SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "hsv:(MEDICIN OCH HÄLSOVETENSKAP) hsv:(Medicinska och farmaceutiska grundvetenskaper) hsv:(Neurovetenskaper) ;pers:(Lannfelt Lars)"

Sökning: hsv:(MEDICIN OCH HÄLSOVETENSKAP) hsv:(Medicinska och farmaceutiska grundvetenskaper) hsv:(Neurovetenskaper) > Lannfelt Lars

  • Resultat 1-10 av 60
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Näsström, Thomas (författare)
  • Characterization of α-synuclein oligomers : Implications for Lewy Body Disorders
  • 2011
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Parkinson’s disease, dementia with Lewy bodies and multiple system atrophy are disorders featuring accumulation of Lewy bodies in brain. The main component of these large insoluble intracellular inclusions is the presynaptic protein alpha-synuclein (α-synuclein). It is generally believed that α-synuclein monomers adopt an abnormal conformation that favors the formation of soluble oligomers or protofibrils and, eventually, insoluble fibrils depositing as Lewy bodies. Notably, the intermediately sized oligomers/protofibrils seem to have particular neurotoxic effects. Several factors may influence the formation of α-synuclein oligomers/protofibrils, e.g. the reactive aldehydes 4-hydroxy-2-nonenal (HNE) and 4-oxo-2-nonenal (ONE) formed during oxidative stress. The overall aims of this thesis were to investigate biophysical and biochemical properties of in vitro generated α-synuclein oligomers, characterize their functional effects on cell and animal disease models as well as to explore whether their formation could be prevented in a cell culture model for oligomerization.  Here, it was found that α-synuclein rapidly formed oligomers after incubation with both ONE and HNE. The resulting oligomers were stable and did not continue to form insoluble fibrils. By comparing HNE- and ONE induced α-synuclein oligomers biochemically they were both found to exhibit extensive β-beta sheet structure and had a molecular size of ~2000 kDa. However, they differed in morphology; the ONE induced α-synuclein oligomers described round amorphous species whereas the HNE induced α-synuclein oligomers appeared as elongated protofibril-like structures. Both these oligomers were cell internalized to varying degrees and induced toxicity in neuroblastoma cells. In addition, the ONE induced α-synuclein oligomers seemed to initiate aggregation of monomeric α-synuclein in vitro, but failed to do so in vivo. Finally, treatment of α-synuclein overexpressing cells with monoclonal antibodies specific for α-synuclein significantly reduced aggregation and lowered levels of the protein, suggesting increased turnover in these cells.  To conclude, this thesis has characterized different oligomeric α-synuclein species, which may have properties similar to soluble species central to the pathogenesis of Parkinson’s disease and other disorders with α-synuclein pathology. For therapeutic strategies it is important to selectively target such harmful protein species and avoid interaction with other forms of α-synuclein, which may have vital physiological cellular functions.
  •  
2.
  •  
3.
  • Fang, Xiaotian T., 1990- (författare)
  • Preclinical PET imaging of Alzheimer's disease progression
  • 2017
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Amyloid PET imaging with [11C]PIB enabled detection of Aβ for the first time in vivo. However, [11C]PIB is a small molecule that binds only the insoluble Aβ plaque. Rather, the soluble Aβ aggregates are considered the cause of Alzheimer’s disease (AD). As such, a more sensitive and specific PET tracer is needed for tracking longitudinal AD pathology.Soluble Aβ aggregates likely interact with the metabotropic glutamate receptor 5 (mGluR5) to cause neurotoxic effects. However, with [11C]ABP688 PET we were unable to detect aberrant mGluR5 binding in AD mouse models, although we find elevated mGluR5 protein levels with immunoblotting.Antibodies are highly specific large molecules that can bind specifically to soluble Aβ aggregates, thus they can be a good marker for AD pathology. Unfortunately, due to their large size they cannot cross the blood-brain barrier (BBB). However, it is possible to shuttle antibodies into the brain by taking advantage of endogenous transporter systems on the BBB. By creating bispecific antibodies binding both to soluble Aβ aggregates and to the transferrin receptor (BBB target), we successfully transported the antibody into the brain and could visually detect soluble Aβ aggregates with PET.Recombinant expression further improved and optimized antibody design, creating smaller bispecific antibody-based constructs that had better pharmacokinetic properties allowing for earlier PET scanning (1 day instead of 3), and more sensitive signal.Lastly, using TCO-tetrazine click chemistry, we indirectly labeled our antibodies with fluorine-18, and could successfully perform PET already 11 h post-injection with a fluorine-18 labeled antibody.
  •  
4.
  • Pagnon de la Vega, María, 1994-, et al. (författare)
  • Mutation analysis of disease causing genes in patients with early onset or familial forms of Alzheimer’s disease and frontotemporal dementia
  • 2022
  • Ingår i: BMC Genomics. - : Springer Nature. - 1471-2164. ; 23
  • Tidskriftsartikel (refereegranskat)abstract
    • Background: Most dementia disorders have a clear genetic background and a number of disease genes have beenidentified. Mutations in the tau gene (MAPT) lead to frontotemporal dementia (FTD), whereas mutations in the genesfor the amyloid-β precursor protein (APP) and the presenilins (PSEN1, PSEN2) cause early-onset, dominantly inheritedforms of Alzheimer’s disease (AD).Even if mutations causing Mendelian forms of these diseases are uncommon, elucidation of the pathogenic effects ofsuch mutations have proven important for understanding the pathogenic processes. Here, we performed a screen toidentify novel pathogenic mutations in known disease genes among patients undergoing dementia investigation.Results: Using targeted exome sequencing we have screened all coding exons in eleven known dementia genes(PSEN1, PSEN2, APP, MAPT, APOE, GRN, TARDBP, CHMP2B, TREM2, VCP and FUS) in 102 patients with AD, FTD, otherdementia diagnoses or mild cognitive impairment.We found three AD patients with two previously identified pathogenic mutations in PSEN1 (Pro264Leu and Met-146Val). In this screen, we also identified the recently reported APP mutation in two siblings with AD. This mutation,named the Uppsala mutation, consists of a six amino acid intra-amyloid β deletion.In addition, we found several potentially pathogenic mutations in PSEN2, FUS, MAPT, GRN and APOE. Finally, APOE ε4was prevalent in this patient group with an allele frequency of 54%Conclusions: Among the 102 screened patients, we found two disease causing mutations in PSEN1 and one in APP,as well as several potentially pathogenic mutations in other genes related to neurodegenerative disorders. Apart fromgiving important information to the clinical investigation, the identification of disease mutations can contribute to anincreased understanding of disease mechanisms.
  •  
5.
  • Fagerqvist, Therese, et al. (författare)
  • Off-pathway alpha-synuclein oligomers seem to alter alpha-synuclein turnover in a cell model but lack seeding capability in vivo
  • 2013
  • Ingår i: Amyloid. - : Informa UK Limited. - 1350-6129 .- 1744-2818. ; 20:4, s. 233-244
  • Tidskriftsartikel (refereegranskat)abstract
    • Aggregated α-synuclein is the major component of Lewy bodies, protein inclusions observed in the brain in neurodegenerative disorders such as Parkinson’s disease and dementia with Lewy bodies. Experimental evidence indicates that α-synuclein potentially can be transferred between cells and act as a seed to accelerate the aggregation process. Here, we investigated in vitro and in vivo seeding effects of α-synuclein oligomers induced by the reactive aldehyde 4-oxo-2-nonenal (ONE). As measured by a Thioflavin-T based fibrillization assay, there was an earlier onset of aggregation when α-synuclein oligomers were added to monomeric α-synuclein. In contrast, exogenously added α-synuclein oligomers did not induce aggregation in a cell model. However, cells overexpressing α-synuclein that were treated with the oligomers displayed reduced α-synuclein levels, indicating that internalized oligomers either decreased the expression or accelerated the degradation of transfected α-synuclein. Also in vivo there were no clear seeding effects, as intracerebral injections of α-synuclein oligomers into the neocortex of α-synuclein transgenic mice did not induce formation of Proteinase K resistant α-synuclein pathology. Taken together, we could observe a seeding effect of the ONE-induced α-synuclein oligomers in a fibrillization assay, but neither in a cell nor in a mouse model.
  •  
6.
  •  
7.
  • Näsström, Thomas, et al. (författare)
  • Antibodies against alpha-synuclein reduce oligomerization in living cells
  • 2011
  • Ingår i: PLOS ONE. - : Public Library of Science (PLoS). - 1932-6203. ; 6:10, s. e27230-
  • Tidskriftsartikel (refereegranskat)abstract
    • Recent research implicates soluble aggregated forms of α-synuclein as neurotoxic species with a central role in the pathogenesis of Parkinson’s disease and related disorders. The pathway by which α-synuclein aggregates is believed to follow a step-wise pattern, in which dimers and smaller oligomers are initially formed. Here, we used H4 neuroglioma cells expressing α-synuclein fused to hemi:GFP to study the effects of α-synuclein monoclonal antibodies on the early stages of aggregation, as quantified by Bimolecular Fluorescence Complementation assay. Widefield and confocal microscopy revealed that cells treated for 48 h with monoclonal antibodies internalized antibodies to various degrees. Oligomer-selective and C-terminal specific α-synuclein antibodies reduced the extent of α-synuclein dimerization/oligomerization, as indicated by decreased GFP fluorescence signal. Furthermore, ELISA measurements on lysates and conditioned media from antibody treated cells displayed lower α-synuclein levels compared to untreated cells, suggesting increased protein turnover. Taken together, our results propose that extracellular administration of monoclonal antibodies can modify or inhibit early steps in the aggregation process of α-synuclein, thus providing further support for passive immunization against diseases with α-synuclein pathology.
  •  
8.
  • O'Callaghan, Paul (författare)
  • Heparan Sulfate in the Amyloidosis and Inflammation of Alzheimer’s Disease
  • 2011
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Alzheimer’s disease (AD) is a neurodegenerative disorder, with extensive evidence implicating the misfolding, aggregation and deposition of the amyloid-β (Aβ) peptide as central to the pathogenesis. Heparan sulfate (HS) is an interactive glycosaminoglycan, attached to core proteins as HS proteoglycans (HSPGs). HSPGs are present on cell surfaces and in the extracellular matrix where they facilitate multiple signaling functions, but HS is also consistently present in all amyloid deposits, including those of AD. In amyloidosis HS has been studied as an aggregation template, promoting fibril formation and serving a scaffold function in the resulting deposits. The objective of this thesis was to assess how cell surface HS is potentially implicated in Aβ amyloidosis and the associated neuroinflammation of AD.  In AD brain we determined that HS predominantly accumulated in Aβ deposits with dense cores and found glial-expressed HSPGs within these deposits. Aβ elevated HSPG levels in primary glial cultures, implicating activated glia as one source of the Aβ-associated HS. Next, we determined that microglial HSPGs are critical for the upregulation of interleukin-1β and tumor necrosis factor-α following exposure to lipopolysaccharide, an established inflammatory insult. Together these results raise the possibility that Aβ-induced expression of microglial HSPGs may promote neuroinflammation.  Multiple mechanisms of Aβ toxicity have been proposed and different Aβ assemblies exert their toxicity through alternative routes. We found that three different preparations of Aβ aggregates all exhibited HS-dependent cytotoxicity, which in part correlated with Aβ internalization. Furthermore, heparin treatment attenuated Aβ cytotoxicity and uptake. In Aβ-positive AD microvasculature, HS deposited with Apolipoprotein E (ApoE) and its receptor, the low density lipoprotein receptor-related protein 1 (LRP1). In cell culture, HS and LRP1 co-operated in Aβ interactions and the addition of ApoE increased the levels of cell-associated Aβ in a HS- and LRP1-dependent manner. This ApoE-mediated increase in cell-associated Aβ may promote toxicity and vascular degeneration, but equally HS-mediated internalization of Aβ could represent a clearance route across the blood-brain-barrier.The findings presented here illustrate multiple roles for cell-surface HSPGs in interactions relevant to the pathogenesis of AD.
  •  
9.
  • O'Callaghan, Paul, et al. (författare)
  • Microglial heparan sulfate proteoglycans mediate pro-inflammatory signaling
  • Annan publikation (övrigt vetenskapligt/konstnärligt)abstract
    • Microglia are the central nervous system’s (CNS) first line of defense against pathogenic insults and acute inflammatory responses are necessary for the resolution of infection. However, unregulated and/or chronic activation of microglia is associated with neurodegeneration. Heparan sulfate proteoglycans (HSPGs) have been attributed various roles in inflammation, but the possibility that HSPGs are integral to pro-inflammatory signaling mechanisms has not been fully explored. To analyze the relevance of microglial HSPGs in the pro-inflammatory response we isolated primary microglia from mice overexpressing human heparanase (Hpa-tg), the HS-degrading endoglucuronidase, and challenged them with the pro-inflammatory endotoxin lipopolysaccharide (LPS). The LPS-induced upregulation of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) was inhibited in Hpa-tg microglia, as was upregulation of the LPS-receptor CD14. Analysis of HSPG structures revealed that Hpa-tg microglia produced truncated HS chains. Importantly, co-treatment of microglia with heparin attenuated LPS-induced cytokine upregulation. Together these findings implicate microglial HSPGs as key facilitators of the pro-inflammatory response. Astrocytes constitute a critical support network in the CNS, but are also implicated in inflammation. LPS induced comparable levels of TNF-α in Hpa-tg and Ctrl astrocytes, indicating that the mechanism of HSPG-dependent inflammation is specific to microglia.  We conclude that microglial HSPGs are required for pro-inflammatory signaling events and that heparanase, through its HS-degrading activity, can regulate this mechanism.
  •  
10.
  • Zhang, Xiao, et al. (författare)
  • Heparanase overexpression impedes perivascular clearance of amyloid-beta from murine brain : relevance to Alzheimer's disease
  • 2021
  • Ingår i: Acta neuropathologica communications. - : BioMed Central (BMC). - 2051-5960. ; 9
  • Tidskriftsartikel (refereegranskat)abstract
    • Defective amyloid-beta (A beta) clearance from the brain is a major contributing factor to the pathophysiology of Alzheimer's disease (AD). A beta clearance is mediated by macrophages, enzymatic degradation, perivascular drainage along the vascular basement membrane (VBM) and transcytosis across the blood-brain barrier (BBB). AD pathology is typically associated with cerebral amyloid angiopathy due to perivascular accumulation of A beta. Heparan sulfate (HS) is an important component of the VBM, thought to fulfill multiple roles in AD pathology. We previously showed that macrophage-mediated clearance of intracortically injected A beta was impaired in the brains of transgenic mice overexpressing heparanase (Hpa-tg). This study revealed that perivascular drainage was impeded in the Hpa-tg brain, evidenced by perivascular accumulation of the injected A beta in the thalamus of Hpa-tg mice. Furthermore, endogenous A beta accumulated at the perivasculature of Hpa-tg thalamus, but not in control thalamus. This perivascular clearance defect was confirmed following intracortical injection of dextran that was largely retained in the perivasculature of Hpa-tg brains, compared to control brains. Hpa-tg brains presented with thicker VBMs and swollen perivascular astrocyte endfeet, as well as elevated expression of the BBB-associated water-pump protein aquaporin 4 (AQP4). Elevated levels of both heparanase and AQP4 were also detected in human AD brain. These findings indicate that elevated heparanase levels alter the organization and composition of the BBB, likely through increased fragmentation of BBB-associated HS, resulting in defective perivascular drainage. This defect contributes to perivascular accumulation of A beta in the Hpa-tg brain, highlighting a potential role for heparanase in the pathogenesis of AD.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 60
Typ av publikation
tidskriftsartikel (49)
annan publikation (5)
doktorsavhandling (4)
konferensbidrag (2)
Typ av innehåll
refereegranskat (50)
övrigt vetenskapligt/konstnärligt (10)
Författare/redaktör
Ingelsson, Martin (28)
Blennow, Kaj, 1958 (11)
Basun, Hans (10)
Zetterberg, Henrik, ... (8)
Söderberg, Linda (8)
visa fler...
Sehlin, Dag, 1976- (7)
Möller, Christer (7)
Minthon, Lennart (5)
Nilsson, Peter (4)
Kultima, Kim (4)
Hansson, Oskar (4)
Eriksson, Fredrik (4)
Brinkmalm, Gunnar (3)
Alafuzoff, Irina (3)
Schiöth, Helgi B. (3)
Emami Khoonsari, Pay ... (3)
Shevchenko, Ganna (3)
Gkanatsiou, Eleni (3)
Kim, S. H. (2)
Almkvist, Ove (2)
Fratiglioni, Laura (2)
Boada, Mercè (2)
Nilsson, Lars (2)
Tsolaki, Magda (2)
Pasquier, Florence (2)
Powell, John F. (2)
Bergquist, Jonas (2)
Li, Jin-Ping (2)
Lindahl, Ulf (2)
Karlsson, Mikael (2)
Hardy, J (2)
Bogdanovic, Nenad (2)
Portelius, Erik, 197 ... (2)
Clarke, Robert (2)
Aguilar, Ximena (2)
Olsson, Bob, 1969 (2)
Långström, Bengt (2)
Ramirez, Alfredo (2)
Ikram, M. Arfan (2)
Amin, Najaf (2)
van Duijn, Cornelia ... (2)
Hiltunen, Mikko (2)
Zhang, Xiao (2)
Rotter, Jerome I. (2)
Soininen, Hilkka (2)
Van Broeckhoven, Chr ... (2)
Clarimon, Jordi (2)
Cruchaga, Carlos (2)
visa färre...
Lärosäte
Uppsala universitet (59)
Karolinska Institutet (15)
Göteborgs universitet (13)
Stockholms universitet (5)
Lunds universitet (5)
Kungliga Tekniska Högskolan (3)
visa fler...
Linköpings universitet (1)
Högskolan Dalarna (1)
visa färre...
Språk
Engelska (59)
Svenska (1)
Forskningsämne (UKÄ/SCB)
Medicin och hälsovetenskap (60)
Naturvetenskap (4)
Samhällsvetenskap (1)

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