Rosanne L. van den Berg, Casper de Boer, Marissa D. Zwan, Roos J. Jutten, Mariska van Liere, Marie-Christine A.B.J. van de Glind, Mark A. Dubbelman, Lisa Marie Schlüter, Argonde C. van Harten, Charlotte E. Teunissen, Elsmarieke van de Giessen, Frederik Barkhof, Lyduine E. Collij, Jessica Robin, William Simpson, John E Harrison, Wiesje M. van der Flier, Sietske A.M. Sikkes
{"title":"Digital remote assessment of speech acoustics in cognitively unimpaired adults: feasibility, reliability and associations with amyloid pathology","authors":"Rosanne L. van den Berg, Casper de Boer, Marissa D. Zwan, Roos J. Jutten, Mariska van Liere, Marie-Christine A.B.J. van de Glind, Mark A. Dubbelman, Lisa Marie Schlüter, Argonde C. van Harten, Charlotte E. Teunissen, Elsmarieke van de Giessen, Frederik Barkhof, Lyduine E. Collij, Jessica Robin, William Simpson, John E Harrison, Wiesje M. van der Flier, Sietske A.M. Sikkes","doi":"10.1186/s13195-024-01543-3","DOIUrl":"https://doi.org/10.1186/s13195-024-01543-3","url":null,"abstract":"Digital speech assessment has potential relevance in the earliest, preclinical stages of Alzheimer’s disease (AD). We evaluated the feasibility, test-retest reliability, and association with AD-related amyloid-beta (Aβ) pathology of speech acoustics measured over multiple assessments in a remote setting. Fifty cognitively unimpaired adults (Age 68 ± 6.2 years, 58% female, 46% Aβ-positive) completed remote, tablet-based speech assessments (i.e., picture description, journal-prompt storytelling, verbal fluency tasks) for five days. The testing paradigm was repeated after 2–3 weeks. Acoustic speech features were automatically extracted from the voice recordings, and mean scores were calculated over the 5-day period. We assessed feasibility by adherence rates and usability ratings on the System Usability Scale (SUS) questionnaire. Test-retest reliability was examined with intraclass correlation coefficients (ICCs). We investigated the associations between acoustic features and Aβ-pathology, using linear regression models, adjusted for age, sex and education. The speech assessment was feasible, indicated by 91.6% adherence and usability scores of 86.0 ± 9.9. High reliability (ICC ≥ 0.75) was found across averaged speech samples. Aβ-positive individuals displayed a higher pause-to-word ratio in picture description (B = -0.05, p = 0.040) and journal-prompt storytelling (B = -0.07, p = 0.032) than Aβ-negative individuals, although this effect lost significance after correction for multiple testing. Our findings support the feasibility and reliability of multi-day remote assessment of speech acoustics in cognitively unimpaired individuals with and without Aβ-pathology, which lays the foundation for the use of speech biomarkers in the context of early AD.","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":9.0,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141863789","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Noor Abdulhameed, Alice Babin, Kim Hansen, Riley Weaver, William A Banks, Konrad Talbot, Elizabeth M Rhea
{"title":"Comparing regional brain uptake of incretin receptor agonists after intranasal delivery in CD-1 mice and the APP/PS1 mouse model of Alzheimer's disease.","authors":"Noor Abdulhameed, Alice Babin, Kim Hansen, Riley Weaver, William A Banks, Konrad Talbot, Elizabeth M Rhea","doi":"10.1186/s13195-024-01537-1","DOIUrl":"10.1186/s13195-024-01537-1","url":null,"abstract":"<p><p>Targeting brain insulin resistance (BIR) has become an attractive alternative to traditional therapeutic treatments for Alzheimer's disease (AD). Incretin receptor agonists (IRAs), targeting either or both of the glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors, have proven to reverse BIR and improve cognition in mouse models of AD. We previously showed that many, but not all, IRAs can cross the blood-brain barrier (BBB) after intravenous (IV) delivery. Here we determined if widespread brain uptake of IRAs could be achieved by circumventing the BBB using intranasal (IN) delivery, which has the added advantage of minimizing adverse gastrointestinal effects of systemically delivered IRAs. Of the 5 radiolabeled IRAs tested (exenatide, dulaglutide, semaglutide, DA4-JC, and DA5-CH) in CD-1 mice, exenatide, dulaglutide, and DA4-JC were successfully distributed throughout the brain following IN delivery. We observed significant sex differences in uptake for DA4-JC. Dulaglutide and DA4-JC exhibited high uptake by the hippocampus and multiple neocortical areas. We further tested and found the presence of AD-associated Aβ pathology minimally affected uptake of dulaglutide and DA4-JC. Of the 5 tested IRAs, dulaglutide and DA4-JC are best capable of accessing brain regions most vulnerable in AD (neocortex and hippocampus) after IN administration. Future studies will need to be performed to determine if IN IRA delivery can reduce BIR in AD or animal models of that disorder.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11293113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141858764","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Efficacy of acoustic stimulation techniques on cognitive functions in individuals with Alzheimer's disease-a scoping review.","authors":"Leelavathi Thamizhmani, Kanaka Ganapathy, Hari Prakash Palaniswamy, Divya Sussana Patil, Suzanne Carolyn Purdy","doi":"10.1186/s13195-024-01544-2","DOIUrl":"10.1186/s13195-024-01544-2","url":null,"abstract":"<p><strong>Background: </strong>Alzheimer's disease (AD) is a progressive neurodegenerative disorder that severely affects cognitive functions and social behaviors, leading to a significant decline in an individual's quality of life. Auditory processing deficits often precede the clinical symptoms of AD, prompting interest in auditory-based interventions as potential treatments. This scoping review aimed to compile the existing evidence on active and passive auditory-based interventions for individuals with AD and its prodromal stages.</p><p><strong>Method and results: </strong>This scoping review followed Arksey and O'Malley's five-step framework to identify the existing evidence on auditory-based interventions for AD. Four databases (PubMed, Web of Science, CINAHL, and Embase) were used to search for studies on auditory stimulation techniques to treat cognitive decline in AD patients. In total, 14 studies were included in the analysis. Seven studies explored active auditory stimulation techniques, such as the Brain Fitness Program (BrainHQ), aiming to improve cognitive function in individuals with Mild Cognitive Impairment (MCI). The other seven studies focused on passive auditory stimulation, often combined with other sensory stimuli such as light or tactile inputs. Passive stimulation studies have focused mainly on Gamma Entrainment Using Sensory Stimulation (GENUS). The intervention frequency and duration varied across studies, ranging from one session lasting 8 h to a year. Both active and passive auditory stimulation showed potential for enhancing cognitive function in individuals with AD.</p><p><strong>Conclusion: </strong>The literature suggests that auditory stimulation may positively influence cortical wiring and enhance cognitive abilities. Multimodal interventions that combine auditory stimulation with other sensory or behavioural approaches could yield more substantial effects on global cognition. However, the study design, intervention characteristics and outcome measures varied across studies, underscoring the necessity for standardised reporting. Well-designed studies using standard cognitive assessment protocols are recommended.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11293131/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141858765","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lukas Preis, Kersten Villringer, Frederic Brosseron, Emrah Düzel, Frank Jessen, Gabor C Petzold, Alfredo Ramirez, Annika Spottke, Jochen B Fiebach, Oliver Peters
{"title":"Assessing blood-brain barrier dysfunction and its association with Alzheimer's pathology, cognitive impairment and neuroinflammation.","authors":"Lukas Preis, Kersten Villringer, Frederic Brosseron, Emrah Düzel, Frank Jessen, Gabor C Petzold, Alfredo Ramirez, Annika Spottke, Jochen B Fiebach, Oliver Peters","doi":"10.1186/s13195-024-01529-1","DOIUrl":"10.1186/s13195-024-01529-1","url":null,"abstract":"<p><strong>Background: </strong>Blood-brain barrier (BBB) alterations may contribute to AD pathology through various mechanisms, including impaired amyloid-β (Aβ) clearance and neuroinflammation. Soluble platelet-derived growth factor receptor beta (sPDGFRβ) has emerged as a potential biomarker for BBB integrity. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) offers a direct assessment of BBB permeability. However, the relationship between BBB dysfunction, cognitive impairment, and AD pathology remains unclear, with inconsistent findings in the literature.</p><p><strong>Methods: </strong>We conducted a cross-sectional study using data from the DELCODE and DESCRIBE cohorts to investigate BBB dysfunction in participants with normal cognition (NC), mild cognitive impairment (MCI), and AD dementia. BBB function was assessed using DCE-MRI and sPDGFRβ levels in cerebrospinal fluid and AD biomarkers Aβ and tau were measured. In a subset of patients, the CSF/plasma-ratio of albumin (QAlb) as a standard marker of BBB integrity and markers of neuroinflammation were analyzed.</p><p><strong>Results: </strong>91 participants (NC: 44, MCI: 21, AD: 26) were included in the analysis. The average age was 74.4 years, 42% were female. Increased hippocampal BBB disruption was observed in the AD-group (K<sup>trans</sup>: 0.55 × 10<sup>- 3</sup> min<sup>- 1</sup> ± 0.74 × 10<sup>- 3</sup> min<sup>- 1</sup>) but not the MCI-group (K<sup>trans</sup>: 0.177 × 10<sup>- 3</sup> min<sup>- 1</sup> ± 0.22 × 10<sup>- 3</sup> min<sup>- 1</sup>), compared to the NC group (K<sup>trans</sup>: 0.19 × 10<sup>- 3</sup> min<sup>- 1</sup> ± 0.37 × 10<sup>- 3</sup> min<sup>- 1</sup>, p < .01). sPDGFRβ was not significantly different between the cognitive groups. However, sPDGFRβ levels were significantly associated with age (r = .33, p < .01), independent of vascular risk factors. Further, sPDGFRβ showed significant positive associations with soluble Aβ levels (Aβ40: r = .57, p < .01; Aβ42: r = .39, p < .01) and YKL-40 (r = .53, p < .01), a marker of neuroinflammation. sPDGFRβ/DCE-MRI was not associated with overall AD biomarker positivity or APOE-status.</p><p><strong>Conclusion: </strong>In dementia, but not MCI, hippocampal BBB disruption was observed. sPDGFRβ increased with age and was associated with neuroinflammation independent of cognitive impairment. The association between Aβ and sPDGFRβ may indicate a bidirectional relationship reflecting pericytes' clearance of soluble Aβ and/or vasculotoxic properties of Aβ.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11290219/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141858758","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Shahzad Ahmad, Wei Yang, Adelina Orellana, Lutz Frölich, Itziar de Rojas, Amanda Cano, Mercè Boada, Isabel Hernández, Lucrezia Hausner, Amy C Harms, Margot H M Bakker, Alfredo Cabrera-Socorro, Najaf Amin, Alfredo Ramírez, Agustín Ruiz, Cornelia M Van Duijn, Thomas Hankemeier
{"title":"Association of oxidative stress and inflammatory metabolites with Alzheimer's disease cerebrospinal fluid biomarkers in mild cognitive impairment.","authors":"Shahzad Ahmad, Wei Yang, Adelina Orellana, Lutz Frölich, Itziar de Rojas, Amanda Cano, Mercè Boada, Isabel Hernández, Lucrezia Hausner, Amy C Harms, Margot H M Bakker, Alfredo Cabrera-Socorro, Najaf Amin, Alfredo Ramírez, Agustín Ruiz, Cornelia M Van Duijn, Thomas Hankemeier","doi":"10.1186/s13195-024-01542-4","DOIUrl":"10.1186/s13195-024-01542-4","url":null,"abstract":"<p><strong>Background: </strong>Isoprostanes and prostaglandins are biomarkers for oxidative stress and inflammation. Their role in Alzheimer's disease (AD) pathophysiology is yet unknown. In the current study, we aim to identify the association of isoprostanes and prostaglandins with the Amyloid, Tau, Neurodegeneration (ATN) biomarkers (Aβ-42, p-tau, and t-tau) of AD pathophysiology in mild cognitive impairment (MCI) subjects.</p><p><strong>Methods: </strong>Targeted metabolomics profiling was performed using liquid chromatography-mass spectrometry (LCMS) in 147 paired plasma-CSF samples from the Ace Alzheimer Center Barcelona and 58 CSF samples of MCI patients from the Mannheim/Heidelberg cohort. Linear regression was used to evaluate the association of metabolites with CSF levels of ATN biomarkers in the overall sample and stratified by Aβ-42 pathology and APOE genotype. We further evaluated the role of metabolites in MCI to AD dementia progression.</p><p><strong>Results: </strong>Increased CSF levels of PGF2α, 8,12-iso-iPF2α VI, and 5-iPF2α VI were significantly associated (False discovery rate (FDR) < 0.05) with higher p-tau levels. Additionally, 8,12-iso-iPF2α VI was associated with increased total tau levels in CSF. In MCI due to AD, PGF2α was associated with both p-tau and total tau, whereases 8,12-iso-iPF2α VI was specifically associated with p-tau levels. In APOE stratified analysis, association of PGF2α with p-tau and t-tau was observed in only APOE ε4 carriers while 5-iPF2α VI showed association with both p-tau and t-tau in APOE ε33 carriers. CSF levels of 8,12- iso-iPF2α VI showed association with p-tau and t-tau in APOE ε33/APOE ε4 carriers and with t-tau in APOE ε3 carriers. None of the metabolites showed evidence of association with MCI to AD progression.</p><p><strong>Conclusions: </strong>Oxidative stress (8,12-iso-iPF2α VI) and inflammatory (PGF2α) biomarkers are correlated with biomarkers of AD pathology during the prodromal stage of AD and relation of PGF2α with tau pathology markers may be influenced by APOE genotype.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11287840/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854529","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Adele Sainsily-Cesarus, Elise Schmitt, Lionel Landre, Anne Botzung, Lucie Rauch, Catherine Demuynck, Nathalie Philippi, Paulo Loureiro de Sousa, Catherine Mutter, Benjamin Cretin, Catherine Martin-Hunyadi, Frederic Blanc
{"title":"Dementia with Lewy bodies and gait neural basis: a cross-sectional study.","authors":"Adele Sainsily-Cesarus, Elise Schmitt, Lionel Landre, Anne Botzung, Lucie Rauch, Catherine Demuynck, Nathalie Philippi, Paulo Loureiro de Sousa, Catherine Mutter, Benjamin Cretin, Catherine Martin-Hunyadi, Frederic Blanc","doi":"10.1186/s13195-024-01539-z","DOIUrl":"10.1186/s13195-024-01539-z","url":null,"abstract":"<p><strong>Background: </strong>Dementia with Lewy Bodies (DLB) is responsible for cognitive-behavioural disorders but also for gait disorders. The latter are thought to be related to parkinsonism, but the neural bases of these disorders are not well known, especially in the early stages. The aim of this study was to investigate by volumetric Magnetic Resonance Imaging the neuronal basis of gait disorders in DLB patients, compared to Healthy Elderly Controls and Alzheimer's Disease patients.</p><p><strong>Methods: </strong>Clinical examination with motor assessment including 10-meter walking speed, one-leg balance and Timed Up and Go test, a comprehensive neuropsychological evaluation and 3D brain Magnetic Resonance Imaging were performed on 84 DLB patients, 39 Alzheimer's Disease patients and 22 Healthy Elderly Controls. We used Statistical Parametric Mapping 12 to perform a one-sample t-test to investigate the correlation between each gait score and gray matter volume (P ≤ 0.05 corrected for family-wise error).</p><p><strong>Results: </strong>We found a correlation for DLB patients between walking speed and gray matter decrease (P < 0.05, corrected for family-wise error) in caudate nuclei, anterior cingulate cortex, mid-cingulate cortex, hippocampi, supplementary motor area, right cerebellar cortex and left parietal operculum. We found no correlation with Timed Up and Go test and one-leg balance.</p><p><strong>Conclusion: </strong>Gait disorders are underpinned by certain classical regions such as the cerebellum and the supplementary motor area. Our results suggest there may be a motivational and emotional component of voluntary gait in DLB subjects, underpinned by the cingulate cortex, a spatial orientation component, underpinned by hippocampi and suggest the involvement of brain processing speed and parkinsonism, underpinned by the caudate nuclei.</p><p><strong>Trial registration: </strong>The study protocol has been registered on ClinicalTrials.gov. (NCT01876459) on June 12, 2013.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11287986/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anna Bonaterra-Pastra, Montse Solé, Silvia Lope-Piedrafita, Maria Lucas-Parra, Laura Castellote, Paula Marazuela, Olalla Pancorbo, David Rodríguez-Luna, Mar Hernández-Guillamon
{"title":"The presence of circulating human apolipoprotein J reduces the occurrence of cerebral microbleeds in a transgenic mouse model with cerebral amyloid angiopathy.","authors":"Anna Bonaterra-Pastra, Montse Solé, Silvia Lope-Piedrafita, Maria Lucas-Parra, Laura Castellote, Paula Marazuela, Olalla Pancorbo, David Rodríguez-Luna, Mar Hernández-Guillamon","doi":"10.1186/s13195-024-01541-5","DOIUrl":"10.1186/s13195-024-01541-5","url":null,"abstract":"<p><strong>Background: </strong>Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β (Aβ) deposition in cerebral vessels, leading to lobar cerebral microbleeds (CMB) and intracerebral hemorrhages (ICH). Apolipoprotein J (ApoJ) is a multifunctional chaperone related to Aβ aggregation and clearance. Our study investigated the vascular impact of chronic recombinant human Apolipoprotein J (rhApoJ) treatment in a transgenic mouse model of β-amyloidosis with prominent CAA.</p><p><strong>Methods: </strong>Twenty-month-old APP23 C57BL/6 mice received 25 doses of rhApoJ (1 mg/kg) (n = 9) or saline (n = 8) intraperitoneally for 13 weeks, while Wild-type (WT) mice received saline (n = 13). Postmortem brains underwent T2*-weighted magnetic resonance imaging (MRI) to detect hemorrhagic lesions. Aβ levels and distribution, cerebral fibrinogen leakage, brain smooth muscle actin (sma), and plasma matrix metalloproteinases and inflammatory markers were analyzed after treatments. Additionally, plasma samples from 22 patients with lobar ICH were examined to determine the clinical relevance of the preclinical findings.</p><p><strong>Results: </strong>rhApoJ-treated APP23 presented fewer cortical CMBs (50-300 μm diameter) (p = 0.012) and cortical larger hemorrhages (> 300 μm) (p = 0.002) than saline-treated mice, independently of Aβ brain levels. MRI-detected hemorrhagic lesions correlated with fibrinogen cerebral extravasation (p = 0.011). Additionally, rhApoJ-treated mice presented higher number of sma-positive vessels than saline-treated mice (p = 0.038). In rhApoJ-treated mice, human ApoJ was detected in plasma and in occasional leptomeningeal vessels, but not in the parenchyma, suggesting that its mechanism of action operates through the periphery. The administration of rhApoJ induced an increase in plasma Groα (p = 0.035) and MIP-1α (p = 0.035) levels, while lower MMP-12 (p = 0.046) levels, compared to the saline-treated group. In acute lobar ICH patients, MMP-12 plasma levels correlated with larger hemorrhage volume (p = 0.040) and irregular ICH shape (p = 0.036).</p><p><strong>Conclusions: </strong>Chronic rhApoJ treatment in aged APP23 mice ameliorated CAA-related neurovascular damage by reducing the occurrence of CMB. We propose that rhApoJ may prevent blood-brain barrier (BBB) leakage and CMB appearance partly through circulating MMP-12 modulation.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11285315/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141786988","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Anne-Brita Knapskog, Trine Holt Edwin, Per Magne Ueland, Arve Ulvik, Evandro Fei Fang, Rannveig Sakshaug Eldholm, Nathalie Bodd Halaas, Lasse M. Giil, Ingvild Saltvedt, Leiv Otto Watne, Mari Aksnes
{"title":"Sex-specific associations of kynurenic acid with neopterin in Alzheimer’s disease","authors":"Anne-Brita Knapskog, Trine Holt Edwin, Per Magne Ueland, Arve Ulvik, Evandro Fei Fang, Rannveig Sakshaug Eldholm, Nathalie Bodd Halaas, Lasse M. Giil, Ingvild Saltvedt, Leiv Otto Watne, Mari Aksnes","doi":"10.1186/s13195-024-01531-7","DOIUrl":"https://doi.org/10.1186/s13195-024-01531-7","url":null,"abstract":"Sex differences in neuroinflammation could contribute to women’s increased risk of Alzheimer’s disease (AD), providing rationale for exploring sex-specific AD biomarkers. In AD, dysregulation of the kynurenine pathway (KP) contributes to neuroinflammation and there is some evidence of sex differences in KP metabolism. However, the sex-specific associations between KP metabolism and biomarkers of AD and neuroinflammation need to be explored further. Here we investigate sex differences in cerebrospinal fluid concentrations of seven KP metabolites and sex-specific associations with established AD biomarkers and neopterin, an indicator of neuroinflammation. This study included 311 patients with symptomatic AD and 105 age-matched cognitively unimpaired (CU) controls, followed for up to 5 years. We found sex differences in KP metabolites in the AD group, with higher levels of most metabolites in men, while there were no sex differences in the CU group. In line with this, more KP metabolites were significantly altered in AD men compared to CU men, and there was a trend in the same direction in AD women. Furthermore, we found sex-specific associations between kynurenic acid and the kynurenic acid/quinolinic acid ratio with neopterin, but no sex differences in the associations between KP metabolites and clinical progression. In our cohort, sex differences in KP metabolites were restricted to AD patients. Our results suggest that dysregulation of the KP due to increased inflammation could contribute to higher AD risk in women.","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":9.0,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141780027","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Javier Arranz, Nuole Zhu, Sara Rubio-Guerra, Íñigo Rodríguez-Baz, Rosa Ferrer, María Carmona-Iragui, Isabel Barroeta, Ignacio Illán-Gala, Miguel Santos-Santos, Juan Fortea, Alberto Lleó, Mireia Tondo, Daniel Alcolea
{"title":"Correction: Diagnostic performance of plasma pTau217, pTau181, Aβ1‑42 and Aβ1‑40 in the LUMIPULSE automated platform for the detection of Alzheimer disease","authors":"Javier Arranz, Nuole Zhu, Sara Rubio-Guerra, Íñigo Rodríguez-Baz, Rosa Ferrer, María Carmona-Iragui, Isabel Barroeta, Ignacio Illán-Gala, Miguel Santos-Santos, Juan Fortea, Alberto Lleó, Mireia Tondo, Daniel Alcolea","doi":"10.1186/s13195-024-01538-0","DOIUrl":"https://doi.org/10.1186/s13195-024-01538-0","url":null,"abstract":"<p><b>Correction:</b><b><i> Alz Res Therapy</i></b><b> 16, 139 (2024)</b></p><p><b>https://doi.org/10.1186/s13195-024-01513-9</b>.</p><p>Following publication of the original article [1], the authors corrected the Funding section to mention “project PI22/00611”.</p><p>The original article [1] has been corrected.</p><ol data-track-component=\"outbound reference\" data-track-context=\"references section\"><li data-counter=\"1.\"><p>Arranz J, Zhu N, Rubio-Guerra S, et al. Diagnostic performance of plasma pTau<sub>217</sub>, pTau<sub>181</sub>, Aβ<sub>1-42</sub> and Aβ<sub>1-40</sub> in the LUMIPULSE automated platform for the detection of Alzheimer disease. Alz Res Therapy. 2024;16:139. https://doi.org/10.1186/s13195-024-01513-9.</p><p>Article CAS Google Scholar </p></li></ol><p>Download references<svg aria-hidden=\"true\" focusable=\"false\" height=\"16\" role=\"img\" width=\"16\"><use xlink:href=\"#icon-eds-i-download-medium\" xmlns:xlink=\"http://www.w3.org/1999/xlink\"></use></svg></p><h3>Authors and Affiliations</h3><ol><li><p>Sant Pau Memory Unit, Department of Neurology, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, C/Sant Quintí 89, 08041, Barcelona, Spain</p><p>Javier Arranz, Nuole Zhu, Sara Rubio-Guerra, Íñigo Rodríguez-Baz, María Carmona-Iragui, Isabel Barroeta, Ignacio Illán-Gala, Miguel Santos-Santos, Juan Fortea, Alberto Lleó & Daniel Alcolea</p></li><li><p>Department of Neurology, Unidad Alzheimer-Down, IR SANT PAU, Hospital de La Santa Creu I Sant Pau; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain</p><p>Javier Arranz, Íñigo Rodríguez-Baz, María Carmona-Iragui, Isabel Barroeta & Juan Fortea</p></li><li><p>Universitat Autònoma de Barcelona, Barcelona, Spain</p><p>Javier Arranz, Nuole Zhu & Sara Rubio-Guerra</p></li><li><p>Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain</p><p>Nuole Zhu, María Carmona-Iragui, Isabel Barroeta, Ignacio Illán-Gala, Miguel Santos-Santos, Juan Fortea, Alberto Lleó & Daniel Alcolea</p></li><li><p>Servei de Bioquímica I Biologia Molecular, IR SANT PAU, Hospital de La Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, C/Sant Quintí 89, 08041, Barcelona, Spain</p><p>Rosa Ferrer & Mireia Tondo</p></li><li><p>Centro de Investigación Biomédica en Red en Diabetes y Enfermedades Metabólicas, CIBERDEM, Madrid, Spain</p><p>Mireia Tondo</p></li></ol><span>Authors</span><ol><li><span>Javier Arranz</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Nuole Zhu</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Sara Rubio-Guerra</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Íñigo Rodríguez-Baz</span>View author publications<p>You can also search for this author in","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":9.0,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141780026","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Virginie Dauphinot, Marie Laurent, Martin Prodel, Alexandre Civet, Alexandre Vainchtock, Claire Moutet, Pierre Krolak-Salmon, Antoine Garnier-Crussard
{"title":"Identification of profiles associated with conversions between the Alzheimer's disease stages, using a machine learning approach.","authors":"Virginie Dauphinot, Marie Laurent, Martin Prodel, Alexandre Civet, Alexandre Vainchtock, Claire Moutet, Pierre Krolak-Salmon, Antoine Garnier-Crussard","doi":"10.1186/s13195-024-01533-5","DOIUrl":"10.1186/s13195-024-01533-5","url":null,"abstract":"<p><strong>Background: </strong>The identification of factors involved in the conversion across the different Alzheimer's disease (AD) stages is crucial to prevent or slow the disease progression. We aimed to assess the factors and their combination associated with the conversion across the AD stages, from mild cognitive impairment to dementia, at a mild, moderate or severe stage and to identify profiles associated with earliest/latest conversion across the AD stages.</p><p><strong>Methods: </strong>In this study conducted on the real-life MEMORA cohort data collected from January 1, 2013, and December 31, 2019, three cohorts were selected depending on the baseline neurocognitive stage from a consecutive sample of patients attending a memory center, aged between 50 and 90 years old, with a diagnosis of AD during the follow-up, and with at least 2 visits at 6 months to 1 year of interval. A machine learning approach was used to assess the relationship between factors including socio-demographic characteristics, comorbidities and history of diseases, prescription of drugs, and geriatric hospitalizations, and the censored time to conversion from mild cognitive impairment to AD dementia, from the mild stage of dementia to the moderate or severe stages of AD dementia, and from the moderate stage of AD dementia to the severe stage. Profiles of earliest/latest conversion compared to median time to conversion across stages were identified. The median time to conversion was estimated with a Kaplan-Meier estimator.</p><p><strong>Results: </strong>Overall, 2891 patients were included (mean age 77±9 years old, 65% women). The median time of follow-up was 28 months for mild cognitive impairment (MCI) patients, 33 months for mild AD dementia and 30 months for moderate AD dementia. Among the 1264 patients at MCI stage, 61% converted to AD dementia (median time to conversion: 25 months). Among the 1142 patients with mild AD dementia, 59% converted to moderate/severe stage (median time: 23 months) and among the 1332 patients with moderate AD dementia, 23% converted to severe stage (Q3 time to conversion: 22 months). Among the studied factors, cardiovascular comorbidities, anxiety, social isolation, osteoporosis, and hearing disorders were identified as being associated with earlier conversion across stages. Symptomatic treatment i.e. cholinesterase inhibitors for AD was associated with later conversion from mild stage of dementia to moderate/severe stages.</p><p><strong>Conclusion: </strong>This study based on a machine learning approach allowed to identify potentially modifiable factors associated with conversion across AD stages for which timely interventions may be implemented to delay disease progression.</p>","PeriodicalId":7516,"journal":{"name":"Alzheimer's Research & Therapy","volume":null,"pages":null},"PeriodicalIF":7.9,"publicationDate":"2024-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11282744/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141764815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}