Network


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

Hotspot


Dive into the research topics where Nikolett Lénárt is active.

Publication


Featured researches published by Nikolett Lénárt.


Nature Communications | 2016

Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke

Gergely Szalay; Bernadett Martinecz; Nikolett Lénárt; Zsuzsanna Környei; Barbara Orsolits; Linda Judák; Eszter Császár; Rebeka Fekete; Brian L. West; Gergely Katona; Balázs Rózsa; Adam Denes

Microglia are the main immune cells of the brain and contribute to common brain diseases. However, it is unclear how microglia influence neuronal activity and survival in the injured brain in vivo. Here we develop a precisely controlled model of brain injury induced by cerebral ischaemia combined with fast in vivo two-photon calcium imaging and selective microglial manipulation. We show that selective elimination of microglia leads to a striking, 60% increase in infarct size, which is reversed by microglial repopulation. Microglia-mediated protection includes reduction of excitotoxic injury, since an absence of microglia leads to dysregulated neuronal calcium responses, calcium overload and increased neuronal death. Furthermore, the incidence of spreading depolarization (SD) is markedly reduced in the absence of microglia. Thus, microglia are involved in changes in neuronal network activity and SD after brain injury in vivo that could have important implications for common brain diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2015

AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain injury independently of NLRP3

Adam Denes; Graham Coutts; Nikolett Lénárt; Sheena M. Cruickshank; Pablo Pelegrín; Joanne Skinner; Nancy J. Rothwell; Stuart M. Allan; David Brough

Significance Cerebral ischemia (CI; stroke, brain injury, vascular dementia, neonatal hypoxia, and many other conditions) affects people at all stages of life. Of the many diseases associated with CI, stroke alone causes up to 10% of deaths worldwide and is a leading cause of disability; yet treatment options are extremely limited, so this represents an area of massive unmet clinical need. Inflammation involving the cytokine interleukin-1 is a major contributor to cell death in the ischemic brain. Inflammation can be regulated by large protein complexes called inflammasomes. Here we show that the NLRC4 (NLR family, CARD domain containing 4) and AIM2 (absent in melanoma 2) inflammasomes, but not the NLRP3 (NLR family, pyrin domain containing 3) inflammasome, contribute to inflammation and injury in an ischemic brain and are thus potential therapeutic targets for these devastating diseases. Inflammation that contributes to acute cerebrovascular disease is driven by the proinflammatory cytokine interleukin-1 and is known to exacerbate resulting injury. The activity of interleukin-1 is regulated by multimolecular protein complexes called inflammasomes. There are multiple potential inflammasomes activated in diverse diseases, yet the nature of the inflammasomes involved in brain injury is currently unknown. Here, using a rodent model of stroke, we show that the NLRC4 (NLR family, CARD domain containing 4) and AIM2 (absent in melanoma 2) inflammasomes contribute to brain injury. We also show that acute ischemic brain injury is regulated by mechanisms that require ASC (apoptosis-associated speck-like protein containing a CARD), a common adaptor protein for several inflammasomes, and that the NLRP3 (NLR family, pyrin domain containing 3) inflammasome is not involved in this process. These discoveries identify the NLRC4 and AIM2 inflammasomes as potential therapeutic targets for stroke and provide new insights into how the inflammatory response is regulated after an acute injury to the brain.


Cell Stress & Chaperones | 2013

Overexpression of Hsp27 ameliorates symptoms of Alzheimer's disease in APP/PS1 mice

Melinda E. Tóth; Viktor Szegedi; Edina Varga; Gábor Juhász; János Horváth; Emőke Borbély; Balázs Csibrány; Róbert Alföldi; Nikolett Lénárt; Botond Penke; Miklós Sántha

Hsp27 belongs to the small heat shock protein family, which are ATP-independent chaperones. The most important function of Hsp27 is based on its ability to bind non-native proteins and inhibit the aggregation of incorrectly folded proteins maintaining them in a refolding-competent state. Additionally, it has anti-apoptotic and antioxidant activities. To study the effect of Hsp27 on memory and synaptic functions, amyloid-β (Aβ) accumulation, and neurodegeneration, we generated transgenic mice overexpressing human Hsp27 protein and crossed with APPswe/PS1dE9 mouse strain, a mouse model of Alzheimers disease (AD). Using different behavioral tests, we found that spatial learning was impaired in AD model mice and was rescued by Hsp27 overexpression. Electrophysiological recordings have revealed that excitability of neurons was significantly increased, and long-term potentiation (LTP) was impaired in AD model mice, whereas they were normalized in Hsp27 overexpressing AD model mice. Using anti-amyloid antibody, we counted significantly less amyloid plaques in the brain of APPswe/PS1dE9/Hsp27 animals compared to AD model mice. These results suggest that overexpression of Hsp27 protein might ameliorate certain symptoms of AD.


Journal of Cerebral Blood Flow and Metabolism | 2016

Inflammasomes link vascular disease with neuroinflammation and brain disorders.

Nikolett Lénárt; David Brough; Adam Denes

The role of inflammation in neurological disorders is increasingly recognised. Inflammatory processes are associated with the aetiology and clinical progression of migraine, psychiatric conditions, epilepsy, cerebrovascular diseases, dementia and neurodegeneration, such as seen in Alzheimer’s or Parkinson’s disease. Both central and systemic inflammatory actions have been linked with the development of brain diseases, suggesting that complex neuro-immune interactions could contribute to pathological changes in the brain across multiple temporal and spatial scales. However, the mechanisms through which inflammation impacts on neurological disease are improperly defined. To develop effective therapeutic approaches, it is imperative to understand how detrimental inflammatory processes could be blocked selectively, or controlled for prolonged periods, without compromising essential immune defence mechanisms. Increasing evidence indicates that common risk factors for brain disorders, such as atherosclerosis, diabetes, hypertension, obesity or infection involve the activation of NLRP3, NLRP1, NLRC4 or AIM2 inflammasomes, which are also associated with various neurological diseases. This review focuses on the mechanisms whereby inflammasomes, which integrate diverse inflammatory signals in response to pathogen-driven stimuli, tissue injury or metabolic alterations in multiple cell types and different organs of the body, could functionally link vascular- and neurological diseases and hence represent a promising therapeutic target.


Brain Behavior and Immunity | 2016

Brain injury induces specific changes in the caecal microbiota of mice via altered autonomic activity and mucoprotein production

Ashley Houlden; Marie Goldrick; David Brough; E S Vizi; Nikolett Lénárt; Bernadett Martinecz; Ian S. Roberts; Adam Denes

Highlights • Ischaemic brain injury drives profound changes in the gut microbiota.• The effects of brain injury on the gut microbiota include changes in goblet cells and noradrenaline.• Traumatic brain injury (TBI) also changes the gut microbiota.


Neuropharmacology | 2015

Low dosage of rimonabant leads to anxiolytic-like behavior via inhibiting expression levels and G-protein activity of kappa opioid receptors in a cannabinoid receptor independent manner

Ferenc Zádor; Nikolett Lénárt; Balázs Csibrány; Miklós Sántha; Máté Molnár; Reza Samavati; Péter Klivényi; László Vécsei; Annamária Marton; Csaba Vizler; György M. Nagy; Anna Borsodi; Sándor Benyhe; Eszter Páldy

WHAT IS KNOWN There is an increasing number of studies demonstrating the direct effect of the cannabinoid receptor 1 (CB1) antagonist/inverse agonist rimonabant on the opioid system. The kappa opioid receptors (KORs) are well known to mediate depression- and anxiety-like behavior. Clinical studies on chronic rimonabant administration have revealed that rimonabant leads to a very similar pathophysiology, suggesting a potential impact of rimonabant on KORs. OBJECTIVES Our objectives were to examine the putative effects of rimonabant on KOR ligand binding, G-protein activity, protein expression and how all these contribute to the development of depression- and anxiety-like behavior. RESULTS In Chinese hamster ovary (CHO) cell membranes transfected with rat KOR (CHO-rKOR) rimonabant inhibited KOR agonist [3H]U69593 binding in the micromolar range in competition binding experiments and specifically reduced KOR basal activity at lower micromolar concentrations in [35S]GTPγS binding assays. Rimonabant significantly inhibited dynorphin (1-11)-induced [35S]GTPγS binding in micromolar range in CHO-rKOR cells, CB1 knockout (CB1 K.O.) and CB1/CB2 double knockout mouse forebrain membranes. A single dose of i.p. 0.1 mg/kg rimonabant significantly reduced dynorphin (1-11)-induced KOR G-protein activity and KOR protein expression levels 24 h following the administration in both wild type and CB1 K.O. mice forebrain. Furthermore, in elevated plus maze mice showed an anxiolytic-like effect upon rimonabant injection that could be reversed by 1 mg/kg KOR antagonist norbinaltorphimine. The anxiolytic-like effects were further confirmed with the light–dark box test. CONCLUSION Rimonabant reduced KOR ligand binding, receptor mediated G-protein activity and protein expression level, which overall leads to altered anxiety-like behavior.


Journal of Cerebral Blood Flow and Metabolism | 2015

A novel SPECT-based approach reveals early mechanisms of central and peripheral inflammation after cerebral ischemia.

Krisztián Szigeti; Ildiko Horvath; Dániel S. Veres; Bernadett Martinecz; Nikolett Lénárt; Noémi Kovács; Erika Bakcsa; Alexa Marta; Mariann Semjeni; Domokos Máthé; Adam Denes

Inflammation that develops in the brain and peripheral organs after stroke contributes profoundly to poor outcome of patients. However, mechanisms through which inflammation impacts on brain injury and overall outcome are improperly understood, in part because the earliest inflammatory events after brain injury are not revealed by current imaging tools. Here, we show that single-photon emission computed tomography (NanoSPECT/CT Plus) allows visualization of blood brain barrier (BBB) injury after experimental stroke well before changes can be detected with magnetic resonance imaging (MRI). Early 99mTc-DTPA (diethylene triamine pentaacetic acid) signal changes predict infarct development and systemic inflammation preceding experimental stroke leads to very early perfusion deficits and increased BBB injury within 2 hours after the onset of ischemia. Acute brain injury also leads to peripheral inflammation and immunosuppression, which contribute to poor outcome of stroke patients. The SPECT imaging revealed early (within 2 hours) changes in perfusion, barrier function and inflammation in the lungs and the gut after experimental stroke, with good predictive value for the development of histopathologic changes at later time points. Collectively, visualization of early inflammatory changes after stroke could open new translational research avenues to elucidate the interactions between central and peripheral inflammation and to evaluate in vivo ‘multi-system’ effects of putative anti-inflammatory treatments.


Fluids and Barriers of the CNS | 2015

Cultured cells of the blood–brain barrier from apolipoprotein B-100 transgenic mice: effects of oxidized low-density lipoprotein treatment

Nikolett Lénárt; Fruzsina R. Walter; Alexandra Bocsik; Petra Sántha; Melinda E. Tóth; András Harazin; Andrea E. Tóth; Csaba Vizler; Zsolt Török; Ana Maria Pilbat; László Vígh; László G. Puskás; Miklós Sántha; Mária A. Deli

BackgroundThe apolipoprotein B-100 (ApoB-100) transgenic mouse line is a model of human atherosclerosis. Latest findings suggest the importance of ApoB-100 in the development of neurodegenerative diseases and microvascular/perivascular localization of ApoB-100 protein was demonstrated in the cerebral cortex of ApoB-100 transgenic mice. The aim of the study was to characterize cultured brain endothelial cells, pericytes and glial cells from wild-type and ApoB-100 transgenic mice and to study the effect of oxidized low-density lipoprotein (oxLDL) on these cells.MethodsMorphology of cells isolated from brains of wild type and ApoB-100 transgenic mice was characterized by immunohistochemistry and the intensity of immunolabeling was quantified by image analysis. Toxicity of oxLDL treatment was monitored by real-time impedance measurement and lactate dehydrogenase release. Reactive oxygen species and nitric oxide production, barrier permeability in triple co-culture blood–brain barrier model and membrane fluidity were also determined after low-density lipoprotein (LDL) or oxLDL treatment.ResultsThe presence of ApoB-100 was confirmed in brain endothelial cells, while no morphological change was observed between wild type and transgenic cells. Oxidized but not native LDL exerted dose-dependent toxicity in all three cell types, induced barrier dysfunction and increased reactive oxygen species (ROS) production in both genotypes. A partial protection from oxLDL toxicity was seen in brain endothelial and glial cells from ApoB-100 transgenic mice. Increased membrane rigidity was measured in brain endothelial cells from ApoB-100 transgenic mice and in LDL or oxLDL treated wild type cells.ConclusionThe morphological and functional properties of cultured brain endothelial cells, pericytes and glial cells from ApoB-100 transgenic mice were characterized and compared to wild type cells for the first time. The membrane fluidity changes in ApoB-100 transgenic cells related to brain microvasculature indicate alterations in lipid composition which may be linked to the partial protection against oxLDL toxicity.


PLOS ONE | 2012

Increased Tau Phosphorylation and Impaired Presynaptic Function in Hypertriglyceridemic ApoB-100 Transgenic Mice

Nikolett Lénárt; Viktor Szegedi; Gábor Juhász; Aniko Kasztner; János Horváth; Erika Bereczki; Melinda E. Tóth; Botond Penke; Miklós Sántha

Aims ApoB-100 is the major protein component of cholesterol- and triglyceride-rich LDL and VLDL lipoproteins in the serum. Previously, we generated and partially described transgenic mice overexpressing the human ApoB-100 protein. Here, we further characterize this transgenic strain in order to reveal a possible link between hypeprlipidemia and neurodegeneration. Methods and Results We analyzed the serum and cerebral lipid profiles, tau phosphorylation patterns, amyloid plaque-formation, neuronal apoptosis and synaptic plasticity of young (3 month old), adult (6 month old) and aging (10–11 month old) transgenic mice. We show that ApoB-100 transgenic animals present i) elevated serum and cerebral levels of triglycerides and ApoB-100, ii) increased cerebral tau phosphorylation at phosphosites Ser199, Ser199/202, Ser396 and Ser404. Furthermore, we demonstrate, that tau hyperphosphorylation is accompanied by impaired presynaptic function, long-term potentiation and widespread hippocampal neuronal apoptosis. Conclusions The results presented here indicate that elevated ApoB-100 level and the consequent chronic hypertriglyceridemia may lead to impaired neuronal function and neurodegeneration, possibly via hyperphosphorylation of tau protein. On account of their specific phenotype, ApoB-100 transgenic mice may be considered a versatile model of hyperlipidemia-induced age-related neurodegeneration.


Frontiers in Cellular Neuroscience | 2018

Cerebrovascular Pathology in Hypertriglyceridemic APOB-100 Transgenic Mice

Zsófia Hoyk; Melinda E. Tóth; Nikolett Lénárt; Dóra Nagy; Brigitta Dukay; Alexandra Csefová; Ágnes Zvara; György Seprényi; András Kincses; Fruzsina R. Walter; Szilvia Veszelka; Judit Vígh; Beáta Barabási; András Harazin; Ágnes Kittel; László G. Puskás; Botond Penke; László Vígh; Mária A. Deli; Miklós Sántha

Hypertriglyceridemia is not only a serious risk factor in the development of cardiovascular diseases, but it is linked to neurodegeneration, too. Previously, we generated transgenic mice overexpressing the human APOB-100 protein, a mouse model of human atherosclerosis. In this model we observed high plasma levels of triglycerides, oxidative stress, tau hyperphosphorylation, synaptic dysfunction, cognitive impairment, increased neural apoptosis and neurodegeneration. Neurovascular dysfunction is recognized as a key factor in the development of neurodegenerative diseases, but the cellular and molecular events linking cerebrovascular pathology and neurodegeneration are not fully understood. Our aim was to study cerebrovascular changes in APOB-100 transgenic mice. We described the kinetics of the development of chronic hypertriglyceridemia in the transgenic animals. Increased blood-brain barrier permeability was found in the hippocampus of APOB-100 transgenic mice which was accompanied by structural changes. Using transmission electron microscopy, we detected changes in the brain capillary endothelial tight junction structure and edematous swelling of astrocyte endfeet. In brain microvessels isolated from APOB-100 transgenic animals increased Lox-1, Aqp4, and decreased Meox-2, Mfsd2a, Abcb1a, Lrp2, Glut-1, Nos2, Nos3, Vim, and in transgenic brains reduced Cdh2 and Gfap-σ gene expressions were measured using quantitative real-time PCR. We confirmed the decreased P-glycoprotein (ABCB1) and vimentin expression related to the neurovascular unit by immunostaining in transgenic brain sections using confocal microscopy. We conclude that in chronic hypertriglyceridemic APOB-100 transgenic mice both functional and morphological cerebrovascular pathology can be observed, and this animal model could be a useful tool to study the link between cerebrovascular pathology and neurodegeneration.

Collaboration


Dive into the Nikolett Lénárt's collaboration.

Top Co-Authors

Avatar

Adam Denes

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Miklós Sántha

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Bernadett Martinecz

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Melinda E. Tóth

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Botond Penke

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Eszter Császár

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Rebeka Fekete

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Zsuzsanna Környei

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

David Brough

Manchester Academic Health Science Centre

View shared research outputs
Top Co-Authors

Avatar

András Harazin

Hungarian Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge