Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hanna Laudon is active.

Publication


Featured researches published by Hanna Laudon.


Journal of Biological Chemistry | 2003

Presenilin-1 Mutation L271V Results in Altered Exon 8 Splicing and Alzheimer's Disease with Non-cored Plaques and No Neuritic Dystrophy

John B. Kwok; Glenda M. Halliday; William S. Brooks; Georgia Dolios; Hanna Laudon; Ohoshi Murayama; Marianne Hallupp; Renee F. Badenhop; Jc Vickers; Rong Wang; Jan Näslund; Akihiko Takashima; Samuel E. Gandy; Peter R. Schofield

The mutation L271V in exon 8 of the presenilin-1 (PS-1) gene was detected in an Alzheimers disease pedigree. Neuropathological examination of affected individuals identified variant, large, non-cored plaques without neuritic dystrophy, reminiscent of cotton wool plaques. Biochemical analysis of L271V mutation showed that it increased secretion of the 42-amino acid amyloid-β peptide, suggesting a pathogenic mutation. Analysis of PS-1 transcripts from the brains of two mutation carriers revealed a 17–50% increase in PS-1 transcripts with deletion of exon 8 (PS-1Δexon8) compared with unrelated Alzheimers disease brains. Exon trapping analysis confirmed that L271V mutation enhanced the deletion of exon 8. Western blots of brain lysates indicated that PS-1Δexon8 was overexpressed in an affected individual. Biochemical analysis of PS-1Δexon8 in COS and BD8 cells indicate the splice isoform is not intrinsically active but interacts with wild-type PS-1 to generate amyloid-β. Western blots of cell lysates immunoprecipitated with anti-Tau or anti-GSK-3β antibodies indicated that PS-1Δexon8, unlike wild-type PS-1, does not interact directly with Tau or GSK-3β, potential modifiers of neuritic dystrophy. We postulate that variant plaques observed in this family are due in part to the effects of PS-1Δexon8 and that interaction between PS-1 and various protein complexes are necessary for neuritic plaque formation.


Journal of Alzheimer's Disease | 2014

The Murine Version of BAN2401 (mAb158) Selectively Reduces Amyloid-β Protofibrils in Brain and Cerebrospinal Fluid of tg-ArcSwe Mice

Stina Tucker; Christer Möller; Karin Tegerstedt; Anna Lord; Hanna Laudon; Johan Sjödahl; Linda Söderberg; Erika Spens; Charlotte Sahlin; Erik Rollman Waara; Andrew Satlin; Pär Gellerfors; Gunilla Osswald; Lars Lannfelt

Amyloid-β (Aβ) immunotherapy for Alzheimers disease (AD) has good preclinical support from transgenic mouse models and clinical data suggesting that a long-term treatment effect is possible. Soluble Aβ protofibrils have been shown to exhibit neurotoxicity in vitro and in vivo, and constitute an attractive target for immunotherapy. Here, we demonstrate that the humanized antibody BAN2401 and its murine version mAb158 exhibit a strong binding preference for Aβ protofibrils over Aβ monomers. Further, we confirm the presence of the target by showing that both antibodies efficiently immunoprecipitate soluble Aβ aggregates in human AD brain extracts. mAb158 reached the brain and reduced the brain protofibril levels by 42% in an exposure-dependent manner both after long-term and short-term treatment in tg-ArcSwe mice. Notably, a 53% reduction of protofibrils/oligomers in cerebrospinal fluid (CSF) that correlated with reduced brain protofibril levels was observed after long-term treatment, suggesting that CSF protofibrils/oligomers could be used as a potential biomarker. No change in native monomeric Aβ42 could be observed in brain TBS extracts after mAb158-treatment in tg-ArcSwe mice. By confirming the specific ability of mAb158 to selectively bind and reduce soluble Aβ protofibrils, with minimal binding to Aβ monomers, we provide further support in favor of its position as an attractive new candidate for AD immunotherapy. BAN2401 has undergone full phase 1 development, and available data indicate a favorable safety profile in AD patients.


Experimental Cell Research | 2003

APP intracellular domain formation and unaltered signaling in the presence of familial Alzheimer’s disease mutations

Anna Bergman; Dorota Religa; Helena Karlström; Hanna Laudon; Bengt Winblad; Lars Lannfelt; Johan Lundkvist; Jan Näslund

One of the cardinal neuropathological findings in brains from Alzheimers disease (AD) patients is the occurrence of amyloid beta-peptide (Abeta) deposits. The gamma-secretase-mediated intramembrane proteolysis event generating Abeta also results in the release of the APP intracellular domain (AICD), which may mediate nuclear signaling. It was recently shown that AICD starts at a position distal to the site predicted from gamma-secretase cleavage within the membrane. This novel site, the epsilon site, is located close to the inner leaflet of the membrane bilayer. The relationship between proteolysis at the gamma and epsilon sites has not been fully characterized. Here we studied AICD signaling in intact cells using a chimeric C99 molecule and a luciferase reporter system. We show that the release of AICD from the membrane takes place in a compartment downstream of the endoplasmic reticulum, is dependent on presenilin proteins, and can be inhibited by treatment with established gamma-secretase inhibitors. Moreover, we find that AICD signaling remains unaltered from C99 derivatives containing mutations associated with increased Abeta42 production and familial AD. These findings indicate that there are very similar routes for Abeta and AICD formation but that FAD-linked mutations in APP primarily affect gamma-secretase-mediated Abeta42 formation, and not AICD signaling.


Journal of Neurochemistry | 2005

γ‐Secretase complexes containing N‐ and C‐terminal fragments of different presenilin origin retain normal γ‐secretase activity

Kia Strömberg; Emil M. Hansson; Hanna Laudon; Susanne Bergstedt; Jan Näslund; Johan Lundkvist; Urban Lendahl

The γ‐secretase complex processes substrate proteins within membranes and consists of four proteins: presenilin (PS), nicastrin, Aph‐1 and Pen‐2. PS harbours the enzymatic activity of the complex, and there are two mammalian PS homologues: PS1 and PS2. PS undergoes endoproteolysis, generating the N‐ and C‐terminal fragments, NTF and CTF, which represent the active species of PS. To characterize the functional similarity between complexes of various PS composition, we analysed PS1, PS2, and chimeric PS composed of the NTF from PS1 and CTF from PS2, or vice versa, in assembly and function of the γ‐secretase complex. Chimeric PSs, like PS1 and PS2, undergo normal endoproteolysis when introduced into cells devoid of endogenous PS. Furthermore, PS2 CTF can, at least partially, restore processing in a truncated PS1, which cannot undergo endoproteolysis. All PS forms enable maturation of nicastrin and cleave full length Notch receptors, indicating that both PS1 and PS2 are present at the cell surface. Finally, when co‐introduced as separate molecules, NTF and CTF of different PS origin reconstitute γ‐secretase activity. In conclusion, these data show that endoproteolysis, NTF–CTF interactions, and the assembly and activity of γ‐secretase complexes are very conserved between PS1 and PS2.


Physiology & Behavior | 2007

The Alzheimer's disease-associated γ-secretase complex: Functional domains in the presenilin 1 protein

Hanna Laudon; Bengt Winblad; Jan Näslund

Alzheimers disease is neuropathologically characterized by the presence of neurofibrillary tangles and amyloid plaques in the brain. Amyloid plaques are extracellular deposits primarily composed of the amyloid beta-peptide, which is derived from the amyloid beta-precursor protein (APP) by sequential cleavages at the beta-secretase and gamma-secretase sites. gamma-Secretase cleavage is performed by a high molecular weight protein complex containing presenilin (PS), nicastrin, Aph-1 and Pen-2. The gamma-secretase complex is an unusual transmembrane aspartyl protease that cleaves APP within the transmembrane domain. In addition to APP, a large number of other single membrane-spanning proteins have been shown to be cleaved within their transmembrane domains by the gamma-secretase complex in a process referred to as regulated intramembrane proteolysis. Here we review recent research leading to the identification and understanding of the gamma-secretase complex components with emphasis on PS, which harbors the catalytic site. In addition, we summarize our own work focused on identifying and studying domains in PS1 that are critical for mediating gamma-secretase activity. Biochemical understanding of the gamma-secretase complex is important from a basic biological and physiological point of view, and could help in the development of small molecules that modulate gamma-secretase processing in an APP-specific manner.


Journal of Biological Chemistry | 2010

The Large Hydrophilic Loop of Presenilin 1 Is Important for Regulating γ-Secretase Complex Assembly and Dictating the Amyloid β Peptide (Aβ) Profile without Affecting Notch Processing

Johanna Wanngren; Jenny Frånberg; Anne-Lie Svensson; Hanna Laudon; Fredrik Olsson; Bengt Winblad; Frank Liu; Jan Näslund; Johan Lundkvist; Helena Karlström

γ-Secretase is an enzyme complex that mediates both Notch signaling and β-amyloid precursor protein (APP) processing, resulting in the generation of Notch intracellular domain, APP intracellular domain, and the amyloid β peptide (Aβ), the latter playing a central role in Alzheimer disease (AD). By a hitherto undefined mechanism, the activity of γ-secretase gives rise to Aβ peptides of different lengths, where Aβ42 is considered to play a particular role in AD. In this study we have examined the role of the large hydrophilic loop (amino acids 320–374, encoded by exon 10) of presenilin 1 (PS1), the catalytic subunit of γ-secretase, for γ-secretase complex formation and activity on Notch and APP processing. Deletion of exon 10 resulted in impaired PS1 endoproteolysis, γ-secretase complex formation, and had a differential effect on Aβ-peptide production. Although the production of Aβ38, Aβ39, and Aβ40 was severely impaired, the effect on Aβ42 was affected to a lesser extent, implying that the production of the AD-related Aβ42 peptide is separate from the production of the Aβ38, Aβ39, and Aβ40 peptides. Interestingly, formation of the intracellular domains of both APP and Notch was intact, implying a differential cleavage activity between the ϵ/S3 and γ sites. The most C-terminal amino acids of the hydrophilic loop were important for regulating APP processing. In summary, the large hydrophilic loop of PS1 appears to differentially regulate the relative production of different Aβ peptides without affecting Notch processing, two parameters of significance when considering γ-secretase as a target for pharmaceutical intervention in AD.


Journal of Neurochemistry | 2006

Caspase cleaved presenilin-1 is part of active γ-secretase complexes

Camilla A. Hansson; Bogdan O. Popescu; Hanna Laudon; Angel Cedazo-Minguez; Laurentiu M. Popescu; Bengt Winblad; Maria Ankarcrona

γ‐Secretase is a key enzyme involved in the processing of the β‐amyloid precursor protein into amyloid β‐peptides (Aβ). Aβ accumulates and forms plaques in Alzheimers disease (AD) brains. A progressive neurodegeneration and cognitive decline occurs during the course of the disease, and Aβ is believed to be central for the molecular pathogenesis of AD. Apoptosis has been implicated as one of the mechanisms behind the neuronal cell loss seen in AD. We have studied preservation and activity of the γ‐secretase complex during apoptosis in neuroblastoma cells (SH‐SY5Y) exposed to staurosporine (STS). We report that the known components (presenilin, Nicastrin, Aph‐1 and Pen‐2) interact and form active γ‐secretase complexes in apoptotic cells. In addition, the fragments corresponding to the PS1 N‐terminal fragment and the caspase‐cleaved PS1 C‐terminal fragment (PS1‐caspCTF) were found to form active γ‐secretase complexes when co‐expressed in presenilin (PS) knockout cells. Interestingly, PS1‐caspCTF replaced the normal PS1 C‐terminal fragment and was co‐immunoprecipitated with the γ‐secretase complex in SH‐SY5Y cells exposed to STS. In addition, Aβ was detected in medium from apoptotic HEK APPswe cells. Together, the data show that γ‐secretase complexes containing PS1‐caspCTF are active, and suggest that this proteolytic activity is also important in dying cells and may affect the progression of AD.


Alzheimer's Research & Therapy | 2018

Efficient clearance of Aβ protofibrils in AβPP-transgenic mice treated with a brain-penetrating bifunctional antibody

Stina Syvänen; Greta Hultqvist; Tobias Gustavsson; Astrid Gumucio; Hanna Laudon; Linda Söderberg; Martin Ingelsson; Lars Lannfelt; Dag Sehlin

BackgroundAmyloid-β (Aβ) immunotherapy is one of the most promising disease-modifying strategies for Alzheimer’s disease (AD). Despite recent progress targeting aggregated forms of Aβ, low antibody brain penetrance remains a challenge. In the present study, we used transferrin receptor (TfR)-mediated transcytosis to facilitate brain uptake of our previously developed Aβ protofibril-selective mAb158, with the aim of increasing the efficacy of immunotherapy directed toward soluble Aβ protofibrils.MethodsAβ protein precursor (AβPP)-transgenic mice (tg-ArcSwe) were given a single dose of mAb158, modified for TfR-mediated transcytosis (RmAb158-scFv8D3), in comparison with an equimolar dose or a tenfold higher dose of unmodified recombinant mAb158 (RmAb158). Soluble Aβ protofibrils and total Aβ in the brain were measured by enzyme-linked immunosorbent assay (ELISA). Brain distribution of radiolabeled antibodies was visualized by positron emission tomography (PET) and ex vivo autoradiography.ResultsELISA analysis of Tris-buffered saline brain extracts demonstrated a 40% reduction of soluble Aβ protofibrils in both RmAb158-scFv8D3- and high-dose RmAb158-treated mice, whereas there was no Aβ protofibril reduction in mice treated with a low dose of RmAb158. Further, ex vivo autoradiography and PET imaging revealed different brain distribution patterns of RmAb158-scFv8D3 and RmAb158, suggesting that these antibodies may affect Aβ levels by different mechanisms.ConclusionsWith a combination of biochemical and imaging analyses, this study demonstrates that antibodies engineered to be transported across the blood-brain barrier can be used to increase the efficacy of Aβ immunotherapy. This strategy may allow for decreased antibody doses and thereby reduced side effects and treatment costs.


Journal of Biological Chemistry | 2005

A nine-transmembrane domain topology for presenilin 1.

Hanna Laudon; Emil M. Hansson; Karin Melén; Anna Bergman; Mark R. Farmery; Bengt Winblad; Urban Lendahl; Gunnar von Heijne; Jan Näslund


Journal of Neurochemistry | 2004

Co-expressed presenilin 1 NTF and CTF form functional γ-secretase complexes in cells devoid of full-length protein

Hanna Laudon; Paul M. Mathews; Helena Karlström; Anna Bergman; Mark R. Farmery; Ralph A. Nixon; Bengt Winblad; Samuel E. Gandy; Urban Lendahl; Johan Lundkvist; Jan Näslund

Collaboration


Dive into the Hanna Laudon's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Samuel E. Gandy

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge