SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "LAR1:gu ;pers:(Blennow Kaj 1958)"

Sökning: LAR1:gu > Blennow Kaj 1958

  • Resultat 481-490 av 1860
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
481.
  • Falcon, Carles, et al. (författare)
  • Time-encoded ASL reveals lower cerebral blood flow in the early AD continuum
  • 2024
  • Ingår i: ALZHEIMERS & DEMENTIA. - 1552-5260 .- 1552-5279.
  • Tidskriftsartikel (refereegranskat)abstract
    • INTRODUCTION Cerebral blood flow (CBF) is reduced in cognitively impaired (CI) Alzheimer's disease (AD) patients. We checked the sensitivity of time-encoded arterial spin labeling (te-ASL) in measuring CBF alterations in individuals with positive AD biomarkers and associations with relevant biomarkers in cognitively unimpaired (CU) individuals. METHODS We compared te-ASL with single-postlabel delay (PLD) ASL in measuring CBF in 59 adults across the AD continuum, classified as CU amyloid beta (A beta) negative (-), CU A beta positive (+), and CI A beta+. We sought associations of CBF with biomarkers of AD, cerebrovascular disease, synaptic dysfunction, neurodegeneration, and cognition in CU participants. RESULTS te-ASL was more sensitive at detecting CBF reduction in the CU A beta+ and CI A beta+ groups. In CU participants, lower CBF was associated with altered biomarkers of A beta, tau, synaptic dysfunction, and neurodegeneration. DISCUSSION CBF reduction occurs early in the AD continuum. te-ASL is more sensitive than single-PLD ASL at detecting CBF changes in AD.
  •  
482.
  • Falgas, N., et al. (författare)
  • Contribution of CSF biomarkers to early-onset Alzheimer's disease and frontotemporal dementia neuroimaging signatures
  • 2020
  • Ingår i: Human Brain Mapping. - : Wiley. - 1065-9471 .- 1097-0193. ; 41:8, s. 2004-2013
  • Tidskriftsartikel (refereegranskat)abstract
    • Prior studies have described distinct patterns of brain gray matter and white matter alterations in Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD), as well as differences in their cerebrospinal fluid (CSF) biomarkers profiles. We aim to investigate the relationship between early‐onset AD (EOAD) and FTLD structural alterations and CSF biomarker levels. We included 138 subjects (64 EOAD, 26 FTLD, and 48 controls), all of them with a 3T MRI brain scan and CSF biomarkers available (the 42 amino acid‐long form of the amyloid‐beta protein [Aβ42], total‐tau protein [T‐tau], neurofilament light chain [NfL], neurogranin [Ng], and 14‐3‐3 levels). We used FreeSurfer and FSL to obtain cortical thickness (CTh) and fraction anisotropy (FA) maps. We studied group differences in CTh and FA and described the “AD signature” and “FTLD signature.” We tested multiple regression models to find which CSF‐biomarkers better explained each disease neuroimaging signature. CTh and FA maps corresponding to the AD and FTLD signatures were in accordance with previous literature. Multiple regression analyses showed that the biomarkers that better explained CTh values within the AD signature were Aβ and 14‐3‐3; whereas NfL and 14‐3‐3 levels explained CTh values within the FTLD signature. Similarly, NfL levels explained FA values in the FTLD signature. Ng levels were not predictive in any of the models. Biochemical markers contribute differently to structural (CTh and FA) changes typical of AD and FTLD.
  •  
483.
  • Fang, C., et al. (författare)
  • Buntanetap, a Novel Translational Inhibitor of Multiple Neurotoxic Proteins, Proves to Be Safe and Promising in Both Alzheimer’s and Parkinson’s Patients
  • 2023
  • Ingår i: Journal of Prevention of Alzheimer's Disease. - : SERDI. - 2274-5807 .- 2426-0266. ; 10:1, s. 25-33
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Previously we reported the clinical safety and pharmacological activity of buntanetap (known as Posiphen or ANVS401) in healthy volunteers and mild cognitive impaired (MCI) patients (21). The data supported continued clinical evaluation of buntanetap for treating Alzheimer’s Disease (AD). Neurodegenerative diseases such as AD and Parkinson’s disease (PD) share several pathological manifestations, including increased levels of multiple neurotoxic protein aggregates. Therefore, a treatment strategy that targets toxic species common to both disorders can potentially provide better clinical outcomes than attacking one neurotoxic protein alone. To test this hypothesis, we recently completed a clinical study in early AD and early PD participants and report the data here. Objectives: We evaluated safety, pharmacokinetics, biomarkers, and efficacy of buntanetap in treating early AD and PD patients. Design: Double-blind, placebo-controlled, multi-center study. Setting: 13 sites in the US participated in this clinical trial. The registration number is NCT04524351 at ClinicalTrials.gov. Participants: 14 early AD patients and 54 early PD patients. Intervention: AD patients were given either 80mg buntanetap or placebo QD. PD patients were given 5mg, 10mg, 20mg, 40mg, 80mg buntanetap or placebo QD. Measurements: Primary endpoint is safety and tolerability; secondary endpoint is pharmacokinetics of buntanetap in plasma; exploratory endpoints are 1) biomarkers in cerebrospinal fluid (CSF) in both AD and PD patients 2) psychometric tests specific for AD (ADAS-Cogs & WAIS coding test) or PD (MDS-UPDRS & WAIS coding test). Results: Buntanetap was safe and well tolerated. Biomarker data indicated a trend in lowering levels of neurotoxic proteins and inflammatory factors and improving axonal integrity and synaptic function in both AD and PD cohorts. Psychometric tests showed statistically significant improvements in ADAS-Cog11 and WAIS coding in AD patients and MDS-UPDRS and WAIS coding in PD patients. Conclusions: Buntanetap is well tolerated and safe at doses up to 80mg QD in both AD and PD patients. Cmax and AUC increase with dose without evidence for a plateau up to 80mg QD. The drug shows promising evidence in exploratory biomarker and efficacy measures. Further evaluation of buntanetap in larger, longer-term clinical trials for the treatment of AD and PD are warranted. © 2022, The Authors.
  •  
484.
  • Farid, Karim, et al. (författare)
  • Case Report of Complex Amyotrophic Lateral Sclerosis with Cognitive Impairment and Cortical Amyloid Deposition.
  • 2015
  • Ingår i: Journal of Alzheimer's disease : JAD. - 1875-8908 .- 1387-2877. ; 47:3, s. 661-7
  • Tidskriftsartikel (refereegranskat)abstract
    • Amyotrophic lateral sclerosis (ALS), a fatal disease of unknown origin, affects motor neurons in the primary motor cortex, brainstem, and spinal cord. Cognitive impairment may occur before the motor symptoms. We present a patient who was initially diagnosed with mild cognitive impairment (MCI) due to Alzheimer's disease (AD) but who developed ALS-like symptoms during follow-up and died shortly thereafter. A 60-year-old subject with cognitive impairment underwent neuropsychological testing, cerebrospinal fluid (CSF) analysis, structural imaging (computed tomography and magnetic resonance imaging) and functional imaging [11C]-Pittsburgh compound B (PIB) positron emission tomography (PET), [18F]-fluorodeoxyglucose (FDG) PET, and [11C]-deuterium-L-deprenyl (DED) PET. Neuropsychological testing showed episodic memory impairment. CSF P-tau and T-tau levels were elevated. CSF amyloid-β (Aβ)42 levels were initially normal but became pathological during follow-up. MCI was diagnosed. [18F]-FDG PET showed hypometabolism in the left temporal and prefrontal cortices and [11C]-PIB PET demonstrated amyloid plaque deposition in the prefrontal, posterior cingulate, and parietal cortices. [11C]-DED PET showed high brain accumulation consistent with astrocytosis. The memory impairment progressed and AD was diagnosed. Motor impairments developed subsequently and, following additional neurological evaluation, ALS was diagnosed. The disease progressed rapidly and the patient died with pronounced motor symptoms three years after the initial cognitive assessment. Since relatives refused autopsy, postmortem analysis was not possible.
  •  
485.
  • Fatima, Tahzeeb, et al. (författare)
  • Association between serum urate and CSF markers of Alzheimer's disease pathology in a population-based sample of 70-year-olds
  • 2021
  • Ingår i: Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring. - : Wiley. - 2352-8729. ; 13:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Introduction: The relationship between urate and biomarkers for Alzheimer's disease (AD) pathophysiology has not been investigated. Methods: We examined whether serum concentration of urate was associated with cerebrospinal fluid biomarkers, amyloid beta (A beta)(42), A beta(40), phosphorylated tau (p-tau), total tau (t-tau), neurofilament light (NfL), and A beta(42)/A beta(40) ratio, in cognitively unimpaired 70-year-old individuals from Gothenburg, Sweden. We also evaluated whether possible associations were modulated by the apolipoprotein E (APOE) epsilon 4 allele. Results: Serum urate was positively associated with A beta(42) in males (beta = 0.55 pg/mL, P = .04). There was a positive urate-APOE epsilon 4 interaction (1.24 pg/mL, P-interaction = .02) in relation to A beta(42) association. The positive urate and A beta(42) association strengthened in male APOE epsilon 4 carriers (beta = 1.28 pg/mL, P = .01). Discussion: The positive association between urate and A beta(42) in cognitively healthy men may suggest a protective effect of urate against deposition of amyloid protein in the brain parenchyma, and in the longer term, maybe against AD dementia.
  •  
486.
  • Fauria, K., et al. (författare)
  • Exploring cognitive and biological correlates of sleep quality and their potential links with Alzheimer's disease (ALFASleep project): protocol for an observational study
  • 2022
  • Ingår i: Bmj Open. - : BMJ. - 2044-6055. ; 12:12
  • Tidskriftsartikel (refereegranskat)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 3years 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.
  •  
487.
  • Faux, Noel G, et al. (författare)
  • PBT2 Rapidly Improves Cognition in Alzheimer's Disease : Additional Phase II Analyses
  • 2010
  • Ingår i: Journal of Alzheimer's Disease. - 1387-2877 .- 1875-8908. ; 20:2, s. 509-516
  • Tidskriftsartikel (refereegranskat)abstract
    • PBT2 is a copper/zinc ionophore that rapidly restores cognition in mouse models of Alzheimer's disease (AD). A recent Phase IIa double-blind, randomized, placebo-controlled trial found that the 250 mg dose of PBT2 was well-tolerated, significantly lowered cerebrospinal fluid (CSF) levels of amyloid-beta_{42}, and significantly improved executive function on a Neuro-psychological Test Battery (NTB) within 12 weeks of treatment in patients with AD. In the post-hoc analysis reported here, the cognitive, blood marker, and CSF neurochemistry outcomes from the trial were subjected to further analysis. Ranking the responses to treatment after 12 weeks with placebo, PBT2 50 mg, and PBT2 250 mg revealed that the proportions of patients showing improvement on NTB Composite or Executive Factor z-scores were significantly greater in the PBT2 250 mg group than in the placebo group. Receiver-operator characteristic analyses revealed that the probability of an improver at any level coming from the PBT2 250 mg group was significantly greater, compared to placebo, for Composite z-scores (Area Under the Curve [AUC] =0.76, p=0.0007), Executive Factor z-scores (AUC =0.93, p=1.3 x 10;{-9}), and near-significant for the ADAS-cog (AUC =0.72, p=0.056). There were no correlations between changes in CSF amyloid-beta or tau species and cognitive changes. These findings further encourage larger-scale testing of PBT2 for AD.
  •  
488.
  • Fernandes Gomes, Bárbara, et al. (författare)
  • α-Synuclein seed amplification assay as a diagnostic tool for parkinsonian disorders.
  • 2023
  • Ingår i: Parkinsonism & related disorders. - 1873-5126. ; 117
  • Tidskriftsartikel (refereegranskat)abstract
    • Synucleinopathies such as Parkinson's disease (PD) and multiple system atrophy (MSA) can be challenging to diagnose due to the symptom overlap with, for example, atypical parkinsonisms like progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). Seed amplification assays (SAA), developed for the detection of α-synuclein (αSyn) aggregates in CSF, have been successful when used as a biomarker evaluation for synucleinopathies. In this study, we investigated the potential of this assay to not only detect αSyn seeds in CSF, but also discriminate between movement disorders.The αSyn-SAA was tested in a Scandinavian cohort composed of 129 CSF samples from patients with PD (n=55), MSA (n=27), CBD (n=7), and PSP (n=16), as well as healthy controls (HC, n=24).The αSyn seed amplification assay (αSyn-SAA) was able to correctly identify all PD samples as positive (sensitivity of 100%) while also discriminating the PD group from HC (70.8% specificity, p<0.0001) and tauopathies [CBD (71% specificity) and PSP (75% specificity), p<0.0001)]. The αSyn-SAA was also able to identify almost all MSA samples as positive for αSyn aggregation (sensitivity of 92.6%). In general, this assay is able to discriminate between the synucleinopathies and tauopathies analyzed herein (p<0.0001) despite the overlapping symptoms in these diseases.These findings suggest the αSyn-SAA is a useful diagnostic tool for differentiating between different parkinsonian disorders, although further optimization may be needed.
  •  
489.
  • Fernström, Erik, et al. (författare)
  • Cerebrospinal fluid markers of extracellular matrix remodelling, synaptic plasticity and neuroinflammation before and after cranial radiotherapy
  • 2018
  • Ingår i: Journal of Internal Medicine. - : Wiley. - 0954-6820. ; 284:2, s. 211-225
  • Tidskriftsartikel (refereegranskat)abstract
    • Background Advances in the treatment of brain tumours have increased the number of long-term survivors, but at the cost of side effects following cranial radiotherapy ranging from neurocognitive deficits to outright tissue necrosis. At present, there are no tools reflecting the molecular mechanisms underlying such side effects, and thus no means to evaluate interventional effects after cranial radiotherapy. Therefore, fluid biomarkers are of great clinical interest. Objective Cerebrospinal fluid (CSF) levels of proteins involved in inflammatory signalling, synaptic plasticity and extracellular matrix (ECM) integrity were investigated following radiotherapy to the brain. Methods Patients with small-cell lung cancer (SCLC) eligible for prophylactic cranial irradiation (PCI) were asked to participate in the study. PCI was prescribed either as 2 Gy/fraction to a total dose of 30 Gy (limited disease) or 4 Gy/fraction to 20 Gy (extensive disease). CSF was collected by lumbar puncture at baseline, 3 months and 1 year following PCI. Protein concentrations were measured using immunobased assays or mass spectrometry. Results The inflammatory markers IL-15, IL-16 and MCP-1/CCL2 were elevated in CSF 3 months following PCI compared to baseline. The plasticity marker GAP-43 was elevated 3 months following PCI, and the same trend was seen for SNAP-25, but not for SYT1. The investigated ECM proteins, brevican and neurocan, showed a decline following PCI. There was a strong correlation between the progressive decline of soluble APP and brevican levels. Conclusion To our knowledge, this is the first time ECM-related proteins have been shown to be affected by cranial radiotherapy in patients with cancer. These findings may help us to get a better understanding of the mechanisms behind side effects following radiotherapy.
  •  
490.
  • Ferrari-Souza, J. P., et al. (författare)
  • APOEε4 associates with microglial activation independently of Aβ plaques and tau tangles
  • 2023
  • Ingår i: Science Advances. - 2375-2548. ; 9:14
  • Tidskriftsartikel (refereegranskat)abstract
    • Animal studies suggest that the apolipoprotein E epsilon 4 (APOE epsilon 4) allele is a culprit of early microglial activation in Alzheimer's disease (AD). Here, we tested the association between APOE epsilon 4 status and microglial activation in living individuals across the aging and AD spectrum. We studied 118 individuals with positron emission tomog-raphy for amyloid-beta (A beta; [18F]AZD4694), tau ([18F]MK6240), and microglial activation ([11C]PBR28). We found that APOE epsilon 4 carriers presented increased microglial activation relative to noncarriers in early Braak stage regions within the medial temporal cortex accounting for A beta and tau deposition. Furthermore, microglial acti-vation mediated the A beta-independent effects of APOE epsilon 4 on tau accumulation, which was further associated with neurodegeneration and clinical impairment. The physiological distribution of APOE mRNA expression predicted the patterns of APOE epsilon 4-related microglial activation in our population, suggesting that APOE gene expression may regulate the local vulnerability to neuroinflammation. Our results support that the APOE epsilon 4 genotype exerts A beta-independent effects on AD pathogenesis by activating microglia in brain regions associated with early tau deposition.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 481-490 av 1860
Typ av publikation
tidskriftsartikel (1745)
forskningsöversikt (64)
konferensbidrag (27)
bokkapitel (16)
bok (7)
samlingsverk (redaktörskap) (1)
visa fler...
visa färre...
Typ av innehåll
refereegranskat (1786)
övrigt vetenskapligt/konstnärligt (74)
Författare/redaktör
Zetterberg, Henrik, ... (1521)
Ashton, Nicholas J. (205)
Andreasson, Ulf, 196 ... (163)
Hansson, Oskar (155)
Portelius, Erik, 197 ... (126)
visa fler...
Karikari, Thomas (114)
Wallin, Anders, 1950 (104)
Skoog, Ingmar, 1954 (102)
Minthon, Lennart (100)
Brinkmalm, Gunnar (85)
Mattsson, Niklas, 19 ... (80)
Kern, Silke (68)
Andreasen, Niels (68)
Gobom, Johan (66)
Hampel, Harald (60)
Lessa Benedet, André ... (59)
Zettergren, Anna, 19 ... (54)
Lantero Rodriguez, J ... (52)
Stomrud, Erik (45)
Höglund, Kina, 1976 (45)
Rosengren, Lars, 195 ... (44)
Brinkmalm-Westman, A ... (43)
Janelidze, Shorena (43)
Scheltens, Philip (42)
Teunissen, Charlotte ... (39)
Molinuevo, J. L. (38)
Palmqvist, Sebastian (37)
Engelborghs, Sebasti ... (37)
Johnson, Sterling C (36)
Shaw, Leslie M (36)
Hampel, H. (36)
Kollmorgen, G. (35)
Molinuevo, José Luis (35)
Johnson, S. C. (34)
Lleó, Alberto (34)
Scheltens, P (34)
Vanmechelen, Eugeen (33)
Kvartsberg, Hlin, 19 ... (32)
Simrén, Joel, 1996 (31)
Trojanowski, John Q (31)
Schöll, Michael, 198 ... (31)
Svensson, Johan, 196 ... (30)
Mattsson-Carlgren, N ... (30)
Asthana, S. (30)
Carlsson, Cynthia M (30)
Visser, Pieter Jelle (30)
Montoliu-Gaya, Laia (29)
Rosa-Neto, Pedro (29)
Hanrieder, Jörg, 198 ... (29)
visa färre...
Lärosäte
Göteborgs universitet (1860)
Karolinska Institutet (328)
Lunds universitet (231)
Uppsala universitet (85)
Linköpings universitet (41)
Chalmers tekniska högskola (37)
visa fler...
Örebro universitet (29)
Umeå universitet (28)
Stockholms universitet (15)
Luleå tekniska universitet (11)
Kungliga Tekniska Högskolan (7)
Jönköping University (5)
Högskolan Kristianstad (3)
Linnéuniversitetet (3)
Sveriges Lantbruksuniversitet (1)
VTI - Statens väg- och transportforskningsinstitut (1)
Sophiahemmet Högskola (1)
visa färre...
Språk
Engelska (1837)
Svenska (21)
Odefinierat språk (2)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (1782)
Naturvetenskap (49)
Samhällsvetenskap (33)
Teknik (4)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy