51
|
Villemagne VL, Chételat G. Neuroimaging biomarkers in Alzheimer's disease and other dementias. Ageing Res Rev 2016; 30:4-16. [PMID: 26827785 DOI: 10.1016/j.arr.2016.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/16/2022]
Abstract
In vivo imaging of β-amyloid (Aβ) has transformed the assessment of Aβ pathology and its changes over time, extending our insight into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional or global Aβ burden in the brain. This knowledge is essential for therapeutic trial recruitment and for the evaluation of anti-Aβ treatments. Although cross sectional evaluation of Aβ burden does not strongly correlate with cognitive impairment, it does correlate with cognitive (especially memory) decline and with a higher risk for conversion to AD in the aging population and MCI subjects. This suggests that Aβ deposition is a protracted pathological process starting well before the onset of symptoms. Longitudinal observations, coupled with different disease-specific biomarkers to assess potential downstream effects of Aβ are required to confirm this hypothesis and further elucidate the role of Aβ deposition in the course of Alzheimer's disease.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Molecular Imaging & Therapy, Centre for PET, Austin Health, Victoria 3084, Australia; Department of Medicine, University of Melbourne, Austin Health, Victoria 3084, Australia; The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia; Institut National de la Santé et de la Recherche Médicale (Inserm), Unité, 1077 Caen, France.
| | - Gaël Chételat
- The Florey Institute of Neuroscience and Mental Health, Victoria 3052, Australia; Institut National de la Santé et de la Recherche Médicale (Inserm), Unité, 1077 Caen, France; Université de Caen Basse-Normandie, Unité Mixte de Recherche (UMR), S1077 Caen, France; Ecole Pratique des Hautes Etudes, UMR-S1077, 14000 Caen, France; Unité 1077, Centre Hospitalier Universitaire de Caen, 14000 Caen, France
| |
Collapse
|
52
|
Witte AV, Köbe T, Graunke A, Schuchardt JP, Hahn A, Tesky VA, Pantel J, Flöel A. Impact of leptin on memory function and hippocampal structure in mild cognitive impairment. Hum Brain Mapp 2016; 37:4539-4549. [PMID: 27511061 DOI: 10.1002/hbm.23327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 06/08/2016] [Accepted: 07/18/2016] [Indexed: 12/12/2022] Open
Abstract
Metabolic changes have been suggested to contribute to dementia and its precursor mild cognitive impairment (MCI), yet previous results particularly for the "satiety hormone" leptin are mixed. Therefore, we aimed to determine if MCI patients show systematic differences in leptin, independent of sex, adipose mass, age, and glucose and lipid metabolism, and whether leptin levels correlated with memory performance and hippocampal integrity. Forty MCI patients (20 females, aged 67 years ± 7 SD) were compared to 40 healthy controls (HC) that were pair-wise matched for sex, age, and body fat. Memory performance was assessed using the auditory verbal learning test. Volume and microstructure of the hippocampus were determined using 3T-neuroimaging. Fasting serum markers of leptin, glucose and lipid metabolism, and other confounding factors were assayed. MCI patients, compared with HC, showed lower serum leptin, independent of sex, age, and body fat (P < 0.001). Glucose and lipid markers did not attenuate these results. Moreover, MCI patients exhibited poorer memory and lower volume and microstructural integrity within hippocampal subfields. While leptin and memory were not significantly correlated, mediation analyses indicated that lower leptin contributed to poorer memory through its negative effect on right hippocampus volume and left hippocampus microstructure. We demonstrated that MCI is associated with lower serum leptin independent of sex, age, body fat, glucose, and lipid metabolism. Our data further suggest that inefficient leptin signaling could partly contribute to decreases in memory performance through changes in hippocampus structure, a hypothesis that should now be verified in longitudinal studies. Hum Brain Mapp 37:4539-4549, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- A Veronica Witte
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Max-Planck-Institute of Human Cognitive and Brain Sciences, Stephanstraße 1A, 04103, Leipzig, Germany
| | - Theresa Köbe
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Anders Graunke
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jan Philipp Schuchardt
- Department of Nutrition Physiology and Human Nutrition, Gottfried Wilhelm Leibniz University, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Andreas Hahn
- Department of Nutrition Physiology and Human Nutrition, Gottfried Wilhelm Leibniz University, Am Kleinen Felde 30, 30167, Hannover, Germany
| | - Valentina A Tesky
- Institute of General Practice, Goethe-University, Frankfurt Am Main, 60590, Germany
| | - Johannes Pantel
- Institute of General Practice, Goethe-University, Frankfurt Am Main, 60590, Germany
| | - Agnes Flöel
- Department of Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
53
|
Naro A, Corallo F, De Salvo S, Marra A, Di Lorenzo G, Muscarà N, Russo M, Marino S, De Luca R, Bramanti P, Calabrò RS. Promising Role of Neuromodulation in Predicting the Progression of Mild Cognitive Impairment to Dementia. J Alzheimers Dis 2016; 53:1375-88. [DOI: 10.3233/jad-160305] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
54
|
Jiang DP, Li JH, Zhang J, Xu SL, Kuang F, Lang HY, Wang YF, An GZ, Li J, Guo GZ. Long-term electromagnetic pulse exposure induces Abeta deposition and cognitive dysfunction through oxidative stress and overexpression of APP and BACE1. Brain Res 2016; 1642:10-19. [DOI: 10.1016/j.brainres.2016.02.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 02/05/2016] [Accepted: 02/14/2016] [Indexed: 11/30/2022]
|
55
|
Cover KS, van Schijndel RA, Versteeg A, Leung KK, Mulder ER, Jong RA, Visser PJ, Redolfi A, Revillard J, Grenier B, Manset D, Damangir S, Bosco P, Vrenken H, van Dijk BW, Frisoni GB, Barkhof F. Reproducibility of hippocampal atrophy rates measured with manual, FreeSurfer, AdaBoost, FSL/FIRST and the MAPS-HBSI methods in Alzheimer's disease. Psychiatry Res Neuroimaging 2016; 252:26-35. [PMID: 27179313 DOI: 10.1016/j.pscychresns.2016.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 02/16/2016] [Accepted: 04/08/2016] [Indexed: 11/23/2022]
Abstract
The purpose of this study is to assess the reproducibility of hippocampal atrophy rate measurements of commonly used fully-automated algorithms in Alzheimer disease (AD). The reproducibility of hippocampal atrophy rate for FSL/FIRST, AdaBoost, FreeSurfer, MAPS independently and MAPS combined with the boundary shift integral (MAPS-HBSI) were calculated. Back-to-back (BTB) 3D T1-weighted MPRAGE MRI from the Alzheimer's Disease Neuroimaging Initiative (ADNI1) study at baseline and year one were used. Analysis on 3 groups of subjects was performed - 562 subjects at 1.5T, a 75 subject group that also had manual segmentation and 111 subjects at 3T. A simple and novel statistical test based on the binomial distribution was used that handled outlying data points robustly. Median hippocampal atrophy rates were -1.1%/year for healthy controls, -3.0%/year for mildly cognitively impaired and -5.1%/year for AD subjects. The best reproducibility was observed for MAPS-HBSI (1.3%), while the other methods tested had reproducibilities at least 50% higher at 1.5T and 3T which was statistically significant. For a clinical trial, MAPS-HBSI should require less than half the subjects of the other methods tested. All methods had good accuracy versus manual segmentation. The MAPS-HBSI method has substantially better reproducibility than the other methods considered.
Collapse
Affiliation(s)
- Keith S Cover
- VU University Medical Center, Amsterdam, Netherlands.
| | | | | | | | - Emma R Mulder
- VU University Medical Center, Amsterdam, Netherlands
| | - Remko A Jong
- VU University Medical Center, Amsterdam, Netherlands
| | | | | | | | | | | | | | - Paolo Bosco
- IRCCS San Giovanni di Dio Fatebenefratelli, Italy
| | - Hugo Vrenken
- VU University Medical Center, Amsterdam, Netherlands
| | | | - Giovanni B Frisoni
- IRCCS San Giovanni di Dio Fatebenefratelli, Italy; University Hospitals and University of Geneva, Switzerland
| | | |
Collapse
|
56
|
Chiang HS, Mudar RA, Pudhiyidath A, Spence JS, Womack KB, Cullum CM, Tanner JA, Eroh J, Kraut MA, Hart J. Altered Neural Activity during Semantic Object Memory Retrieval in Amnestic Mild Cognitive Impairment as Measured by Event-Related Potentials. J Alzheimers Dis 2016; 46:703-17. [PMID: 25835419 DOI: 10.3233/jad-142781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Deficits in semantic memory in individuals with amnestic mild cognitive impairment (aMCI) have been previously reported, but the underlying neurobiological mechanisms remain to be clarified. We examined event-related potentials (ERPs) associated with semantic memory retrieval in 16 individuals with aMCI as compared to 17 normal controls using the Semantic Object Retrieval Task (EEG SORT). In this task, subjects judged whether pairs of words (object features) elicited retrieval of an object (retrieval trials) or not (non-retrieval trials). Behavioral findings revealed that aMCI subjects had lower accuracy scores and marginally longer reaction time compared to controls. We used a multivariate analytical technique (STAT-PCA) to investigate similarities and differences in ERPs between aMCI and control groups. STAT-PCA revealed a left fronto-temporal component starting at around 750 ms post-stimulus in both groups. However, unlike controls, aMCI subjects showed an increase in the frontal-parietal scalp potential that distinguished retrieval from non-retrieval trials between 950 and 1050 ms post-stimulus negatively correlated with the performance on the logical memory subtest of the Wechsler Memory Scale-III. Thus, individuals with aMCI were not only impaired in their behavioral performance on SORT relative to controls, but also displayed alteration in the corresponding ERPs. The altered neural activity in aMCI compared to controls suggests a more sustained and effortful search during object memory retrieval, which may be a potential marker indicating disease processes at the pre-dementia stage.
Collapse
Affiliation(s)
- Hsueh-Sheng Chiang
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Raksha A Mudar
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Speech and Hearing Science, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Athula Pudhiyidath
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
| | - Jeffrey S Spence
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA
| | - Kyle B Womack
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA.,Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - C Munro Cullum
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeremy A Tanner
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Justin Eroh
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Michael A Kraut
- Department of Radiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John Hart
- Center for BrainHealth, The University of Texas at Dallas, Dallas, TX, USA.,School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
57
|
A semi-mechanism approach based on MRI and proteomics for prediction of conversion from mild cognitive impairment to Alzheimer's disease. Sci Rep 2016; 6:26712. [PMID: 27273250 PMCID: PMC4896009 DOI: 10.1038/srep26712] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/04/2016] [Indexed: 01/18/2023] Open
Abstract
Mild cognitive impairment (MCI) is a precursor phase of Alzheimer’s disease (AD). As current treatments may be effective only at the early stages of AD, it is important to track MCI patients who will convert to AD. The aim of this study is to develop a high performance semi-mechanism based approach to predict the conversion from MCI to AD and improve our understanding of MCI-to-AD conversion mechanism. First, analysis of variance (ANOVA) test and lasso regression are employed to identify the markers related to the conversion. Then the Bayesian network based on selected markers is established to predict MCI-to-AD conversion. The structure of Bayesian network suggests that the conversion may start with fibrin clot formation, verbal memory impairment, eating pattern changing and hyperinsulinemia. The Bayesian network achieves a high 10-fold cross-validated prediction performance with 96% accuracy, 95% sensitivity, 65% specificity, area under the receiver operating characteristic curve of 0.82 on data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database. The semi-mechanism based approach provides not only high prediction performance but also clues of mechanism for MCI-to-AD conversion.
Collapse
|
58
|
Abstract
Genetic characterization of individuals at risk of Alzheimer's disease (AD), i.e. people having amyloid deposits in the brain without symptoms, people suffering from subjective cognitive decline (SCD) or mild cognitive impairment (MCI), has spurred the interests of researchers. However, their pre-dementia genetic profile remains mostly unexplored. In this study, we reviewed the loci related to phenotypes of AD, MCI and SCD from literature and performed the first meta-analyses evaluating the role of apolipoprotein E (APOE) in the risk of conversion from a healthy status to MCI and SCD. For AD dementia risk, an increased number of loci have been identified; to date, 28 genes have been associated with Late Onset AD. In MCI syndrome, APOE is confirmed as a pheno-conversion factor leading from MCI to AD, and clusterin is a promising candidate. Additionally, our meta-analyses revealed APOE as genetic risk factor to convert from a healthy status to MCI [OR = 1.849 (1.587-2.153); P = 2.80 × 10-15] and to a lesser extent from healthy status to SCD [OR = 1.151 (1.015-1.304); P = 0.028]. Thus, we believe that genetic studies in longitudinal SCD and MCI series may provide new therapeutic targets and improve the existing knowledge of AD. This type of studies must be completed on healthy subjects to better understand the natural disease resistance to brain insults and neurodegeneration.
Collapse
|
59
|
Brayet P, Petit D, Frauscher B, Gagnon JF, Gosselin N, Gagnon K, Rouleau I, Montplaisir J. Quantitative EEG of Rapid-Eye-Movement Sleep: A Marker of Amnestic Mild Cognitive Impairment. Clin EEG Neurosci 2016; 47:134-41. [PMID: 26323578 DOI: 10.1177/1550059415603050] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/07/2015] [Indexed: 11/15/2022]
Abstract
The basal forebrain cholinergic system, which is impaired in early Alzheimer's disease, is more crucial for the activation of rapid-eye-movement (REM) sleep electroencephalogram (EEG) than it is for wakefulness. Quantitative EEG from REM sleep might thus provide an earlier and more accurate marker of the development of Alzheimer's disease in subjects with mild cognitive impairment (MCI) subjects than that from wakefulness. To assess the superiority of the REM sleep EEG as a screening tool for preclinical Alzheimer's disease, 22 subjects with amnestic MCI (a-MCI; 63.9±7.7 years), 10 subjects with nonamnestic MCI (na-MCI; 64.1±4.5 years) and 32 controls (63.7±6.6 years) participated in the study. Spectral analyses of the waking EEG and REM sleep EEG were performed and the [(delta+theta)/(alpha+beta)] ratio was used to assess between-group differences in EEG slowing. The a-MCI subgroup showed EEG slowing in frontal lateral regions compared to both na-MCI and control groups. This EEG slowing was present in wakefulness (compared to controls) but was much more prominent in REM sleep. Moreover, the comparison between amnestic and nonamnestic subjects was found significant only for the REM sleep EEG. There was no difference in EEG power ratio between na-MCI and controls for any of the 7 cortical regions studied. These findings demonstrate the superiority of the REM sleep EEG in the discrimination between a-MCI and both na-MCI and control subjects.
Collapse
Affiliation(s)
- Pauline Brayet
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montreal, Montreal, Quebec, Canada Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada
| | - Dominique Petit
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montreal, Montreal, Quebec, Canada Department of Psychiatry, Université de Montréal, Montreal, Quebec, Canada
| | - Birgit Frauscher
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jean-François Gagnon
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montreal, Montreal, Quebec, Canada Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montreal, Montreal, Quebec, Canada Department of Psychology, Université de Montréal, Montreal, Quebec, Canada
| | - Katia Gagnon
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montreal, Montreal, Quebec, Canada Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada
| | - Isabelle Rouleau
- Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada Neurology Service, Hôpital Notre-Dame du CHUM, Montreal, Quebec, Canada
| | - Jacques Montplaisir
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montreal, Montreal, Quebec, Canada Department of Psychiatry, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
60
|
Schröder J, Pantel J. Neuroimaging of hippocampal atrophy in early recognition of Alzheimer's disease--a critical appraisal after two decades of research. Psychiatry Res Neuroimaging 2016; 247:71-78. [PMID: 26774855 DOI: 10.1016/j.pscychresns.2015.08.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 08/27/2015] [Indexed: 01/27/2023]
Abstract
As a characteristic feature of Alzheimer's disease (AD) hippocampal atrophy (HA) can be demonstrated in the majority of patients by using neuroimaging techniques in particular magnetic resonance imaging (MRI). Hippocampal atrophy is associated with declarative memory deficits and can also be associated with changes of adjacent medial temporal substructures such as the parahippocampal gyrus or the the entorhinal cortex. Similar findings are present in patients with mild cognitive impairment (MCI) albeit to a lesser extent. While these finding facilitate the diagnostic process in patients with clinical suspicious AD, the metric properties of hippocampal atrophy for delineating healthy aging from MCI and mild AD still appear to be rather limited; as such it is not sufficient to establish the diagnosis of AD (and even more so of MCI). This limitation partly refers to methodological issues and partly to the fact that hippocampal tissue integrity is subject to various pathogenetic influences other than AD. Moreover,the effects of hippocampal atrophy on the behavioral level (e.g. cognitive deficits) are modulated by the individual's cognitive reserve. From a clinical standpoint these observations are in line with the hypothesis that the onset and course of AD is influenced by a number of peristatic factors which are partly conceptualized in the concepts of brain and/or cognitive reserve. These complex interactions have to be considered when using the presence of hippocampal atrophy in the routine diagnostic procedure of AD.
Collapse
Affiliation(s)
- Johannes Schröder
- Section of Geriatric Psychiatry & Institute of Gerontology University of Heidelberg, Germany.
| | - Johannes Pantel
- Department of General Medicine, University of Frankfurt/M, Germany
| |
Collapse
|
61
|
Trebbastoni A, Pichiorri F, D’Antonio F, Campanelli A, Onesti E, Ceccanti M, de Lena C, Inghilleri M. Altered Cortical Synaptic Plasticity in Response to 5-Hz Repetitive Transcranial Magnetic Stimulation as a New Electrophysiological Finding in Amnestic Mild Cognitive Impairment Converting to Alzheimer's Disease: Results from a 4-year Prospective Cohort Study. Front Aging Neurosci 2016; 7:253. [PMID: 26793103 PMCID: PMC4709411 DOI: 10.3389/fnagi.2015.00253] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/21/2015] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION To investigate cortical excitability and synaptic plasticity in amnestic mild cognitive impairment (aMCI) using 5 Hz repetitive transcranial magnetic stimulation (5 Hz-rTMS) and to assess whether specific TMS parameters predict conversion time to Alzheimer's disease (AD). MATERIALS AND METHODS Forty aMCI patients (single- and multi-domain) and 20 healthy controls underwent, at baseline, a neuropsychological examination and 5 Hz-rTMS delivered in trains of 10 stimuli and 120% of resting motor threshold (rMT) intensity over the dominant motor area. The rMT and the ratio between amplitude of the 1st and the 10th motor-evoked potential elicited by the train (X/I-MEP ratio) were calculated as measures of cortical excitability and synaptic plasticity, respectively. Patients were followed up annually over a period of 48 months. Analysis of variance for repeated measures was used to compare TMS parameters in patients with those in controls. Spearman's correlation was performed by considering demographic variables, aMCI subtype, neuropsychological test scores, TMS parameters, and conversion time. RESULTS Thirty-five aMCI subjects completed the study; 60% of these converted to AD. The baseline rMT and X/I-MEP ratio were significantly lower in patients than in controls (p = 0.04 and p = 0.01). Spearman's analysis showed that conversion time correlated with the rMT (0.40) and X/I-MEP ratio (0.51). DISCUSSION aMCI patients displayed cortical hyperexcitability and altered synaptic plasticity to 5 Hz-rTMS when compared with healthy subjects. The extent of these changes correlated with conversion time. These alterations, which have previously been observed in AD, are thus present in the early stages of disease and may be considered as potential neurophysiological markers of conversion from aMCI to AD.
Collapse
Affiliation(s)
| | - Floriana Pichiorri
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
- Neuroelectrical Imaging and Brain Computer Interface Laboratory, Fondazione Santa Lucia – Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Fabrizia D’Antonio
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | | | - Emanuela Onesti
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | - Marco Ceccanti
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | - Carlo de Lena
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | - Maurizio Inghilleri
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
62
|
Eisenmenger LB, Huo EJ, Hoffman JM, Minoshima S, Matesan MC, Lewis DH, Lopresti BJ, Mathis CA, Okonkwo DO, Mountz JM. Advances in PET Imaging of Degenerative, Cerebrovascular, and Traumatic Causes of Dementia. Semin Nucl Med 2016; 46:57-87. [DOI: 10.1053/j.semnuclmed.2015.09.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
63
|
Hellwig K, Kvartsberg H, Portelius E, Andreasson U, Oberstein TJ, Lewczuk P, Blennow K, Kornhuber J, Maler JM, Zetterberg H, Spitzer P. Neurogranin and YKL-40: independent markers of synaptic degeneration and neuroinflammation in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2015; 7:74. [PMID: 26698298 PMCID: PMC4690296 DOI: 10.1186/s13195-015-0161-y] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/28/2015] [Indexed: 12/17/2022]
Abstract
Introduction Neuroinflammation and synaptic degeneration are major neuropathological hallmarks in Alzheimer’s disease (AD). Neurogranin and YKL-40 in cerebrospinal fluid (CSF) are newly discovered markers indicating synaptic damage and microglial activation, respectively. Methods CSF samples from 95 individuals including 39 patients with AD dementia (AD-D), 13 with mild cognitive impairment (MCI) due to AD (MCI-AD), 29 with MCI not due to AD (MCI-o) and 14 patients with non-AD dementias (non-AD-D) were analyzed for neurogranin and YKL-40. Results Patients with dementia or MCI due to AD showed elevated levels of CSF neurogranin (p < 0.001 for AD-D and p < 0.05 for MCI-AD) and YKL-40 (p < 0.05 for AD-D and p = 0.15 for MCI-AD) compared to mildly cognitively impaired subjects not diagnosed with AD. CSF levels of neurogranin and YKL-40 did not differ between MCI not due to AD and non-AD dementias. In AD subjects no correlation between YKL-40 and neurogranin was found. The CSF neurogranin levels correlated moderately with tau and p-tau but not with Aβ42 or the MMSE in AD samples. No relevant associations between YKL-40 and MMSE or the core AD biomarkers, Aβ42, t-tau and p-tau were found in AD subjects. Conclusions Neurogranin and YKL-40 are promising AD biomarkers, independent of and complementary to the established core AD biomarkers, reflecting additional pathological changes in the course of AD. Electronic supplementary material The online version of this article (doi:10.1186/s13195-015-0161-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Konstantin Hellwig
- Department of Psychiatry and Psychotherapy, University clinic Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Hlin Kvartsberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,AlzeCure Foundation, Karolinska Institutet Science Park, Huddinge, Sweden.
| | - Erik Portelius
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Timo Jan Oberstein
- Department of Psychiatry and Psychotherapy, University clinic Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, University clinic Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany. .,Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland.
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University clinic Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Juan Manuel Maler
- Department of Psychiatry and Psychotherapy, University clinic Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Department of Molecular Neuroscience, UCL Institute of Neurology, London, WC1N 3BG, UK.
| | - Philipp Spitzer
- Department of Psychiatry and Psychotherapy, University clinic Erlangen and Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| |
Collapse
|
64
|
Stomrud E, Minthon L, Zetterberg H, Blennow K, Hansson O. Longitudinal cerebrospinal fluid biomarker measurements in preclinical sporadic Alzheimer's disease: A prospective 9-year study. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2015; 1:403-11. [PMID: 27239521 PMCID: PMC4879483 DOI: 10.1016/j.dadm.2015.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction Ascertainment of the pattern and temporal change of biomarkers in preclinical (asymptomatic) sporadic Alzheimer's disease (AD) will increase knowledge about early pathogenesis and facilitate interventional therapeutic trials. Methods In this prospective longitudinal study, repeated cerebrospinal fluid (CSF) collections and cognitive evaluations were performed in cognitively healthy elderly individuals during a 9-year period. Results Low CSF β-amyloid (Aβ)42 levels predicted subsequent development of clinical AD 9 years later. Noteworthy, one-third of individuals with pathologically low baseline Aβ42 levels remained cognitively intact during follow-up. No further decrease in Aβ42 was seen in those with low levels already at baseline. Discussion CSF Aβ42 predicts sporadic AD at least 9 years before dementia onset and has plateaued already at this time. However, many individuals can harbor brain amyloid accumulation over a decade without signs of cognitive deterioration, which could implicate how CSF biomarkers are used to identify preclinical AD in future interventional therapeutic trials.
Collapse
Affiliation(s)
- Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Skåne University Hospital, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Lennart Minthon
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Skåne University Hospital, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Skåne University Hospital, Malmö, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
65
|
Abstract
Symbols and signs have been suggested to improve the orientation of patients suffering from Alzheimer disease (AD). However, there are hardly any studies that confirm whether AD patients benefit from signs or symbols and which symbol characteristics might improve or impede their symbol comprehension. To address these issues, 30 AD patients and 30 matched healthy controls performed a symbol processing task (SPT) with 4 different item categories. A repeated-measures analysis of variance was run to identify impact of different item categories on performance accuracy in both the experimental groups. Moreover, SPT scores were correlated with neuropsychological test scores in a broad range of other cognitive domains. Finally, diagnostic accuracy of the SPT was calculated by a receiver-operating characteristic curve analysis. Results revealed a global symbol processing dysfunction in AD that was associated with semantic memory and executive deficits. Moreover, AD patients showed a disproportional performance decline at SPT items with visual distraction. Finally, the SPT total score showed high sensitivity and specificity in differentiating between AD patients and healthy controls. The present findings suggest that specific symbol features impede symbol processing in AD and argue for a diagnostic benefit of the SPT in neuropsychological assessment.
Collapse
|
66
|
Gispert JD, Rami L, Sánchez-Benavides G, Falcon C, Tucholka A, Rojas S, Molinuevo JL. Nonlinear cerebral atrophy patterns across the Alzheimer's disease continuum: impact of APOE4 genotype. Neurobiol Aging 2015; 36:2687-701. [PMID: 26239178 DOI: 10.1016/j.neurobiolaging.2015.06.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 06/26/2015] [Accepted: 06/30/2015] [Indexed: 01/11/2023]
Abstract
The progression of Alzheimer's disease (AD) is characterized by complex trajectories of cerebral atrophy that are affected by interactions with age and apolipoprotein E allele ε4 (APOE4) status. In this article, we report the nonlinear volumetric changes in gray matter across the full biological spectrum of the disease, represented by the AD-cerebrospinal fluid (CSF) index. This index reflects the subject's level of pathology and position along the AD continuum. We also evaluated the associated impact of the APOE4 genotype. The atrophy pattern associated with the AD-CSF index was highly symmetrical and corresponded with the typical AD signature. Medial temporal structures showed different atrophy dynamics along the progression of the disease. The bilateral parahippocampal cortices and a parietotemporal region extending from the middle temporal to the supramarginal gyrus presented an initial increase in volume which later reverted. Similarly, a portion of the precuneus presented a rather linear inverse association with the AD-CSF index whereas some other clusters did not show significant atrophy until index values corresponded to positive CSF tau values. APOE4 carriers showed steeper hippocampal volume reductions with AD progression. Overall, the reported atrophy patterns are in close agreement with those mentioned in previous findings. However, the detected nonlinearities suggest that there may be different pathological processes taking place at specific moments during AD progression and reveal the impact of the APOE4 allele.
Collapse
Affiliation(s)
- J D Gispert
- Clinical and Neuroimaging Departments, Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - L Rami
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - C Falcon
- Clinical and Neuroimaging Departments, Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - A Tucholka
- Clinical and Neuroimaging Departments, Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - S Rojas
- Clinical and Neuroimaging Departments, Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Department of Morphological Sciences, Anatomy and Embriology Unit, Faculty of Medicine, Autonomous University of Barcelona
| | - J L Molinuevo
- Clinical and Neuroimaging Departments, Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
67
|
Kishi T, Matsunaga S, Oya K, Ikuta T, Iwata N. Protection against Brain Atrophy by Anti-dementia Medication in Mild Cognitive Impairment and Alzheimer's Disease: Meta-Analysis of Longitudinal Randomized Placebo-Controlled Trials. Int J Neuropsychopharmacol 2015; 18:pyv070. [PMID: 26091818 PMCID: PMC4675981 DOI: 10.1093/ijnp/pyv070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 06/16/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND There has not been conclusive evidence for prevention of brain atrophy by anti-dementia drugs in mild cognitive impairment and Alzheimer's Disease. METHODS Relevant studies were identified through searches of PubMed, databases of the Cochrane Library, and PsycINFO citations up to 16 May, 2015. Only double-blind, randomized, placebo-controlled clinical trials of anti-dementia drugs in patients with mild cognitive impairment or Alzheimer's Disease were included. Primary outcomes were annualized percent change of total brain volume (%TBV/y), annualized percent change of hippocampal volume (%HV/y), and annualized percent change of ventricular volume (%VV/y) measured by magnetic resonance imaging. Standardized mean difference (SMD) and 95% confidence intervals (CI) were calculated for relevant outcomes. RESULTS Seven randomized, placebo-controlled clinical trials (n=1708) were found to meet the inclusion criteria, including 4 mild cognitive impairment studies (n=1327) and 3 Alzheimer's Disease studies (n=381) [3 donepezil studies (2 mild cognitive impairment studies and 1 Alzheimer's Disease study), 1 galantaime study for mild cognitive impairment, 2 mementine studies for Alzheimer's Disease, and 1 rivastigmine study for mild cognitive impairment]. Pooled anti-dementia drugs showed superior protective outcomes compared with placebo regarding %TBV/y (SMD=-0.21, 95%CI=-0.37 to -0.04, P=.01, N=4, n=624) and %VV/y (SMD=-0.79, 95%CI=-1.40 to -0.19, P=.01, N=3, n=851). However, %HV/y failed to show difference between both groups. Among anti-dementia drugs, donepezil showed significantly greater protective effects than placebo regarding %TBV/y (SMD=-0.43, 95%CI=-0.74 to -0.12, P=.007, N=1, n=164) and %VV/y (SMD=-0.51, 95%CI=-0.73 to -0.29, P<.00001, N=2, n=338). Rivastigmine was also superior to placebo regarding %VV/y (SMD=-1.33, 95%CI=-1.52 to -1.14, P<.00001). CONCLUSIONS The results favored the hypothesis that anti-dementia drugs may prevent brain atrophy in patients with mild cognitive impairment and Alzheimer's Disease.
Collapse
Affiliation(s)
- Taro Kishi
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Drs Kishi, Matsunaga, Oya, and Iwata); Department of Communication Sciences and Disorders, School of Applied Sciences, University of Mississippi, University, Mississippi (Dr Ikuta).
| | | | | | | | | |
Collapse
|
68
|
Ciblis AS, Butler ML, Bokde ALW, Mullins PG, McNulty JP. The use of neuroimaging in dementia by Irish general practitioners. Ir J Med Sci 2015; 185:597-602. [PMID: 26048643 DOI: 10.1007/s11845-015-1315-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/16/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND More than 48,000 people in Ireland are living with dementia, and the number is likely to rise to 130,000 by 2041. Dementia frequently remains undiagnosed, depriving many of early interventions and the opportunity to plan for the future. Neuroimaging is helpful in the diagnosis of dementia, yet it is often insufficiently utilised. General practitioners (GPs) often decide which patients should be referred on for specialist assessment and as such play a crucial role in dementia diagnosis. AIMS To establish the accessibility of neuroimaging in dementia by GPs, current referral patterns, confidence in referral and opinions on radiology reports. METHODS The research design was a postal survey among GPs in single and group practices in urban, rural and semi-rural areas in the east and southeast of Ireland. GPs were identified from the Irish Medical Directory and posted individual anonymous questionnaires. RESULTS A third of participants reported that they had no direct access to neuroimaging. Access differed between public and private patients. GPs primarily referred to computed tomography and magnetic resonance imaging, but only 14.6 % based these referrals on published guidelines. A total of 47.8 % of participants were not very confident in their ability to choose the most appropriate modality. CONCLUSION Access to neuroimaging investigations for suspected cases of dementia varies between locations and public and private systems. To improve diagnostic rates and ensure appropriate utilisation of imaging resources, GPs require access to clinical and referral guidelines to ensure appropriate use of neuroimaging and the best possible patient outcomes.
Collapse
Affiliation(s)
- A S Ciblis
- UCD School of Medicine and Medical Science, Diagnostic Imaging, Room 223, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland.
| | - M-L Butler
- UCD School of Medicine and Medical Science, Diagnostic Imaging, Room 223, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - A L W Bokde
- Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - P G Mullins
- School of Psychology, Bangor University, Bangor, UK
| | - J P McNulty
- UCD School of Medicine and Medical Science, Diagnostic Imaging, Room 223, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
69
|
Castorina A, Szychlinska MA, Marzagalli R, Musumeci G. Mesenchymal stem cells-based therapy as a potential treatment in neurodegenerative disorders: is the escape from senescence an answer? Neural Regen Res 2015; 10:850-8. [PMID: 26199588 PMCID: PMC4498333 DOI: 10.4103/1673-5374.158352] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2015] [Indexed: 01/09/2023] Open
Abstract
Aging is the most prominent risk factor contributing to the development of neurodegenerative disorders. In the United States, over 35 million of elderly people suffer from age-related diseases. Aging impairs the self-repair ability of neuronal cells, which undergo progressive deterioration. Once initiated, this process hampers the already limited regenerative power of the central nervous system, making the search for new therapeutic strategies particularly difficult in elderly affected patients. So far, mesenchymal stem cells have proven to be a viable option to ameliorate certain aspects of neurodegeneration, as they possess high proliferative rate and differentiate in vitro into multiple lineages. However, accumulating data have demonstrated that during long-term culture, mesenchymal stem cells undergo spontaneous transformation. Transformed mesenchymal stem cells show typical features of senescence, including the progressive shortening of telomers, which results in cell loss and, as a consequence, hampered regenerative potential. These evidences, in line with those observed in mesenchymal stem cells isolated from old donors, suggest that senescence may represent a limit to mesenchymal stem cells exploitation in therapy, prompting scholars to either find alternative sources of pluripotent cells or to arrest the age-related transformation. In the present review, we summarize findings from recent literature, and critically discuss some of the major hurdles encountered in the search of appropriate sources of mesenchymal stem cells, as well as benefits arising from their use in neurodegenerative diseases. Finally, we provide some insights that may aid in the development of strategies to arrest or, at least, delay the aging of mesenchymal stem cells to improve their therapeutic potential.
Collapse
Affiliation(s)
- Alessandro Castorina
- Department of Biomedical and Biotechnological Science, Section of Human Anatomy and Histology, School of Medicine, University of Catania, Via S. Sofia 87, Catania, Italy
| | - Marta Anna Szychlinska
- Department of Biomedical and Biotechnological Science, Section of Human Anatomy and Histology, School of Medicine, University of Catania, Via S. Sofia 87, Catania, Italy
| | - Rubina Marzagalli
- Department of Biomedical and Biotechnological Science, Section of Human Anatomy and Histology, School of Medicine, University of Catania, Via S. Sofia 87, Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Science, Section of Human Anatomy and Histology, School of Medicine, University of Catania, Via S. Sofia 87, Catania, Italy
| |
Collapse
|
70
|
Durazzo TC, Mattsson N, Weiner MW. Interaction of Cigarette Smoking History With APOE Genotype and Age on Amyloid Level, Glucose Metabolism, and Neurocognition in Cognitively Normal Elders. Nicotine Tob Res 2015; 18:204-11. [PMID: 25847292 DOI: 10.1093/ntr/ntv075] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/22/2015] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Chronic cigarette smoking is associated with increased risk for Alzheimer's disease (AD). The goal of this study was to determine if smoking history moderated the associations of age and APOE genotype (the most robust risk factors for AD) on brain amyloid deposition, glucose metabolism, and neurocognition in cognitively-normal elders. METHODS Participants (n = 264) were grouped according to their APOE ε4 carrier status (ε4 carrier: APOE4+; non-ε4 carrier: APOE4-) and smoking status (smokers: at least 1 year of smoking during lifetime; never-smokers: no history of smoking). Approximately 89% of the smoking sample was former-smokers. We specifically tested for interactions of smoking status with APOE ε4 carrier status and age on measures of cortical amyloid deposition, glucose metabolism, and neurocognition. RESULTS (1) smoking status interacted with APOE ε4 carrier status, where smoker APOE4+ showed lower glucose metabolism and poorer auditory-verbal learning and memory than never-smoking APOE4-, never-smoking APOE4+, and smoking APOE4-; (2) smoking status interacted with age on measures of semantic fluency, processing speed/set-shifting and global neurocognition; smokers, irrespective of APOE ε4 carrier status, demonstrated poorer performance with increasing age than never-smokers; and (3) smoking APOE4+ and never-smoking APOE4+ showed greater cortical amyloid deposition than never-smoking APOE4- and smoking APOE4-. CONCLUSIONS The findings indicate consideration of smoking history is essential to both better understand the factors associated with neurobiological and neurocognitive abnormalities in elders, and the risk for development of AD-related neuropathology.
Collapse
Affiliation(s)
- Timothy C Durazzo
- Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center, San Francisco, CA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA;
| | - Niklas Mattsson
- Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center, San Francisco, CA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA; Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Mölndal, Sweden
| | - Michael W Weiner
- Center for Imaging of Neurodegenerative Diseases, San Francisco VA Medical Center, San Francisco, CA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA; Departments of Psychiatry, Medicine, and Neurology, University of California, San Francisco, CA
| | | |
Collapse
|
71
|
Ciblis AS, Butler ML, Bokde AL, Mullins PG, O'Neill D, McNulty JP. Neuroimaging referral for dementia diagnosis: The specialist's perspective in Ireland. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2015; 1:41-7. [PMID: 27239490 PMCID: PMC4876894 DOI: 10.1016/j.dadm.2014.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Neuroimaging is an increasingly important tool in the diagnostic workup of dementia. Neurologists, geriatricians, and old-age psychiatrists are involved in key tasks in the diagnostic process, frequently referring patients with suspected dementia for neuroimaging. METHODS The research design was a postal survey of all geriatricians, old-age psychiatrists, and neurologists in the Republic of Ireland (N = 176) as identified by the Irish Medical Directory 2011-2012 and supplementary listings. RESULTS Almost 65% of specialists did not have access to 2-[18F]fluoro-2-deoxy-D-glucose positron emission (FDG-PET) or FDG-PET/computed tomography (CT), and 80.3% did not have access to perfusion hexamethylpropyleneamine oxime single-photon emission computed tomography (SPECT) or dopaminergic iodine-123-radiolabeled 2β-carbomethoxy-3β-(4-iodophenyl)-N-(3-fluoropropyl) nortropane SPECT. Most specialists (88.7%) referred patients with mild cognitive impairment or suspected dementia for magnetic resonance imaging (MRI), 81.7% referred for CT, and 26.8% for FDG-PET or FDG-PET/CT. Only 44.6% of respondents were aware of dementia-specific protocols for referrals for neuroimaging. CONCLUSION Specialist access to imaging modalities other than CT and MRI is restricted. Improved access may affect patient treatment and care.
Collapse
Affiliation(s)
- Aurelia S. Ciblis
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Marie-Louise Butler
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Arun L.W. Bokde
- Cognitive Systems Group, Discipline of Psychiatry, School of Medicine and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | - Desmond O'Neill
- Centre for Ageing, Neuroscience and the Humanities, Trinity College Dublin, Dublin, Ireland
| | - Jonathan P. McNulty
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
72
|
Ungar L, Altmann A, Greicius MD. Apolipoprotein E, gender, and Alzheimer's disease: an overlooked, but potent and promising interaction. Brain Imaging Behav 2014; 8:262-73. [PMID: 24293121 DOI: 10.1007/s11682-013-9272-x] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is an increasingly prevalent, fatal neurodegenerative disease that has proven resistant, thus far, to all attempts to prevent it, forestall it, or slow its progression. The ε4 allele of the Apolipoprotein E gene (APOE4) is a potent genetic risk factor for sporadic and late-onset familial AD. While the link between APOE4 and AD is strong, many expected effects, like increasing the risk of conversion from MCI to AD, have not been widely replicable. One critical, and commonly overlooked, feature of the APOE4 link to AD is that several lines of evidence suggest it is far more pronounced in women than in men. Here we review previous literature on the APOE4 by gender interaction with a particular focus on imaging-related studies.
Collapse
Affiliation(s)
- Leo Ungar
- Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA,
| | | | | |
Collapse
|
73
|
Cavedo E, Lista S, Khachaturian Z, Aisen P, Amouyel P, Herholz K, Jack CR, Sperling R, Cummings J, Blennow K, O'Bryant S, Frisoni GB, Khachaturian A, Kivipelto M, Klunk W, Broich K, Andrieu S, de Schotten MT, Mangin JF, Lammertsma AA, Johnson K, Teipel S, Drzezga A, Bokde A, Colliot O, Bakardjian H, Zetterberg H, Dubois B, Vellas B, Schneider LS, Hampel H. The Road Ahead to Cure Alzheimer's Disease: Development of Biological Markers and Neuroimaging Methods for Prevention Trials Across all Stages and Target Populations. J Prev Alzheimers Dis 2014; 1:181-202. [PMID: 26478889 PMCID: PMC4606938 DOI: 10.14283/jpad.2014.32] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is a slowly progressing non-linear dynamic brain disease in which pathophysiological abnormalities, detectable in vivo by biological markers, precede overt clinical symptoms by many years to decades. Use of these biomarkers for the detection of early and preclinical AD has become of central importance following publication of two international expert working group's revised criteria for the diagnosis of AD dementia, mild cognitive impairment (MCI) due to AD, prodromal AD and preclinical AD. As a consequence of matured research evidence six AD biomarkers are sufficiently validated and partly qualified to be incorporated into operationalized clinical diagnostic criteria and use in primary and secondary prevention trials. These biomarkers fall into two molecular categories: biomarkers of amyloid-beta (Aβ) deposition and plaque formation as well as of tau-protein related hyperphosphorylation and neurodegeneration. Three of the six gold-standard ("core feasible) biomarkers are neuroimaging measures and three are cerebrospinal fluid (CSF) analytes. CSF Aβ1-42 (Aβ1-42), also expressed as Aβ1-42 : Aβ1-40 ratio, T-tau, and P-tau Thr181 & Thr231 proteins have proven diagnostic accuracy and risk enhancement in prodromal MCI and AD dementia. Conversely, having all three biomarkers in the normal range rules out AD. Intermediate conditions require further patient follow-up. Magnetic resonance imaging (MRI) at increasing field strength and resolution allows detecting the evolution of distinct types of structural and functional abnormality pattern throughout early to late AD stages. Anatomical or volumetric MRI is the most widely used technique and provides local and global measures of atrophy. The revised diagnostic criteria for "prodromal AD" and "mild cognitive impairment due to AD" include hippocampal atrophy (as the fourth validated biomarker), which is considered an indicator of regional neuronal injury. Advanced image analysis techniques generate automatic and reproducible measures both in regions of interest, such as the hippocampus and in an exploratory fashion, observer and hypothesis-indedendent, throughout the entire brain. Evolving modalities such as diffusion-tensor imaging (DTI) and advanced tractography as well as resting-state functional MRI provide useful additionally useful measures indicating the degree of fiber tract and neural network disintegration (structural, effective and functional connectivity) that may substantially contribute to early detection and the mapping of progression. These modalities require further standardization and validation. The use of molecular in vivo amyloid imaging agents (the fifth validated biomarker), such as the Pittsburgh Compound-B and markers of neurodegeneration, such as fluoro-2-deoxy-D-glucose (FDG) (as the sixth validated biomarker) support the detection of early AD pathological processes and associated neurodegeneration. How to use, interpret, and disclose biomarker results drives the need for optimized standardization. Multimodal AD biomarkers do not evolve in an identical manner but rather in a sequential but temporally overlapping fashion. Models of the temporal evolution of AD biomarkers can take the form of plots of biomarker severity (degree of abnormality) versus time. AD biomarkers can be combined to increase accuracy or risk. A list of genetic risk factors is increasingly included in secondary prevention trials to stratify and select individuals at genetic risk of AD. Although most of these biomarker candidates are not yet qualified and approved by regulatory authorities for their intended use in drug trials, they are nonetheless applied in ongoing clinical studies for the following functions: (i) inclusion/exclusion criteria, (ii) patient stratification, (iii) evaluation of treatment effect, (iv) drug target engagement, and (v) safety. Moreover, novel promising hypothesis-driven, as well as exploratory biochemical, genetic, electrophysiological, and neuroimaging markers for use in clinical trials are being developed. The current state-of-the-art and future perspectives on both biological and neuroimaging derived biomarker discovery and development as well as the intended application in prevention trials is outlined in the present publication.
Collapse
Affiliation(s)
- E Cavedo
- Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Institut du Cerveau et de la Moelle épinière (ICM), UMR S 1127, Hôpital de la Pitié-Salpétrière Paris & CATI multicenter neuroimaging platform, France; Laboratory of Epidemiology, Neuroimaging and Telemedicine, IRCCS San Giovanni di Dio Fatebenefratelli Brescia, Italy
| | - S Lista
- AXA Research Fund & UPMC Chair; Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Inserm U1127 Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpétrière Paris, France
| | - Z Khachaturian
- The Campaign to Prevent Alzheimer's Disease by 2020 (PAD2020), Potomac, MD, USA
| | - P Aisen
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - P Amouyel
- Inserm, U744, Lille, 59000, France; Université Lille 2, Lille, 59000, France; Institut Pasteur de Lille, Lille, 59000, France; Centre Hospitalier Régional Universitaire de Lille, Lille, 59000, France
| | - K Herholz
- Institute of Brain, Behaviour and Mental Health, University of Manchester, UK
| | - C R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - R Sperling
- Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, 888 West Bonneville Avenue, Las Vegas, Nevada 89106, USA
| | - K Blennow
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - S O'Bryant
- Department of Internal Medicine, Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - G B Frisoni
- IRCCS Istituto Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy; University Hospitals and University of Geneva, Geneva, Switzerland
| | | | - M Kivipelto
- Karolinska Institutet Alzheimer Research Center, NVS, Stockholm, Sweden
| | - W Klunk
- Department of Psychiatry, University of Pittsburgh School of Medicine, USA; Department of Neurology, University of Pittsburgh School of Medicine, USA
| | - K Broich
- Federal Institute of Drugs and Medical Devices (BfArM), Bonn, Germany
| | - S Andrieu
- Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France; Public health department, CHU de Toulouse
| | - M Thiebaut de Schotten
- Natbrainlab, Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, King's College London, London, UK; Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière (ICM), UMRS 1127 Paris, France; Inserm, U 1127, Paris, France; CNRS, UMR 7225, Paris, France
| | - J-F Mangin
- CEA UNATI, Neurospin, CEA Gif-sur-Yvette, France & CATI multicenter neuroimaging platform
| | - A A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - K Johnson
- Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - S Teipel
- Department of Psychosomatic Medicine, University of Rostock, and DZNE, German Center for Neurodegenerative Diseases, Rostock, Germany
| | - A Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne Germany
| | - A Bokde
- Cognitive Systems Group, Discipline of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland and Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - O Colliot
- Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013, Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, Inserm, U1127, F-75013, Paris, France; CNRS, UMR 7225 ICM, 75013, Paris, France; Inria, Aramis project-team, Centre de Recherche Paris-Rocquencourt, France
| | - H Bakardjian
- Institute of Memory and Alzheimer's Disease (IM2A), Pitié-Salpétrière University Hospital, Paris, France; IHU-A-ICM - Paris Institute of Translational Neurosciences, Paris, France
| | - H Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology, Queen Square, London, UK
| | - B Dubois
- Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Inserm U1127 Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpétrière Paris, France
| | - B Vellas
- Inserm UMR1027, University of Toulouse, Toulouse, France
| | - L S Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - H Hampel
- AXA Research Fund & UPMC Chair; Sorbonne Universités, Université Pierre et Marie Curie, Paris 06, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A) Hôpital de la Pitié-Salpétrière & Inserm U1127 Institut du Cerveau et de la Moelle épinière (ICM), Hôpital de la Pitié-Salpétrière Paris, France
| |
Collapse
|
74
|
Jovicich J, Marizzoni M, Bosch B, Bartrés-Faz D, Arnold J, Benninghoff J, Wiltfang J, Roccatagliata L, Picco A, Nobili F, Blin O, Bombois S, Lopes R, Bordet R, Chanoine V, Ranjeva JP, Didic M, Gros-Dagnac H, Payoux P, Zoccatelli G, Alessandrini F, Beltramello A, Bargalló N, Ferretti A, Caulo M, Aiello M, Ragucci M, Soricelli A, Salvadori N, Tarducci R, Floridi P, Tsolaki M, Constantinidis M, Drevelegas A, Rossini PM, Marra C, Otto J, Reiss-Zimmermann M, Hoffmann KT, Galluzzi S, Frisoni GB. Multisite longitudinal reliability of tract-based spatial statistics in diffusion tensor imaging of healthy elderly subjects. Neuroimage 2014; 101:390-403. [DOI: 10.1016/j.neuroimage.2014.06.075] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/30/2014] [Accepted: 06/28/2014] [Indexed: 12/13/2022] Open
|
75
|
Adlard PA, Tran BA, Finkelstein DI, Desmond PM, Johnston LA, Bush AI, Egan GF. A review of β-amyloid neuroimaging in Alzheimer's disease. Front Neurosci 2014; 8:327. [PMID: 25400539 PMCID: PMC4215612 DOI: 10.3389/fnins.2014.00327] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 09/27/2014] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. As advancing age is the greatest risk factor for developing AD, the number of those afflicted is expected to increase markedly with the aging of the world's population. The inability to definitively diagnose AD until autopsy remains an impediment to establishing effective targeted treatments. Neuroimaging has enabled in vivo visualization of pathological changes in the brain associated with the disease, providing a greater understanding of its pathophysiological development and progression. However, neuroimaging biomarkers do not yet offer clear advantages over current clinical diagnostic criteria for them to be accepted into routine clinical use. Nonetheless, current insights from neuroimaging combined with the elucidation of biochemical and molecular processes in AD are informing the ongoing development of new imaging techniques and their application. Much of this research has been greatly assisted by the availability of transgenic mouse models of AD. In this review we summarize the main efforts of neuroimaging in AD in humans and in mouse models, with a specific focus on β-amyloid, and discuss the potential of new applications and novel approaches.
Collapse
Affiliation(s)
- Paul A. Adlard
- Division of Mental Health, The Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Bob A. Tran
- Department of Radiology, University of MelbourneParkville, VIC, Australia
| | - David I. Finkelstein
- Division of Mental Health, The Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Patricia M. Desmond
- Department of Radiology, University of MelbourneParkville, VIC, Australia
- Department of Radiology, The Royal Melbourne HospitalParkville, VIC, Australia
| | - Leigh A. Johnston
- Division of Mental Health, The Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
- Department of Electrical and Electronic Engineering, University of MelbourneParkville, VIC, Australia
| | - Ashley I. Bush
- Division of Mental Health, The Florey Institute of Neuroscience and Mental Health, University of MelbourneParkville, VIC, Australia
| | - Gary F. Egan
- Monash Biomedical Imaging, Monash UniversityClayton, VIC, Australia
- School of Psychology and Psychiatry, Monash UniversityClayton, VIC, Australia
| |
Collapse
|
76
|
Garn H, Waser M, Deistler M, Benke T, Dal-Bianco P, Ransmayr G, Schmidt H, Sanin G, Santer P, Caravias G, Seiler S, Grossegger D, Fruehwirt W, Schmidt R. Quantitative EEG markers relate to Alzheimer's disease severity in the Prospective Dementia Registry Austria (PRODEM). Clin Neurophysiol 2014; 126:505-13. [PMID: 25091343 DOI: 10.1016/j.clinph.2014.07.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 06/26/2014] [Accepted: 07/07/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To investigate which single quantitative electro-encephalographic (QEEG) marker or which combination of markers correlates best with Alzheimer's disease (AD) severity as measured by the Mini-Mental State Examination (MMSE). METHODS We compared quantitative EEG markers for slowing (relative band powers), synchrony (coherence, canonical correlation, Granger causality) and complexity (auto-mutual information, Shannon/Tsallis entropy) in 118 AD patients from the multi-centric study PRODEM Austria. Signal spectra were determined using an indirect spectral estimator. Analyses were adjusted for age, sex, duration of dementia, and level of education. RESULTS For the whole group (39 possible, 79 probable AD cases) MMSE scores explained 33% of the variations in relative theta power during face encoding, and 31% of auto-mutual information in resting state with eyes closed. MMSE scores explained also 25% of the overall QEEG factor. This factor was thus subordinate to individual markers. In probable AD, QEEG coefficients of determination were always higher than in the whole group, where MMSE scores explained 51% of the variations in relative theta power. CONCLUSIONS Selected QEEG markers show strong associations with AD severity. Both cognitive and resting state should be used for QEEG assessments. SIGNIFICANCE Our data indicate theta power measured during face-name encoding to be most closely related to AD severity.
Collapse
Affiliation(s)
- Heinrich Garn
- AIT Austrian Institute of Technology GmbH, Vienna, Austria.
| | - Markus Waser
- AIT Austrian Institute of Technology GmbH, Vienna, Austria
| | | | - Thomas Benke
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Peter Dal-Bianco
- Department of Neurology, Vienna Medical University, Vienna, Austria
| | | | - Helena Schmidt
- Department of Neurology, Graz Medical University, Graz, Austria
| | - Guenter Sanin
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Peter Santer
- Department of Neurology, Vienna Medical University, Vienna, Austria
| | - Georg Caravias
- Department of Neurology, Linz General Hospital, Linz, Austria
| | - Stephan Seiler
- Department of Neurology, Graz Medical University, Graz, Austria
| | | | | | | |
Collapse
|
77
|
A systematic review of the evidence that brain structure is related to muscle structure and their relationship to brain and muscle function in humans over the lifecourse. BMC Geriatr 2014; 14:85. [PMID: 25011478 PMCID: PMC4105796 DOI: 10.1186/1471-2318-14-85] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 07/01/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND An association between cognition and physical function has been shown to exist but the roles of muscle and brain structure in this relationship are not fully understood. A greater understanding of these relationships may lead to identification of the underlying mechanisms in this important area of research. This systematic review examines the evidence for whether: a) brain structure is related to muscle structure; b) brain structure is related to muscle function; and c) brain function is related to muscle structure in healthy children and adults. METHODS Medline, Embase, CINAHL and PsycINFO were searched on March 6th 2014. A grey literature search was performed using Google and Google Scholar. Hand searching through citations and references of relevant articles was also undertaken. RESULTS 53 articles were included in the review; mean age of the subjects ranged from 8.8 to 85.5 years old. There is evidence of a positive association between both whole brain volume and white matter (WM) volume and muscle size. Total grey matter (GM) volume was not associated with muscle size but some areas of regional GM volume were associated with muscle size (right temporal pole and bilateral ventromedial prefrontal cortex). No evidence was found of a relationship between grip strength and whole brain volume however there was some evidence of a positive association with WM volume. Conversely, there is evidence that gait speed is positively associated with whole brain volume; this relationship may be driven by total WM volume or regional GM volumes, specifically the hippocampus. Markers of brain ageing, that is brain atrophy and greater accumulation of white matter hyperintensities (WMH), were associated with grip strength and gait speed. The location of WMH is important for gait speed; periventricular hyperintensities and brainstem WMH are associated with gait speed but subcortical WMH play less of a role. Cognitive function does not appear to be associated with muscle size. CONCLUSION There is evidence that brain structure is associated with muscle structure and function. Future studies need to follow these interactions longitudinally to understand potential causal relationships.
Collapse
|
78
|
Abstract
Mild cognitive impairment is the term applied to the cognitive state that lies between normal aging and dementia. There has been significant controversy around describing, defining and characterizing mild cognitive impairment. This review will cover current understanding of the condition and discuss clinical features, research strategies and future directions.
Collapse
Affiliation(s)
- Craig Gordon
- ST5 Old Age Psychiatry, NHS Greater Glasgow and Clyde, Glasgow, UK University of Glasgow, MHW, 1055 Great Western Road, Glasgow, UK
| | | |
Collapse
|
79
|
Caravaglios G, Muscoso EG, Di Maria G, Costanzo E. Patients with mild cognitive impairment have an abnormal upper-alpha event-related desynchronization/synchronization (ERD/ERS) during a task of temporal attention. J Neural Transm (Vienna) 2014; 122:441-53. [PMID: 24947877 DOI: 10.1007/s00702-014-1262-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/11/2014] [Indexed: 10/25/2022]
Abstract
There are several evidences indicating that an impairment in attention-executive functions is present in prodromal Alzheimer's disease and predict future global cognitive decline. In particular, the issue of temporal orienting of attention in patients with mild cognitive impairment (MCI) due to Alzheimer's disease has been overlooked. The present research aimed to explore whether subtle deficits of cortical activation are present in these patients early in the course of the disease. We studied the upper-alpha event-related synchronization/desynchronization phenomenon during a paradigm of temporal orientation of attention. MCI patients (n = 27) and healthy elderly controls (n = 15) performed a task in which periodically omitted tones had to be predicted and their virtual onset time had to be marked by pressing a button. Single-trial responses were measured, respectively, before and after the motor response. Then, upper-alpha responses were compared to upper-alpha power during eyes-closed resting state. The time course of the task was characterized by two different behavioral conditions: (1) a pre-event epoch, in which the subject awaited the virtual onset of the omitted tone, (2) a post-event epoch (after button pressing), in which the subject was in a post-motor response condition. The principal findings are: (1) during the waiting epoch, only healthy elderly had an upper-alpha ERD at the level of both temporal and posterior brain regions; (2) during the post-motor epoch, the aMCI patients had a weaker upper-alpha ERS on prefrontal regions; (3) only healthy elderly showed a laterality effect: (a) during the waiting epoch, the upper-alpha ERD was greater at the level of the right posterior-temporal lead; during the post-motor epoch, the upper alpha ERS was greater on the left prefrontal lead. The relevance of these findings is that the weaker upper-alpha response observed in aMCI patients is evident even if the accuracy of the behavioral performance (i.e., button pressing) is still spared. This abnormal upper-alpha response might represent an early biomarker of the attention-executive network impairment in MCI due to Alzheimer's disease.
Collapse
Affiliation(s)
- Giuseppe Caravaglios
- Azienda Ospedaliera Cannizzaro, U.O.C. di Neurologia, Via Messina, 829, 95126, Catania, Italy,
| | | | | | | |
Collapse
|
80
|
Quantitative EEG in Alzheimer's disease: cognitive state, resting state and association with disease severity. Int J Psychophysiol 2014; 93:390-7. [PMID: 24933410 DOI: 10.1016/j.ijpsycho.2014.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Revised: 06/03/2014] [Accepted: 06/06/2014] [Indexed: 11/20/2022]
Abstract
BACKGROUND Quantitative electroencephalogram (qEEG) recorded during cognitive tasks has been shown to differentiate between patients with Alzheimer's disease (AD) and healthy individuals. However, the association between various qEEG markers recorded during mnestic paradigms and clinical measures of AD has not been studied in detail. OBJECTIVE To evaluate if 'cognitive' qEEG is a useful diagnostic option, particularly if memory paradigms are used as cognitive stimulators. METHODS This study is part of the Prospective Registry on Dementia in Austria (PRODEM), a multicenter dementia research project. A cohort of 79 probable AD patients was included in a cross-sectional analysis. qEEG recordings performed in resting states were compared with recordings during cognitively active states. Cognition was evoked with a face-name paradigm and a paired-associate word list task, respectively. Relative band powers, coherence and auto-mutual information were computed as functions of MMSE scores for the memory paradigms and during rest. Analyses were adjusted for the co-variables age, sex, duration of dementia and educational level. RESULTS MMSE scores explained 36-51% of the variances of qEEG-markers. Face-name encoding with eyes open was superior to resting state with eyes closed in relative theta and beta1 power as well as coherence, whereas relative alpha power and auto-mutual information yielded more significant results during resting state with eyes closed. The face-name task yielded stronger correlations with MMSE scores than the verbal memory task. CONCLUSION qEEG alterations recorded during mnestic activity, particularly face-name encoding showed the highest association with the MMSE and may serve as a clinically valuable marker for disease severity.
Collapse
|
81
|
Solodkin A, Chen EE, Van Hoesen GW, Heimer L, Shereen A, Kruggel F, Mastrianni J. In vivo parahippocampal white matter pathology as a biomarker of disease progression to Alzheimer's disease. J Comp Neurol 2014; 521:4300-17. [PMID: 23839862 DOI: 10.1002/cne.23418] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 06/14/2013] [Accepted: 06/19/2013] [Indexed: 01/18/2023]
Abstract
Noninvasive diagnostic tests for Alzheimer's disease (AD) are limited. Postmortem diagnosis is based on density and distribution of neurofibrillary tangles (NFTs) and amyloid-rich neuritic plaques. In preclinical stages of AD, the cells of origin for the perforant pathway within the entorhinal cortex are among the first to display NFTs, indicating its compromise in early stages of AD. We used diffusion tensor imaging (DTI) to assess the integrity of the parahippocampal white matter in mild cognitive impairment (MCI) and AD, as a first step in developing a noninvasive tool for early diagnosis. Subjects with AD (N = 9), MCI (N = 8), or no cognitive impairment (NCI; N = 20) underwent DTI-MRI. Fractional anisotropy (FA) and mean (MD) and radial (RD) diffusivity measured from the parahippocampal white matter in AD and NCI subjects differed greatly. Discriminant analysis in the MCI cases assigned statistical membership of 38% of MCI subjects to the AD group. Preliminary data 1 year later showed that all MCI cases assigned to the AD group either met the diagnostic criteria for probable AD or showed significant cognitive decline. Voxelwise analysis in the parahippocampal white matter revealed a progressive change in the DTI patterns in MCI and AD subjects: whereas converted MCI cases showed structural changes restricted to the anterior portions of this region, in AD the pathology was generalized along the entire anterior-posterior axis. The use of DTI for in vivo assessment of the parahippocampal white matter may be useful for identifying individuals with MCI at highest risk for conversion to AD and for assessing disease progression.
Collapse
Affiliation(s)
- Ana Solodkin
- Department of Anatomy and Neurobiology, UC Irvine Medical School, Irvine, California, 92697-3940; Department of Neurology, UC Irvine Medical School, Irvine, California, 92697-3940
| | | | | | | | | | | | | |
Collapse
|
82
|
Durazzo TC, Mattsson N, Weiner MW. Smoking and increased Alzheimer's disease risk: a review of potential mechanisms. Alzheimers Dement 2014; 10:S122-45. [PMID: 24924665 PMCID: PMC4098701 DOI: 10.1016/j.jalz.2014.04.009] [Citation(s) in RCA: 261] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Cigarette smoking has been linked with both increased and decreased risk for Alzheimer's disease (AD). This is relevant for the US military because the prevalence of smoking in the military is approximately 11% higher than in civilians. METHODS A systematic review of published studies on the association between smoking and increased risk for AD and preclinical and human literature on the relationships between smoking, nicotine exposure, and AD-related neuropathology was conducted. Original data from comparisons of smoking and never-smoking cognitively normal elders on in vivo amyloid imaging are also presented. RESULTS Overall, literature indicates that former/active smoking is related to a significantly increased risk for AD. Cigarette smoke/smoking is associated with AD neuropathology in preclinical models and humans. Smoking-related cerebral oxidative stress is a potential mechanism promoting AD pathology and increased risk for AD. CONCLUSIONS A reduction in the incidence of smoking will likely reduce the future prevalence of AD.
Collapse
Affiliation(s)
- Timothy C Durazzo
- Center for Imaging of Neurodegenerative Diseases (CIND), San Francisco VA Medical Center, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
| | - Niklas Mattsson
- Center for Imaging of Neurodegenerative Diseases (CIND), San Francisco VA Medical Center, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA; Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Michael W Weiner
- Center for Imaging of Neurodegenerative Diseases (CIND), San Francisco VA Medical Center, San Francisco, CA, USA; Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
83
|
Ma CC, Liu AJ, Liu AH, Zhou XY, Zhou SN. Electroencephalogram global field synchronization analysis: a new method for assessing the progress of cognitive decline in Alzheimer's disease. Clin EEG Neurosci 2014; 45:98-103. [PMID: 23986293 DOI: 10.1177/1550059413489669] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Global field synchronization (GFS) can measure functional synchronization in frequency-domain electroencephalogram (EEG) data. The aim of this study is to explore GFS values and its clinical significance for severity of cognitive decline in AD. EEGs were recorded from 37 AD patients and 37 age-matched healthy individuals. GFS values were calculated in delta, theta, alpha, beta 1, beta 2, beta 3, gamma, and full frequency bands. The Montreal Cognitive Assessment (MoCA) and Clinical Dementia Rating scale (CDR) were employed to assess symptom severity in AD patients. Correlation analysis, clustering analysis, and concordance analysis were performed to analyze the relationship between GFS values and MoCA scores in AD patients. GFS values of the beta 1, beta 2, beta 3, and full bands were lower in AD patients than in healthy individuals, and positively correlated with MoCA and CDR scores in the combined group (AD patients and healthy individuals). GFS values were positively correlated with MoCA socres in 3 beta bands and full bands, and with CDR scores in the delta band. There was a good concordance between K-means clustering algorithm calculating of GFS values and MoCA scoring (κ = .913, P < .001). In conclusion, the present results indicated that GFS can serve as an indicator of cognitive decline or impairment in AD patients. Furthermore, the GFS method of EEG holds considerable promise to distinguish mild cognitive impairment from serious cognitive impairment in patients with AD.
Collapse
|
84
|
Fani N, King TZ, Reiser E, Binder EB, Jovanovic T, Bradley B, Ressler KJ. FKBP5 genotype and structural integrity of the posterior cingulum. Neuropsychopharmacology 2014; 39:1206-13. [PMID: 24253961 PMCID: PMC3957115 DOI: 10.1038/npp.2013.322] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/27/2013] [Accepted: 11/11/2013] [Indexed: 11/09/2022]
Abstract
Alterations in the microarchitecture of the posterior cingulum (PC), a white matter tract proximal to the hippocampus that facilitates communication between the entorhinal and cingulate cortices, have been observed in individuals with psychiatric disorders, such as depression and post-traumatic stress disorder (PTSD). PC decrements may be a heritable source of vulnerability for the development of affective disorders; however, genetic substrates for these white matter abnormalities have not been identified. The FKBP5 gene product modulates glucocorticoid receptor function and has been previously associated with differential hippocampal structure, function, and affect disorder risk. Thus, FKBP5 is an attractive genetic target for investigations of PC integrity. We examined associations between PC integrity, measured through diffusion tensor imaging (DTI) and fractional anisotropy (FA; an index of white matter integrity), and polymorphisms in the FKBP5 SNP rs1360780 in a sample of 82 traumatized female civilians. Findings indicated that, compared with individuals without this allele, individuals who carried two 'risk' alleles for this FKBP5 SNP (T allele; previously associated with mood and anxiety disorder risk) demonstrated significantly lower FA in the left PC, even after statistically controlling for variance associated with age, trauma exposure, and PTSD symptoms. These data suggest that specific allelic variants for an FKBP5 polymorphism are associated with decrements in the left PC microarchitecture. These white matter abnormalities may be a heritable biological marker that indicates increased vulnerability for the development of psychiatric disorders, such as PTSD.
Collapse
Affiliation(s)
- Negar Fani
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Tricia Z King
- Department of Psychology, Georgia State University, Atlanta, GA, USA
| | - Emily Reiser
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Elisabeth B Binder
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Tanja Jovanovic
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Kerry J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
85
|
Mandas A, Dessì S. Mononuclear cells in dementia. Clin Chim Acta 2014; 431:278-87. [DOI: 10.1016/j.cca.2014.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 02/15/2014] [Accepted: 02/18/2014] [Indexed: 12/14/2022]
|
86
|
Fjell AM, Amlien IK, Sneve MH, Grydeland H, Tamnes CK, Chaplin TA, Rosa MGP, Walhovd KB. The Roots of Alzheimer's Disease: Are High-Expanding Cortical Areas Preferentially Targeted?†. Cereb Cortex 2014; 25:2556-65. [PMID: 24658616 DOI: 10.1093/cercor/bhu055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is regarded a human-specific condition, and it has been suggested that brain regions highly expanded in humans compared with other primates are selectively targeted. We calculated shared and unique variance in the distribution of AD atrophy accounted for by cortical expansion between macaque and human, affiliation to the default mode network (DMN), ontogenetic development and normal aging. Cortical expansion was moderately related to atrophy, but a critical discrepancy was seen in the medial temporo-parietal episodic memory network. Identification of "hotspots" and "coldspots" of expansion across several primate species did not yield compelling evidence for the hypothesis that highly expanded regions are specifically targeted. Controlling for distribution of atrophy in aging substantially attenuated the expansion-AD relationship. A path model showed that all variables explained unique variance in AD atrophy but were generally mediated through aging. This supports a systems-vulnerability model, where critical networks are subject to various negative impacts, aging in particular, rather than being selectively targeted in AD. An alternative approach is suggested, focused on the interplay of the phylogenetically old and preserved medial temporal lobe areas with more highly expanded association cortices governed by different principles of plasticity and stability.
Collapse
Affiliation(s)
- Anders M Fjell
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Norway Department of Physical Medicine and Rehabilitation, Unit of Neuropsychology, Oslo University Hospital, 0424 Norway
| | - Inge K Amlien
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Norway
| | - Markus H Sneve
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Norway
| | - Håkon Grydeland
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Norway
| | - Christian K Tamnes
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Norway
| | - Tristan A Chaplin
- Department of Physiology and Monash Vision Group, Monash University, Clayton, Victoria 3800, Australia ARC Centre of Excellence for Integrative Brain Function, Clayton, Victoria 3800, Australia
| | - Marcello G P Rosa
- Department of Physiology and Monash Vision Group, Monash University, Clayton, Victoria 3800, Australia ARC Centre of Excellence for Integrative Brain Function, Clayton, Victoria 3800, Australia
| | - Kristine B Walhovd
- Research Group for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373 Norway Department of Physical Medicine and Rehabilitation, Unit of Neuropsychology, Oslo University Hospital, 0424 Norway
| |
Collapse
|
87
|
Mulder ER, de Jong RA, Knol DL, van Schijndel RA, Cover KS, Visser PJ, Barkhof F, Vrenken H. Hippocampal volume change measurement: quantitative assessment of the reproducibility of expert manual outlining and the automated methods FreeSurfer and FIRST. Neuroimage 2014; 92:169-81. [PMID: 24521851 DOI: 10.1016/j.neuroimage.2014.01.058] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 01/23/2014] [Accepted: 01/31/2014] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND To measure hippocampal volume change in Alzheimer's disease (AD) or mild cognitive impairment (MCI), expert manual delineation is often used because of its supposed accuracy. It has been suggested that expert outlining yields poorer reproducibility as compared to automated methods, but this has not been investigated. AIM To determine the reproducibilities of expert manual outlining and two common automated methods for measuring hippocampal atrophy rates in healthy aging, MCI and AD. METHODS From the Alzheimer's Disease Neuroimaging Initiative (ADNI), 80 subjects were selected: 20 patients with AD, 40 patients with mild cognitive impairment (MCI) and 20 healthy controls (HCs). Left and right hippocampal volume change between baseline and month-12 visit was assessed by using expert manual delineation, and by the automated software packages FreeSurfer (longitudinal processing stream) and FIRST. To assess reproducibility of the measured hippocampal volume change, both back-to-back (BTB) MPRAGE scans available for each visit were analyzed. Hippocampal volume change was expressed in μL, and as a percentage of baseline volume. Reproducibility of the 1-year hippocampal volume change was estimated from the BTB measurements by using linear mixed model to calculate the limits of agreement (LoA) of each method, reflecting its measurement uncertainty. Using the delta method, approximate p-values were calculated for the pairwise comparisons between methods. Statistical analyses were performed both with inclusion and exclusion of visibly incorrect segmentations. RESULTS Visibly incorrect automated segmentation in either one or both scans of a longitudinal scan pair occurred in 7.5% of the hippocampi for FreeSurfer and in 6.9% of the hippocampi for FIRST. After excluding these failed cases, reproducibility analysis for 1-year percentage volume change yielded LoA of ±7.2% for FreeSurfer, ±9.7% for expert manual delineation, and ±10.0% for FIRST. Methods ranked the same for reproducibility of 1-year μL volume change, with LoA of ±218 μL for FreeSurfer, ±319 μL for expert manual delineation, and ±333 μL for FIRST. Approximate p-values indicated that reproducibility was better for FreeSurfer than for manual or FIRST, and that manual and FIRST did not differ. Inclusion of failed automated segmentations led to worsening of reproducibility of both automated methods for 1-year raw and percentage volume change. CONCLUSION Quantitative reproducibility values of 1-year microliter and percentage hippocampal volume change were roughly similar between expert manual outlining, FIRST and FreeSurfer, but FreeSurfer reproducibility was statistically significantly superior to both manual outlining and FIRST after exclusion of failed segmentations.
Collapse
Affiliation(s)
- Emma R Mulder
- Image Analysis Center, VU University Medical Center, Amsterdam, The Netherlands; Department of Radiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Remko A de Jong
- Image Analysis Center, VU University Medical Center, Amsterdam, The Netherlands; Department of Radiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Dirk L Knol
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| | - Ronald A van Schijndel
- Image Analysis Center, VU University Medical Center, Amsterdam, The Netherlands; Department of Information and Communication Technology, VU University Medical Center, Amsterdam, The Netherlands
| | - Keith S Cover
- Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands
| | - Pieter J Visser
- Department of Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Image Analysis Center, VU University Medical Center, Amsterdam, The Netherlands; Department of Radiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Hugo Vrenken
- Department of Radiology, VU University Medical Center, Amsterdam, The Netherlands; Department of Physics and Medical Technology, VU University Medical Center, Amsterdam, The Netherlands.
| | | |
Collapse
|
88
|
Costa A, Monaco M, Zabberoni S, Peppe A, Perri R, Fadda L, Iannarelli F, Caltagirone C, Carlesimo GA. Free and cued recall memory in Parkinson's disease associated with amnestic mild cognitive impairment. PLoS One 2014; 9:e86233. [PMID: 24465977 PMCID: PMC3900509 DOI: 10.1371/journal.pone.0086233] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 12/09/2013] [Indexed: 11/29/2022] Open
Abstract
The hypothesis has been advanced that memory disorders in individuals with Parkinson's disease (PD) are related to either retrieval or consolidation failure. However, the characteristics of the memory impairments of PD patients with amnestic mild cognitive impairment have not been clarified. This study was aimed at investigating whether memory deficits in PD patients with amnestic mild cognitive impairment (PDaMCI) are due to failure of retrieval or consolidation processes. Sixteen individuals with PDaMCI, 20 with amnestic mild cognitive impairment without PD (aMCINPD), and 20 healthy controls were recruited. Participants were administered the Free and Cued Selective Reminding Test. An index of cueing was computed for each subject to capture the advantage in retrieval of cued compared to free recall. Individuals with PDaMCI performed worse than healthy controls on the free recall (p<0.01) but not the cued recall (p>0.10) task, and they performed better than aMCINPD subjects on both recall measures (p<0.01). The index of cueing of subjects with PD was comparable to that of healthy controls (p>0.10) but it was significantly higher than that of the aMCINPD sample (p<0.01). Moreover, PD patients' performance on free recall trials was significantly predicted by scores on a test investigating executive functions (i.e., the Modified Card Sorting Test; p = 0.042). Findings of the study document that, in subjects with amnestic mild cognitive impairment associated to PD, episodic memory impairment is related to retrieval rather than to consolidation failure. The same data suggest that, in these individuals, memory deficits might be due to altered frontal-related executive functioning.
Collapse
Affiliation(s)
- Alberto Costa
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
| | - Marco Monaco
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
| | - Silvia Zabberoni
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
| | - Antonella Peppe
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
| | - Roberta Perri
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
| | - Lucia Fadda
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
- Dipartimento di Medicina dei Sistemi, Rome University “Tor Vergata”, Rome, Italy
| | - Francesca Iannarelli
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
| | - Carlo Caltagirone
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
- Dipartimento di Medicina dei Sistemi, Rome University “Tor Vergata”, Rome, Italy
| | - Giovanni A. Carlesimo
- Behavioural and Clinical Neurology Laboratory, I.R.C.C.S. Fondazione S. Lucia, Rome, Italy
- Dipartimento di Medicina dei Sistemi, Rome University “Tor Vergata”, Rome, Italy
| |
Collapse
|
89
|
Pereira MLF, Camargo MVZA, Aprahamian I, Forlenza OV. Eye movement analysis and cognitive processing: detecting indicators of conversion to Alzheimer's disease. Neuropsychiatr Dis Treat 2014; 10:1273-85. [PMID: 25031536 PMCID: PMC4096446 DOI: 10.2147/ndt.s55371] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A great amount of research has been developed around the early cognitive impairments that best predict the onset of Alzheimer's disease (AD). Given that mild cognitive impairment (MCI) is no longer considered to be an intermediate state between normal aging and AD, new paths have been traced to acquire further knowledge about this condition and its subtypes, and to determine which of them have a higher risk of conversion to AD. It is now known that other deficits besides episodic and semantic memory impairments may be present in the early stages of AD, such as visuospatial and executive function deficits. Furthermore, recent investigations have proven that the hippocampus and the medial temporal lobe structures are not only involved in memory functioning, but also in visual processes. These early changes in memory, visual, and executive processes may also be detected with the study of eye movement patterns in pathological conditions like MCI and AD. In the present review, we attempt to explore the existing literature concerning these patterns of oculomotor changes and how these changes are related to the early signs of AD. In particular, we argue that deficits in visual short-term memory, specifically in iconic memory, attention processes, and inhibitory control, may be found through the analysis of eye movement patterns, and we discuss how they might help to predict the progression from MCI to AD. We add that the study of eye movement patterns in these conditions, in combination with neuroimaging techniques and appropriate neuropsychological tasks based on rigorous concepts derived from cognitive psychology, may highlight the early presence of cognitive impairments in the course of the disease.
Collapse
Affiliation(s)
- Marta Lg Freitas Pereira
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Marina von Zuben A Camargo
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Ivan Aprahamian
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Orestes V Forlenza
- Laboratory of Neuroscience (LIM-27), Department and Institute of Psychiatry, Faculty of Medicine, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
90
|
Brain morphometry reproducibility in multi-center 3T MRI studies: A comparison of cross-sectional and longitudinal segmentations. Neuroimage 2013; 83:472-84. [DOI: 10.1016/j.neuroimage.2013.05.007] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 03/25/2013] [Accepted: 05/01/2013] [Indexed: 11/24/2022] Open
|
91
|
Hampel H, Lista S, Teipel SJ, Garaci F, Nisticò R, Blennow K, Zetterberg H, Bertram L, Duyckaerts C, Bakardjian H, Drzezga A, Colliot O, Epelbaum S, Broich K, Lehéricy S, Brice A, Khachaturian ZS, Aisen PS, Dubois B. Perspective on future role of biological markers in clinical therapy trials of Alzheimer's disease: a long-range point of view beyond 2020. Biochem Pharmacol 2013; 88:426-49. [PMID: 24275164 DOI: 10.1016/j.bcp.2013.11.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/13/2013] [Accepted: 11/13/2013] [Indexed: 10/26/2022]
Abstract
Recent advances in understanding the molecular mechanisms underlying various paths toward the pathogenesis of Alzheimer's disease (AD) has begun to provide new insight for interventions to modify disease progression. The evolving knowledge gained from multidisciplinary basic research has begun to identify new concepts for treatments and distinct classes of therapeutic targets; as well as putative disease-modifying compounds that are now being tested in clinical trials. There is a mounting consensus that such disease modifying compounds and/or interventions are more likely to be effectively administered as early as possible in the cascade of pathogenic processes preceding and underlying the clinical expression of AD. The budding sentiment is that "treatments" need to be applied before various molecular mechanisms converge into an irreversible pathway leading to morphological, metabolic and functional alterations that characterize the pathophysiology of AD. In light of this, biological indicators of pathophysiological mechanisms are desired to chart and detect AD throughout the asymptomatic early molecular stages into the prodromal and early dementia phase. A major conceptual development in the clinical AD research field was the recent proposal of new diagnostic criteria, which specifically incorporate the use of biomarkers as defining criteria for preclinical stages of AD. This paradigm shift in AD definition, conceptualization, operationalization, detection and diagnosis represents novel fundamental opportunities for the modification of interventional trial designs. This perspective summarizes not only present knowledge regarding biological markers but also unresolved questions on the status of surrogate indicators for detection of the disease in asymptomatic people and diagnosis of AD.
Collapse
Affiliation(s)
- Harald Hampel
- Université Pierre et Marie Curie, Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Pavillon François Lhermitte, Hôpital de la Salpêtrière, Paris, France.
| | - Simone Lista
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany.
| | - Stefan J Teipel
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany; German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Francesco Garaci
- Department of Diagnostic Imaging, Molecular Imaging, Interventional Radiology, and Radiotherapy, University of Rome "Tor Vergata", Rome, Italy; IRCCS San Raffaele Pisana, Rome and San Raffaele Cassino, Cassino, Italy
| | - Robert Nisticò
- Department of Physiology and Pharmacology, University of Rome "La Sapienza", Rome, Italy; IRCSS Santa Lucia Foundation, Rome, Italy
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; University College London Institute of Neurology, Queen Square, London, UK
| | - Lars Bertram
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Charles Duyckaerts
- Laboratoire de Neuropathologie Raymond-Escourolle, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France
| | - Hovagim Bakardjian
- IM2A - Institute of Memory and Alzheimer's Disease, Paris, France; IHU-A-ICM - Paris Institute of Translational Neurosciences Pitié-Salpêtrière University Hospital, Paris, France
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| | - Olivier Colliot
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épinière, UMR-S975 Paris, France; Inserm, U975, Paris, France; CNRS, UMR 7225, Paris, France; ICM - Institut du Cerveau et de la Moelle Épinière, Paris, France; INRIA, Aramis Team, Centre de Recherche Paris-Rocquencourt, France
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié Salpêtrière, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Karl Broich
- Federal Institute of Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Stéphane Lehéricy
- IHU-A-ICM - Paris Institute of Translational Neurosciences Pitié-Salpêtrière University Hospital, Paris, France; Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épinière, UMR-S975 Paris, France; Inserm, U975, Paris, France; CNRS, UMR 7225, Paris, France; ICM - Institut du Cerveau et de la Moelle Épinière, Paris, France
| | - Alexis Brice
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épinière, UMR-S975 Paris, France; Inserm, U975, Paris, France; CNRS, UMR 7225, Paris, France; ICM - Institut du Cerveau et de la Moelle Épinière, Paris, France; AP-HP, Hôpital de la Salpêtrière, Département de Génétique et Cytogénétique, Paris, France
| | | | - Paul S Aisen
- Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié Salpêtrière, Paris, France; Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
92
|
Abstract
Knowledge of aging and dementia is rapidly evolving with the aim of identifying individuals in the earliest stages of disease processes. Biomarkers allow clinicians to show the presence of a pathologic process and resultant synapse dysfunction and neurodegeneration, even in the earliest stages. This article focuses on biomarkers for mild cognitive impairment caused by Alzheimer disease, structural magnetic resonance imaging, fluorodeoxyglucose positron emission tomography (PET) or single-photon emission computed tomography, and PET with dopamine ligands. Although these biomarkers are useful, several limitations exist. Several new biomarkers are emerging and a more biological characterization of underlying pathophysiologic spectra may become possible.
Collapse
Affiliation(s)
- Meredith Wicklund
- Fellow, Division of Behavioral Neurology, Department of Neurology, Mayo Clinic, Rochester, MN
| | - Ronald C. Petersen
- Cora Kanow Professor of Alzheimer's Disease Research, Director, Mayo Alzheimer's Disease Research Center, Division of Behavioral Neurology, Department of Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
93
|
Bosco P, Ferri R, Salluzzo MG, Castellano S, Signorelli M, Nicoletti F, Nuovo SD, Drago F, Caraci F. Role of the Transforming-Growth-Factor-β1 Gene in Late-Onset Alzheimer's Disease: Implications for the Treatment. Curr Genomics 2013; 14:147-56. [PMID: 24082824 PMCID: PMC3637679 DOI: 10.2174/1389202911314020007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 01/31/2013] [Accepted: 02/01/2013] [Indexed: 11/22/2022] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common form of dementia in the elderly. LOAD has a complex and largely unknown etiology with strong genetic determinants. Genetics of LOAD is known to involve several genetic risk factors among which the Apolipoprotein E (APOE) gene seems to be the major recognized genetic determinant. Recent efforts have been made to identify other genetic factors involved in the pathophysiology of LOAD such as genes associated with a deficit of neurotrophic factors in the AD brain. Genetic variations of neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), and transforming-growth-factor-β1 (TGF-β1) are known to increase the risk to develop LOAD and have also been related to depression susceptibility in LOAD. Transforming-Growth-Factor-β1 (TGF-β1) is a neurotrophic factor that exerts neuroprotective effects against ß-amyloid-induced neurodegeneration. Recent evidence suggests that a specific impairment in the signaling of TGF-β is an early event in the pathogenesis of AD. TGF-β1 protein levels are predominantly under genetic control, and the TGF-β1 gene, located on chromosome 19q13.1–3, con-tains several single nucleotide polymorphisms (SNPs) upstream and in the transcript region, such as the SNP at codon +10 (T/C) and +25 (G/C), which is known to influence the level of expression of TGF-β1. In the present review, we summarize the current literature on genetic risk factors for LOAD, focusing on the role of the TGF-β1 gene, finally discussing the possible implications of these genetic studies for the selection of patients eligible for neuroprotective strategies in AD.
Collapse
Affiliation(s)
- Paolo Bosco
- IRCCS Associazione Oasi Maria S.S. - Institute for Research on Mental Retardation and Brain Aging, 94018 Troina, Enna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Morellini F. Spatial memory tasks in rodents: what do they model? Cell Tissue Res 2013; 354:273-86. [PMID: 23793547 DOI: 10.1007/s00441-013-1668-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/23/2013] [Indexed: 02/08/2023]
Abstract
The analysis of spatial learning and memory in rodents is commonly used to investigate the mechanisms underlying certain forms of human cognition and to model their dysfunction in neuropsychiatric and neurodegenerative diseases. Proper interpretation of rodent behavior in terms of spatial memory and as a model of human cognitive functions is only possible if various navigation strategies and factors controlling the performance of the animal in a spatial task are taken into consideration. The aim of this review is to describe the experimental approaches that are being used for the study of spatial memory in rats and mice and the way that they can be interpreted in terms of general memory functions. After an introduction to the classification of memory into various categories and respective underlying neuroanatomical substrates, I explain the concept of spatial memory and its measurement in rats and mice by analysis of their navigation strategies. Subsequently, I describe the most common paradigms for spatial memory assessment with specific focus on methodological issues relevant for the correct interpretation of the results in terms of cognitive function. Finally, I present recent advances in the use of spatial memory tasks to investigate episodic-like memory in mice.
Collapse
Affiliation(s)
- Fabio Morellini
- AG Experimentelle Neuropädiatrie, Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany,
| |
Collapse
|
95
|
Association between Sirtuin 2 gene rs10410544 polymorphism and depression in Alzheimer's disease in two independent European samples. J Neural Transm (Vienna) 2013; 120:1709-15. [PMID: 23712749 DOI: 10.1007/s00702-013-1045-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/07/2013] [Indexed: 01/07/2023]
Abstract
Among the several genes associated with late-onset Alzheimer's disease (LOAD), recently, Sirtuin genes have roused a growing interest because of their involvement in metabolic homeostasis and in brain aging. Particularly SIRT2 gene has been associated with Alzheimer's disease (AD) as well as with mood disorders. The aim of this study is to investigate the possible associations between Sirtuin 2 gene (SIRT2) rs10410544 polymorphism and AD as well as depression in AD. In addition, we performed some exploratory analyses to investigate possible associations between the rs10410544 genotype and clinical features. We investigated these associations in two independent samples: the first one was composed of 275 Greek inhabitants and 117 patients; the second sample counted 181 Italian people and 43 patients. All patients were affected by LOAD. We failed to find any association between rs10410544 genotype and AD in the two samples. On the other hand, we found an association between the single nucleotide polymorphism (SNP) and depressive symptomatology (in the total sample p = 0.002), which was modulated by the tumor necrosis factor (TNF) values. Particularly, TT genotype seems to be protective versus depression. Finally, in the exploratory analyses, we found that the TT genotype was associated with earlier AD onset and a longer duration of the illness. In conclusion, we confirmed the association between SIRT2 gene and mood disturbances, although in AD patients. Further, we provided evidence that the TT genotype may be protective versus depressive symptoms, allowing an easier and thus earlier diagnosis of AD. This awareness may lead to a more detailed approach to these patients concerning diagnosis and therapy.
Collapse
|
96
|
Liu Y, Mattila J, Ruiz MÁM, Paajanen T, Koikkalainen J, van Gils M, Herukka SK, Waldemar G, Lötjönen J, Soininen H. Predicting AD conversion: comparison between prodromal AD guidelines and computer assisted PredictAD tool. PLoS One 2013; 8:e55246. [PMID: 23424625 PMCID: PMC3570420 DOI: 10.1371/journal.pone.0055246] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 12/27/2012] [Indexed: 11/28/2022] Open
Abstract
Purpose To compare the accuracies of predicting AD conversion by using a decision support system (PredictAD tool) and current research criteria of prodromal AD as identified by combinations of episodic memory impairment of hippocampal type and visual assessment of medial temporal lobe atrophy (MTA) on MRI and CSF biomarkers. Methods Altogether 391 MCI cases (158 AD converters) were selected from the ADNI cohort. All the cases had baseline cognitive tests, MRI and/or CSF levels of Aβ1–42 and Tau. Using baseline data, the status of MCI patients (AD or MCI) three years later was predicted using current diagnostic research guidelines and the PredictAD software tool designed for supporting clinical diagnostics. The data used were 1) clinical criteria for episodic memory loss of the hippocampal type, 2) visual MTA, 3) positive CSF markers, 4) their combinations, and 5) when the PredictAD tool was applied, automatically computed MRI measures were used instead of the visual MTA results. The accuracies of diagnosis were evaluated with the diagnosis made 3 years later. Results The PredictAD tool achieved the overall accuracy of 72% (sensitivity 73%, specificity 71%) in predicting the AD diagnosis. The corresponding number for a clinician’s prediction with the assistance of the PredictAD tool was 71% (sensitivity 75%, specificity 68%). Diagnosis with the PredictAD tool was significantly better than diagnosis by biomarkers alone or the combinations of clinical diagnosis of hippocampal pattern for the memory loss and biomarkers (p≤0.037). Conclusion With the assistance of PredictAD tool, the clinician can predict AD conversion more accurately than the current diagnostic criteria.
Collapse
Affiliation(s)
- Yawu Liu
- Department of Neurology, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
- Department of Clinical Radiology, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Jussi Mattila
- VTT Technical Research Centre of Finland, Tampere, Finland
| | - Miguel Ángel Muñoz Ruiz
- Department of Neurology, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Teemu Paajanen
- Department of Neurology, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | | | - Mark van Gils
- VTT Technical Research Centre of Finland, Tampere, Finland
| | - Sanna-Kaisa Herukka
- Department of Neurology, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Gunhild Waldemar
- Department of Neurology, Memory Disorders Research Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jyrki Lötjönen
- VTT Technical Research Centre of Finland, Tampere, Finland
| | - Hilkka Soininen
- Department of Neurology, University of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
- * E-mail:
| | | |
Collapse
|
97
|
Hertze J, Palmqvist S, Minthon L, Hansson O. Tau pathology and parietal white matter lesions have independent but synergistic effects on early development of Alzheimer's disease. Dement Geriatr Cogn Dis Extra 2013; 3:113-22. [PMID: 23687506 PMCID: PMC3656673 DOI: 10.1159/000348353] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND White matter lesions (WMLs) are a common finding in patients with dementia. This study investigates the relationship between WMLs, hyperphosphorylated tau (P-tau) in cerebrospinal fluid (CSF) and apolipoprotein E (APOE) ε4 genotype in prodromal Alzheimer's disease (AD). METHODS Baseline levels of tau, P-tau and β-amyloid 1-42 in CSF, the presence of WMLs in the brain, and the APOE genotype were ascertained in 159 patients with mild cognitive impairment (MCI) and 38 cognitively healthy controls. RESULTS After 5.7 years, 58 patients had developed AD. In this group, patients with normal levels of CSF P-tau had higher levels of WMLs in the parietal regions than those with pathological P-tau levels (p < 0.05). Also, patients without APOE ε4 alleles had more WMLs in the parietal lobes than those with at least one allele (p < 0.05). MCI patients with pathological P-tau levels and parietal WMLs showed a greater risk of developing AD than those with just one of the two pathological parameters. CONCLUSIONS We suggest that WMLs in parietal lobes and tau pathology likely have independent but synergistic effects on the reduction of the cognitive reserve capacity of the brain. In patients with a more low-grade AD pathology, WMLs in the parietal lobes might increase the risk of developing dementia.
Collapse
Affiliation(s)
- Joakim Hertze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Lennart Minthon
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Lund, Sweden
| |
Collapse
|
98
|
Peng Y, Huang S, Cheng B, Nie X, Enhe J, Feng C, Fu X. Mesenchymal stem cells: a revolution in therapeutic strategies of age-related diseases. Ageing Res Rev 2013; 12:103-15. [PMID: 22569401 DOI: 10.1016/j.arr.2012.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/11/2012] [Accepted: 04/23/2012] [Indexed: 02/06/2023]
Abstract
The great evolutionary biologist Theodosius Dobzhansky once said: "Nothing in biology makes sense except in the light of evolution". Aging is a complex biological phenomenon and the factors governing the process of aging and age-related diseases are only beginning to be understood, oxidative stress, telomere shortening in DNA components and genetic changes were shown to be the mainly regulating mechanisms during the recent decades. Although a considerable amount of both animal and clinical data that demonstrate the extensive and safe use of mesenchymal stromal cells (MSCs) is available, the precise summarization and identification of MSCs in age-related diseases remains a challenge. Along this line, this review discussed several typical age-related diseases for which MSCs have been proved to confer protection and put forward a hypothesis for the association among MSCs and age-related diseases from an evolutionary perspective. Above all, we hope further and more research efforts could be aroused to elucidate the role and mechanisms that MSCs involved in the age-related diseases.
Collapse
|
99
|
Lee S, Tong M, Hang S, Deochand C, de la Monte S. CSF and Brain Indices of Insulin Resistance, Oxidative Stress and Neuro-Inflammation in Early versus Late Alzheimer's Disease. ACTA ACUST UNITED AC 2013; 3:128. [PMID: 25035815 PMCID: PMC4096626 DOI: 10.4172/2161-0460.1000128] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer’s disease (AD) is characterized by progressive impairments in cognitive and behavioral functions with deficits in learning, memory and executive reasoning. Growing evidence points toward brain insulin and insulin-like growth factor (IGF) resistance-mediated metabolic derangements as critical etiologic factors in AD. This suggests that indices of insulin/IGF resistance and their consequences, i.e. oxidative stress, neuro-inflammation, and reduced neuronal plasticity, should be included in biomarker panels for AD. Herein, we examine a range of metabolic, inflammatory, stress, and neuronal plasticity related proteins in early AD, late AD, and aged control postmortem brain, postmortem ventricular fluid (VF), and clinical cerebrospinal fluid (CSF) samples. In AD brain, VF, and CSF samples the trends with respect to alterations in metabolic, neurotrophin, and stress indices were similar, but for pro-inflammatory cytokines, the patterns were discordant. With the greater severities of dementia and neurodegeneration, the differences from control were more pronounced for late AD (VF and brain) than early or moderate AD (brain, VF and CSF). The findings suggest that the inclusion of metabolic, neurotrophin, stress biomarkers in AβPP-Aβ+pTau CSF-based panels could provide more information about the status and progression of neurodegeneration, as well as aid in predicting progression from early- to late-stage AD. Furthermore, standardized multi-targeted molecular assays of neurodegeneration could help streamline postmortem diagnoses, including assessments of AD severity and pathology.
Collapse
Affiliation(s)
- Sarah Lee
- Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Providence, RI, USA
| | - Steven Hang
- Department of Medicine, Warren Alpert Medical School, Providence, RI, USA
| | - Chetram Deochand
- Departments of Medicine, Rhode Island Hospital, Brown University, Providence, RI, USA
| | - Suzanne de la Monte
- Department of Medicine, Pathology (Neuropathology), Neurology and Neurosurgery, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
100
|
Hardenacke K, Kuhn J, Lenartz D, Maarouf M, Mai JK, Bartsch C, Freund HJ, Sturm V. Stimulate or degenerate: deep brain stimulation of the nucleus basalis Meynert in Alzheimer dementia. World Neurosurg 2012; 80:S27.e35-43. [PMID: 23246738 DOI: 10.1016/j.wneu.2012.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 11/16/2012] [Accepted: 12/07/2012] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Deep brain stimulation (DBS) is a therapeutically effective neurosurgical method originally applied in movement disorders. Over time, the application of DBS has increasingly been considered as a therapeutic option for several neuropsychiatric disorders, including Gilles de la Tourette syndrome, obsessive compulsive disorder, major depression and addiction. Latest research suggests beneficial effects of DBS in Alzheimer dementia (AD). Because of the high prevalence and the considerable burden of the disease, we endeavored to discuss and reveal the challenges of DBS in AD. METHODS Recent literature on the pathophysiology of AD, including translational data and human studies, has been studied to generate a fundamental hypothesis regarding the effects of electrical stimulation on cognition and to facilitate our ongoing pilot study regarding DBS of the nucleus basalis Meynert (NBM) in patients with AD. RESULTS It is hypothesized that DBS in the nucleus basalis Meynert could probably improve or at least stabilize memory and cognitive functioning in patients with AD by facilitating neural oscillations and by enhancing the synthesis of nerve growth factors. CONCLUSIONS Considering the large number of patients suffering from AD, there is a great need for novel and effective treatment methods. Our research provides insights into the theoretical background of DBS in AD. Providing that our hypothesis will be validated by our ongoing pilot study, DBS could be an opportunity in the treatment of AD.
Collapse
Affiliation(s)
- Katja Hardenacke
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|