 30784923 PMCID: PMC6714976 DOI: 10.1016/j.biopha.2019.108628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 11/19/2022] Open
Abstract
Purpose: Hyperglycemia affects FDG uptake in the brain, potentially emulating
Alzheimer’s disease in normal individuals. This study investigates
global and regional cerebral FDG uptake as a function of plasma glucose in a
cohort of patients. Methods: 120 consecutive male patients with FDG PET/CT for initial oncologic
staging (July-Dee 2015) were reviewed. Patients with dementia,
cerebrovascular accident, structural brain lesion, prior oncology treatment
or high metabolic tumor burden (recently shown affecting brain FDG uptake)
were excluded. 53 (24 nondiabetic) eligible patients (age 65.7 ± 2.8
mean ± SE) were analyzed with parametric computer software,
MIMneuro™. Regional Z-scores were evaluated as a function of plasma
glucose and age using multi variable linear mixed effects models with false
discovery analysis adjusting for multiple comparisons. If the regression
slope was significantly (p < 0.05) different than zero, hyperglycemia
effect was present. Results: There was a negative inverse relationship (p < 0.001) between
global brain FDG uptake and hyperglycemia. No regional hyperglycemia effect
on uptake were present when subjects were normalized using pons or
cerebellum. However, regional hyperglycemia effects were seen (p <
0.047–0.001) when normalizing by the whole brain. No obvious pattern
was seen in the regions affected. Age had a significant effect using whole
brain normalization (p < 0.04–0.01). Conclusions: Cortical variation in FDG uptake were identified when subjects were
hyperglycemic. However, these variations didn’t fit a particular
pattern of dementia and the severity of the affect is not likely to alter
clinical interpretation.
Collapse
Affiliation(s)
- Benjamin L Viglianti
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Nuclear Medicine Service, Department of Veterans Affairs Healthcare System, Ann Arbor, MI, USA.
| | - Daniel J Wale
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Nuclear Medicine Service, Department of Veterans Affairs Healthcare System, Ann Arbor, MI, USA
| | - Tianwen Ma
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Timothy D Johnson
- Department of Biostatistics, University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Nicolaas I Bohnen
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Nuclear Medicine Service, Department of Veterans Affairs Healthcare System, Ann Arbor, MI, USA
| | - Ka Kit Wong
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Christy Ky
- University of Michigan School of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kirk A Frey
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Danyelle M Townsend
- Department of Physiology, Division of New Drugs Development, University of Southern Carolina, USA
| | - Domenico Rubello
- Department of Nuclear Medicine, Santa Maria della Misericordia Hospital, Rovigo, Italy.
| | - Milton D Gross
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA; Nuclear Medicine Service, Department of Veterans Affairs Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
93
|
Ding Y, Sohn JH, Kawczynski MG, Trivedi H, Harnish R, Jenkins NW, Lituiev D, Copeland TP, Aboian MS, Mari Aparici C, Behr SC, Flavell RR, Huang SY, Zalocusky KA, Nardo L, Seo Y, Hawkins RA, Hernandez Pampaloni M, Hadley D, Franc BL. A Deep Learning Model to Predict a Diagnosis of Alzheimer Disease by Using 18F-FDG PET of the Brain. Radiology 2019; 290:456-464. [PMID: 30398430 PMCID: PMC6358051 DOI: 10.1148/radiol.2018180958] [Citation(s) in RCA: 270] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/24/2018] [Accepted: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Purpose To develop and validate a deep learning algorithm that predicts the final diagnosis of Alzheimer disease (AD), mild cognitive impairment, or neither at fluorine 18 (18F) fluorodeoxyglucose (FDG) PET of the brain and compare its performance to that of radiologic readers. Materials and Methods Prospective 18F-FDG PET brain images from the Alzheimer's Disease Neuroimaging Initiative (ADNI) (2109 imaging studies from 2005 to 2017, 1002 patients) and retrospective independent test set (40 imaging studies from 2006 to 2016, 40 patients) were collected. Final clinical diagnosis at follow-up was recorded. Convolutional neural network of InceptionV3 architecture was trained on 90% of ADNI data set and tested on the remaining 10%, as well as the independent test set, with performance compared to radiologic readers. Model was analyzed with sensitivity, specificity, receiver operating characteristic (ROC), saliency map, and t-distributed stochastic neighbor embedding. Results The algorithm achieved area under the ROC curve of 0.98 (95% confidence interval: 0.94, 1.00) when evaluated on predicting the final clinical diagnosis of AD in the independent test set (82% specificity at 100% sensitivity), an average of 75.8 months prior to the final diagnosis, which in ROC space outperformed reader performance (57% [four of seven] sensitivity, 91% [30 of 33] specificity; P < .05). Saliency map demonstrated attention to known areas of interest but with focus on the entire brain. Conclusion By using fluorine 18 fluorodeoxyglucose PET of the brain, a deep learning algorithm developed for early prediction of Alzheimer disease achieved 82% specificity at 100% sensitivity, an average of 75.8 months prior to the final diagnosis. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Larvie in this issue.
Collapse
Affiliation(s)
- Yiming Ding
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Jae Ho Sohn
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Michael G. Kawczynski
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Hari Trivedi
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Roy Harnish
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Nathaniel W. Jenkins
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Dmytro Lituiev
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Timothy P. Copeland
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Mariam S. Aboian
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Carina Mari Aparici
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Spencer C. Behr
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Robert R. Flavell
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Shih-Ying Huang
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Kelly A. Zalocusky
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Lorenzo Nardo
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Youngho Seo
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Randall A. Hawkins
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Miguel Hernandez Pampaloni
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Dexter Hadley
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Benjamin L. Franc
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| |
Collapse
|
94
|
Floeter MK, Gendron TF. Biomarkers for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia Associated With Hexanucleotide Expansion Mutations in C9orf72. Front Neurol 2018; 9:1063. [PMID: 30568632 PMCID: PMC6289985 DOI: 10.3389/fneur.2018.01063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022] Open
Abstract
Now that genetic testing can identify persons at risk for developing amyotrophic lateral sclerosis (ALS) many decades before symptoms begin, there is a critical need for biomarkers that signal the onset and progression of degeneration. The search for candidate disease biomarkers in patients with mutations in the gene C9orf72 has included imaging, physiology, and biofluid measurements. In cross-sectional imaging studies, C9+ ALS patients display diffuse reductions of gray and white matter integrity compared to ALS patients without mutations. This structural imaging signature overlaps with frontotemporal dementia (FTD), reflecting the frequent co-occurrence of cognitive impairment, even frank FTD, in C9+ ALS patients. Changes in functional connectivity occur as critical components of the networks associated with cognition and behavior degenerate. In presymptomatic C9+carriers, subtle differences in volumes of subcortical structures and functional connectivity can be detected, often decades before the typical family age of symptom onset. Dipeptide repeat proteins produced by the repeat expansion mutation are also measurable in the cerebrospinal fluid (CSF) of presymptomatic gene carriers, possibly throughout their lives. In contrast, a rise in the level of neurofilament proteins in the CSF appears to presage the onset of degeneration in presymptomatic carriers in one longitudinal study. Cross-sectional studies indicate that neurofilament protein levels may provide prognostic information for survival in C9+ ALS patients. Longitudinal studies will be needed to validate the candidate biomarkers discussed here. Understanding how these candidate biomarkers change over time is critical if they are to be used in future therapeutic decisions.
Collapse
Affiliation(s)
- Mary Kay Floeter
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
95
|
Wei L, Guo K, Li Y, Guo Z, Gao C, Yuan M, Zhang M. Construction of a novel Chinese normal brain database using 18F-FDG PET images and MIMneuro software, the initial application in epilepsy. Int J Neurosci 2018; 129:417-422. [PMID: 30375250 DOI: 10.1080/00207454.2018.1538138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE To create a standard Western Chinese normal functional brain database for quantitative analysis using 2-deoxy-2-[18F] fluoro-d-glucose (18F-FDG) positron emission tomography (PET) images and MIMneuro software. METHODS 78 healthy right-handed Chinese volunteers from Tangdu Hospital were scanned using 18F-FDG PET to evaluate brain metabolism between March and October 2016. All PET images were processed using MIMneuro software to create a normal database platform. The platform included anatomical optimization to facilitate spatial localization of abnormalities and a statistical comparison with normal cases utilizing the Z-scores, which represent the number of standard deviations from the mean of the normal controls in the database. RESULTS The novel Chinese brain metabolism database platform including 78 healthy volunteers (male: female 40:38; age 3-78 years, mean age, 45 years) was constructed based on the MIMneuro software, which increased the diagnostic confidence in the test patient by quantifying and emphasizing the abnormality. The BrainAlignTM deformation algorithm of MIMneuro matched the size, shape, and orientation of the patient's brain scan to a template brain for comparison against a database of normal controls. The quantitative analysis performed on a voxel and regional level was useful in assessing the areas of abnormalities. CONCLUSIONS A novel Chinese 18F-FDG PET-based normal brain function database was created to highlight the local regions of abnormal metabolic activity through quantitative comparisons against the normal database. The Z-scores obtained by MIMneuro potentially aid in visualizing and quantifying the subtle lesions on 18FDG-PET scan images as observed in a patient diagnosed with epilepsy.
Collapse
Affiliation(s)
- Longxiao Wei
- a Department of Radiology , The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shanxi , China
| | - Kun Guo
- b Department of Nuclear Medicine , The second affiliated Hospital of Air Force Medical University , Xi'an, Shanxi , China
| | - Yunbo Li
- b Department of Nuclear Medicine , The second affiliated Hospital of Air Force Medical University , Xi'an, Shanxi , China
| | - Zhirui Guo
- b Department of Nuclear Medicine , The second affiliated Hospital of Air Force Medical University , Xi'an, Shanxi , China
| | - Chengcheng Gao
- b Department of Nuclear Medicine , The second affiliated Hospital of Air Force Medical University , Xi'an, Shanxi , China
| | - Menghui Yuan
- b Department of Nuclear Medicine , The second affiliated Hospital of Air Force Medical University , Xi'an, Shanxi , China
| | - Ming Zhang
- a Department of Radiology , The First Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shanxi , China
| |
Collapse
|
96
|
Rajkumar R, Farrher E, Mauler J, Sripad P, Régio Brambilla C, Rota Kops E, Scheins J, Dammers J, Lerche C, Langen KJ, Herzog H, Biswal B, Shah NJ, Neuner I. Comparison of EEG microstates with resting state fMRI and FDG-PET measures in the default mode network via simultaneously recorded trimodal (PET/MR/EEG) data. Hum Brain Mapp 2018; 42:4122-4133. [PMID: 30367727 DOI: 10.1002/hbm.24429] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022] Open
Abstract
Simultaneous trimodal positron emission tomography/magnetic resonance imaging/electroencephalography (PET/MRI/EEG) resting state (rs) brain data were acquired from 10 healthy male volunteers. The rs-functional MRI (fMRI) metrics, such as regional homogeneity (ReHo), degree centrality (DC) and fractional amplitude of low-frequency fluctuations (fALFFs), as well as 2-[18F]fluoro-2-desoxy-d-glucose (FDG)-PET standardised uptake value (SUV), were calculated and the measures were extracted from the default mode network (DMN) regions of the brain. Similarly, four microstates for each subject, showing the diverse functional states of the whole brain via topographical variations due to global field power (GFP), were estimated from artefact-corrected EEG signals. In this exploratory analysis, the GFP of microstates was nonparametrically compared to rs-fMRI metrics and FDG-PET SUV measured in the DMN of the brain. The rs-fMRI metrics (ReHO, fALFF) and FDG-PET SUV did not show any significant correlations with any of the microstates. The DC metric showed a significant positive correlation with microstate C (rs = 0.73, p = .01). FDG-PET SUVs indicate a trend for a negative correlation with microstates A, B and C. The positive correlation of microstate C with DC metrics suggests a functional relationship between cortical hubs in the frontal and occipital lobes. The results of this study suggest further exploration of this method in a larger sample and in patients with neuropsychiatric disorders. The aim of this exploratory pilot study is to lay the foundation for the development of such multimodal measures to be applied as biomarkers for diagnosis, disease staging, treatment response and monitoring of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ravichandran Rajkumar
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Ezequiel Farrher
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Jörg Mauler
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Praveen Sripad
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Cláudia Régio Brambilla
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Elena Rota Kops
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Jürgen Scheins
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Jürgen Dammers
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Christoph Lerche
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.,Department of Nuclear Medicine, RWTH Aachen University, Aachen, Germany
| | - Hans Herzog
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Bharat Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey
| | - N Jon Shah
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany.,Institute of Neuroscience and Medicine 11, INM-11, Forschungszentrum Jülich, Jülich, Germany.,Department of Neurology, RWTH Aachen University, Aachen, Germany.,Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| | - Irene Neuner
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany
| |
Collapse
|
97
|
Heckmann JG, Niedermeier W, Büchner M, Scher B. Distinctive FDG-PET/CT Findings in Acute Neurological Hospital Care. Neurohospitalist 2018; 9:93-99. [PMID: 30915187 DOI: 10.1177/1941874418805339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A compilation of 6 distinctive 18F-fluorodeoxyglucose positron emission tomography (PET) combined with computed tomography (CT) findings in the acute setting of neurohospital care is presented. In case 1, PET/CT allowed the final diagnosis of circumscribed ischemic infarction by demonstrating a clear pattern of luxury perfusion. In case 2, diagnosis of thalamic abscess was made, whereby PET/CT demonstrated an empty zone. Hypermetabolic enlarged hilar lymph nodes and hypermetabolic spinal lumbar roots in PET/CT led to the diagnosis of neurosarcoidosis in case 3. In case 4, a hypermetabolic brain focus in PET/CT identified the seizure focus in epilepsia partialis continua. A cerebral hemispheric hypometabolism in PET/CT in case 5 supported the diagnosis of Creutzfeldt-Jakob disease, which initially mimicked acute stroke. In case 6, PET/CT detected infective endocarditis as a source of multiple cerebral ischemic lesions. In conclusion, PET/CT can contribute importantly to find the correct diagnosis in acute neurohospital patients.
Collapse
Affiliation(s)
| | - Wolfgang Niedermeier
- Department of Radiology and Nuclear Medicine, Municipal Hospital Waid, Zurich, Switzerland
| | - Markus Büchner
- Department of Nuclear Medicine, Municipal Hospital Landshut, Germany
| | - Bernhard Scher
- Department of Nuclear Medicine, Municipal Hospital Landshut, Germany
| |
Collapse
|
98
|
Duignan J, Healy GM, Hughes NM, Redmond CE, McGuigan C, Beausang A, Killeen RP. FDG-PET diagnoses of sporadic Creutzfeldt-Jakob disease: radiology-pathology correlation. QJM 2018; 111:669-670. [PMID: 29617881 DOI: 10.1093/qjmed/hcy071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- J Duignan
- Department of Radiology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - G M Healy
- Department of Radiology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - N M Hughes
- Department of Radiology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - C E Redmond
- Department of Radiology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - C McGuigan
- Department of Neurology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| | - A Beausang
- Department of Neuropathology, Beaumont Hospital, Beaumont Road, Dublin 9, Ireland
| | - R P Killeen
- Department of Radiology, St Vincent's University Hospital, Elm Park, Dublin 4, Ireland
| |
Collapse
|
99
|
Chen Z, Jamadar SD, Li S, Sforazzini F, Baran J, Ferris N, Shah NJ, Egan GF. From simultaneous to synergistic MR-PET brain imaging: A review of hybrid MR-PET imaging methodologies. Hum Brain Mapp 2018; 39:5126-5144. [PMID: 30076750 DOI: 10.1002/hbm.24314] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022] Open
Abstract
Simultaneous Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET) scanning is a recent major development in biomedical imaging. The full integration of the PET detector ring and electronics within the MR system has been a technologically challenging design to develop but provides capacity for simultaneous imaging and the potential for new diagnostic and research capability. This article reviews state-of-the-art MR-PET hardware and software, and discusses future developments focusing on neuroimaging methodologies for MR-PET scanning. We particularly focus on the methodologies that lead to an improved synergy between MRI and PET, including optimal data acquisition, PET attenuation and motion correction, and joint image reconstruction and processing methods based on the underlying complementary and mutual information. We further review the current and potential future applications of simultaneous MR-PET in both systems neuroscience and clinical neuroimaging research. We demonstrate a simultaneous data acquisition protocol to highlight new applications of MR-PET neuroimaging research studies.
Collapse
Affiliation(s)
- Zhaolin Chen
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Clayton, Victoria, Australia
| | - Sharna D Jamadar
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| | - Shenpeng Li
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Clayton, Victoria, Australia
| | | | - Jakub Baran
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Department of Biophysics, Faculty of Mathematics and Natural Sciences, University of Rzeszów, Rzeszów, Poland
| | - Nicholas Ferris
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Imaging, Monash Health, Clayton, Victoria, Australia
| | - Nadim Jon Shah
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum, Jülich, Germany
| | - Gary F Egan
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia.,Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Clayton, Victoria, Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
100
|
Sarikaya I, Sarikaya A, Elgazzar AH. Current Status of 18F-FDG PET Brain Imaging in Patients with Dementia. J Nucl Med Technol 2018; 46:362-367. [DOI: 10.2967/jnmt.118.210237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/07/2018] [Indexed: 11/16/2022] Open
|