184 results on '"Marc Suárez-Calvet"'
Search Results
2. Plasma brain-derived tau is an amyloid-associated neurodegeneration biomarker in Alzheimer’s disease
- Author
-
Fernando Gonzalez-Ortiz, Bjørn-Eivind Kirsebom, José Contador, Jordan E. Tanley, Per Selnes, Berglind Gísladóttir, Lene Pålhaugen, Mathilde Suhr Hemminghyth, Jonas Jarholm, Ragnhild Skogseth, Geir Bråthen, Gøril Grøndtvedt, Atle Bjørnerud, Sandra Tecelao, Knut Waterloo, Dag Aarsland, Aida Fernández-Lebrero, Greta García-Escobar, Irene Navalpotro-Gómez, Michael Turton, Agnes Hesthamar, Przemyslaw R. Kac, Johanna Nilsson, Jose Luchsinger, Kathleen M. Hayden, Peter Harrison, Albert Puig-Pijoan, Henrik Zetterberg, Timothy M. Hughes, Marc Suárez-Calvet, Thomas K. Karikari, Tormod Fladby, and Kaj Blennow
- Subjects
Science - Abstract
Abstract Staging amyloid-beta (Aβ) pathophysiology according to the intensity of neurodegeneration could identify individuals at risk for cognitive decline in Alzheimer’s disease (AD). In blood, phosphorylated tau (p-tau) associates with Aβ pathophysiology but an AD-type neurodegeneration biomarker has been lacking. In this multicenter study (n = 1076), we show that brain-derived tau (BD-tau) in blood increases according to concomitant Aβ (“A”) and neurodegeneration (“N”) abnormalities (determined using cerebrospinal fluid biomarkers); We used blood-based A/N biomarkers to profile the participants in this study; individuals with blood-based p-tau+/BD-tau+ profiles had the fastest cognitive decline and atrophy rates, irrespective of the baseline cognitive status. Furthermore, BD-tau showed no or much weaker correlations with age, renal function, other comorbidities/risk factors and self-identified race/ethnicity, compared with other blood biomarkers. Here we show that blood-based BD-tau is a biomarker for identifying Aβ-positive individuals at risk of short-term cognitive decline and atrophy, with implications for clinical trials and implementation of anti-Aβ therapies.
- Published
- 2024
- Full Text
- View/download PDF
3. Omega‐3 blood biomarkers relate to brain glucose uptake in individuals at risk of Alzheimer's disease dementia
- Author
-
Iolanda Lázaro, Oriol Grau‐Rivera, Marc Suárez‐Calvet, Karine Fauria, Carolina Minguillón, Mahnaz Shekari, Carles Falcón, Marina García‐Prat, Jordi Huguet, José Luis Molinuevo, Juan‐Domingo Gispert, Aleix Sala‐Vila, and for the ALFA study
- Subjects
biomarkers ,diet ,fatty fish ,fish oil ,n‐3 fatty acids ,nuts ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract INTRODUCTION Brain glucose hypometabolism is a preclinical feature of Alzheimer's disease (AD). Dietary omega‐3 fatty acids promote brain glucose metabolism, but clinical research is incipient. Circulating omega‐3s objectively reflect their dietary intake. METHODS This was a cross‐sectional study in 320 cognitively unimpaired participants at increased risk of AD dementia. Using lipidomics, we determined blood docosahexaenoic (DHA) and alpha‐linolenic (ALA) acid levels (omega‐3s from marine and plant origin, respectively). We assessed brain glucose metabolism using [18‐F]‐fluorodeoxyglucose (FDG) positron emission tomography (PET). RESULTS Blood ALA directly related to FDG uptake in brain areas known to be affected in AD. Stronger associations were observed in apolipoprotein E ε4 carriers and homozygotes. For DHA, significant direct associations were restricted to amyloid beta–positive tau‐positive participants. DISCUSSION Blood omega‐3 directly relate to preserved glucose metabolism in AD‐vulnerable brain regions in individuals at increased risk of AD dementia. This adds to the benefits of omega‐3 supplementation in the preclinical stage of AD dementia. Highlights Blood omega‐3s were related to brain glucose uptake in participants at risk of Alzheimer's disease (AD) dementia. Complementary associations were observed for omega‐3 from marine and plant sources. Foods rich in omega‐3 might be useful in early features of AD.
- Published
- 2024
- Full Text
- View/download PDF
4. NeuroToolKit Data Hackathon: advancing data collaboration in Alzheimer's disease
- Author
-
Craig Ritchie, Kaj Blennow, Juan Domingo Gispert, Sterling Johnson, Ingrid van Maurik, Lisa Vermunt, Marc Suárez-Calvet, Caitlin P. McHugh, Matthew H. S. Clement, Alexandra Anastasiu, Eugen Rosenfeld, Oana Cosma, Chad A. Logan, Frances-Catherine Quevenco, Mariana Castro Dias, and Margherita Carboni
- Subjects
Alzheimer's disease ,cloud-based workbench ,collaborative research ,NTKApp ,online community ,virtual platform ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Published
- 2024
- Full Text
- View/download PDF
5. Awareness of episodic memory and meta-cognitive profiles: associations with cerebrospinal fluid biomarkers at the preclinical stage of the Alzheimer’s continuum
- Author
-
David López-Martos, Marc Suárez-Calvet, Marta Milà-Alomà, Juan Domingo Gispert, Carolina Minguillon, Clara Quijano-Rubio, Gwendlyn Kollmorgen, Henrik Zetterberg, Kaj Blennow, Oriol Grau-Rivera, and Gonzalo Sánchez-Benavides
- Subjects
Alzheimer’s disease ,preclinical ,awareness ,episodic memory ,biomarkers ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
IntroductionThe lack of cognitive awareness, anosognosia, is a clinical deficit in Alzheimer’s disease (AD) dementia. However, an increased awareness of cognitive function, hypernosognosia, may serve as a marker in the preclinical stage. Subjective cognitive decline (SCD) might correspond to the initial symptom in the dynamic trajectory of awareness, but SCD might be absent along with low awareness of actual cognitive performance in the preclinical stage. We hypothesized that distinct meta-cognitive profiles, both hypernosognosia and anosognosia, might be identified in preclinical-AD. This research evaluated the association between cerebrospinal fluid (CSF) AD biomarkers and the awareness of episodic memory, further exploring dyadic (participant-partner) SCD reports, in the preclinical Alzheimer’s continuum.MethodsWe analyzed 314 cognitively unimpaired (CU) middle-aged individuals (mean age: 60, SD: 4) from the ALFA+ cohort study. Episodic memory was evaluated with the delayed recall from the Memory Binding Test (MBT). Awareness of episodic memory, meta-memory, was defined as the normalized discrepancy between objective and subjective performance. SCD was defined using self-report, and dyadic SCD profiles incorporated the study partner’s report using parallel SCD-Questionnaires. The relationship between CSF Aβ42/40 and CSF p-tau181 with meta-memory was evaluated with multivariable regression models. The role of SCD and the dyadic contingency was explored with the corresponding stratified analysis.ResultsCSF Aβ42/40 was non-linearly associated with meta-memory, showing an increased awareness up to Aβ-positivity and a decreased awareness beyond this threshold. In the non-SCD subset, the non-linear association between CSF Aβ42/40 and meta-memory persisted. In the SCD subset, higher Aβ-pathology was linearly associated with increased awareness. Individuals presenting only study partner’s SCD, defined as unaware decliners, exhibited higher levels of CSF p-tau181 correlated with lower meta-memory performance.DiscussionThese results suggested that distinct meta-cognitive profiles can be identified in preclinical-AD. While most individuals might experience an increased awareness associated with the entrance in the AD continuum, hypernosognosia, some might be already losing insight and stepping into the anosognosic trajectory. This research reinforced that an early anosognosic profile, although at increased risk of AD-related decline, might be currently overlooked considering actual diagnostic criteria, and therefore its medical attention delayed.
- Published
- 2024
- Full Text
- View/download PDF
6. The mediating role of neuroimaging-derived biological brain age in the association between risk factors for dementia and cognitive decline in middle-aged and older individuals without cognitive impairment: a cohort study
- Author
-
Irene Cumplido-Mayoral, MSc, Anna Brugulat-Serrat, PhD, Gonzalo Sánchez-Benavides, PhD, Armand González-Escalante, MSc, Federica Anastasi, PhD, Marta Milà-Alomà, PhD, David López-Martos, MSc, Muge Akinci, MSc, Carles Falcón, PhD, Mahnaz Shekari, MSc, Raffaele Cacciaglia, PhD, Eider M Arenaza-Urquijo, PhD, Carolina Minguillón, PhD, Karine Fauria, PhD, José Luis Molinuevo, MD PhD, Marc Suárez-Calvet, MD PhD, Oriol Grau-Rivera, MD PhD, Verónica Vilaplana, PhD, Juan Domingo Gispert, PhD, R AQUITE AGUILAR, A BETETA GORRITI, A BRUGULAT SERRAT, R E CACCIAGLIA, L CANALS GISPERT, A CAÑAS MARTINEZ, M DEL CAMPO MILAN, C DEULOFEU GOMEZ, R DOMINGUEZ IGLESIAS, M EMILIO, K M E FAURIA, A FERNANDEZ, S D FUENTES JULIAN, P GENIUS SERRA, J D GISPERT LOPEZ, A GONZALEZ ESCALANTE, O GRAU RIVERA, L HERNANDEZ PENAS, G HUESA RODRÍGUEZ, J HUGUET NINOU, L IGLESIAS GAMEZ, I KNEZEVIC, P MARNE ALVAREZ, T MENCHON DIAZ, C MINGUILLON GIL, E PALACIOS, M PASCUAL, W PELKMANS, A POLO BALLESTER, S PRADAS MENDEZ, I A RADOI, B RODRIGUEZ FERNANDEZ, L ROS FREIXEDES, A SALA-VILA, G A SANCHEZ BENAVIDES, M SHEKARI, L SOLSONA HARSTER, A SOTERAS PRAT, L STANKEVICIUTE, M SUAREZ CALVET, M VILANOVA JARAMILLO, and N VILOR TEJEDOR
- Subjects
Geriatrics ,RC952-954.6 ,Medicine - Abstract
Summary: Background: Neuroimaging-based brain-age delta has been shown to be a mediator linking cardiovascular risk factors to cognitive function. We aimed to assess the mediating role of brain-age delta in the association between modifiable risk factors of dementia and longitudinal cognitive decline in middle-aged and older individuals who are asymptomatic, stratified by Alzheimer's disease pathology. We also explored whether the mediation effect is specific to cognitive domain. Methods: In this cohort study, we included participants from the ALFA+ cohort aged between 45 years and 65 years who were cognitively unimpaired and who had available structural MRI, cerebrospinal fluid β-amyloid (Aβ)42 and Aβ40 measurements obtained within 1 year of each other, modifiable risk factors assessment, and cognitive evaluation over 3 years. Participants were recruited from the Barcelonaβeta Brain Research Center (Barcelona, Spain). Included individuals underwent a first assessment between Oct 25, 2016, and Jan 28, 2020, and a follow-up cognitive assessment 3·28 (SD 0·27) years later. We computed brain-age delta and composites of different cognitive function domains (preclinical Alzheimer's cognitive composite [PACC], attention, executive function, episodic memory, visual processing, and language). We used partial least squares path modelling to explore mediation effects in the associations between modifiable risk factors (including cardiovascular, mental health, mood, metabolic or endocrine history, and alcohol use) and changes in cognitive composites. To assess the role of Alzheimer's disease pathology, we computed separate models for Aβ-negative and Aβ-positive individuals. Findings: Of the 419 participants enrolled in ALFA+, 302 met our inclusion criteria, of which 108 participants were classified as Aβ-positive and 194 as Aβ-negative. In Aβ-positive individuals, brain-age delta partially mediated (percent mediation proportion 15·73% [95% CI 14·22–16·66]) the association between modifiable risk factors and decline in overall cognition (across cognitive domains). Brain-age delta fully mediated (mediation proportion 28·03% [26·25–29·21]) the effect of modifiable risk factors on the PACC, wherein increased values for risk factors correlated with an older brain-age delta, and, consequently, an older brain-age delta was linked to greater PACC decline. This effect appears to be primarily driven by memory decline. Mediation was not significant in Aβ-negative individuals (3·52% [0·072–4·17]) on PACC, although path coefficients were not significantly different from those in the Aβ-positive group. Interpretation: Our findings suggest that brain-age delta captures the association between modifiable risk factors and longitudinal cognitive decline in middle-aged and older people. In asymptomatic middle-aged and older individuals who are Aβ-positive, the pathology might be the strongest driver of cognitive decline, whereas the effect of risk factors is smaller. Our results highlight the potential of brain-age delta as an objective outcome measure for preventive lifestyle interventions targeting cognitive decline. Funding: La Caixa Foundation, the TriBEKa Imaging Platform, and the Universities and Research Secretariat of the Catalan Government. Translation: For the Spanish translation of the abstract see Supplementary Materials section.
- Published
- 2024
- Full Text
- View/download PDF
7. Clinical performance and head-to-head comparison of CSF p-tau235 with p-tau181, p-tau217 and p-tau231 in two memory clinic cohorts
- Author
-
Juan Lantero-Rodriguez, Agathe Vrillon, Aida Fernández-Lebrero, Paula Ortiz-Romero, Anniina Snellman, Laia Montoliu-Gaya, Wagner S. Brum, Emmanuel Cognat, Julien Dumurgier, Albert Puig-Pijoan, Irene Navalpotro-Gómez, Greta García-Escobar, Thomas K. Karikari, Eugeen Vanmechelen, Nicholas J. Ashton, Henrik Zetterberg, Marc Suárez-Calvet, Claire Paquet, and Kaj Blennow
- Subjects
Alzheimer’s disease ,CSF ,Biomarkers ,p-tau235 ,p-tau181 ,p-tau217 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Cerebrospinal fluid (CSF) p-tau235 is a novel biomarker highly specific of Alzheimer’s disease (AD). However, CSF p-tau235 has only been studied in well-characterized research cohorts, which do not fully reflect the patient landscape found in clinical settings. Therefore, in this multicentre study, we investigated the performance of CSF p-tau235 to detect symptomatic AD in clinical settings and compared it with CSF p-tau181, p-tau217 and p-tau231. Methods CSF p-tau235 was measured using an in-house single molecule array (Simoa) assay in two independent memory clinic cohorts: Paris cohort (Lariboisière Fernand-Widal University Hospital Paris, France; n=212) and BIODEGMAR cohort (Hospital del Mar, Barcelona, Spain; n=175). Patients were classified by the syndromic diagnosis (cognitively unimpaired [CU], mild cognitive impairment [MCI] or dementia) and their biological diagnosis (amyloid-beta [Aβ]+ or Aβ $$-$$ - ). Both cohorts included detailed cognitive assessments and CSF biomarker measurements (clinically validated core AD biomarkers [Lumipulse CSF Aβ1–42/40 ratio, p-tau181 and t-tau] and in-house developed Simoa CSF p-tau181, p-tau217 and p-tau231). Results High CSF p-tau235 levels were strongly associated with CSF amyloidosis regardless of the clinical diagnosis, being significantly increased in MCI Aβ+ and dementia Aβ+ when compared with all other Aβ− groups (Paris cohort: P ˂0.0001 for all; BIODEGMAR cohort: P ˂0.05 for all). CSF p-tau235 was pronouncedly increased in the A+T+ profile group compared with A−T− and A+T− groups (P ˂0.0001 for all). Moreover, CSF p-tau235 demonstrated high diagnostic accuracies identifying CSF amyloidosis in symptomatic cases (AUCs=0.86 to 0.96) and discriminating AT groups (AUCs=0.79 to 0.98). Overall, CSF p-tau235 showed similar performances to CSF p-tau181 and CSF p-tau231 when discriminating CSF amyloidosis in various scenarios, but lower than CSF p-tau217. Finally, CSF p-tau235 associated with global cognition and memory domain in both cohorts. Conclusions CSF p-tau235 was increased with the presence of CSF amyloidosis in two independent memory clinic cohorts. CSF p-tau235 accurately identified AD in both MCI and dementia patients. Overall, the diagnostic performance of CSF p-tau235 was comparable to that of other CSF p-tau measurements, indicating its suitability to support a biomarker-based AD diagnosis in clinical settings.
- Published
- 2023
- Full Text
- View/download PDF
8. APOE -ε4 modulates the association between regional amyloid deposition and cognitive performance in cognitively unimpaired middle-aged individuals
- Author
-
Anna Brugulat-Serrat, Gonzalo Sánchez-Benavides, Raffaele Cacciaglia, Gemma Salvadó, Mahnaz Shekari, Lyduine E. Collij, Christopher Buckley, Bart N. M. van Berckel, Andrés Perissinotti, Aida Niñerola-Baizán, Marta Milà-Alomà, Natàlia Vilor-Tejedor, Grégory Operto, Carles Falcon, Oriol Grau-Rivera, Eider M. Arenaza-Urquijo, Carolina Minguillón, Karine Fauria, José Luis Molinuevo, Marc Suárez-Calvet, Juan Domingo Gispert, and the ALFA Study
- Subjects
Alzheimer’s disease ,Amyloid PET ,Visual read ,Memory ,Executive function ,APOE-ε4 ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 - Abstract
Abstract Purpose To determine whether the APOE-ε4 allele modulates the relationship between regional β-amyloid (Aβ) accumulation and cognitive change in middle-aged cognitively unimpaired (CU) participants. Methods The 352 CU participants (mean aged 61.1 [4.7] years) included completed two cognitive assessments (average interval 3.34 years), underwent [18F]flutemetamol Aβ positron emission tomography (PET), T1w magnetic resonance imaging (MRI), as well as APOE genotyping. Global and regional Aβ PET positivity was assessed across five regions-of-interest by visual reading (VR) and regional Centiloids. Linear regression models were developed to examine the interaction between regional and global Aβ PET positivity and APOE-ε4 status on longitudinal cognitive change assessed with the Preclinical Alzheimer’s Cognitive Composite (PACC), episodic memory, and executive function, after controlling for age, sex, education, cognitive baseline scores, and hippocampal volume. Results In total, 57 participants (16.2%) were VR+ of whom 41 (71.9%) were APOE-ε4 carriers. No significant APOE-ε4*global Aβ PET interactions were associated with cognitive change for any cognitive test. However, APOE-ε4 carriers who were VR+ in temporal areas (n = 19 [9.81%], p = 0.04) and in the striatum (n = 8 [4.14%], p = 0.01) exhibited a higher decline in the PACC. The temporal areas findings were replicated when regional PET positivity was determined with Centiloid values. Regionally, VR+ in the striatum was associated with higher memory decline. As for executive function, interactions between APOE-ε4 and regional VR+ were found in temporal and parietal regions, and in the striatum. Conclusion CU APOE-ε4 carriers with a positive Aβ PET VR in regions known to accumulate amyloid at later stages of the Alzheimer’s disease (AD) continuum exhibited a steeper cognitive decline. This work supports the contention that regional VR of Aβ PET might convey prognostic information about future cognitive decline in individuals at higher risk of developing AD. ClinicalTrials.gov Identifier: NCT02485730. Registered 20 June 2015 https://clinicaltrials.gov/ct2/show/NCT02485730 and ClinicalTrials.gov Identifier:NCT02685969. Registered 19 February 2016 https://clinicaltrials.gov/ct2/show/NCT02685969 .
- Published
- 2023
- Full Text
- View/download PDF
9. sTREM2 is associated with amyloid‐related p‐tau increases and glucose hypermetabolism in Alzheimer's disease
- Author
-
Davina Biel, Marc Suárez‐Calvet, Paul Hager, Anna Rubinski, Anna Dewenter, Anna Steward, Sebastian Roemer, Michael Ewers, Christian Haass, Matthias Brendel, Nicolai Franzmeier, and for the Alzheimer's Disease Neuroimaging Initiative (ADNI)
- Subjects
Alzheimer's disease ,beta‐amyloid ,glucose metabolism ,p‐tau ,sTREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Microglial activation occurs early in Alzheimer's disease (AD) and previous studies reported both detrimental and protective effects of microglia on AD progression. Here, we used CSF sTREM2 to investigate disease stage‐dependent drivers of microglial activation and to determine downstream consequences on AD progression. We included 402 patients with measures of earliest beta‐amyloid (CSF Aβ1‐42) and late‐stage fibrillary Aβ pathology (amyloid‐PET centiloid), as well as sTREM2, p‐tau181, and FDG‐PET. To determine disease stage, we stratified participants into early Aβ‐accumulators (Aβ CSF+/PET−; n = 70) or late Aβ‐accumulators (Aβ CSF+/PET+; n = 201) plus 131 controls. In early Aβ‐accumulators, higher centiloid was associated with cross‐sectional/longitudinal sTREM2 and p‐tau181 increases. Further, higher sTREM2 mediated the association between centiloid and cross‐sectional/longitudinal p‐tau181 increases and higher sTREM2 was associated with FDG‐PET hypermetabolism. In late Aβ‐accumulators, we found no association between centiloid and sTREM2 but a cross‐sectional association between higher sTREM2, higher p‐tau181 and glucose hypometabolism. Our findings suggest that a TREM2‐related microglial response follows earliest Aβ fibrillization, manifests in inflammatory glucose hypermetabolism and may facilitate subsequent p‐tau181 increases in earliest AD.
- Published
- 2023
- Full Text
- View/download PDF
10. Identifying clinically useful biomarkers in neurodegenerative disease through a collaborative approach: the NeuroToolKit
- Author
-
Sterling C. Johnson, Marc Suárez-Calvet, Ivonne Suridjan, Carolina Minguillón, Juan Domingo Gispert, Erin Jonaitis, Agata Michna, Margherita Carboni, Tobias Bittner, Christina Rabe, Gwendlyn Kollmorgen, Henrik Zetterberg, and Kaj Blennow
- Subjects
Alzheimer’s disease ,Amyloid-β ,Cerebrospinal fluid biomarkers ,Glial activation ,Inflammation ,Neurodegeneration ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Alzheimer’s disease (AD) is a complex and heterogeneous disease, which requires reliable biomarkers for diagnosis and monitoring disease activity. Preanalytical protocol and technical variability associated with biomarker immunoassays makes comparability of biomarker data across multiple cohorts difficult. This study aimed to compare cerebrospinal fluid (CSF) biomarker results across independent cohorts, including participants spanning the AD continuum. Methods Measured on the NeuroToolKit (NTK) prototype panel of immunoassays, 12 CSF biomarkers were evaluated from three cohorts (ALFA+, Wisconsin, and Abby/Blaze). A correction factor was applied to biomarkers found to be affected by preanalytical procedures (amyloid-β1–42, amyloid-β1–40, and alpha-synuclein), and results between cohorts for each disease stage were compared. The relationship between CSF biomarker concentration and cognitive scores was evaluated. Results Biomarker distributions were comparable across cohorts following correction. Correlations of biomarker values were consistent across cohorts, regardless of disease stage. Disease stage differentiation was highest for neurofilament light (NfL), phosphorylated tau, and total tau, regardless of the cohort. Correlation between biomarker concentration and cognitive scores was comparable across cohorts, and strongest for NfL, chitinase-3-like protein-1 (YKL40), and glial fibrillary acidic protein. Discussion The precision of the NTK enables merging of biomarker datasets, after correction for preanalytical confounders. Assessment of multiple cohorts is crucial to increase power in future studies into AD pathogenesis.
- Published
- 2023
- Full Text
- View/download PDF
11. Genetically predicted telomere length and Alzheimer’s disease endophenotypes: a Mendelian randomization study
- Author
-
Blanca Rodríguez-Fernández, Natalia Vilor-Tejedor, Eider M. Arenaza-Urquijo, Gonzalo Sánchez-Benavides, Marc Suárez-Calvet, Grégory Operto, Carolina Minguillón, Karine Fauria, Gwendlyn Kollmorgen, Ivonne Suridjan, Manuel Castro de Moura, David Piñeyro, Manel Esteller, Kaj Blennow, Henrik Zetterberg, Immaculata De Vivo, José Luis Molinuevo, Arcadi Navarro, Juan Domingo Gispert, Aleix Sala-Vila, Marta Crous-Bou, and for the ALFA study
- Subjects
Alzheimer’s disease ,Cerebrospinal fluid biomarkers ,Mendelian randomization ,Neuroimaging ,Polygenic risk score ,Telomere length ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Telomere length (TL) is associated with biological aging, consequently influencing the risk of age-related diseases such as Alzheimer’s disease (AD). We aimed to evaluate the potential causal role of TL in AD endophenotypes (i.e., cognitive performance, N = 2233; brain age and AD-related signatures, N = 1134; and cerebrospinal fluid biomarkers (CSF) of AD and neurodegeneration, N = 304) through a Mendelian randomization (MR) analysis. Our analysis was conducted in the context of the ALFA (ALzheimer and FAmilies) study, a population of cognitively healthy individuals at risk of AD. A total of 20 single nucleotide polymorphisms associated with TL were used to determine the effect of TL on AD endophenotypes. Analyses were adjusted by age, sex, and years of education. Stratified analyses by APOE-ɛ4 status and polygenic risk score of AD were conducted. MR analysis revealed significant associations between genetically predicted longer TL and lower levels of CSF Aβ and higher levels of CSF NfL only in APOE-ɛ4 non-carriers. Moreover, inheriting longer TL was associated with greater cortical thickness in age and AD-related brain signatures and lower levels of CSF p-tau among individuals at a high genetic predisposition to AD. Further observational analyses are warranted to better understand these associations. Graphical Abstract
- Published
- 2022
- Full Text
- View/download PDF
12. Subjective cognitive decline and anxious/depressive symptoms during the COVID-19 pandemic: what is the role of stress perception, stress resilience, and β-amyloid?
- Author
-
Muge Akinci, Gonzalo Sánchez-Benavides, Anna Brugulat-Serrat, Cleofé Peña-Gómez, Eleni Palpatzis, Mahnaz Shekari, Carme Deulofeu, Sherezade Fuentes-Julian, Gemma Salvadó, José Maria González-de-Echávarri, Marc Suárez-Calvet, Carolina Minguillón, Karine Fauria, José Luis Molinuevo, Juan Domingo Gispert, Oriol Grau-Rivera, Eider M. Arenaza-Urquijo, and for the ALFA Study
- Subjects
Subjective cognitive decline ,Stress ,Anxiety ,Depression ,COVID-19 confinement ,Alzheimer’s disease ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The COVID-19 pandemic may worsen the mental health of people reporting subjective cognitive decline (SCD) and therefore their clinical prognosis. We aimed to investigate the association between the intensity of SCD and anxious/depressive symptoms during confinement and the underlying mechanisms. Methods Two hundred fifty cognitively unimpaired participants completed the Hospital Anxiety and Depression Scale (HADS) and SCD-Questionnaire (SCD-Q) and underwent amyloid-β positron emission tomography imaging with [18F] flutemetamol (N = 205) on average 2.4 (± 0.8) years before the COVID-19 confinement. During the confinement, participants completed the HADS, Perceived Stress Scale (PSS), Brief Resilience Scale (BRS), and an ad hoc questionnaire on worries (access to primary products, self-protection materials, economic situation) and lifestyle changes (sleep duration, sleep quality, eating habits). We investigated stress-related measurements, worries, and lifestyle changes in relation to SCD. We then conducted an analysis of covariance to investigate the association of SCD-Q with HADS scores during the confinement while controlling for pre-confinement anxiety/depression scores and demographics. Furthermore, we introduced amyloid-β positivity, PSS, and BRS in the models and performed mediation analyses to explore the mechanisms explaining the association between SCD and anxiety/depression. Results In the whole sample, the average SCD-Q score was 4.1 (± 4.4); 70 (28%) participants were classified as SCD, and 26 (12.7%) were amyloid-β-positive. During the confinement, participants reporting SCD showed higher PSS (p = 0.035) but not BRS scores (p = 0.65) than those that did not report SCD. No differences in worries or lifestyle changes were observed. Higher SCD-Q scores showed an association with greater anxiety/depression scores irrespective of pre-confinement anxiety/depression levels (p = 0.002). This association was not significant after introducing amyloid-β positivity and stress-related variables in the model (p = 0.069). Amyloid-β positivity and PSS were associated with greater HADS irrespective of pre-confinement anxiety/depression scores (p = 0.023; p < 0.001). The association of SCD-Q with HADS was mediated by PSS (p = 0.01). Conclusions Higher intensity of SCD, amyloid-β positivity, and stress perception showed independent associations with anxious/depressive symptoms during the COVID-19 confinement irrespective of pre-confinement anxiety/depression levels. The association of SCD intensity with anxiety/depression was mediated by stress perception, suggesting stress regulation as a potential intervention to reduce affective symptomatology in the SCD population in the face of stressors.
- Published
- 2022
- Full Text
- View/download PDF
13. Precision medicine in neurodegeneration: the IHI-PROMINENT project
- Author
-
Ashley Tate, Marc Suárez-Calvet, Mats Ekelund, Sven Eriksson, Maria Eriksdotter, Wiesje M. Van Der Flier, Jean Georges, Miia Kivipelto, Milica G. Kramberger, Peter Lindgren, Juan Domingo Gispert López, Jyrki Lötjönen, Sofie Persson, Sandra Pla, Alina Solomon, Lennart Thurfjell, Anders Wimo, Bengt Winblad, Linus Jönsson, and on behalf of the PROMINENT consortium
- Subjects
Alzheimer’s disease ,dementia ,precision medicine ,biomarkers ,clinical decision support ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Neurodegenerative diseases are one of the most important contributors to morbidity and mortality in the elderly. In Europe, over 14 million people are currently living with dementia, at a cost of over 400 billion EUR annually. Recent advances in diagnostics and approval for new pharmaceutical treatments for Alzheimer’s disease (AD), the most common etiology of dementia, heralds the beginning of precision medicine in this field. However, their implementation will challenge an already over-burdened healthcare systems. There is a need for innovative digital solutions that can drive the related clinical pathways and optimize and personalize care delivery. Public-private partnerships are ideal vehicles to tackle these challenges. Here we describe the Innovative Health Initiative (IHI) public-private partnership project PROMINENT that has been initiated by connecting leading dementia researchers, medical professionals, dementia patients and their care partners with the latest innovative health technologies using a precision medicine based digital platform. The project builds upon the knowledge and already implemented digital tools from several collaborative initiatives that address new models for early detection, diagnosis, and monitoring of AD and other neurodegenerative disorders. The project aims to provide support to improvement efforts to each aspect of the care pathway including diagnosis, prognosis, treatment, and data collection for real world evidence and cost effectiveness studies. Ultimately the PROMINENT project is expected to lead to cost-effective care and improved health outcomes.
- Published
- 2023
- Full Text
- View/download PDF
14. Biological brain age prediction using machine learning on structural neuroimaging data: Multi-cohort validation against biomarkers of Alzheimer’s disease and neurodegeneration stratified by sex
- Author
-
Irene Cumplido-Mayoral, Marina García-Prat, Grégory Operto, Carles Falcon, Mahnaz Shekari, Raffaele Cacciaglia, Marta Milà-Alomà, Luigi Lorenzini, Silvia Ingala, Alle Meije Wink, Henk JMM Mutsaerts, Carolina Minguillón, Karine Fauria, José Luis Molinuevo, Sven Haller, Gael Chetelat, Adam Waldman, Adam J Schwarz, Frederik Barkhof, Ivonne Suridjan, Gwendlyn Kollmorgen, Anna Bayfield, Henrik Zetterberg, Kaj Blennow, Marc Suárez-Calvet, Verónica Vilaplana, Juan Domingo Gispert, ALFA study, EPAD study, ADNI study, and OASIS study
- Subjects
brain age prediction ,neuroimaging ,alzheimer's disease ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Brain-age can be inferred from structural neuroimaging and compared to chronological age (brain-age delta) as a marker of biological brain aging. Accelerated aging has been found in neurodegenerative disorders like Alzheimer’s disease (AD), but its validation against markers of neurodegeneration and AD is lacking. Here, imaging-derived measures from the UK Biobank dataset (N=22,661) were used to predict brain-age in 2,314 cognitively unimpaired (CU) individuals at higher risk of AD and mild cognitive impaired (MCI) patients from four independent cohorts with available biomarker data: ALFA+, ADNI, EPAD, and OASIS. Brain-age delta was associated with abnormal amyloid-β, more advanced stages (AT) of AD pathology and APOE-ε4 status. Brain-age delta was positively associated with plasma neurofilament light, a marker of neurodegeneration, and sex differences in the brain effects of this marker were found. These results validate brain-age delta as a non-invasive marker of biological brain aging in non-demented individuals with abnormal levels of biomarkers of AD and axonal injury.
- Published
- 2023
- Full Text
- View/download PDF
15. Exploring cognitive and biological correlates of sleep quality and their potential links with Alzheimer’s disease (ALFASleep project): protocol for an observational study
- Author
-
Henrik Zetterberg, Kaj Blennow, José Luis Molinuevo, Andreea Radoi, Carolina Minguillón, Karine Fauria, Carme Deulofeu, Marc Suárez-Calvet, Oriol Grau-Rivera, Alex Iranzo, Iva Knezevic, Núria Tort-Colet, Laura Stankeviciute, Laura Hernández, Sherezade Fuentes-Julián, Israel Turull, David Fusté, Gonzalo Sánchez-Benavides, Eider M Arenaza-Urquijo, Sebastian C Holst, Pilar Garcés, Thomas Mueggler, Aurora Arqueros, and Juan Domingo Gispert
- Subjects
Medicine - Abstract
Introduction The growing worldwide prevalence of Alzheimer’s disease (AD) and the lack of effective treatments pose a dire medical challenge. Sleep disruption is also prevalent in the ageing population and is increasingly recognised as a risk factor and an early sign of AD. The ALFASleep project aims to characterise sleep with subjective and objective measurements in cognitively unimpaired middle/late middle-aged adults at increased risk of AD who are phenotyped with fluid and neuroimaging AD biomarkers. This will contribute to a better understanding of the pathophysiological mechanisms linking sleep with AD, thereby paving the way for the development of non-invasive biomarkers and preventive strategies targeting sleep.Methods and analysis We will invite 200 participants enrolled in the ALFA+ (for ALzheimer and FAmilies) prospective observational study to join the ALFASleep study. ALFA+ participants are cognitively unimpaired middle-aged/late middle-aged adults who are followed up every 3 years with a comprehensive set of evaluations including neuropsychological tests, blood and cerebrospinal fluid (CSF) sampling, and MRI and positron emission tomography acquisition. ALFASleep participants will be additionally characterised with actigraphy and CSF–orexin-A measurements, and a subset (n=90) will undergo overnight polysomnography. We will test associations of sleep measurements and CSF–orexin-A with fluid biomarkers of AD and glial activation, neuroimaging outcomes and cognitive performance. In case we found any associations, we will test whether changes in AD and/or glial activation markers mediate the association between sleep and neuroimaging or cognitive outcomes and whether sleep mediates associations between CSF–orexin-A and AD biomarkers.Ethics and dissemination The ALFASleep study protocol has been approved by the independent Ethics Committee Parc de Salut Mar, Barcelona (2018/8207/I). All participants have signed a written informed consent before their inclusion (approved by the same ethics committee). Study findings will be presented at national and international conferences and submitted for publication in peer-reviewed journals.Trial registration number NCT04932473.
- Published
- 2022
- Full Text
- View/download PDF
16. P‐tau235: a novel biomarker for staging preclinical Alzheimer’s disease
- Author
-
Juan Lantero‐Rodriguez, Anniina Snellman, Andrea L Benedet, Marta Milà‐Alomà, Elena Camporesi, Laia Montoliu‐Gaya, Nicholas J Ashton, Agathe Vrillon, Thomas K Karikari, Juan Domingo Gispert, Gemma Salvadó, Mahnaz Shekari, Christina E Toomey, Tammaryn L Lashley, Henrik Zetterberg, Marc Suárez‐Calvet, Gunnar Brinkmalm, Pedro Rosa Neto, and Kaj Blennow
- Subjects
Alzheimer’s disease ,biomarkers ,CSF ,p‐tau235 ,tau ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Alzheimer’s disease (AD) is characterised by a long preclinical phase. Although phosphorylated tau (p‐tau) species such as p‐tau217 and p‐tau231 provide accurate detection of early pathological changes, other biomarkers capable of staging disease progression during preclinical AD are still needed. Combining exploratory and targeted mass spectrometry methods in neuropathologically confirmed brain tissue, we observed that p‐tau235 is a prominent feature of AD pathology. In addition, p‐tau235 seemed to be preceded by p‐tau231, in what appeared to be a sequential phosphorylation event. To exploit its biomarker potential in cerebrospinal fluid (CSF), we developed and validated a new p‐tau235 Simoa assay. Using three clinical cohorts, we demonstrated that (i) CSF p‐235 increases early in AD continuum, and (ii) changes in CSF p‐tau235 and p‐tau231 levels during preclinical AD are consistent with the sequential phosphorylation evidence in AD brain. In conclusion, CSF p‐tau235 appears to be not only a highly specific biomarker of AD but also a promising staging biomarker for the preclinical phase. Thus, it could prove useful tracking disease progression and help enriching clinical trial recruitment.
- Published
- 2021
- Full Text
- View/download PDF
17. Perivascular spaces are associated with tau pathophysiology and synaptic dysfunction in early Alzheimer’s continuum
- Author
-
Natalia Vilor-Tejedor, Iacopo Ciampa, Grégory Operto, Carles Falcón, Marc Suárez-Calvet, Marta Crous-Bou, Mahnaz Shekari, Eider M. Arenaza-Urquijo, Marta Milà-Alomà, Oriol Grau-Rivera, Carolina Minguillon, Gwendlyn Kollmorgen, Henrik Zetterberg, Kaj Blennow, Roderic Guigo, José Luis Molinuevo, Juan Domingo Gispert, and for the ALFA study
- Subjects
Alzheimer’s disease ,MRI ,CSF biomarkers ,Perivascular spaces ,Tau pathophysiology ,Virchow-Robin spaces ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Perivascular spaces (PVS) have an important role in the elimination of metabolic waste from the brain. It has been hypothesized that the enlargement of PVS (ePVS) could be affected by pathophysiological mechanisms involved in Alzheimer’s disease (AD), such as abnormal levels of CSF biomarkers. However, the relationship between ePVS and these pathophysiological mechanisms remains unknown. Objective We aimed to investigate the association between ePVS and CSF biomarkers of several pathophysiological mechanisms for AD. We hypothesized that ePVS will be associated to CSF biomarkers early in the AD continuum (i.e., amyloid positive cognitively unimpaired individuals). Besides, we explored associations between ePVS and demographic and cardiovascular risk factors. Methods The study included 322 middle-aged cognitively unimpaired participants from the ALFA + study, many within the Alzheimer’s continuum. NeuroToolKit and Elecsys® immunoassays were used to measure CSF Aβ42, Aβ40, p-tau and t-tau, NfL, neurogranin, TREM2, YKL40, GFAP, IL6, S100, and α-synuclein. PVS in the basal ganglia (BG) and centrum semiovale (CS) were assessed based on a validated 4-point visual rating scale. Odds ratios were calculated for associations of cardiovascular and AD risk factors with ePVS using logistic and multinomial models adjusted for relevant confounders. Models were stratified by Aβ status (positivity defined as Aβ42/40
- Published
- 2021
- Full Text
- View/download PDF
18. Cognitively unimpaired individuals with a low burden of Aβ pathology have a distinct CSF biomarker profile
- Author
-
Marta Milà-Alomà, Mahnaz Shekari, Gemma Salvadó, Juan Domingo Gispert, Eider M. Arenaza-Urquijo, Grégory Operto, Carles Falcon, Natalia Vilor-Tejedor, Oriol Grau-Rivera, Aleix Sala-Vila, Gonzalo Sánchez-Benavides, José Maria González-de-Echávarri, Carolina Minguillon, Karine Fauria, Aida Niñerola-Baizán, Andrés Perissinotti, Maryline Simon, Gwendlyn Kollmorgen, Henrik Zetterberg, Kaj Blennow, Marc Suárez-Calvet, José Luis Molinuevo, and for the ALFA study
- Subjects
Preclinical ,Alzheimer’s disease ,CSF ,Biomarkers ,Subthreshold ,Cognitively unimpaired ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Understanding the changes that occur in the transitional stage between absent and overt amyloid-β (Aβ) pathology within the Alzheimer’s continuum is crucial to develop therapeutic and preventive strategies. The objective of this study is to test whether cognitively unimpaired individuals with a low burden of Aβ pathology have a distinct CSF, structural, and functional neuroimaging biomarker profile. Methods Cross-sectional study of 318 middle-aged, cognitively unimpaired individuals from the ALFA+ cohort. We measured CSF Aβ42/40, phosphorylated tau (p-tau), total tau (t-tau), neurofilament light (NfL), neurogranin, sTREM2, YKL40, GFAP, IL6, S100B, and α-synuclein. Participants also underwent cognitive assessments, APOE genotyping, structural MRI, [18F]-FDG, and [18F]-flutemetamol PET. To ensure the robustness of our results, we used three definitions of low burden of Aβ pathology: (1) positive CSF Aβ42/40 and < 30 Centiloids in Aβ PET, (2) positive CSF Aβ42/40 and negative Aβ PET visual read, and (3) 20–40 Centiloid range in Aβ PET. We tested CSF and neuroimaging biomarker differences between the low burden group and the corresponding Aβ-negative group, adjusted by age and sex. Results The prevalence and demographic characteristics of the low burden group differed between the three definitions. CSF p-tau and t-tau were increased in the low burden group compared to the Aβ-negative in all definitions. CSF neurogranin was increased in the low burden group definitions 1 and 3, while CSF NfL was only increased in the low burden group definition 1. None of the defined low burden groups showed signs of atrophy or glucose hypometabolism. Instead, we found slight increases in cortical thickness and metabolism in definition 2. Conclusions There are biologically meaningful Aβ-downstream effects in individuals with a low burden of Aβ pathology, while structural and functional changes are still subtle or absent. These findings support considering individuals with a low burden of Aβ pathology for clinical trials. Trial registration NCT02485730
- Published
- 2021
- Full Text
- View/download PDF
19. Association of weight change with cerebrospinal fluid biomarkers and amyloid positron emission tomography in preclinical Alzheimer’s disease
- Author
-
Oriol Grau-Rivera, Irene Navalpotro-Gomez, Gonzalo Sánchez-Benavides, Marc Suárez-Calvet, Marta Milà-Alomà, Eider M. Arenaza-Urquijo, Gemma Salvadó, Aleix Sala-Vila, Mahnaz Shekari, José Maria González-de-Echávarri, Carolina Minguillón, Aida Niñerola-Baizán, Andrés Perissinotti, Maryline Simon, Gwendlyn Kollmorgen, Henrik Zetterberg, Kaj Blennow, Juan Domingo Gispert, José Luis Molinuevo, and for the ALFA Study
- Subjects
Alzheimer’s disease ,Preclinical ,Cognitively unimpaired ,Weight loss ,Biomarkers ,Risk factors ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Recognizing clinical manifestations heralding the development of Alzheimer’s disease (AD)-related cognitive impairment could improve the identification of individuals at higher risk of AD who may benefit from potential prevention strategies targeting preclinical population. We aim to characterize the association of body weight change with cognitive changes and AD biomarkers in cognitively unimpaired middle-aged adults. Methods This prospective cohort study included data from cognitively unimpaired adults from the ALFA study (n = 2743), a research platform focused on preclinical AD. Cognitive and anthropometric data were collected at baseline between April 2013 and November 2014. Between October 2016 and February 2020, 450 participants were visited in the context of the nested ALFA+ study and underwent cerebrospinal fluid (CSF) extraction and acquisition of positron emission tomography images with [18F]flutemetamol (FTM-PET). From these, 408 (90.1%) were included in the present study. We used data from two visits (average interval 4.1 years) to compute rates of change in weight and cognitive performance. We tested associations between these variables and between weight change and categorical and continuous measures of CSF and neuroimaging AD biomarkers obtained at follow-up. We classified participants with CSF data according to the AT (amyloid, tau) system and assessed between-group differences in weight change. Results Weight loss predicted a higher likelihood of positive FTM-PET visual read (OR 1.27, 95% CI 1.00–1.61, p = 0.049), abnormal CSF p-tau levels (OR 1.50, 95% CI 1.19–1.89, p = 0.001), and an A+T+ profile (OR 1.64, 95% CI 1.25–2.20, p = 0.001) and was greater among participants with an A+T+ profile (p
- Published
- 2021
- Full Text
- View/download PDF
20. Association between polygenic risk score of Alzheimer’s disease and plasma phosphorylated tau in individuals from the Alzheimer’s Disease Neuroimaging Initiative
- Author
-
Anna Zettergren, Jodie Lord, Nicholas J. Ashton, Andrea L. Benedet, Thomas K. Karikari, Juan Lantero Rodriguez, the Alzheimer’s Disease Neuroimaging Initiative, Anniina Snellman, Marc Suárez-Calvet, Petroula Proitsi, Henrik Zetterberg, and Kaj Blennow
- Subjects
Alzheimer’s disease ,Polygenic risk score ,Plasma phosphorylated tau 181 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Recent studies suggest that plasma phosphorylated tau181 (p-tau181) is a highly specific biomarker for Alzheimer’s disease (AD)-related tau pathology. It has great potential for the diagnostic and prognostic evaluation of AD, since it identifies AD with the same accuracy as tau PET and CSF p-tau181 and predicts the development of AD dementia in cognitively unimpaired (CU) individuals and in those with mild cognitive impairment (MCI). Plasma p-tau181 may also be used as a biomarker in studies exploring disease pathogenesis, such as genetic or environmental risk factors for AD-type tau pathology. The aim of the present study was to investigate the relation between polygenic risk scores (PRSs) for AD and plasma p-tau181. Methods Data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) was used to examine the relation between AD PRSs, constructed based on findings in recent genome-wide association studies, and plasma p-tau181, using linear regression models. Analyses were performed in the total sample (n = 818), after stratification on diagnostic status (CU (n = 236), MCI (n = 434), AD dementia (n = 148)), and after stratification on Aβ pathology status (Aβ positives (n = 322), Aβ negatives (n = 409)). Results Associations between plasma p-tau181 and APOE PRSs (p = 3e−18–7e−15) and non-APOE PRSs (p = 3e−4–0.03) were seen in the total sample. The APOE PRSs were associated with plasma p-tau181 in all diagnostic groups (CU, MCI, and AD dementia), while the non-APOE PRSs were associated only in the MCI group. The APOE PRSs showed similar results in amyloid-β (Aβ)-positive and negative individuals (p = 5e−5–1e−3), while the non-APOE PRSs were associated with plasma p-tau181 in Aβ positives only (p = 0.02). Conclusions Polygenic risk for AD including APOE was found to associate with plasma p-tau181 independent of diagnostic and Aβ pathology status, while polygenic risk for AD beyond APOE was associated with plasma p-tau181 only in MCI and Aβ-positive individuals. These results extend the knowledge about the relation between genetic risk for AD and p-tau181, and further support the usefulness of plasma p-tau181 as a biomarker of AD.
- Published
- 2021
- Full Text
- View/download PDF
21. CSF p-tau231: A biomarker for early preclinical Alzheimer?
- Author
-
Marc Suárez-Calvet
- Subjects
Medicine ,Medicine (General) ,R5-920 - Published
- 2022
- Full Text
- View/download PDF
22. Novel tau biomarkers phosphorylated at T181, T217 or T231 rise in the initial stages of the preclinical Alzheimer’s continuum when only subtle changes in Aβ pathology are detected
- Author
-
Marc Suárez‐Calvet, Thomas K Karikari, Nicholas J Ashton, Juan Lantero Rodríguez, Marta Milà‐Alomà, Juan Domingo Gispert, Gemma Salvadó, Carolina Minguillon, Karine Fauria, Mahnaz Shekari, Oriol Grau‐Rivera, Eider M Arenaza‐Urquijo, Aleix Sala‐Vila, Gonzalo Sánchez‐Benavides, José Maria González‐de‐Echávarri, Gwendlyn Kollmorgen, Erik Stoops, Eugeen Vanmechelen, Henrik Zetterberg, Kaj Blennow, José Luis Molinuevo, for the ALFA Study, Annabella Beteta, Raffaele Cacciaglia, Alba Cañas, Carme Deulofeu, Irene Cumplido, Ruth Dominguez, Maria Emilio, Carles Falcon, Sherezade Fuentes, Laura Hernandez, Gema Huesa, Jordi Huguet, Paula Marne, Tania Menchón, Grégory Operto, Albina Polo, Sandra Pradas, Anna Soteras, Marc Vilanova, and Natalia Vilor‐Tejedor
- Subjects
Alzheimer’s disease ,biomarker ,cerebrospinal fluid ,plasma ,tau ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract In Alzheimer’s disease (AD), tau phosphorylation in the brain and its subsequent release into cerebrospinal fluid (CSF) and blood is a dynamic process that changes during disease evolution. The main aim of our study was to characterize the pattern of changes in phosphorylated tau (p‐tau) in the preclinical stage of the Alzheimer’s continuum. We measured three novel CSF p‐tau biomarkers, phosphorylated at threonine‐181 and threonine‐217 with an N‐terminal partner antibody and at threonine‐231 with a mid‐region partner antibody. These were compared with an automated mid‐region p‐tau181 assay (Elecsys) as the gold standard p‐tau measure. We demonstrate that these novel p‐tau biomarkers increase more prominently in preclinical Alzheimer, when only subtle changes of amyloid‐β (Aβ) pathology are detected, and can accurately differentiate Aβ‐positive from Aβ‐negative cognitively unimpaired individuals. Moreover, we show that the novel plasma N‐terminal p‐tau181 biomarker is mildly but significantly increased in the preclinical stage. Our results support the idea that early changes in neuronal tau metabolism in preclinical Alzheimer, likely in response to Aβ exposure, can be detected with these novel p‐tau assays.
- Published
- 2020
- Full Text
- View/download PDF
23. Higher CSF sTREM2 and microglia activation are associated with slower rates of beta‐amyloid accumulation
- Author
-
Michael Ewers, Gloria Biechele, Marc Suárez‐Calvet, Christian Sacher, Tanja Blume, Estrella Morenas‐Rodriguez, Yuetiva Deming, Laura Piccio, Carlos Cruchaga, Gernot Kleinberger, Leslie Shaw, John Q Trojanowski, Jochen Herms, Martin Dichgans, the Alzheimer's Disease Neuroimaging Initiative (ADNI), Matthias Brendel, Christian Haass, and Nicolai Franzmeier
- Subjects
beta‐amyloid accumulation ,microglia ,protective ,tau ,TREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Microglia activation is the brain's major immune response to amyloid plaques in Alzheimer's disease (AD). Both cerebrospinal fluid (CSF) levels of soluble TREM2 (sTREM2), a biomarker of microglia activation, and microglia PET are increased in AD; however, whether an increase in these biomarkers is associated with reduced amyloid‐beta (Aβ) accumulation remains unclear. To address this question, we pursued a two‐pronged translational approach. Firstly, in non‐demented and demented individuals, we tested CSF sTREM2 at baseline to predict (i) amyloid PET changes over ∼2 years and (ii) tau PET cross‐sectionally assessed in a subset of patients. We found higher CSF sTREM2 associated with attenuated amyloid PET increase and lower tau PET. Secondly, in the AppNL‐G-F mouse model of amyloidosis, we studied baseline 18F‐GE180 microglia PET and longitudinal amyloid PET to test the microglia vs. Aβ association, without any confounding co‐pathologies often present in AD patients. Higher microglia PET at age 5 months was associated with a slower amyloid PET increase between ages 5‐to‐10 months. In conclusion, higher microglia activation as determined by CSF sTREM2 or microglia PET shows protective effects on subsequent amyloid accumulation.
- Published
- 2020
- Full Text
- View/download PDF
24. Association between insomnia and cognitive performance, gray matter volume, and white matter microstructure in cognitively unimpaired adults
- Author
-
Oriol Grau-Rivera, Grégory Operto, Carles Falcón, Gonzalo Sánchez-Benavides, Raffaele Cacciaglia, Anna Brugulat-Serrat, Nina Gramunt, Gemma Salvadó, Marc Suárez-Calvet, Carolina Minguillon, Álex Iranzo, Juan Domingo Gispert, José Luis Molinuevo, and for the ALFA Study
- Subjects
Sleep ,Insomnia ,Neurocognitive disorders ,Alzheimer disease ,Inflammation ,Neuropsychology ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Mounting evidence links poor sleep quality with a higher risk of late-life dementia. However, the structural and cognitive correlates of insomnia are still not well understood. The study aims were to characterize the cognitive performance and brain structural pattern of cognitively unimpaired adults at increased risk for Alzheimer’s disease (AD) with insomnia. Methods This cross-sectional study included 1683 cognitively unimpaired middle/late-middle-aged adults from the ALFA (ALzheimer and FAmilies) study who underwent neuropsychological assessment, T1-weighted structural imaging (n = 366), and diffusion-weighted imaging (n = 334). The World Health Organization’s World Mental Health Survey Initiative version of the Composite International Diagnostic Interview was used to define the presence or absence of insomnia. Multivariable regression models were used to evaluate differences in cognitive performance between individuals with and without insomnia, as well as potential interactions between insomnia and the APOE genotype. Voxel-based morphometry and tract-based spatial statistics were used to assess between-group differences and potential interactions between insomnia and the APOE genotype in gray matter volume and white matter diffusion metrics. Results Insomnia was reported by 615 out of 1683 participants (36.5%), including 137 out of 366 (37.4%) with T1-weighted structural imaging available and 119 out of 334 (35.6%) with diffusion-weighted imaging. Individuals with insomnia (n = 615) performed worse in executive function tests than non-insomniacs and displayed lower gray matter volume in left orbitofrontal and right middle temporal cortex, bilateral precuneus, posterior cingulate cortex and thalamus, higher gray matter volume in the left caudate nucleus, and widespread reduction of mean and axial diffusivity in right hemisphere white matter tracts. Insomnia interacted with the APOE genotype, with APOE-ε4 carriers displaying lower gray matter volumes when insomnia was present, but higher volumes when insomnia was not present, in several gray matter regions, including the left angular gyrus, the bilateral superior frontal gyri, the thalami, and the right hippocampus. Conclusions Insomnia in cognitively unimpaired adults at increased risk for AD is associated to poorer performance in some executive functions and volume changes in cortical and subcortical gray matter, including key areas involved in Alzheimer’s disease, as well as decreased white matter diffusivity.
- Published
- 2020
- Full Text
- View/download PDF
25. Leveraging large multi-center cohorts of Alzheimer disease endophenotypes to understand the role of Klotho heterozygosity on disease risk.
- Author
-
Muhammad Ali, Yun Ju Sung, Fengxian Wang, Maria V Fernández, John C Morris, Anne M Fagan, Kaj Blennow, Henrik Zetterberg, Amanda Heslegrave, Per M Johansson, Johan Svensson, Bengt Nellgård, Alberto Lleó, Daniel Alcolea, Jordi Clarimon, Lorena Rami, José Luis Molinuevo, Marc Suárez-Calvet, Estrella Morenas-Rodríguez, Gernot Kleinberger, Christian Haass, Michael Ewers, Johannes Levin, Martin R Farlow, Richard J Perrin, Alzheimer’s Disease Neuroimaging Initiative (ADNI), Dominantly Inherited Alzheimer Network (DIAN), and Carlos Cruchaga
- Subjects
Medicine ,Science - Abstract
Two genetic variants in strong linkage disequilibrium (rs9536314 and rs9527025) in the Klotho (KL) gene, encoding a transmembrane protein, implicated in longevity and associated with brain resilience during normal aging, were recently shown to be associated with Alzheimer disease (AD) risk in cognitively normal participants who are APOE ε4 carriers. Specifically, the participants heterozygous for this variant (KL-SVHET+) showed lower risk of developing AD. Furthermore, a neuroprotective effect of KL-VSHET+ has been suggested against amyloid burden for cognitively normal participants, potentially mediated via the regulation of redox pathways. However, inconsistent associations and a smaller sample size of existing studies pose significant hurdles in drawing definitive conclusions. Here, we performed a well-powered association analysis between KL-VSHET+ and five different AD endophenotypes; brain amyloidosis measured by positron emission tomography (PET) scans (n = 5,541) or cerebrospinal fluid Aβ42 levels (CSF; n = 5,093), as well as biomarkers associated with tau pathology: the CSF Tau (n = 5,127), phosphorylated Tau (pTau181; n = 4,778) and inflammation: CSF soluble triggering receptor expressed on myeloid cells 2 (sTREM2; n = 2,123) levels. Our results found nominally significant associations of KL-VSHET+ status with biomarkers for brain amyloidosis (e.g., CSF Aβ positivity; odds ratio [OR] = 0.67 [95% CI, 0.55-0.78], β = 0.72, p = 0.007) and tau pathology (e.g., biomarker positivity for CSF Tau; OR = 0.39 [95% CI, 0.19-0.77], β = -0.94, p = 0.007, and pTau; OR = 0.50 [95% CI, 0.27-0.96], β = -0.68, p = 0.04) in cognitively normal participants, 60-80 years old, who are APOE e4-carriers. Our work supports previous findings, suggesting that the KL-VSHET+ on an APOE ε4 genotype background may modulate Aβ and tau pathology, thereby lowering the intensity of neurodegeneration and incidence of cognitive decline in older controls susceptible to AD.
- Published
- 2022
- Full Text
- View/download PDF
26. Associations between air pollution and biomarkers of Alzheimer’s disease in cognitively unimpaired individuals
- Author
-
Silvia Alemany, Marta Crous-Bou, Natalia Vilor-Tejedor, Marta Milà-Alomà, Marc Suárez-Calvet, Gemma Salvadó, Marta Cirach, Eider M. Arenaza-Urquijo, Gonzalo Sanchez-Benavides, Oriol Grau-Rivera, Carolina Minguillon, Karine Fauria, Gwendlyn Kollmorgen, Juan Domingo Gispert, Mireia Gascón, Mark Nieuwenhuijsen, Henrik Zetterberg, Kaj Blennow, Jordi Sunyer, and José Luis Molinuevo
- Subjects
Air pollution ,Nitrogen dioxide ,Particulate matter ,Neurodegeneration ,AD biomarkers ,Amyloid-β ,Environmental sciences ,GE1-350 - Abstract
Background: Air quality contributes to incidence of Alzheimer’s disease (AD) although the underlying neurobiological mechanisms are unclear. This study was aimed to examine the association between air pollution and concentrations of cerebrospinal fluid (CSF) AD biomarkers and amyloid-β (Aβ) deposition.Participants and methodsThe sample included 156 cognitively unimpaired adults aged 57 years (61 at biomarkers assessment) with increased risk of AD from the ALFA + Study. We examined CSF levels of Aβ42, Aβ40, p-Tau, t-Tau, neurofilament light (NfL) and cerebral amyloid load (Centiloid). A Land Use Regression model from 2009 was used to estimate residential exposure to air pollutants including nitrogen dioxide (NO2,) and particulate matter (PM2.5, PM2.5 abs, PM10). This model was considered a surrogate of long-term exposure until time of data collection in 2013–2014. Participants have resided in the same residence for at least the previous 3 years. Multiple linear regression models were used to estimate associations between air pollutants and biomarkers. The effect modification by CSF Aβ status and APOE-ε4 carriership was also assessed. Results: A consistent pattern of results indicated that greater exposure to NO2 and PM2.5 absorbance was associated with higher levels of brain Aβ deposition, while greater exposure to PM10 and PM2.5was associated with higher levels of CSF NfL. Most associations were driven by individuals that were Aβ-positive. Although APOE-ε4 status did not significantly modify these associations, the effect of air pollutants exposure on CSF NfL levels was stronger in APOE-ε4 carriers. Conclusion: In a population of cognitively unimpaired adults with increased risk of AD, long-term exposure to air pollution was associated with higher levels in biomarkers of AD pathology. While further research is granted to elucidate the mechanisms involved in such associations, our results reinforce the role of air pollution as an environmental risk factor for AD.
- Published
- 2021
- Full Text
- View/download PDF
27. Plasma levels of soluble TREM2 and neurofilament light chain in TREM2 rare variant carriers
- Author
-
Nicholas J. Ashton, Marc Suárez-Calvet, Amanda Heslegrave, Abdul Hye, Cristina Razquin, Pau Pastor, Raquel Sanchez-Valle, José L. Molinuevo, Pieter Jelle Visser, Kaj Blennow, Angela K. Hodges, and Henrik Zetterberg
- Subjects
Alzheimer’s disease ,sTREM2 ,Blood ,Biomarkers ,Neurofilament light chain ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Results from recent clinical studies suggest that cerebrospinal fluid (CSF) biomarkers that are indicative of Alzheimer’s disease (AD) can be replicated in blood, e.g. amyloid-beta peptides (Aβ42 and Aβ40) and neurofilament light chain (NFL). Such data proposes that blood is a rich source of potential biomarkers reflecting central nervous system pathophysiology and should be fully explored for biomarkers that show promise in CSF. Recently, soluble fragments of the triggering receptor expressed on myeloid cells 2 (sTREM2) protein in CSF have been reported to be increased in prodromal AD and also in individuals with TREM2 rare genetic variants that increase the likelihood of developing dementia. Methods In this study, we measured the levels of plasma sTREM2 and plasma NFL using the MesoScale Discovery and single molecule array platforms, respectively, in 48 confirmed TREM2 rare variant carriers and 49 non-carriers. Results Our results indicate that there are no changes in plasma sTREM2 and NFL concentrations between TREM2 rare variant carriers and non-carriers. Furthermore, plasma sTREM2 is not different between healthy controls, mild cognitive impairment (MCI) or AD. Conclusion Concentrations of plasma sTREM2 do not mimic the recent changes found in CSF sTREM2.
- Published
- 2019
- Full Text
- View/download PDF
28. Centiloid cut-off values for optimal agreement between PET and CSF core AD biomarkers
- Author
-
Gemma Salvadó, José Luis Molinuevo, Anna Brugulat-Serrat, Carles Falcon, Oriol Grau-Rivera, Marc Suárez-Calvet, Javier Pavia, Aida Niñerola-Baizán, Andrés Perissinotti, Francisco Lomeña, Carolina Minguillon, Karine Fauria, Henrik Zetterberg, Kaj Blennow, Juan Domingo Gispert, and for the Alzheimer’s Disease Neuroimaging Initiative, for the ALFA Study
- Subjects
AD pathophysiology ,Biomarker concordance ,Threshold ,Positivity ,Preclinical ,Early detection ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background The Centiloid scale has been developed to standardize measurements of amyloid PET imaging. Reference cut-off values of this continuous measurement enable the consistent operationalization of decision-making for multicentre research studies and clinical trials. In this study, we aimed at deriving reference Centiloid thresholds that maximize the agreement against core Alzheimer’s disease (AD) cerebrospinal fluid (CSF) biomarkers in two large independent cohorts. Methods A total of 516 participants of the ALFA+ Study (N = 205) and ADNI (N = 311) underwent amyloid PET imaging ([18F]flutemetamol and [18F]florbetapir, respectively) and core AD CSF biomarker determination using Elecsys® tests. Tracer uptake was quantified in Centiloid units (CL). Optimal Centiloid cut-offs were sought that maximize the agreement between PET and dichotomous determinations based on CSF levels of Aβ42, tTau, pTau, and their ratios, using pre-established reference cut-off values. To this end, a receiver operating characteristic analysis (ROC) was conducted, and Centiloid cut-offs were calculated as those that maximized the Youden’s J Index or the overall percentage agreement recorded. Results All Centiloid cut-offs fell within the range of 25–35, except for CSF Aβ42 that rendered an optimal cut-off value of 12 CL. As expected, the agreement of tau/Aβ42 ratios was higher than that of CSF Aβ42. Centiloid cut-off robustness was confirmed even when established in an independent cohort and against variations of CSF cut-offs. Conclusions A cut-off of 12 CL matches previously reported values derived against postmortem measures of AD neuropathology. Together with these previous findings, our results flag two relevant inflection points that would serve as boundary of different stages of amyloid pathology: one around 12 CL that marks the transition from the absence of pathology to subtle pathology and another one around 30 CL indicating the presence of established pathology. The derivation of robust and generalizable cut-offs for core AD biomarkers requires cohorts with adequate representation of intermediate levels. Trial registration ALFA+ Study, NCT02485730 ALFA PET Sub-study, NCT02685969
- Published
- 2019
- Full Text
- View/download PDF
29. Spatial patterns of white matter hyperintensities associated with Alzheimer’s disease risk factors in a cognitively healthy middle-aged cohort
- Author
-
Gemma Salvadó, Anna Brugulat-Serrat, Carole H. Sudre, Oriol Grau-Rivera, Marc Suárez-Calvet, Carles Falcon, Karine Fauria, M. Jorge Cardoso, Frederik Barkhof, José Luis Molinuevo, Juan Domingo Gispert, and for the ALFA Study
- Subjects
Vascular ,Lesions ,Aging ,Brain ,Prevention ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background White matter hyperintensities (WMH) of presumed vascular origin have been associated with an increased risk of Alzheimer’s disease (AD). This study aims to describe the patterns of WMH associated with dementia risk estimates and individual risk factors in a cohort of middle-aged/late middle-aged individuals (mean 58 (interquartile range 51–64) years old). Methods Magnetic resonance imaging and AD risk factors were collected from 575 cognitively unimpaired participants. WMH load was automatically calculated in each brain lobe and in four equidistant layers from the ventricular surface to the cortical interface. Global volumes and regional patterns of WMH load were analyzed as a function of the Cardiovascular Risk Factors, Aging and Incidence of Dementia (CAIDE) dementia risk score, as well as family history of AD and Apolipoprotein E (APOE) genotype. Additional analyses were performed after correcting for the effect of age and hypertension. Results The studied cohort showed very low WMH burden (median 1.94 cm3) and 20-year dementia risk estimates (median 1.47 %). Even so, higher CAIDE scores were significantly associated with increased global WMH load. The main drivers of this association were age and hypertension, with hypercholesterolemia and body mass index also displaying a minor, albeit significant, influence. Regionally, CAIDE scores were positively associated with WMH in anterior areas, mostly in the frontal lobe. Age and hypertension showed significant association with WMH in almost all regions analyzed. The APOE-ε2 allele showed a protective effect over global WMH with a pattern that comprised juxtacortical temporo-occipital and fronto-parietal deep white matter regions. Participants with maternal family history of AD had higher WMH load than those without, especially in temporal and occipital lobes. Conclusions WMH load is associated with AD risk factors even in cognitively unimpaired subjects with very low WMH burden and dementia risk estimates. Our results suggest that tight control of modifiable risk factors in middle-age/late middle-age could have a significant impact on late-life dementia.
- Published
- 2019
- Full Text
- View/download PDF
30. Early increase of CSF sTREM2 in Alzheimer’s disease is associated with tau related-neurodegeneration but not with amyloid-β pathology
- Author
-
Marc Suárez-Calvet, Estrella Morenas-Rodríguez, Gernot Kleinberger, Kai Schlepckow, Miguel Ángel Araque Caballero, Nicolai Franzmeier, Anja Capell, Katrin Fellerer, Brigitte Nuscher, Erden Eren, Johannes Levin, Yuetiva Deming, Laura Piccio, Celeste M. Karch, Carlos Cruchaga, Leslie M. Shaw, John Q. Trojanowski, Michael Weiner, Michael Ewers, Christian Haass, and for the Alzheimer’s Disease Neuroimaging Initiative
- Subjects
Alzheimer’s disease ,Biomarkers ,Microglia ,Neurodegeneration ,Neuroinflammation ,Shedding ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Background TREM2 is a transmembrane receptor that is predominantly expressed by microglia in the central nervous system. Rare variants in the TREM2 gene increase the risk for late-onset Alzheimer’s disease (AD). Soluble TREM2 (sTREM2) resulting from shedding of the TREM2 ectodomain can be detected in the cerebrospinal fluid (CSF) and is a surrogate measure of TREM2-mediated microglia function. CSF sTREM2 has been previously reported to increase at different clinical stages of AD, however, alterations in relation to Amyloid β-peptide (Aβ) deposition or additional pathological processes in the amyloid cascade (such as tau pathology or neurodegeneration) remain unclear. In the current cross-sectional study, we employed the biomarker-based classification framework recently proposed by the NIA-AA consensus guidelines, in combination with clinical staging, in order to examine the CSF sTREM2 alterations at early asymptomatic and symptomatic stages of AD. Methods A cross-sectional study of 1027 participants of the Alzheimer’s Disease Imaging Initiative (ADNI) cohort, including 43 subjects carrying TREM2 rare genetic variants, was conducted to measure CSF sTREM2 using a previously validated enzyme-linked immunosorbent assay (ELISA). ADNI participants were classified following the A/T/N framework, which we implemented based on the CSF levels of Aβ1-42 (A), phosphorylated tau (T) and total tau as a marker of neurodegeneration (N), at different clinical stages defined by the clinical dementia rating (CDR) score. Results CSF sTREM2 differed between TREM2 variants, whereas the p.R47H variant had higher CSF sTREM2, p.L211P had lower CSF sTREM2 than non-carriers. We found that CSF sTREM2 increased in early symptomatic stages of late-onset AD but, unexpectedly, we observed decreased CSF sTREM2 levels at the earliest asymptomatic phase when only abnormal Aβ pathology (A+) but no tau pathology or neurodegeneration (TN-), is present. Conclusions Aβ pathology (A) and tau pathology/neurodegeneration (TN) have differing associations with CSF sTREM2. While tau-related neurodegeneration is associated with an increase in CSF sTREM2, Aβ pathology in the absence of downstream tau-related neurodegeneration is associated with a decrease in CSF sTREM2.
- Published
- 2019
- Full Text
- View/download PDF
31. Brain and cognitive correlates of subjective cognitive decline-plus features in a population-based cohort
- Author
-
Gonzalo Sánchez-Benavides, Oriol Grau-Rivera, Marc Suárez-Calvet, Carolina Minguillon, Raffaele Cacciaglia, Nina Gramunt, Carles Falcon, ALFA Study, Juan Domingo Gispert, and José Luis Molinuevo
- Subjects
Subjective cognitive decline ,Memory ,Voxel-based morphometry ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Subjective cognitive decline (SCD) consists of self-perceived decline in cognition over time. The occurrence of specific additional features in SCD (so-called SCDplus) confers a higher risk of future cognitive decline. However, it is not known whether SCDplus patients have a distinct cognitive and neuroimaging profile. Therefore, we aimed to study the associations between SCDplus features and cognitive and neuroimaging profiles in a population-based cohort. Methods A total of 2670 individuals from the ALFA cohort underwent clinical, cognitive, and MRI (n = 532) explorations. Subjects were classified as self-reporting cognitive decline (SCD) or not self-reporting cognitive decline (non-SCD). Within the SCD group, participants were also classified according to the number of SCDplus features they met (SCD+, > 3; SCD–, ≤ 3). Results The prevalence of SCD in the cohort was 21.4% (55.8% SCD–, 44.2% SCD+). SCD+ subjects performed worse than non-SCD and SCD– subjects in memory and executive function. Among the SCDplus features, confirmation of decline by an informant was the best predictor of worse cognitive performance and lower gray matter volumes. Conclusions Our findings show that individuals with SCDplus features have a distinct cognitive and brain volumetric profile similar to that found in Alzheimer’s disease and therefore support the use of the SCDplus concept as an enrichment criterion in population-based cohorts.
- Published
- 2018
- Full Text
- View/download PDF
32. Effects of pre‐analytical procedures on blood biomarkers for Alzheimer's pathophysiology, glial activation, and neurodegeneration
- Author
-
Nicholas J. Ashton, Marc Suárez‐Calvet, Thomas K. Karikari, Juan Lantero‐Rodriguez, Anniina Snellman, Mathias Sauer, Joel Simrén, Carolina Minguillon, Karine Fauria, Kaj Blennow, and Henrik Zetterberg
- Subjects
amyloid beta ,biomarkers ,blood ,citrate ,ethylenediaminetetraacetic acid ,glial fibrillary acidic protein ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Introduction We tested how tube types (ethylenediaminetetraacetic acid [EDTA], serum, lithium heparin [LiHep], and citrate) and freeze–thaw cycles affect levels of blood biomarkers for Alzheimer's disease (AD) pathophysiology, glial activation, and neuronal injury. Methods Amyloid beta (Aβ)42, Aβ40, phosphorylated tau181 (p‐tau181), glial fibrillary acidic protein, total tau (t‐tau), neurofilament light, and phosphorylated neurofilament heavy protein were measured using single molecule arrays. Results LiHep demonstrated the highest mean value for all biomarkers. Tube types were highly correlated for most biomarkers (r > 0.95) but gave significantly different absolute concentrations. Weaker correlations between tube types were found for Aβ42/40 (r = 0.63–0.86) and serum t‐tau (r = 0.46–0.64). Freeze–thaw cycles highly influenced levels of serum Aβ and t‐tau (P 3 should be avoided for p‐tau181.
- Published
- 2021
- Full Text
- View/download PDF
33. Quantitative informant‐ and self‐reports of subjective cognitive decline predict amyloid beta PET outcomes in cognitively unimpaired individuals independently of age and APOE ε4
- Author
-
Gonzalo Sánchez‐Benavides, Gemma Salvadó, Eider M. Arenaza‐Urquijo, Oriol Grau‐Rivera, Marc Suárez‐Calvet, Marta Milà‐Alomà, José María González‐de‐Echávarri, Carolina Minguillon, Marta Crous‐Bou, Aida Niñerola‐Baizán, Andrés Perissinotti, Juan Domingo Gispert, José Luis Molinuevo, and for the ALFA Study
- Subjects
Alzheimer's disease ,amyloid ,informant reports ,preclinical ,subjective cognitive decline ,Neurology. Diseases of the nervous system ,RC346-429 ,Geriatrics ,RC952-954.6 - Abstract
Abstract Introduction Amyloid beta (Aβ) pathology is an Alzheimer's disease early hallmark. Here we assess the value of longitudinal self‐ and informant reports of cognitive decline to predict Aβ positron emission tomography (PET) outcome in cognitively unimpaired middle‐aged individuals. Methods A total of 261 participants from the ALFA+ study underwent [18F]flutemetamol PET and Subjective Cognitive Decline Questionnaire (SCD‐Q) concurrently, and 3 years before scan. We used logistic regressions to evaluate the ability of SCD‐Q scores (self and informant) to predict Aβ PET visual read, and repeated analysis of variance to assess whether changes in SCD‐Q scores relate to Aβ status. Results Self‐perception of decline in memory (odds ratio [OR] = 1.2), and informant perception of executive decline (OR = 1.6), increased the probability of a positive scan. Informant reports 3 years before scanning predicted Aβ PET outcome. Longitudinal increase of self‐reported executive decline was predictive of Aβ in women (P = .003). Discussion Subjective reports of cognitive decline are useful to predict Aβ and may improve recruitment strategies.
- Published
- 2020
- Full Text
- View/download PDF
34. CSF progranulin increases in the course of Alzheimer's disease and is associated with sTREM2, neurodegeneration and cognitive decline
- Author
-
Marc Suárez‐Calvet, Anja Capell, Miguel Ángel Araque Caballero, Estrella Morenas‐Rodríguez, Katrin Fellerer, Nicolai Franzmeier, Gernot Kleinberger, Erden Eren, Yuetiva Deming, Laura Piccio, Celeste M Karch, Carlos Cruchaga, Katrina Paumier, Randall J Bateman, Anne M Fagan, John C Morris, Johannes Levin, Adrian Danek, Mathias Jucker, Colin L Masters, Martin N Rossor, John M Ringman, Leslie M Shaw, John Q Trojanowski, Michael Weiner, Michael Ewers, Christian Haass, the Dominantly Inherited Alzheimer Network, and the Alzheimer's Disease Neuroimaging Initiative
- Subjects
Alzheimer's disease ,biomarker ,microglia ,progranulin ,TREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Progranulin (PGRN) is predominantly expressed by microglia in the brain, and genetic and experimental evidence suggests a critical role in Alzheimer's disease (AD). We asked whether PGRN expression is changed in a disease severity‐specific manner in AD. We measured PGRN in cerebrospinal fluid (CSF) in two of the best‐characterized AD patient cohorts, namely the Dominant Inherited Alzheimer's Disease Network (DIAN) and the Alzheimer's Disease Neuroimaging Initiative (ADNI). In carriers of AD causing dominant mutations, cross‐sectionally assessed CSF PGRN increased over the course of the disease and significantly differed from non‐carriers 10 years before the expected symptom onset. In late‐onset AD, higher CSF PGRN was associated with more advanced disease stages and cognitive impairment. Higher CSF PGRN was associated with higher CSF soluble TREM2 (triggering receptor expressed on myeloid cells 2) only when there was underlying pathology, but not in controls. In conclusion, we demonstrate that, although CSF PGRN is not a diagnostic biomarker for AD, it may together with sTREM2 reflect microglial activation during the disease.
- Published
- 2018
- Full Text
- View/download PDF
35. Latest advances in cerebrospinal fluid and blood biomarkers of Alzheimer’s disease
- Author
-
Marta Milà-Alomà, Marc Suárez-Calvet, and José Luís Molinuevo
- Subjects
Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease and its diagnosis has classically been based on clinical symptoms. Recently, a biological rather than a syndromic definition of the disease has been proposed that is based on biomarkers that reflect neuropathological changes. In AD, there are two main biomarker categories, namely neuroimaging and fluid biomarkers [cerebrospinal fluid (CSF) and blood]. As a complex and multifactorial disease, AD biomarkers are important for an accurate diagnosis and to stage the disease, assess the prognosis, test target engagement, and measure the response to treatment. In addition, biomarkers provide us with information that, even if it does not have a current clinical use, helps us to understand the mechanisms of the disease. In addition to the pathological hallmarks of AD, which include amyloid-β and tau deposition, there are multiple concomitant pathological events that play a key role in the disease. These include, but are not limited to, neurodegeneration, inflammation, vascular dysregulation or synaptic dysfunction. In addition, AD patients often have an accumulation of other proteins including α-synuclein and TDP-43, which may have a pathogenic effect on AD. In combination, there is a need to have biomarkers that reflect different aspects of AD pathogenesis and this will be important in the future to establish what are the most suitable applications for each of these AD-related biomarkers. It is unclear whether sex, gender, or both have an effect on the causes of AD. There may be differences in fluid biomarkers due to sex but this issue has often been neglected and warrants further research. In this review, we summarize the current state of the principal AD fluid biomarkers and discuss the effect of sex on these biomarkers.
- Published
- 2019
- Full Text
- View/download PDF
36. Glial Activation Markers in CSF and Serum From Patients With Primary Progressive Multiple Sclerosis: Potential of Serum GFAP as Disease Severity Marker?
- Author
-
Ahmed Abdelhak, Tilman Hottenrott, Estrella Morenas-Rodríguez, Marc Suárez-Calvet, Uwe K. Zettl, Christian Haass, Sven G. Meuth, Sebastian Rauer, Markus Otto, Hayrettin Tumani, and André Huss
- Subjects
SiMoA ,GFAP ,PPMS ,glial activation ,progressive multiple sclerosis ,neurofilaments ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Background: In progressive multiple sclerosis (MS), glial activation is thought to be a relevant mechanism of disability progression. Therefore, in vivo assessment of the glial cell activity is, in the emerging treatment era of primary progressive MS (PPMS), more important than ever.Objectives: To test the association of cerebrospinal fluid (CSF) and serum markers of glial activation in PPMS patients; including glial fibrillary acidic protein (GFAP), chitinase-3-like protein 1 (CHI3L1), soluble variant of triggering receptor expressed on myeloid cells 2 (sTREM2), and marker of neuroaxonal damage (Neurofilament light chain, NfL) as well as clinical severity.Methods: CSF and serum samples from PPMS patients were collected in the MS-centers at Universities of Freiburg (n = 49), Ulm (n = 27), Muenster (n = 11), and Rostock (n = 6). sTREM2 and CHI3L1 levels were measured using the previously reported ELISA assays, while NfL and GFAP were measured using SIMOA assays. Clinical data included age, gender, disease duration, treatment status, and Expanded Disability Status Scale (EDSS).Results: 93 CSF samples and 71 matching serum samples were analyzed. The median age of patients was 49 years and disease duration 4.5 years. GFAPserum correlated with EDSS after correction for age (β = 0.3, p = 0.001). Furthermore, EDSS was higher in patients with a GFAPserum level ≥ 151.7 pg/ml compared to patients with GFAPserum below this cut-off (5.5 vs. 4.0, p = 0.009). Other markers did not correlate with the clinical severity. Moreover, we found a correlation between NfLCSF and GFAPCSF, sTREM2 and CHI3L1 (ρ = 0.4 for GFAPCSF and sTREM2, ρ = 0.3 for CHI3L1, p < 0.01 for sTREM2 and CHI3L1 and
- Published
- 2019
- Full Text
- View/download PDF
37. CSF glial biomarkers YKL40 and sTREM2 are associated with longitudinal volume and diffusivity changes in cognitively unimpaired individuals
- Author
-
Carles Falcon, Gemma C. Monté-Rubio, Oriol Grau-Rivera, Marc Suárez-Calvet, Raquel Sánchez-Valle, Lorena Rami, Beatriz Bosch, Christian Haass, Juan Domingo Gispert, and José Luis Molinuevo
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Cerebrospinal fluid (CSF) YKL40 and sTREM2 are astroglial and microglial activity biomarkers, respectively. We assessed whether CSF YKL40 and sTREM2 baseline levels are associated with longitudinal brain volume and diffusivity changes in cognitively unimpaired adults. Two brain MRI scans of 36 participants (57 to 78-years old, 12 male) were acquired in a 2-year interval. Aβ42, p-tau, YKL40 and sTREM2 concentrations in CSF were determined at baseline. We calculated gray and white matter volume changes per year maps (ΔGM and ΔWM, respectively) by means of longitudinal pairwise registration, and mean diffusivity variation per year (ΔMD) by subtraction. We checked voxel-wise for associations between ΔGM, ΔWM and ΔMD and baseline CSF level of YKL40 and sTREM2 and verified to what extent these associations were modulated by age (YKL40xAGE and sTREM2xAGE interactions). We found a positive association between ΔGM and YKL40 in the left inferior parietal region and no association between sTREM2 and ΔGM. Negative associations were also observed between ΔGM and YKL40xAGE (bilateral frontal areas, left precuneus and left postcentral and supramarginal gyri) and sTREM2xAGE (bilateral temporal and frontal cortex, putamen and left middle cingulate gyrus). We found negative associations between ΔWM and YKL40xAGE (bilateral superior longitudinal fasciculus) and sTREM2xAGE (bilateral superior longitudinal fasciculus, left superior corona radiata, retrolenticular external capsule and forceps minor, among other regions) but none between ΔWM and neither YKL40 nor sTREM2. ΔMD was positively correlated with YKL40 in right orbital region and negatively with sTREM2 in left lingual gyrus and precuneus. In addition, significant associations were found between ΔMD and YKL40xAGE (tail of left hippocampus and surrounding areas and right anterior cingulate gyrus) and sTREM2xAGE (right superior temporal gyrus). Areas showing statistically significant differences were disjoint in analyses involving YKL40 and sTREM2. These results suggest that glial biomarkers exert a relevant and distinct influence in longitudinal brain macro- and microstructural changes in cognitively unimpaired adults, which appears to be modulated by age. In younger subjects increased glial markers (both YKL40 and sTREM2) predict a better outcome, as indicated by a decrease in ΔGM and ΔWM and an increase in ΔMD, whereas in older subjects this association is inverted and higher levels of glial markers are associated with a poorer neuroimaging outcome. Keywords: TREM2, YKL40, Preclinical Alzheimer's disease, Longitudinal analysis, Mean diffusivity
- Published
- 2019
- Full Text
- View/download PDF
38. Interactive effect of age and APOE-ε4 allele load on white matter myelin content in cognitively normal middle-aged subjects
- Author
-
Grégory Operto, José Luis Molinuevo, Raffaele Cacciaglia, Carles Falcon, Anna Brugulat-Serrat, Marc Suárez-Calvet, Oriol Grau-Rivera, Nuria Bargalló, Sebastián Morán, Manel Esteller, and Juan Domingo Gispert
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
The apolipoprotein E gene (APOE) ε4 allele has a strong and manifold impact on cognition and neuroimaging phenotypes in cognitively normal subjects, including alterations in the white matter (WM) microstructure. Such alterations have often been regarded as a reflection of potential thinning of the myelin sheath along axons, rather than pure axonal degeneration. Considering the main role of APOE in brain lipid transport, characterizing the impact of APOE on the myelin coating is therefore of crucial interest, especially in healthy APOE-ε4 homozygous individuals, who are exposed to a twelve-fold higher risk of developing Alzheimer's disease (AD), compared to the rest of the population.We examined T1w/T2w ratio maps in 515 cognitively healthy middle-aged participants from the ALFA study (ALzheimer and FAmilies) cohort, a single-site population-based study enriched for AD risk (68 APOE-ε4 homozygotes, 197 heterozygotes, and 250 non-carriers). Using tract-based spatial statistics, we assessed the impact of age and APOE genotype on this ratio taken as an indirect descriptor of myelin content.Healthy APOE-ε4 carriers display decreased T1w/T2w ratios in extensive regions in a dose-dependent manner. These differences were found to interact with age, suggesting faster changes in individuals with more ε4 alleles.These results obtained with T1w/T2w ratios, confirm the increased vulnerability of WM tracts in APOE-ε4 healthy carriers. Early alterations of myelin content could be the result of the impaired function of the ε4 isoform of the APOE protein in cholesterol transport. These findings help to clarify the possible interactions between the APOE-dependent non-pathological burden and age-related changes potentially at the source of the AD pathological cascade. Keywords: Myelination, T1w/T2w ratio, Apolipoprotein E, White matter integrity, Aging, Cognitively normal subjects, Alzheimer
- Published
- 2019
- Full Text
- View/download PDF
39. sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early‐stage Alzheimer's disease and associate with neuronal injury markers
- Author
-
Marc Suárez‐Calvet, Gernot Kleinberger, Miguel Ángel Araque Caballero, Matthias Brendel, Axel Rominger, Daniel Alcolea, Juan Fortea, Alberto Lleó, Rafael Blesa, Juan Domingo Gispert, Raquel Sánchez‐Valle, Anna Antonell, Lorena Rami, José L Molinuevo, Frederic Brosseron, Andreas Traschütz, Michael T Heneka, Hanne Struyfs, Sebastiaan Engelborghs, Kristel Sleegers, Christine Van Broeckhoven, Henrik Zetterberg, Bengt Nellgård, Kaj Blennow, Alexander Crispin, Michael Ewers, and Christian Haass
- Subjects
Alzheimer's disease ,biomarkers ,microglia ,neurodegeneration ,TREM2 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract TREM2 is an innate immune receptor expressed on the surface of microglia. Loss‐of‐function mutations of TREM2 are associated with increased risk of Alzheimer's disease (AD). TREM2 is a type‐1 protein with an ectodomain that is proteolytically cleaved and released into the extracellular space as a soluble variant (sTREM2), which can be measured in the cerebrospinal fluid (CSF). In this cross‐sectional multicenter study, we investigated whether CSF levels of sTREM2 are changed during the clinical course of AD, and in cognitively normal individuals with suspected non‐AD pathology (SNAP). CSF sTREM2 levels were higher in mild cognitive impairment due to AD than in all other AD groups and controls. SNAP individuals also had significantly increased CSF sTREM2 compared to controls. Moreover, increased CSF sTREM2 levels were associated with higher CSF total tau and phospho‐tau181P, which are markers of neuronal degeneration and tau pathology. Our data demonstrate that CSF sTREM2 levels are increased in the early symptomatic phase of AD, probably reflecting a corresponding change of the microglia activation status in response to neuronal degeneration.
- Published
- 2016
- Full Text
- View/download PDF
40. Tau enhances α-synuclein aggregation and toxicity in cellular models of synucleinopathy.
- Author
-
Nahuai Badiola, Rita Machado de Oliveira, Federico Herrera, Cristina Guardia-Laguarta, Susana A Gonçalves, Marta Pera, Marc Suárez-Calvet, Jordi Clarimon, Tiago Fleming Outeiro, and Alberto Lleó
- Subjects
Medicine ,Science - Abstract
BackgroundThe simultaneous accumulation of different misfolded proteins in the central nervous system is a common feature in many neurodegenerative diseases. In most cases, co-occurrence of abnormal deposited proteins is observed in different brain regions and cell populations, but, in some instances, the proteins can be found in the same cellular aggregates. Co-occurrence of tau and α-synuclein (α-syn) aggregates has been described in neurodegenerative disorders with primary deposition of α-syn, such as Parkinson's disease and dementia with Lewy bodies. Although it is known that tau and α-syn have pathological synergistic effects on their mutual fibrillization, the underlying biological effects remain unclear.Methodology/principal findingsWe used different cell models of synucleinopathy to investigate the effects of tau on α-syn aggregation. Using confocal microscopy and FRET-based techniques we observed that tau colocalized and interacted with α-syn aggregates. We also found that tau overexpression changed the pattern of α-syn aggregation, reducing the size and increasing the number of aggregates. This shift was accompanied by an increase in the levels of insoluble α-syn. Furthermore, co-transfection of tau increased secreted α-syn and cytotoxicity.Conclusions/significanceOur data suggest that tau enhances α-syn aggregation and toxicity and disrupts α-syn inclusion formation. This pathological synergistic effect between tau and α-syn may amplify the deleterious process and spread the damage in neurodegenerative diseases that show co-occurrence of both pathologies.
- Published
- 2011
- Full Text
- View/download PDF
41. NEURONORMA Cognitive Battery Associations with Cerebrospinal Fluid Amyloid-β and Tau Levels in the Continuum of Alzheimer’s Disease
- Author
-
Greta García-Escobar, Albert Puig-Pijoan, Víctor Puente-Periz, Aida Fernández-Lebrero, Rosa María Manero, Irene Navalpotro-Gómez, Marc Suárez-Calvet, Oriol Grau-Rivera, José Contador-Muñana, Diego Cascales-Lahoz, Xavier Duran-Jordà, Núncia Boltes, Maria Claustre Pont-Sunyer, Jordi Ortiz-Gil, Sara Carrillo-Molina, María Dolores López-Villegas, María Teresa Abellán-Vidal, María Isabel Martínez-Casamitjana, Juan José Hernández-Sánchez, Anna Padrós-Fluvià, Jordi Peña-Casanova, and Gonzalo Sánchez-Benavides
- Subjects
Psychiatry and Mental health ,Clinical Psychology ,General Neuroscience ,General Medicine ,Geriatrics and Gerontology - Abstract
Background: Neuropsychological assessments are essential to define the cognitive profile and contribute to the diagnosis of Alzheimer’s disease (AD). The progress in knowledge about the pathophysiological process of the disease has allowed conceptualizing AD through biomarkers as a biological continuum that encompasses different clinical stages. Objective: To explore the association between cerebrospinal fluid (CSF) biomarkers of AD and cognition using the NEURONORMA battery, in a sample of cognitively unimpaired (CU), mild cognitive impaired (MCI), and mild dementia of the Alzheimer type (DAT) subjects, and to characterize the cognitive profiles in MCI subjects classified by A/T/N system. Methods: 42 CU, 35 MCI, and 35 mild DAT were assessed using the NEURONORMA battery. Core AD biomarkers [amyloid-β42 (Aβ42) peptide, total tau (t-tau), and phosphorylated tau 181 (p-tau181)] proteins were measured in CSF. Correlation coefficients, multivariate regression, and effect sizes were calculated. We explored the age- and education-adjusted cognitive profiles by A/T/N variants within the MCI group. Results: Cognitive outcomes were directly associated with CSF Aβ42 and inversely with CSF tau measures. We found differences in both biomarkers and cognitive outcomes comparing all pairs except for CSF measures between cognitively impaired groups. The highest effect size was in memory tasks and biomarkers ratios. Lower performances were in memory and executive domains in MCI subjects with AD pathology (A+T+N±) compared to those with normal levels of AD biomarkers (A– T– N). Conclusion: This study provides further evidence of the validity of Spanish NEURONORMA cognitive battery to characterize cognitive impairment in the AD pathological continuum.
- Published
- 2023
42. Reactive astrogliosis is associated with higher cerebral glucose consumption in the early Alzheimer’s continuum
- Author
-
Gemma, Salvadó, Marta, Milà-Alomà, Mahnaz, Shekari, Nicholas J, Ashton, Grégory, Operto, Carles, Falcon, Raffaele, Cacciaglia, Carolina, Minguillon, Karine, Fauria, Aida, Niñerola-Baizán, Andrés, Perissinotti, Andréa L, Benedet, Gwendlyn, Kollmorgen, Ivonne, Suridjan, Norbert, Wild, José Luis, Molinuevo, Henrik, Zetterberg, Kaj, Blennow, Marc, Suárez-Calvet, and Juan Domingo, Gispert
- Subjects
Inflammation ,Amyloid beta-Peptides ,Glucose ,Alzheimer Disease ,Fluorodeoxyglucose F18 ,Glial Fibrillary Acidic Protein ,Humans ,tau Proteins ,Radiology, Nuclear Medicine and imaging ,Gliosis ,General Medicine ,Biomarkers - Abstract
Purpose: glial activation is one of the earliest mechanisms to be altered in Alzheimer's disease (AD). Glial fibrillary acidic protein (GFAP) relates to reactive astrogliosis and can be measured in both cerebrospinal fluid (CSF) and blood. Plasma GFAP has been suggested to become altered earlier in AD than its CSF counterpart. Although astrocytes consume approximately half of the glucose-derived energy in the brain, the relationship between reactive astrogliosis and cerebral glucose metabolism is poorly understood. Here, we aimed to investigate the association between fluorodeoxyglucose ([18F]FDG) uptake and reactive astrogliosis, by means of GFAP quantified in both plasma and CSF for the same participants. Methods: we included 314 cognitively unimpaired participants from the ALFA + cohort, 112 of whom were amyloid-β (Aβ) positive. Associations between GFAP markers and [18F]FDG uptake were studied. We also investigated whether these associations were modified by Aβ and tau status (AT stages). Results: plasma GFAP was positively associated with glucose consumption in the whole brain, while CSF GFAP associations with [18F]FDG uptake were only observed in specific smaller areas like temporal pole and superior temporal lobe. These associations persisted when accounting for biomarkers of Aβ pathology but became negative in Aβ-positive and tau-positive participants (A + T +) in similar areas of AD-related hypometabolism. Conclusions: higher astrocytic reactivity, probably in response to early AD pathological changes, is related to higher glucose consumption. With the onset of tau pathology, the observed uncoupling between astrocytic biomarkers and glucose consumption might be indicative of a failure to sustain the higher energetic demands required by reactive astrocytes. The project leading to these results has received funding from “la Caixa” Foundation (ID 100010434), under agreement LCF/PR/GN17/50300004 and the Alzheimer’s Association and an international anonymous charity foundation through the TriBEKa Imaging Platform project (TriBEKa-17–519007). Additional support has been received from the Universities and Research Secretariat, Ministry of Business and Knowledge of the Catalan Government under the grant no. 2017-SGR-892. JDG is supported by the Spanish Ministry of Science and Innovation (RYC-2013–13054). MSC receives funding from Instituto de Salud Carlos III (PI19/00155) and from the Spanish Ministry of Science, Innovation and Universities (Juan de la Cierva programme grant IJC2018-037478-I). CM receives funding within the context of EURO-FINGERS, an EU Joint Programme—Neurodegenerative Disease Research (JPND) project. The EURO-FINGERS project is supported through the following funding organizations under the aegis of JPND—www.jpnd.eu: Finland, Academy of Finland; Germany, Federal Ministry of Education and Research; Spain, National Institute of Health Carlos III; Luxembourg, National Research Fund; Hungary, National Research, Development and Innovation Office; and the Netherlands, Netherlands Organisation for Health Research and Development (ZonMW-Memorabel #733051102). HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018–02532); the European Research Council (#681712); the Swedish State Support for Clinical Research (#ALFGBG-720931); the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809–2016862); the AD Strategic Fund and the Alzheimer’s Association (#ADSF-21–831376-C, #ADSF-21–831381-C, and #ADSF-21–831377-C); the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2019-0228); the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement no 860197 (MIRIADE), and the UK Dementia Research Institute at UCL. KB is supported by the Swedish Research Council (#2017–00915); the Alzheimer Drug Discovery Foundation (ADDF), USA (#RDAPB-201809–2016615); the Swedish Alzheimer Foundation (#AF-742881); Hjärnfonden, Sweden (#FO2017-0243); the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986); the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466–236); and the National Institute of Health (NIH), USA (grant #1R01AG068398-01).
- Published
- 2022
43. Exploring cognitive and biological correlates of sleep quality and their potential links with Alzheimer's disease (ALFASleep project): protocol for an observational study
- Author
-
Karine Fauria, Carolina Minguillon, Iva Knezevic, Núria Tort-Colet, Laura Stankeviciute, Laura Hernández, Andreea Rădoi, Carme Deulofeu, Sherezade Fuentes-Julián, Israel Turull, David Fusté, Gonzalo Sánchez-Benavides, Eider M Arenaza-Urquijo, Marc Suárez-Calvet, Sebastian C Holst, Pilar Garcés, Thomas Mueggler, Henrik Zetterberg, Kaj Blennow, Aurora Arqueros, Álex Iranzo, Juan Domingo Gispert, José Luis Molinuevo, and Oriol Grau-Rivera
- Subjects
Neurology ,Dementia ,General Medicine ,Sleep medicine - Abstract
Introduction: the growing worldwide prevalence of Alzheimer's disease (AD) and the lack of effective treatments pose a dire medical challenge. Sleep disruption is also prevalent in the ageing population and is increasingly recognised as a risk factor and an early sign of AD. The ALFASleep project aims to characterise sleep with subjective and objective measurements in cognitively unimpaired middle/late middle-aged adults at increased risk of AD who are phenotyped with fluid and neuroimaging AD biomarkers. This will contribute to a better understanding of the pathophysiological mechanisms linking sleep with AD, thereby paving the way for the development of non-invasive biomarkers and preventive strategies targeting sleep. Methods and analysis: we will invite 200 participants enrolled in the ALFA+ (for ALzheimer and FAmilies) prospective observational study to join the ALFASleep study. ALFA+ participants are cognitively unimpaired middle-aged/late middle-aged adults who are followed up every 3 years with a comprehensive set of evaluations including neuropsychological tests, blood and cerebrospinal fluid (CSF) sampling, and MRI and positron emission tomography acquisition. ALFASleep participants will be additionally characterised with actigraphy and CSF-orexin-A measurements, and a subset (n=90) will undergo overnight polysomnography. We will test associations of sleep measurements and CSF-orexin-A with fluid biomarkers of AD and glial activation, neuroimaging outcomes and cognitive performance. In case we found any associations, we will test whether changes in AD and/or glial activation markers mediate the association between sleep and neuroimaging or cognitive outcomes and whether sleep mediates associations between CSF-orexin-A and AD biomarkers. Ethics and dissemination: the ALFASleep study protocol has been approved by the independent Ethics Committee Parc de Salut Mar, Barcelona (2018/8207/I). All participants have signed a written informed consent before their inclusion (approved by the same ethics committee). Study findings will be presented at national and international conferences and submitted for publication in peer-reviewed journals. This work was supported by a grant from the Alzheimer’s Association (2019-AARF-644568) awarded to OG-R. The ALFASleep study also receives funding from the Instituto de Salud Carlos III through the project PI19/00117 (cofunded by European Regional Development Fund, 'A Way to Make Europe') awarded to OG-R. Additional funding has been received from F. Hoffmann-La Roche. The ALFA+ study is funded by 'la Caixa' Foundation (ID 100010434), under agreement LCF/PR/GN17/50300004 and the Alzheimer's Association and an international anonymous charity foundation through the TriBEKa Imaging Platform project (TriBEKa‐17‐519007). OG-R is supported by the Spanish Ministry of Science, Innovation and Universities (IJC2020-043417-I). HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council (#2018-02532), the European Research Council (#681712 and #101053962), Swedish State Support for Clinical Research (#ALFGBG-71320), the Alzheimer Drug Discovery Foundation (ADDF), USA (#201809-2016862), the AD Strategic Fund and the Alzheimer's Association (#ADSF-21-831376-C, #ADSF-21-831381-C and #ADSF-21-831377-C), the Olav Thon Foundation, the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden (#FO2019-0228), the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement number 860197 (MIRIADE), the European Union Joint Programme–Neurodegenerative Disease Research (JPND2021-00694) and the UK Dementia Research Institute at UCL (UKDRI-1003). KB is supported by the Swedish Research Council (#2017-00915), the ADDF, USA (#RDAPB-201809-2016615), the Swedish Alzheimer Foundation (#AF-930351, #AF-939721 and #AF-968270), Hjärnfonden, Sweden (#FO2017-0243 and #ALZ2022-0006), the Swedish state under the agreement between the Swedish government and the County Councils, the ALF-agreement (#ALFGBG-715986 and #ALFGBG-965240), the European Union Joint Program for Neurodegenerative Disorders (JPND2019-466-236), the National Institutes of Health, USA, (grant #1R01AG068398-01) and the Alzheimer’s Association 2021 Zenith Award (ZEN-21-848495).
- Published
- 2022
44. Structural and metabolic brain correlates of excess Aβ accumulation at the earliest AD continuum
- Author
-
Raffaele Cacciaglia, Marta Milà‐Alomà, Mahnaz Shekari, Grégory Operto, Carles Falcon, Carolina Minguillón, Jose Luis Molinuevo, Henrik Zetterberg, Kaj Blennow, Marc Suárez‐Calvet, Frances‐Catherine Quevenco, and Juan Domingo Gispert
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
45. Voxel‐wise Staging of Tau Pathology using [ 18 F]RO‐948 PET in the Early AD Continuum
- Author
-
Mahnaz Shekari, Marta Milà‐Alomà, Carles Falcon, Aida Niñerola‐Baizán, Grégory Operto, Marina Garcia, Gonzalo Sánchez‐Benavides, Anna Brugulat‐Serrat, Matteo Tonietto, Edilio Borroni, Gregory Klein, Nicholas J. Ashton, Thomas K Karikari, Juan Lantero Rodriguez, Anniina Snellman, Paula Ortiz‐Romero, Eugeen Vanmechelen, Carolina Minguillón, Karine Fauria, Andrés Perissinotti, Jose Luis Molinuevo, Henrik Zetterberg, Kaj Blennow, Oriol Grau‐Rivera, Marc Suárez‐Calvet, and Juan Domingo Gispert
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
46. Associations between brain iron deposition and structural Alzheimer’s disease signature in cognitively unimpaired adults
- Author
-
Laura Stankeviciute, Carles Falcon, Grégory Operto, Santiago Rojas, Oriol Grau‐Rivera, Marina Garcia, Mahnaz Shekari, Aida Niñerola‐Baizán, Andrés Perissinotti, Carolina Minguillón, Karine Fauria, Jose Luis Molinuevo, Henrik Zetterberg, Kaj Blennow, Marc Suárez‐Calvet, Raffaele Cacciaglia, and Juan Domingo Gispert
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
47. Derivation of cut‐offs for plasma neurofilament light in clinical routine using healthy subjects aged between 5‐90
- Author
-
Joel Simrén, Johan Gobom, Ulf Andreasson, Marc Suárez‐Calvet, Herman Depypere, Lars Nyberg, Barbara Borroni, Henrik Zetterberg, and Kaj Blennow
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
48. [ 18 F]RO‐948 Tau PET Retention and Correlation with Fluid Tau Biomarkers in the Early AD Continuum
- Author
-
Mahnaz Shekari, Marta Milà‐Alomà, Carles Falcon, Aida Niñerola‐Baizán, Matteo Tonietto, Edilio Borroni, Gregory Klein, Nicholas J. Ashton, Thomas K Karikari, Juan Lantero Rodriguez, Anniina Snellman, Paula Ortiz‐Romero, Eugeen Vanmechelen, Carolina Minguillón, Karine Fauria, Andrés Perissinotti, Jose Luis Molinuevo, Henrik Zetterberg, Kaj Blennow, Oriol Grau‐Rivera, Marc Suárez‐Calvet, and Juan Domingo Gispert
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
49. Stress, anxiety and depression in older adults at increased risk for cognitive decline: association with biomarkers and sex/gender differences
- Author
-
Eider M Arenaza‐Urquijo, Muge Akinci, Eleni Palpatzis, Cleofé Peña‐Gomez, Marc Suárez‐Calvet, Juan Domingo Gispert, Carolina Minguillón, Oriol Grau‐Rivera, Gonzalo Sánchez‐Benavides, and Karine Fauria
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
50. Modifying effect of AD pathology in the association between CSF synaptic biomarkers and brain function and structure in preclinical Alzheimer
- Author
-
Marta Milà‐Alomà, Raffaele Cacciaglia, Mahnaz Shekari, Grégory Operto, Ann Brinkmalm, Hlin Kvartsberg, Nicholas J. Ashton, Gwendlyn Kollmorgen, Norbert Wild, Ivonne Suridjan, Henrik Zetterberg, Kaj Blennow, Jose Luis Molinuevo, Juan Domingo Gispert, and Marc Suárez‐Calvet
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,Neurology (clinical) ,Geriatrics and Gerontology - Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.