Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 193: 106437, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367882

ABSTRACT

TDP-43 pathology is found in several neurodegenerative disorders, collectively referred to as "TDP-43 proteinopathies". Aggregates of TDP-43 are present in the brains and spinal cords of >97% of amyotrophic lateral sclerosis (ALS), and in brains of ∼50% of frontotemporal dementia (FTD) patients. While mutations in the TDP-43 gene (TARDBP) are usually associated with ALS, many clinical reports have linked these mutations to cognitive impairments and/or FTD, but also to other neurodegenerative disorders including Parkinsonism (PD) or progressive supranuclear palsy (PSP). TDP-43 is a ubiquitously expressed, highly conserved RNA-binding protein that is involved in many cellular processes, mainly RNA metabolism. To investigate systemic pathological mechanisms in TDP-43 proteinopathies, aiming to capture the pleiotropic effects of TDP-43 mutations, we have further characterised a mouse model carrying a point mutation (M323K) within the endogenous Tardbp gene. Homozygous mutant mice developed cognitive and behavioural deficits as early as 3 months of age. This was coupled with significant brain structural abnormalities, mainly in the cortex, hippocampus, and white matter fibres, together with progressive cortical interneuron degeneration and neuroinflammation. At the motor level, progressive phenotypes appeared around 6 months of age. Thus, cognitive phenotypes appeared to be of a developmental origin with a mild associated progressive neurodegeneration, while the motor and neuromuscular phenotypes seemed neurodegenerative, underlined by a progressive loss of upper and lower motor neurons as well as distal denervation. This is accompanied by progressive elevated TDP-43 protein and mRNA levels in cortex and spinal cord of homozygous mutant mice from 3 months of age, together with increased cytoplasmic TDP-43 mislocalisation in cortex, hippocampus, hypothalamus, and spinal cord at 12 months of age. In conclusion, we find that Tardbp M323K homozygous mutant mice model many aspects of human TDP-43 proteinopathies, evidencing a dual role for TDP-43 in brain morphogenesis as well as in the maintenance of the motor system, making them an ideal in vivo model system to study the complex biology of TDP-43.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , TDP-43 Proteinopathies , Animals , Child, Preschool , Humans , Mice , Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Cognition , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology
2.
Front Psychol ; 14: 1273608, 2023.
Article in English | MEDLINE | ID: mdl-38034292

ABSTRACT

Introduction: The Addenbrooke's Cognitive Examination III (ACE-III) is a brief test useful for neuropsychological assessment. Several studies have validated the test for the diagnosis of Alzheimer's disease (AD) and frontotemporal dementia (FTD). In this study, we aimed to examine the metabolic correlates associated with the performance of ACE-III in AD and behavioral variant FTD. Methods: We enrolled 300 participants in a cross-sectional study, including 180 patients with AD, 60 with behavioral FTD (bvFTD), and 60 controls. An 18F-Fluorodeoxyglucose positron emission tomography study was performed in all cases. Correlation between the ACE-III and its domains (attention, memory, fluency, language, and visuospatial) with the brain metabolism was estimated. Results: The ACE-III showed distinct neural correlates in bvFTD and AD, effectively capturing the most relevant regions involved in these disorders. Neural correlates differed for each domain, especially in the case of bvFTD. Lower ACE-III scores were associated with more advanced stages in both disorders. The ACE-III exhibited high discrimination between bvFTD vs. HC, and between AD vs. HC. Additionally, it was sensitive to detect hypometabolism in brain regions associated with bvFTD and AD. Conclusion: Our study contributes to the knowledge of the brain regions associated with ACE-III, thereby facilitating its interpretation, and highlighting its suitability for screening and monitoring. This study provides further validation of ACE-III in the context of AD and FTD.

3.
Biology (Basel) ; 12(7)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37508364

ABSTRACT

Alzheimer's disease (AD), the leading cause of dementia, is a growing health issue with very limited treatment options. To meet the need for novel therapeutics, existing drugs with additional preferred pharmacological profiles could be recruited. This strategy is known as 'drug repurposing'. Here, we describe dimethyl fumarate (DMF), a drug approved to treat multiple sclerosis (MS), to be tested as a candidate for other brain diseases. We used an APP-transgenic model (APPtg) of senile ß-amyloidosis mice to further investigate the potential of DMF as a novel AD therapeutic. We treated male and female APPtg mice through drinking water at late stages of ß-amyloid (Aß) deposition. We found that DMF treatment did not result in modulating effects on Aß deposition at this stage. Interestingly, we found that glutathione-modified DMF interacts with the ATP-binding cassette transporter ABCC1, an important gatekeeper at the blood-brain and blood-plexus barriers and a key player for Aß export from the brain. Our findings suggest that ABCC1 prevents the effects of DMF, which makes DMF unsuitable as a novel therapeutic drug against AD. The discovered effects of ABCC1 also have implications for DMF treatment of multiple sclerosis.

4.
J Integr Neurosci ; 22(3): 75, 2023 May 16.
Article in English | MEDLINE | ID: mdl-37258443

ABSTRACT

BACKGROUND: Epilepsy is one of the most common neurologic diseases, and around 30% of all epilepsies, particularly the temporal lobe epilepsy (TLE), are highly refractory to current pharmacological treatments. Abnormal synchronic neuronal activity, brain glucose metabolism alterations, neurodegeneration and neuroinflammation are features of epilepsy. Further, neuroinflammation has been shown to contribute to dysregulation of neuronal excitability and the progression of epileptogenesis. Flufenamic acid (FLU), a non-steroidal anti-inflammatory drug, is also characterized by its wide properties as a dose-dependent ion channel modulator. In this context, in vitro studies have shown that it abolishes seizure-like events in neocortical slices stimulated with a gamma-aminobutyric acid A (GABAA) receptor blocker. However, little is known about its effects in animal models. Thus, our goal was to assess the efficacy and safety of a relatively high dose of FLU in the lithium-pilocarpine rat model of status epilepticus (SE). This animal model reproduces many behavioral and neurobiological features of TLE such as short-term brain hypometabolism, severe hippocampal neurodegeneration and inflammation reflected by a marked reactive astrogliosis. METHODS: FLU (100 mg/kg, i.p.) was administered to adult male rats, 150 min before SE induced by pilocarpine. Three days after the SE, brain glucose metabolism was assessed by 2-deoxy-2-[18F]-fluoro-D-glucose ([18F]FDG) positron emission tomography (PET). Markers of hippocampal integrity, neurodegeneration and reactive astrogliosis were also evaluated. RESULTS: FLU neither prevented the occurrence of the SE nor affected brain glucose hypometabolism as assessed by [18F]FDG PET. Regarding the neurohistochemical studies, FLU neither prevented neuronal damage nor hippocampal reactive astrogliosis. On the contrary, FLU increased the mortality rate and negatively affected body weight in the rats that survived the SE. CONCLUSIONS: Our results do not support an acute anticonvulsant effect of a single dose of FLU. Besides, FLU did not show short-term neuroprotective or anti-inflammatory effects in the rat lithium-pilocarpine model of SE. Moreover, at the dose administered, FLU resulted in deleterious effects.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Status Epilepticus , Rats , Male , Animals , Lithium/adverse effects , Pilocarpine/adverse effects , Flufenamic Acid/metabolism , Flufenamic Acid/pharmacology , Flufenamic Acid/therapeutic use , Rats, Sprague-Dawley , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/pharmacology , Fluorodeoxyglucose F18/therapeutic use , Gliosis/metabolism , Neuroinflammatory Diseases , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Epilepsy/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/drug therapy , Hippocampus/metabolism , Glucose/metabolism , Anti-Inflammatory Agents/adverse effects , Disease Models, Animal
5.
Biomolecules ; 13(2)2023 02 09.
Article in English | MEDLINE | ID: mdl-36830699

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia. Fingolimod has previously shown beneficial effects in different animal models of AD. However, it has shown contradictory effects when it has been applied at early disease stages. Our objective was to evaluate fingolimod in two different treatment paradigms. To address this aim, we treated male and female APP-transgenic mice for 50 days, starting either before plaque deposition at 50 days of age (early) or at 125 days of age (late). To evaluate the effects, we investigated the neuroinflammatory and glial markers, the Aß load, and the concentration of the brain-derived neurotrophic factor (BDNF). We found a reduced Aß load only in male animals in the late treatment paradigm. These animals also showed reduced microglia activation and reduced IL-1ß. No other treatment group showed any difference in comparison to the controls. On the other hand, we detected a linear correlation between BDNF and the brain Aß concentrations. The fingolimod treatment has shown beneficial effects in AD models, but the outcome depends on the neuroinflammatory state at the start of the treatment. Thus, according to our data, a fingolimod treatment would be effective after the onset of the first AD symptoms, mainly affecting the neuroinflammatory reaction to the ongoing Aß deposition.


Subject(s)
Alzheimer Disease , Mice , Animals , Male , Female , Fingolimod Hydrochloride/pharmacology , Amyloid beta-Protein Precursor , Amyloid beta-Peptides , Brain-Derived Neurotrophic Factor , Mice, Transgenic , Disease Models, Animal
6.
Basic Res Cardiol ; 117(1): 52, 2022 10 24.
Article in English | MEDLINE | ID: mdl-36279013

ABSTRACT

Ischemic stroke imparts elevated risk of heart failure though the underlying mechanisms remain poorly described. We aimed to characterize the influence of cerebral ischemic injury on cardiac function using multimodality molecular imaging to investigate brain and cardiac morphology and tissue inflammation in two mouse models of variable stroke severity. Transient middle cerebral artery occlusion (MCAo) generated extensive stroke damage (56.31 ± 40.39 mm3). Positron emission tomography imaging of inflammation targeting the mitochondrial translocator protein (TSPO) revealed localized neuroinflammation at 7 days after stroke compared to sham (3.8 ± 0.8 vs 2.6 ± 0.7 %ID/g max, p < 0.001). By contrast, parenchyma topical application of vasoconstrictor endothelin-1 did not generate significant stroke damage or neuroinflammatory cell activity. MCAo evoked a modest reduction in left ventricle ejection fraction at both 1 weeks and 3 weeks after stroke (LVEF at 3 weeks: 54.3 ± 5.7 vs 66.1 ± 3.5%, p < 0.001). This contractile impairment was paralleled by elevated cardiac TSPO PET signal compared to sham (8.6 ± 2.4 vs 5.8 ± 0.7%ID/g, p = 0.022), but was independent of leukocyte infiltration defined by flow cytometry. Stroke size correlated with severity of cardiac dysfunction (r = 0.590, p = 0.008). Statistical parametric mapping identified a direct association between neuroinflammation at 7 days in a cluster of voxels including the insular cortex and reduced ejection fraction (ρ = - 0.396, p = 0.027). Suppression of microglia led to lower TSPO signal at 7 days which correlated with spared late cardiac function after MCAo (r = - 0.759, p = 0.029). Regional neuroinflammation early after cerebral ischemia influences subsequent cardiac dysfunction. Total body TSPO PET enables monitoring of neuroinflammation, providing insights into brain-heart inter-organ communication and may guide therapeutic intervention to spare cardiac function post-stroke.


Subject(s)
Brain Ischemia , Heart Diseases , Stroke , Animals , Mice , Brain/diagnostic imaging , Brain/metabolism , Brain Ischemia/metabolism , Endothelin-1/metabolism , Heart Diseases/metabolism , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/diagnostic imaging , Inflammation/metabolism , Molecular Imaging/methods , Positron-Emission Tomography/methods , Stroke/metabolism , Vasoconstrictor Agents
7.
Neuroscience ; 499: 142-151, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35878719

ABSTRACT

Synaptic vesicle glycoprotein 2A (SV2A) is a transmembrane protein that binds levetiracetam and is involved in neurotransmission via an unknown mechanism. SV2A-immunoreactivity is reduced in animal models of epilepsy, and in postmortem hippocampi from patients with temporal lobe epilepsy. It is not known if other regions outside the hippocampus are affected in epilepsy, and whether SV2A expression is permanently reduced or regulated over time. In this study, we induced a generalized status epilepticus (SE) by systemic administration of lithium-pilocarpine to adult female rats. The brains from all animals experiencing SE were collected at different time points after the treatment. The radiotracer, [11C]-UCB-J, binds to SV2A with high affinity, and has been used for in vivo imaging as an a-proxy marker for synaptic density. Here we determined the level of tritiated UCB-J binding by semiquantitative autoradiography in the cerebral cortex, hippocampus, thalamus, and hypothalamus, and in cortical subregions. A prominent and highly significant reduction in SV2A binding capacity was observed over the first days after SE in the cerebral cortex and the hippocampus, but not in the thalamus and hypothalamus. The magnitude in reduction was larger and occurred earlier in the hippocampus and the piriform cortex, than in other cortical subregions. Interestingly, in all areas examined, the binding capacity returned to control levels 12 weeks after the SE comparable to the chronic epileptic phase. These data indicate that lithium-pilocarpine-induced epileptogenesis involves both loss and gain of synapses in the in a time-dependent manner.


Subject(s)
Epilepsy , Status Epilepticus , Animals , Brain/metabolism , Epilepsy/metabolism , Female , Hippocampus/metabolism , Lithium , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Pilocarpine , Rats , Status Epilepticus/chemically induced , Status Epilepticus/metabolism
8.
EMBO Rep ; 23(7): e54405, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35620875

ABSTRACT

Findings of early cerebral amyloid-ß deposition in mice after peripheral injection of amyloid-ß-containing brain extracts, and in humans following cadaveric human growth hormone treatment raised concerns that amyloid-ß aggregates and possibly Alzheimer's disease may be transmissible between individuals. Yet, proof that Aß actually reaches the brain from the peripheral injection site is lacking. Here, we use a proteomic approach combining stable isotope labeling of mammals and targeted mass spectrometry. Specifically, we generate 13 C-isotope-labeled brain extracts from mice expressing human amyloid-ß and track 13 C-lysine-labeled amyloid-ß after intraperitoneal administration into young amyloid precursor protein-transgenic mice. We detect injected amyloid-ß in the liver and lymphoid tissues for up to 100 days. In contrast, injected 13 C-lysine-labeled amyloid-ß is not detectable in the brain whereas the mice incorporate 13 C-lysine from the donor brain extracts into endogenous amyloid-ß. Using a highly sensitive and specific proteomic approach, we demonstrate that amyloid-ß does not reach the brain from the periphery. Our study argues against potential transmissibility of Alzheimer's disease while opening new avenues to uncover mechanisms of pathophysiological protein deposition.


Subject(s)
Alzheimer Disease , Prions , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Humans , Isotopes , Lysine , Mammals/metabolism , Mice , Mice, Transgenic , Prions/metabolism , Proteomics
9.
PLoS One ; 16(11): e0260482, 2021.
Article in English | MEDLINE | ID: mdl-34818362

ABSTRACT

PURPOSE: Alterations in brain glucose metabolism detected by 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) may serve as an early predictive biomarker and treatment target for epileptogenesis. Here, we aimed to investigate changes in cerebral glucose metabolism before induction of epileptogenesis, during epileptogenesis as well as during chronic epilepsy. As anesthesia is usually unavoidable for preclinical PET imaging and influences the distribution of the radiotracer, four different protocols were compared. PROCEDURES: We investigated 18F-FDG uptake phase in conscious rats followed by a static scan as well as dynamic scans under continuous isoflurane, medetomidine-midazolam-fentanyl (MMF), or propofol anesthesia. Furthermore, we applied different analysis approaches: atlas-based regional analysis, statistical parametric mapping, and kinetic analysis. RESULTS: At baseline and compared to uptake in conscious rats, isoflurane and propofol anesthesia resulted in decreased cortical 18F-FDG uptake while MMF anesthesia led to a globally decreased tracer uptake. During epileptogenesis, MMF anesthesia was clearly best distinctive for visualization of prominently increased glucometabolism in epilepsy-related brain areas. Kinetic modeling further increased sensitivity, particularly for continuous isoflurane anesthesia. During chronic epilepsy, hypometabolism affecting more or less the whole brain was detectable with all protocols. CONCLUSION: This study reveals evaluation of anesthesia protocols for preclinical 18F-FDG PET imaging as a critical step in the study design. Together with an appropriate data analysis workflow, the chosen anesthesia protocol may uncover otherwise concealed disease-associated regional glucometabolic changes.


Subject(s)
Brain/metabolism , Epilepsy/metabolism , Glucose/metabolism , Positron-Emission Tomography/methods , Anesthesia/methods , Anesthetics, Inhalation/administration & dosage , Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Animals , Brain/diagnostic imaging , Epilepsy/diagnostic imaging , Female , Fluorodeoxyglucose F18/metabolism , Glucose/analysis , Isoflurane/administration & dosage , Isoflurane/pharmacology , Propofol/administration & dosage , Propofol/pharmacology , Rats , Rats, Sprague-Dawley
10.
J Alzheimers Dis ; 84(4): 1677-1690, 2021.
Article in English | MEDLINE | ID: mdl-34719500

ABSTRACT

BACKGROUND: A wide range of techniques has been developed over the past decades to characterize amyloid-ß (Aß) pathology in mice. Until now, no method has been established to quantify spatial changes in Aß plaque deposition due to targeted delivery of substances using ALZET® pumps. OBJECTIVE: Development of a methodology to quantify the local distribution of Aß plaques after intracerebral infusion of compounds. METHODS: We have developed a toolbox to quantify Aß plaques in relation to intracerebral injection channels using Zeiss AxioVision® and Microsoft Excel® software. For the proof of concept, intracerebral stereotactic surgery was performed in 50-day-old APP-transgenic mice injected with PBS. At the age of 100 days, brains were collected for immunhistological analysis. RESULTS: The toolbox can be used to analyze and evaluate Aß plaques (number, size, and coverage) in specific brain areas based on their location relative to the point of the injection or the injection channel. The tool provides classification of Aß plaques in pre-defined distance groups using two different approaches. CONCLUSION: This new analytic toolbox facilitates the analysis of long-term continuous intracerebral experimental compound infusions using ALZET® pumps. This method generates reliable data for Aß deposition characterization in relation to the distribution of experimental compounds.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Brain/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Stereotaxic Techniques
11.
Metab Brain Dis ; 36(8): 2597-2602, 2021 12.
Article in English | MEDLINE | ID: mdl-34570340

ABSTRACT

Status epilepticus (SE) is a clinical emergency with high mortality. SE can trigger neuronal death or injury and alteration of neuronal networks resulting in long-term cognitive decline or epilepsy. Among the multiple factors contributing to this damage, imbalance between oxygen and glucose requirements and brain perfusion during SE has been proposed. Herein, we aimed to quantify by neuroimaging the spatiotemporal course of brain perfusion during and after lithium-pilocarpine-induced SE in rats. To this purpose, animals underwent 99mTc-HMPAO SPECT imaging at different time points during and after SE using a small animal SPECT/CT system. 99mTc-HMPAO regional uptake was normalized to the injected dose. In addition, voxel-based statistical parametric mapping was performed. SPECT imaging showed an increase of cortical perfusion before clinical seizure activity onset followed by regional hypo-perfusion starting with the first convulsive seizure and during SE. Twenty-four hours after SE, brain 99mTc-HMPAO uptake was widely decreased. Finally, chronic epileptic animals showed regionally decreased perfusion affecting hippocampus and cortical sub-regions. Despite elevated energy and oxygen requirements, brain hypo-perfusion is present during SE. Our results suggest that insufficient compensation of required blood flow might contribute to neuronal damage and neuroinflammation, and ultimately to chronic epilepsy generated by SE.


Subject(s)
Status Epilepticus , Tomography, Emission-Computed, Single-Photon , Animals , Brain/blood supply , Brain/diagnostic imaging , Cerebrovascular Circulation/physiology , Neuroimaging , Rats , Status Epilepticus/chemically induced , Status Epilepticus/diagnostic imaging , Technetium Tc 99m Exametazime , Tomography, Emission-Computed, Single-Photon/methods
12.
J Neurosci Methods ; 364: 109371, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34592173

ABSTRACT

BACKGROUND: Interest in artificial intelligence-driven analysis of medical images has seen a steep increase in recent years. Thus, our paper aims to promote and facilitate the use of this state-of-the-art technology to fellow researchers and clinicians. NEW METHOD: We present custom deep learning models generated in DeePathology™ STUDIO without the need for background knowledge in deep learning and computer science underlined by practical suggestions. RESULTS: We describe the general workflow in this commercially available software and present three real-world examples how to detect microglia on IBA1-stained mouse brain sections including their differences, validation results and analysis of a sample slide. COMPARISON WITH EXISTING METHODS: Deep-learning assisted analysis of histological images is faster than classical analysis methods, and offers a wide variety of detection possibilities that are not available using methods based on staining intensity. CONCLUSIONS: Reduced researcher bias, increased speed and extended possibilities make deep-learning assisted analysis of histological images superior to traditional analysis methods for histological images.


Subject(s)
Deep Learning , Animals , Artificial Intelligence , Brain , Image Processing, Computer-Assisted , Mice , Microglia
13.
Brain Res ; 1768: 147579, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34233173

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is the leading cause of dementia and a major global health issue. Currently, only limited treatment options are available to patients. One possibility to expand the treatment repertoire is repurposing of existing drugs such as dimethyl fumarate (DMF). DMF is approved for treatment of multiple sclerosis and previous animal studies have suggested that DMF may also have a beneficial effect for the treatment of AD. METHODS: We used an APPPS1 transgenic model of senile ß-amyloidosis and treated female mice orally with DMF in two treatment paradigms (pre and post onset). We quantified learning and memory parameters, ß-amyloidosis, and neuroinflammation to determine the potential of DMF as AD therapeutics. RESULTS: Treatment with DMF had no influence on water maze performance, ß-amyloid accumulation, plaque formation, microglia activation, and recruitment of immune cells to the brain. Compared to vehicle-treated animals, oral DMF treatment could not halt or retard disease progression in the mice. DISCUSSION: Our results do not favour the use of DMF as treatment for AD. While our results stand in contrast to previous findings in other models, they emphasize the importance of animal model selection and suggest further studies to elucidate the mechanisms leading to conflicting results.


Subject(s)
Amyloidosis/drug therapy , Cognitive Dysfunction/drug therapy , Dimethyl Fumarate/pharmacology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/physiopathology , Animals , Brain/metabolism , Cognitive Dysfunction/physiopathology , Dimethyl Fumarate/metabolism , Disease Models, Animal , Female , Hippocampus/metabolism , Humans , Inflammation/drug therapy , Mice , Mice, Transgenic , Neuroinflammatory Diseases/drug therapy , Peptide Fragments/metabolism
14.
Article in English | MEDLINE | ID: mdl-34977908

ABSTRACT

Adenosine-triphosphate-(ATP)-binding cassette (ABC) transport proteins are ubiquitously present membrane-bound efflux pumps that distribute endo- and xenobiotics across intra- and intercellular barriers. Discovered over 40 years ago, ABC transporters have been identified as key players in various human diseases, such as multidrug-resistant cancer and atherosclerosis, but also neurodegenerative diseases, such as Alzheimer's disease (AD). Most prominent and well-studied are ABCB1, ABCC1, and ABCG2, not only due to their contribution to the multidrug resistance (MDR) phenotype in cancer, but also due to their contribution to AD. However, our understanding of other ABC transporters is limited, and most of the 49 human ABC transporters have been largely neglected as potential targets for novel small-molecule drugs. This is especially true for the ABCA subfamily, which contains several members known to play a role in AD initiation and progression. This review provides up-to-date information on the proposed functional background and pathological role of ABCA transporters in AD. We also provide an overview of small-molecules shown to interact with ABCA transporters as well as potential in silico, in vitro, and in vivo methodologies to gain novel templates for the development of innovative ABC transporter-targeting diagnostics and therapeutics.

15.
J Alzheimers Dis ; 79(2): 597-605, 2021.
Article in English | MEDLINE | ID: mdl-33337377

ABSTRACT

BACKGROUND: Detailed pathology analysis and morphological quantification is tedious and prone to errors. Automatic image analysis can help to increase objectivity and reduce time. Here, we present the evaluation of the DeePathology STUDIO™ for automatic analysis of histological whole-slide images using machine learning/artificial intelligence. OBJECTIVE: To evaluate and validate the use of DeePathology STUDIO for the analysis of histological slides at high resolution. METHODS: We compared the DeePathology STUDIO and our current standard method using macros in AxioVision for the analysis of amyloid-ß (Aß) plaques and microglia in APP-transgenic mice at different ages. We analyzed density variables and total time invested with each approach. In addition, we correlated Aß concentration in brain tissue measured by ELISA with the results of Aß staining analysis. RESULTS: DeePathology STUDIO showed a significant decrease of the time for establishing new analyses and the total analysis time by up to 90%. On the other hand, both approaches showed similar quantitative results in plaque and activated microglia density in the different experimental groups. DeePathology STUDIO showed higher sensitivity and accuracy for small-sized plaques. In addition, DeePathology STUDIO allowed the classification of plaques in diffuse- and dense-packed, which was not possible with our traditional analysis. CONCLUSION: DeePathology STUDIO substantially reduced the effort needed for a new analysis showing comparable quantitative results to the traditional approach. In addition, it allowed including different objects (categories) or cell types in a single analysis, which is not possible with conventional methods.


Subject(s)
Disease Models, Animal , Image Processing, Computer-Assisted , Machine Learning , Microglia/pathology , Plaque, Amyloid/pathology , Alzheimer Disease/pathology , Animals , Mice , Mice, Transgenic
16.
J Alzheimers Dis ; 77(3): 1209-1221, 2020.
Article in English | MEDLINE | ID: mdl-32831204

ABSTRACT

BACKGROUND: The recent failure of clinical trials to treat Alzheimer's disease (AD) indicates that the current approach of modifying disease is either wrong or is too late to be efficient. Mild cognitive impairment (MCI) denotes the phase between the preclinical phase and clinical overt dementia. AD mouse models that overexpress human amyloid-ß (Aß) are used to study disease pathogenesis and to conduct drug development/testing. However, there is no direct correlation between the Aß deposition, the age of onset, and the severity of cognitive dysfunction. OBJECTIVE: To detect and predict MCI when Aß plaques start to appear in the hippocampus of an AD mouse. METHODS: We trained wild-type and AD mice in a Morris water maze (WM) task with different inter-trial intervals (ITI) at 3 months of age and assessed their WM performance. Additionally, we used a classification algorithm to predict the genotype (APPtg versus wild-type) of an individual mouse from their respective WM data. RESULTS: MCI can be empirically detected using a short-ITI protocol. We show that the ITI modulates the spatial learning of AD mice without affecting the formation of spatial memory. Finally, a simple classification algorithm such as logistic regression on WM data can give an accurate prediction of the cognitive dysfunction of a specific mouse. CONCLUSION: MCI can be detected as well as predicted simultaneously with the onset of Aß deposition in the hippocampus in AD mouse model. The mild cognitive impairment prediction can be used for assessing the efficacy of a treatment.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/psychology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/psychology , Alzheimer Disease/pathology , Animals , Cognitive Dysfunction/pathology , Female , Forecasting , Humans , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Drugs R D ; 20(3): 197-207, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32696271

ABSTRACT

Fingolimod is an approved treatment for relapsing-remitting multiple sclerosis (MS), and its properties in different pathways have raised interest in therapy research for other neurodegenerative diseases. Fingolimod is an agonist of sphingosine-1-phosphate (S1P) receptors. Its main pharmacologic effect is immunomodulation by lymphocyte homing, thereby reducing the numbers of T and B cells in circulation. Because of the ubiquitous expression of S1P receptors, other effects have also been described. Here, we review preclinical experiments evaluating the effects of treatment with fingolimod in neurodegenerative diseases other than MS, such as Alzheimer's disease or epilepsy. Fingolimod has shown neuroprotective effects in different animal models of neurodegenerative diseases, summarized here, correlating with increased brain-derived neurotrophic factor and improved disease phenotype (cognition and/or motor abilities). As expected, treatment also induced reductions in different neuroinflammatory markers because of not only inhibition of lymphocytes but also direct effects on astrocytes and microglia. Furthermore, fingolimod treatment exhibited additional effects for specific neurodegenerative disorders, such as reduction of amyloid-ß production, and antiepileptogenic properties. The neuroprotective effects exerted by fingolimod in these preclinical studies are reviewed and support the translation of fingolimod into clinical trials as treatment in neurodegenerative diseases beyond neuroinflammatory conditions (MS).


Subject(s)
Fingolimod Hydrochloride/therapeutic use , Nervous System Diseases/drug therapy , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Alzheimer Disease/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Epilepsy/drug therapy , Humans , Lymphocytes/metabolism , Mice , Multiple Sclerosis/drug therapy , Neuroprotective Agents/therapeutic use , Rats
18.
Neurotherapeutics ; 17(3): 1228-1238, 2020 07.
Article in English | MEDLINE | ID: mdl-31970667

ABSTRACT

Epileptogenesis-associated brain inflammation might be a promising target to prevent or attenuate epileptogenesis. Positron emission tomography (PET) imaging targeting the translocator protein (TSPO) was applied here to quantify effects of different dosing regimens of the anti-inflammatory drug minocycline during the latent phase in two rodent models of epileptogenesis. After induction of epileptogenesis by status epilepticus (SE), rats were treated with minocycline for 7 days (25 or 50 mg/kg) and mice for 5 or 10 days (50 or 100 mg/kg). All animals were subjected to scans at 1 and 2 weeks post-SE. Radiotracer distribution was analyzed and statistical parametric mapping (SPM) was performed, as well as histological analysis of astroglial activation and neuronal cell loss. Atlas-based analysis of [18F]GE180 PET in rats revealed a dose-dependent regional decrease of TSPO expression at 2 weeks post-SE. Results of SPM analysis depicted a treatment effect already at 1 week post-SE in rats treated with the higher minocycline dose. In mice, TSPO PET imaging did not reveal any treatment effects whereas histology identified only a treatment-related reduction in dispersion of dentate gyrus neurons. TSPO PET served as an auspicious tool for temporal monitoring and quantification of anti-inflammatory effects during epileptogenesis. Importantly, the findings underline the need to applying more than one animal model to avoid missing treatment effects. For future studies, the setup is ready to be applied in combination with seizure monitoring to investigate the relationship between individual early treatment response and disease outcome.


Subject(s)
Anti-Inflammatory Agents/metabolism , Carrier Proteins/metabolism , Epilepsy/metabolism , Minocycline/metabolism , Positron-Emission Tomography/methods , Radiopharmaceuticals/metabolism , Receptors, GABA-A/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Epilepsy/diagnostic imaging , Epilepsy/drug therapy , Female , Male , Mice , Minocycline/therapeutic use , Rats , Rats, Sprague-Dawley , Treatment Outcome
19.
J Cereb Blood Flow Metab ; 40(1): 204-213, 2020 01.
Article in English | MEDLINE | ID: mdl-30375913

ABSTRACT

Alterations in metabolism during epileptogenesis may be a therapy target. Recently, an increase in amino acid transport into the brain was proposed to play a role in epileptogenesis. We aimed to characterize alterations of substrate utilization during epileptogenesis and in chronic epilepsy. The lithium-pilocarpine post status epilepticus (SE) rat model was used. We performed longitudinal O-(2-[(18)F]fluoroethyl)-l-tyrosine (18F-FET) and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) and calculated 18F-FET volume of distribution (Vt) and 18F-FDG uptake. Correlation analyses were performed with translocator protein-PET defined neuroinflammation from previously acquired data. We found reduced 18F-FET Vt at 48 h after SE (amygdala: -30.2%, p = 0.014), whereas 18F-FDG showed increased glucose uptake 4 and 24 h after SE (hippocampus: + 43.6% and +42.5%, respectively; p < 0.001) returning to baseline levels thereafter. In chronic epileptic animals, we found a reduction in 18F-FET and 18F-FDG in the hippocampus. No correlation was found for 18F-FET or 18F-FDG to microglial activation at seven days post SE. Whereas metabolic alterations do not reflect higher metabolism associated to activated microglia, they might be partially driven by chronic neuronal loss. However, both metabolisms diverge during early epileptogenesis, pointing to amino acid turnover as a possible biomarker and/or therapeutic target for epileptogenesis.


Subject(s)
Brain Diseases, Metabolic/diagnostic imaging , Brain/metabolism , Epilepsy/metabolism , Positron-Emission Tomography/methods , Amino Acids/pharmacokinetics , Amygdala/diagnostic imaging , Amygdala/metabolism , Animals , Brain Diseases, Metabolic/etiology , Brain Diseases, Metabolic/metabolism , Chronic Disease , Disease Models, Animal , Fluorine Radioisotopes , Fluorodeoxyglucose F18 , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Rats , Substrate Specificity
20.
Neurobiol Dis ; 134: 104664, 2020 02.
Article in English | MEDLINE | ID: mdl-31678583

ABSTRACT

Epilepsy is a complex network phenomenon that, as yet, cannot be prevented or cured. We recently proposed network-based approaches to prevent epileptogenesis. For proof of concept we combined two drugs (levetiracetam and topiramate) for which in silico analysis of drug-protein interaction networks indicated a synergistic effect on a large functional network of epilepsy-relevant proteins. Using the intrahippocampal kainate mouse model of temporal lobe epilepsy, the drug combination was administered during the latent period before onset of spontaneous recurrent seizures (SRS). When SRS were periodically recorded by video-EEG monitoring after termination of treatment, a significant decrease in incidence and frequency of SRS was determined, indicating antiepileptogenic efficacy. Such efficacy was not observed following single drug treatment. Furthermore, a combination of levetiracetam and phenobarbital, for which in silico analysis of drug-protein interaction networks did not indicate any significant drug-drug interaction, was not effective to modify development of epilepsy. Surprisingly, the promising antiepileptogenic effect of the levetiracetam/topiramate combination was obtained in the absence of any significant neuroprotective or anti-inflammatory effects as indicated by multimodal brain imaging and histopathology. High throughput RNA-sequencing (RNA-seq) of the ipsilateral hippocampus of mice treated with the levetiracetam/topiramate combination showed that several genes that have been linked previously to epileptogenesis, were significantly differentially expressed, providing interesting entry points for future mechanistic studies. Overall, we have discovered a novel combination treatment with promise for prevention of epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Brain/drug effects , Drug Therapy, Combination/methods , Epilepsy, Temporal Lobe , Protein Interaction Mapping/methods , Animals , Levetiracetam/pharmacology , Male , Mice , Proof of Concept Study , Topiramate/pharmacology , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...