Network


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

Hotspot


Dive into the research topics where Alberto Lleó is active.

Publication


Featured researches published by Alberto Lleó.


Science Translational Medicine | 2014

TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis

Gernot Kleinberger; Y. Yamanishi; Marc Suárez-Calvet; Eva Czirr; Ebba Lohmann; Elise Cuyvers; Hanne Struyfs; N. Pettkus; Andrea Wenninger-Weinzierl; F. Mazaheri; Sabina Tahirovic; Alberto Lleó; Daniel Alcolea; Juan Fortea; Michael Willem; Sven Lammich; José-Luis Molinuevo; Raquel Sánchez-Valle; Anna Antonell; Alfredo Ramirez; Michael T. Heneka; Kristel Sleegers; J. van der Zee; J. J. Martin; S. Engelborghs; A. Demirtas-Tatlidede; Henrik Zetterberg; C. Van Broeckhoven; Hakan Gurvit; Tony Wyss-Coray

Loss of TREM2 function impairs phagocytosis and correlates with decreased soluble TREM2 in biological fluids of patients with neurodegenerative disorders. TREM2 and Neurodegeneration Little is known about how risk factors facilitate initiation and propagation of neurodegenerative disorders. Rare mutations in TREM2 increase the risk for several neurodegenerative disorders including Alzheimer’s disease (AD), Parkinson’s disease, and frontotemporal dementia (FTD). Kleinberger et al. now show that mutations associated with neurodegenerative diseases interfere with TREM2 function by preventing its maturation, transport to the cell surface, and shedding. Expression of mutant TREM2 led to reduced phagocytic activity by different cell types, suggesting that removal of cellular debris by, for example, microglia in the brain might be affected in patients with TREM2 mutations. In a patient with FTD-like syndrome carrying a homozygous TREM2 mutation, no soluble TREM2 was detected in the cerebrospinal fluid (CSF) and plasma. Patients with sporadic FTD and AD showed slightly reduced concentrations of soluble TREM2 in their CSF. Although much further testing and validation are needed, soluble TREM2 might be useful as a marker of neurodegeneration. Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to Nasu-Hakola disease, Alzheimer’s disease (AD), Parkinson’s disease, amyotrophic lateral sclerosis, frontotemporal dementia (FTD), and FTD-like syndrome without bone involvement. TREM2 is an innate immune receptor preferentially expressed by microglia and is involved in inflammation and phagocytosis. Whether and how TREM2 missense mutations affect TREM2 function is unclear. We report that missense mutations associated with FTD and FTD-like syndrome reduce TREM2 maturation, abolish shedding by ADAM proteases, and impair the phagocytic activity of TREM2-expressing cells. As a consequence of reduced shedding, TREM2 is virtually absent in the cerebrospinal fluid (CSF) and plasma of a patient with FTD-like syndrome. A decrease in soluble TREM2 was also observed in the CSF of patients with AD and FTD, further suggesting that reduced TREM2 function may contribute to increased risk for two neurodegenerative disorders.


Neurobiology of Disease | 2009

A novel GSK-3β inhibitor reduces Alzheimer's pathology and rescues neuronal loss in vivo

L. Serenó; Mireia Coma; M. Rodríguez; P. Sánchez-Ferrer; M.B. Sánchez; I. Gich; J.M. Agulló; Mar Pérez; Jesús Avila; C. Guardia-Laguarta; Jordi Clarimón; Alberto Lleó; T. Gómez-Isla

Amyloid deposits, neurofibrillary tangles, and neuronal cell death in selectively vulnerable brain regions are the chief hallmarks in Alzheimers (AD) brains. Glycogen synthase kinase-3 (GSK-3) is one of the key kinases required for AD-type abnormal hyperphosphorylation of tau, which is believed to be a critical event in neurofibrillary tangle formation. GSK-3 has also been recently implicated in amyloid precursor protein (APP) processing/Abeta production, apoptotic cell death, and learning and memory. Thus, GSK-3 inhibition represents a very attractive drug target in AD and other neurodegenerative disorders. To investigate whether GSK-3 inhibition can reduce amyloid and tau pathologies, neuronal cell death and memory deficits in vivo, double transgenic mice coexpressing human mutant APP and tau were treated with a novel non-ATP competitive GSK-3beta inhibitor, NP12. Treatment with this thiadiazolidinone compound resulted in lower levels of tau phosphorylation, decreased amyloid deposition and plaque-associated astrocytic proliferation, protection of neurons in the entorhinal cortex and CA1 hippocampal subfield against cell death, and prevention of memory deficits in this transgenic mouse model. These results show that this novel GSK-3 inhibitor has a dual impact on amyloid and tau alterations and, perhaps even more important, on neuronal survival in vivo further suggesting that GSK-3 is a relevant therapeutic target in AD.


Nature Medicine | 2004

Nonsteroidal anti-inflammatory drugs lower Aβ42 and change presenilin 1 conformation

Alberto Lleó; Oksana Berezovska; Lauren Herl; Susan Raju; Amy Deng; Brian J. Bacskai; Matthew P. Frosch; Michael C. Irizarry; Bradley T. Hyman

Recent reports suggest that some commonly used nonsteroidal anti-inflammatory drugs (NSAIDs) unexpectedly shift the cleavage products of amyloid precursor protein (APP) to shorter, less fibrillogenic forms, although the underlying mechanism remains unknown. We now demonstrate, using a fluorescence resonance energy transfer method, that Aβ42-lowering NSAIDs specifically affect the proximity between APP and presenilin 1 and alter presenilin 1 conformation both in vitro and in vivo, suggesting a novel allosteric mechanism of action.


The Journal of Neuroscience | 2005

Familial Alzheimer's Disease Presenilin 1 Mutations Cause Alterations in the Conformation of Presenilin and Interactions with Amyloid Precursor Protein

Oksana Berezovska; Alberto Lleó; Lauren Herl; Matthew P. Frosch; Edward A. Stern; Brian J. Bacskai; Bradley T. Hyman

Presenilin 1 (PS1) is a critical component of the γ-secretase complex, an enzymatic activity that cleaves amyloid β (Aβ) from the amyloid precursor protein (APP). More than 100 mutations spread throughout the PS1 molecule are linked to autosomal dominant familial Alzheimers disease (FAD). All of these mutations lead to a similar phenotype: an increased ratio of Aβ42 to Aβ40, increased plaque deposition, and early age of onset. We use a recently developed microscopy approach, fluorescence lifetime imaging microscopy, to monitor the relative molecular distance between PS1 N and C termini in intact cells. We show that FAD-linked missense mutations located near the N and C termini, in the mid-region of PS1, and the exon 9 deletion mutation all change the spatial relationship between PS1 N and C termini in a similar way, increasing proximity of the two epitopes. This effect is opposite of that observed by treatment with Aβ42-lowering nonsteroidal anti-inflammatory drugs (NSAIDs) (Lleo et al., 2004b). Accordingly, treatment of M146L PS1-overexpressing neurons with high-dose NSAIDs somewhat offsets the conformational change associated with the mutation. Moreover, by monitoring the relative distance between a PS1 loop epitope and the APP C terminus, we demonstrate that the FAD PS1 mutations are also associated with a consistent change in the configuration of the PS1-APP complex. The nonpathogenic E318G PS1 polymorphism had no effect on PS1 N terminus-C terminus proximity or PS1-APP interactions. We propose that the conformational change we observed may therefore provide a shared molecular mechanism for FAD pathogenesis caused by a wide range of PS1 mutations.


Annals of Neurology | 2013

Low cerebrospinal fluid concentration of mitochondrial DNA in preclinical Alzheimer disease

Petar Podlesniy; Joana Figueiro-Silva; Albert Lladó; Anna Antonell; Raquel Sánchez-Valle; Daniel Alcolea; Alberto Lleó; José Luis Molinuevo; Nuria Serra; Ramon Trullas

To identify a novel biochemical marker that precedes clinical symptoms in Alzheimer disease (AD).


JAMA Neurology | 2011

Dementia risk in Parkinson disease: disentangling the role of MAPT haplotypes.

Núria Setó-Salvia; Jordi Clarimón; Javier Pagonabarraga; Berta Pascual-Sedano; Antonia Campolongo; Onofre Combarros; Jose Ignacio Mateo; Daniel Regaña; Mercè Martínez-Corral; Marta Marquié; Daniel Alcolea; Marc Suárez-Calvet; Laura Molina-Porcel; Oriol Dols; Teresa Gomez-Isla; Rafael Blesa; Alberto Lleó; Jaime Kulisevsky

BACKGROUND Dementia in Parkinson disease (PD) causes nursing home placement, caregiver distress, higher health care burden, and increased mortality. OBJECTIVE To determine whether the microtubule-associated protein tau (MAPT) H1 haplotype and MAPT subhaplotypes play a role in the risk of PD and Parkinson disease-dementia (PDD) complex. DESIGN Case-control genetic analysis. SETTING Movement Disorders and Memory Units, Hospital de Sant Pau, Barcelona, Spain. PARTICIPANTS Two hundred two patients with PD (48 of whom developed dementia>2 years after disease onset), 41 patients with Lewy body dementia (LBD, pathologically confirmed in 17), 164 patients with Alzheimer disease (AD), and 374 controls. METHODS The MAPT haplotype was determined by testing for a 238-base pair deletion between exons 9 and 10, which is characteristic of the H2 haplotype. Haploview was used to visualize linkage disequilibrium relationships between all genetic variants (5 single-nucleotide polymorphisms and the del-In9 variant) within and surrounding the MAPT region. RESULTS The H1 haplotype was significantly overrepresented in PD patients compared with controls (P=.001). Stratifying the PD sample by the presence of dementia revealed a stronger association in PDD patients (sex- and age-adjusted odds ratio, 3.73; P=.002) than in PD patients without dementia (sex- and age-adjusted odds ratio, 1.89; P=.04). Examination of specific subhaplotypes showed that a rare version of the H1 haplotype (named H1p) was overrepresented in PDD patients compared with controls (2.3% vs 0.1%; P=.003). No positive signals for any of the MAPT variants or H1 subhaplotypes were found in AD or LBD. CONCLUSIONS Our data confirm that MAPT H1 is associated with PD and has a strong influence on the risk of dementia in PD patients. Our results also suggest that none of the MAPT subhaplotypes play a significant role in other neurodegenerative diseases, such as LBD or AD.


American Journal of Geriatric Psychiatry | 2004

Clinical, pathological, and biochemical spectrum of Alzheimer disease associated with PS-1 mutations

Alberto Lleó; Oksana Berezovska; John H. Growdon; Bradley T. Hyman

Three genes have been implicated in the etiology of early-onset autosomal-dominant Alzheimer disease (AD): the amyloid precursor protein, the presenilin-1, and presenilin-2 genes. Approximately half of autosomal-dominant AD cases are associated with mutations in the presenilin-1 (PS-1) gene on the long arm of Chromosome 14. Marked allelic heterogeneity characterizes families with PS-1 gene mutations; more than 100 different mutations have been found in independent families thus far. With the exception of age at onset, the clinical phenotype is similar to late-onset AD, although some rare specific phenotypes have been described. These mutations lead to enhanced deposition of total Abeta and Abeta42 (but not Abeta40) in the brain, compared with sporadic AD. There is a considerable heterogeneity in the histological profiles among brains from patients with different mutations, and although some lead to predominantly parenchymal deposition of Abeta in the form of diffuse and cored plaques, others show predominantly vascular deposition, with severe amyloid angiopathy. Only some mutations are associated with enhanced neurofibrillary tangle formation and increased neuronal loss compared with sporadic AD. However, there is an important clinical and pathological variability even among family members with the same mutation, which suggests the involvement of other genetic or environmental factors that modulate the clinical expression of the disease. This represents a valuable model for identifying such factors and has potential implications for the development of new therapeutic strategies for delaying disease onset.


Movement Disorders | 2012

Glucocerebrosidase mutations confer a greater risk of dementia during Parkinson's disease course.

Núria Setó-Salvia; Javier Pagonabarraga; Henry Houlden; Berta Pascual-Sedano; Oriol Dols-Icardo; Arianna Tucci; Coro Paisán‐Ruiz; Antonia Campolongo; Sofía Antón-Aguirre; Inés Martín; Laia Muñoz; Enric Bufill; Lluïsa Vilageliu; Daniel Grinberg; Mónica Cozar; Rafael Blesa; Alberto Lleó; John Hardy; Jaime Kulisevsky; Jordi Clarimón

Mutations in the glucocerebrosidase gene are associated with Parkinsons disease and Lewy body dementia. However, whether these alterations have any effect on the clinical course of Parkinsons disease is not clear. The glucocerebrosidase coding region was fully sequenced in 225 Parkinsons disease patients, 17 pathologically confirmed Lewy body dementia patients, and 186 controls from Spain. Twenty‐two Parkinsons disease patients (9.8%) and 2 Lewy body dementia patients (11.8%) carried mutations in the glucocerebrosidase gene, compared with only 1 control (0.5%); P = .016 and P = .021 for Parkinsons disease and Lewy body dementia, respectively. The N370S and the L444P mutations represented 50% of the alterations. Two novel variants, L144V and S488T, and 7 previously described alterations were also found. Alterations in glucocerebrosidase were associated with a significant risk of dementia during the clinical course of Parkinsons disease (age at onset, years of evolution, and sex‐adjusted odds ratio, 5.8; P = .001). Mutation carriers did not show worse motor symptoms, had good response to L‐dopa, and tended to present the intermediate parkinsonian phenotype. Our findings suggest that mutations in the glucocerebrosidase gene not only increase the risk of both Parkinsons disease and Lewy body dementia but also strongly influence the course of Parkinsons disease with respect to the appearance of dementia.


Embo Molecular Medicine | 2016

sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early‐stage Alzheimer's disease and associate with neuronal injury markers

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 Luis 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; Christian Haass

TREM2 is an innate immune receptor expressed on the surface of microglia. Loss‐of‐function mutations of TREM2 are associated with increased risk of Alzheimers 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.


Nature Reviews Neurology | 2015

Cerebrospinal fluid biomarkers in trials for Alzheimer and Parkinson diseases

Alberto Lleó; Enrica Cavedo; Lucilla Parnetti; Hugo Vanderstichele; Sanna Kaisa Herukka; Niels Andreasen; Roberta Ghidoni; Piotr Lewczuk; Andreas Jeromin; Bengt Winblad; Magda Tsolaki; Barbara Mroczko; Pieter Jelle Visser; Isabel Santana; Per Svenningsson; Kaj Blennow; Dag Aarsland; José Luis Molinuevo; Henrik Zetterberg; Brit Mollenhauer

Alzheimer disease (AD) and Parkinson disease (PD) are the most common neurodegenerative disorders. For both diseases, early intervention is thought to be essential to the success of disease-modifying treatments. Cerebrospinal fluid (CSF) can reflect some of the pathophysiological changes that occur in the brain, and the number of CSF biomarkers under investigation in neurodegenerative conditions has grown rapidly in the past 20 years. In AD, CSF biomarkers are increasingly being used in clinical practice, and have been incorporated into the majority of clinical trials to demonstrate target engagement, to enrich or stratify patient groups, and to find evidence of disease modification. In PD, CSF biomarkers have not yet reached the clinic, but are being studied in patients with parkinsonism, and are being used in clinical trials either to monitor progression or to demonstrate target engagement and downstream effects of drugs. CSF biomarkers might also serve as surrogate markers of clinical benefit after a specific therapeutic intervention, although additional data are required. It is anticipated that CSF biomarkers will have an important role in trials aimed at disease modification in the near future. In this Review, we provide an overview of CSF biomarkers in AD and PD, and discuss their role in clinical trials.

Collaboration


Dive into the Alberto Lleó's collaboration.

Top Co-Authors

Avatar

Rafael Blesa

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Jordi Clarimón

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Juan Fortea

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Daniel Alcolea

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

María Carmona-Iragui

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge