Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
FEBS J ; 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37921063

ABSTRACT

Vaspin is a serine protease inhibitor that protects against adipose tissue inflammation and insulin resistance, two key drivers of adipocyte dysfunction and metabolic disorders in obesity. Inhibition of target proteases such as KLK7 has been shown to reduce adipose tissue inflammation in obesity, while vaspin binding to cell surface GRP78 has been linked to reduced obesity-induced ER stress and insulin resistance in the liver. However, the molecular mechanisms by which vaspin directly affects cellular processes in adipocytes remain unknown. Using fluorescently labeled vaspin, we found that vaspin is rapidly internalized by mouse and human adipocytes, but less efficiently by endothelial, kidney, liver, and neuronal cells. Internalization occurs by active, clathrin-mediated endocytosis, which is dependent on vaspin binding to the LRP1 receptor, rather than GRP78 as previously thought. This was demonstrated by competition experiments and RNAi-mediated knock-down in adipocytes and by rescuing vaspin internalization in LRP1-deficient Pea13 cells after transfection with a functional LRP1 minireceptor. Vaspin internalization is further increased in mature adipocytes after insulin-stimulated translocation of LRP1. Although vaspin has nanomolar affinity for LRP1 clusters II-IV, binding to cell surface heparan sulfates is required for efficient LRP1-mediated internalization. Native, but not cleaved vaspin, and also vaspin polymers are efficiently endocytosed, and ultimately targeted for lysosomal degradation. Our study provides mechanistic insight into the uptake and degradation of vaspin in adipocytes, thereby broadening our understanding of its functional repertoire. We hypothesize the vaspin-LRP1 axis to be an important mediator of vaspin effects not only in adipose tissue but also in other LRP1-expressing cells.

2.
Int J Mol Sci ; 24(15)2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37569268

ABSTRACT

Along with initiatives to understand the pathophysiology of stroke in detail and to identify neuroprotective targets, cell-stabilizing elements have gained increasing attention. Although cell culture experiments have indicated that tricellulin, α-catenin and microfibrillar-associated protein 5 (MFAP5) contribute to cellular integrity, these elements have not yet been investigated in the ischemic brain. Applying immunofluorescence labeling, this study explored tricellulin, MFAP5 and α-catenin in non-ischemic and ischemic brain areas of mice (24, 4 h of ischemia) and rats (4 h of ischemia), along with collagen IV and fibronectin as vascular and extracellular matrix constituents and microtubule-associated protein 2 (MAP2) and neurofilament light chain (NF-L) as cytoskeletal elements. Immunosignals of tricellulin and notably MFAP5 partially appeared in a fiber-like pattern, and α-catenin appeared more in a dotted pattern. Regional associations with vascular and extracellular constituents were found for tricellulin and α-catenin, particularly in ischemic areas. Due to ischemia, signals of tricellulin, MFAP5 and α-catenin decreased concomitantly with MAP2 and NF-L, whereby MFAP5 provided the most sensitive reaction. For the first time, this study demonstrated ischemia-related alterations in tricellulin, MFAP5 and α-catenin along with the vasculature, extracellular matrix and cytoskeleton. Confirmatory studies are needed, also exploring their role in cellular integrity and the potential for neuroprotective approaches in stroke.


Subject(s)
Brain Ischemia , Stroke , Animals , Mice , Rats , alpha Catenin , Brain Ischemia/metabolism , Cerebral Infarction , Cytoskeleton/metabolism , Ischemia , MARVEL Domain Containing 2 Protein , Stroke/metabolism , Intercellular Signaling Peptides and Proteins , Contractile Proteins
3.
Sci Rep ; 11(1): 23767, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34887476

ABSTRACT

Fibril formation of amyloid ß (Aß) peptides is one of the key molecular events connected to Alzheimer's disease. The pathway of formation and mechanism of action of Aß aggregates in biological systems is still object of very active research. To this end, systematic modifications of the Phe19-Leu34 hydrophobic contact, which has been reported in almost all structural studies of Aß40 fibrils, helps understanding Aß folding pathways and the underlying free energy landscape of the amyloid formation process. In our approach, a series of Aß40 peptide variants with two types of backbone modifications, namely incorporation of (i) a methylene or an ethylene spacer group and (ii) a N-methylation at the amide functional group, of the amino acids at positions 19 or 34 was applied. These mutations are expected to challenge the inter-ß-strand side chain contacts as well as intermolecular backbone ß-sheet hydrogen bridges. Using a multitude of biophysical methods, it is shown that these backbone modifications lead, in most of the cases, to alterations in the fibril formation kinetics, a higher local structural heterogeneity, and a somewhat modified fibril morphology without generally impairing the fibril formation capacity of the peptides. The toxicological profile found for the variants depend on the type and extent of the modification.


Subject(s)
Amino Acids/metabolism , Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , Amyloid/chemistry , Amyloid/genetics , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Spectrum Analysis
4.
J Lipid Res ; 62: 100105, 2021.
Article in English | MEDLINE | ID: mdl-34390703

ABSTRACT

The leptin receptor (Lepr) pathway is important for food intake regulation, energy expenditure, and body weight. Mutations in leptin and the Lepr have been shown to cause early-onset severe obesity in mice and humans. In studies with C57BL/6NCrl mice, we found a mouse with extreme obesity. To identify a putative spontaneous new form of monogenic obesity, we performed backcross studies with this mouse followed by a quantitative trait locus (QTL) analysis and sequencing of the selected chromosomal QTL region. We thereby identified a novel Lepr mutation (C57BL/6N-LeprL536Hfs*6-1NKB), which is located at chromosome 4, exon 11 within the CRH2-leptin-binding site. Compared with C57BL/6N mice, LeprL536Hfs*6 develop early onset obesity and their body weight exceeds that of Leprdb/db mice at an age of 30 weeks. Similar to Leprdb/db mice, the LeprL536Hfs*6 model is characterized by hyperphagia, obesity, lower energy expenditure and activity, hyperglycemia, and hyperinsulinemia compared with C57BL/6N mice. Crossing Leprdb/wt with LeprL536Hfs*6/wt mice results in compound heterozygous LeprL536Hfs*6/db mice, which develop even higher body weight and fat mass than both homozygous Leprdb/db and LeprL536Hfs*6 mice. Compound heterozygous Lepr deficiency affecting functionally different regions of the Lepr causes more severe obesity than the parental homozygous mutations.


Subject(s)
Obesity/genetics , Receptors, Leptin/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Mutation
5.
J Alzheimers Dis ; 81(2): 797-808, 2021.
Article in English | MEDLINE | ID: mdl-33843677

ABSTRACT

BACKGROUND: Circadian rhythm disturbance is commonly observed in Alzheimer's disease (AD). In mammals, these rhythms are orchestrated by the superchiasmatic nucleus (SCN). Our previous study in the Tg2576 AD mouse model suggests that inflammatory responses, most likely manifested by low GABA production, may be one of the underlying perpetrators for the changes in circadian rhythmicity and sleep disturbance in AD. However, the mechanistic connections between SCN dysfunction, GABA modulation, and inflammation in AD is not fully understood. OBJECTIVE: To reveal influences of amyloid pathology in Tg2576 mouse brain on metabolism in SCN and to identify key metabolic sensors that couple SCN dysfunction with GABA modulation and inflammation. METHODS: High resolution magic angle spinning (HR-MAS) NMR in conjunction with multivariate analysis was applied for metabolic profiling in SCN of control and Tg2576 female mice. Immunohistochemical analysis was used to detect neurons, astrocytes, expression of GABA transporter 1 (GAT1) and Bmal1. RESULTS: Metabolic profiling revealed significant metabolic deficits in SCN of Tg2576 mice. Reductions in glucose, glutamate, GABA, and glutamine provide hints toward an impaired GABAergic glucose oxidation and neurotransmitter cycling in SCN of AD mice. In addition, decreased redox co-factor NADPH and glutathione support a redox disbalance. Immunohistochemical examinations showed low expression of the core clock protein, Bmal1, especially in activated astrocytes. Moreover, decreased expression of GAT1 in astrocytes indicates low GABA recycling in this cell type. CONCLUSION: Our results suggest that redox disbalance and compromised GABA signaling are important denominators and connectors between neuroinflammation and clock dysfunction in AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Circadian Rhythm/physiology , Suprachiasmatic Nucleus/metabolism , Amyloidogenic Proteins/metabolism , Animals , Astrocytes/metabolism , Disease Models, Animal , Magnetic Resonance Imaging/methods , Mice , Neurons/metabolism , Suprachiasmatic Nucleus/pathology
6.
Eur J Neurosci ; 53(12): 4034-4050, 2021 06.
Article in English | MEDLINE | ID: mdl-32954591

ABSTRACT

Neurodegenerative disorders are characterised by the activation of brain-resident microglia cells and by the infiltration of peripheral T cells. However, their interplay in disease has not been clarified yet. It is difficult to investigate complex cellular dynamics in living animals, and simple two-dimensional (2D) cell culture models do not resemble the soft 3D structure of brain tissue. Therefore, we developed a biomimetic 3D in vitro culture system for co-cultivation of microglia and T cells. As the activation and/or migration of immune cells in the brain might be affected by components of the extracellular matrix, defined 3D fibrillar collagen I-based matrices were constructed and modified with hyaluronan and/or chondroitin sulphate, resembling aspects of brain extracellular matrix. Murine microglia and spleen-derived T cells were cultured alone or in co-culture on the constructed matrices. Microglia exhibited in vivo-like morphology and T cells showed enhanced survival when co-cultured with microglia or to a minor degree in the presence of glia-conditioned medium. The open and porous fibrillar structure of the matrix allowed for cell invasion and direct cell-cell interaction, with stronger invasion of T cells. Both cell types showed no dependence on the matrix modifications. Microglia could be activated on the matrices by lipopolysaccharide resulting in interleukin-6 and tumour necrosis factor-α secretion. The findings herein indicate that biomimetic 3D matrices allow for co-cultivation and activation of primary microglia and T cells and provide useful tools to study their interaction in vitro.


Subject(s)
Microglia , T-Lymphocytes , Animals , Brain , Cells, Cultured , Coculture Techniques , Extracellular Matrix , Mice
7.
ACS Chem Neurosci ; 11(7): 1038-1047, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32141731

ABSTRACT

The nonproteinogenic amino acid ß-methylamino alarelevant example for environmental hazards are nonnine (BMAA) is a neurotoxin and represents a potential risk factor for neurodegenerative diseases. Despite intense research over the last years, the pathological mechanism of BMAA is still unclear. One of the main open questions is whether BMAA can be misincorporated into proteins, especially as a substitute for serine, and whether this has structural and functional consequences for the afflicted proteins leading to early onset neurodegeneration. In this study, we hypothesize that BMAA was indeed incorporated into Aß40 molecules and study the structural and dynamical consequences of such misincorporation along with the effect such mutated Aß40 peptides have on neuronal cells. We used the synthetic ß-amyloid peptide (Aß40), a known key player in the development of Alzheimer's disease, to incorporate BMAA substitutions at three different positions in the peptide sequence: Ser8BMAA at the peptide's N-terminus, Phe19BMAA in the hydrophobic core region, and S26BMAA in the flexible turn region of Aß40 fibrils. We performed a set of biophysical experiments including fluorescence, circular dichroism, solid-state NMR spectroscopy, transmission electron microscopy, and X-ray diffraction to investigate structural and functional aspects of the mutated peptides compared to wildtype Aß40. All variants showed high structural tolerance to BMAA misincorporation. In contrast, the cellular response and neuronal survival were affected in a mutation site-specific manner. As a consequence, we can state from the physicochemical point of view that, if BMAA was misincorporated into proteins, it could indeed represent a risk factor that could potentially play a role in neurodegeneration. Further research addressing the role of BMAA, especially its protein-associated form, should be performed to obtain a better understanding of neurodegenerative diseases and to develop new therapeutic strategies.


Subject(s)
Amino Acids, Diamino/toxicity , Amyloid beta-Peptides/metabolism , Amyloid/drug effects , Neurotoxins/toxicity , Alzheimer Disease/metabolism , Amino Acid Sequence/physiology , Amyloid/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/pharmacology , Cell Survival/drug effects , Cytoskeleton/metabolism , Humans
8.
Acta Neuropathol Commun ; 7(1): 79, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31109380

ABSTRACT

Pathogenic variants of the huntingtin (HTT) protein and their aggregation have been investigated in great detail in brains of Huntington's disease patients and HTT-transgenic animals. However, little is known about the physiological brain region- and cell type-specific HTT expression pattern in wild type mice and a potential recruitment of endogenous HTT to other pathogenic protein aggregates such as amyloid plaques in cross seeding events. Employing a monoclonal anti-HTT antibody directed against the HTT mid-region and using brain tissue of three different mouse strains, we detected prominent immunoreactivity in a number of brain areas, particularly in cholinergic cranial nerve nuclei, while ubiquitous neuronal staining appeared faint. The region-specific distribution of endogenous HTT was found to be comparable in wild type rat and hamster brain. In human amyloid precursor protein transgenic Tg2576 mice with amyloid plaque pathology, similar neuronal HTT expression patterns and a distinct association of HTT with Abeta plaques were revealed by immunohistochemical double labelling. Additionally, the localization of HTT in reactive astrocytes was demonstrated for the first time in a transgenic Alzheimer's disease animal model. Both, plaque association of HTT and occurrence in astrocytes appeared to be age-dependent. Astrocytic HTT gene and protein expression was confirmed in primary cultures by RT-qPCR and by immunocytochemistry. We provide the first detailed analysis of physiological HTT expression in rodent brain and, under pathological conditions, demonstrate HTT aggregation in proximity to Abeta plaques and Abeta-induced astrocytic expression of endogenous HTT in Tg2576 mice.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Cranial Nerves/metabolism , Huntingtin Protein/metabolism , Plaque, Amyloid/metabolism , Animals , Brain/metabolism , Cricetinae , Disease Models, Animal , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/metabolism , Protein Aggregation, Pathological , Rats, Wistar
9.
J Alzheimers Dis ; 69(2): 363-375, 2019.
Article in English | MEDLINE | ID: mdl-30958376

ABSTRACT

In Alzheimer's disease (AD), disturbances in the circadian rhythm and sleep-wake cycle are frequently observed. Both are controlled by the master clock: the suprachiasmatic nucleus (SCN), which was reported in postmortem studies of AD subjects to be compromised. However, the influence of age and gender on the biophysical integrity and subtle microstructural changes of SCN and mechanistic connections between SCN dysfunction and AD progression in vivo remain to be explored. In the present study, we utilized state-of-the-art in vivo magnetic resonance relaxation measurements in combination with immunohistochemistry to follow microstructural changes in SCN of the Tg2576 mouse model of AD. Longitudinal monitoring of in vivo T2 relaxation with age shows significant shortening of T2 values in the SCN of transgenic mice and more substantially in female transgenic than aged-matched controls. Multiexponential T2 analysis detected a unique long T2 component in SCN of transgenic mice which was absent in wild-type mice. Immunohistochemical examination revealed significantly elevated numbers of activated astrocytes and an increase in the astrocyte to neuron ratio in SCN of transgenic compared to wild-type mice. This increase was more substantial in female than in male transgenic mice. In addition, low GABA production in SCN of transgenic mice was detected. Our results offer a brief appraisal of SCN dysfunction in AD and demonstrate that inflammatory responses may be an underlying perpetrator for the changes in circadian rhythmicity and sleep disturbance in AD and could also be at the root of marked sex disparities observed in AD subjects.


Subject(s)
Alzheimer Disease/diagnostic imaging , Disease Models, Animal , Magnetic Resonance Imaging/methods , Suprachiasmatic Nucleus/chemistry , Suprachiasmatic Nucleus/diagnostic imaging , Alzheimer Disease/pathology , Animals , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Suprachiasmatic Nucleus/pathology
10.
Front Neurosci ; 13: 137, 2019.
Article in English | MEDLINE | ID: mdl-30853883

ABSTRACT

Amyloid precursor protein (APP) transgenic animal models of Alzheimer's disease have become versatile tools for basic and translational research. However, there is great heterogeneity of histological, biochemical, and functional data between transgenic mouse lines, which might be due to different transgene expression patterns. Here, the expression of human APP (hAPP) by GABAergic hippocampal interneurons immunoreactive for the calcium binding proteins parvalbumin, calbindin, calretinin, and for the peptide hormone somatostatin was analyzed in Tg2576 mice by double immunofluorescent microscopy. Overall, there was no GABAergic interneuron subpopulation that did not express the transgene. On the other hand, in no case all neurons of such a subpopulation expressed hAPP. In dentate gyrus molecular layer and in stratum lacunosum moleculare less than 10% of hAPP-positive interneurons co-express any of these interneuron markers, whereas in stratum oriens hAPP-expressing neurons frequently co-express these interneuron markers to different proportions. We conclude that these neurons differentially contribute to deficits in young Tg2576 mice before the onset of Abeta plaque pathology. The detailed analysis of distinct brain region and neuron type-specific APP transgene expression patterns is indispensable to understand particular pathological features and mouse line-specific differences in neuronal and systemic functions.

11.
Glia ; 67(2): 393-403, 2019 02.
Article in English | MEDLINE | ID: mdl-30485540

ABSTRACT

Transgenic Tg2576 mice expressing human amyloid precursor protein (hAPP) with the Swedish mutation are among the most frequently used animal models to study the amyloid pathology related to Alzheimer's disease (AD). The transgene expression in this model is considered to be neuron-specific. Using a novel hAPP-specific antibody in combination with cell type-specific markers for double immunofluorescent labelings and laser scanning microscopy, we here report that-in addition to neurons throughout the brain-astrocytes in the corpus callosum and to a lesser extent in neocortex express hAPP. This astrocytic hAPP expression is already detectable in young Tg2576 mice before the onset of amyloid pathology and still present in aged Tg2576 mice with robust amyloid pathology in neocortex, hippocampus, and corpus callosum. Surprisingly, hAPP immunoreactivity in cortex is restricted to resting astrocytes distant from amyloid plaques but absent from reactive astrocytes in close proximity to amyloid plaques. In contrast, neither microglial cells nor oligodendrocytes of young or aged Tg2576 mice display hAPP labeling. The astrocytic expression of hAPP is substantiated by the analyses of hAPP mRNA and protein expression in primary cultures derived from Tg2576 offspring. We conclude that astrocytes, in particular in corpus callosum, may contribute to amyloid pathology in Tg2576 mice and thus mimic this aspect of AD pathology.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Brain/pathology , Age Factors , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutathione S-Transferase pi/metabolism , Humans , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscle Proteins , Neurons/metabolism , Neurons/pathology , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
12.
Molecules ; 23(4)2018 04 17.
Article in English | MEDLINE | ID: mdl-29673150

ABSTRACT

Oligomeric assemblies of neurotoxic amyloid beta (Abeta) peptides generated by proteolytical processing of the amyloid precursor protein (APP) play a key role in the pathogenesis of Alzheimer's disease (AD). In recent years, a substantial heterogeneity of Abeta peptides with distinct biophysical and cell biological properties has been demonstrated. Among these, a particularly neurotoxic and disease-specific Abeta variant is N-terminally truncated and modified to pyroglutamate (pE-Abeta). Cell biological and animal experimental studies imply the catalysis of this modification by the enzyme glutaminyl cyclase (QC). However, direct histopathological evidence in transgenic animals from comparative brain region and cell type-specific expression of transgenic hAPP and QC, on the one hand, and on the formation of pE-Abeta aggregates, on the other, is lacking. Here, using single light microscopic, as well as triple immunofluorescent, labeling, we report the deposition of pE-Abeta only in the brain regions of APP-transgenic Tg2576 mice with detectable human APP and endogenous QC expression, such as the hippocampus, piriform cortex, and amygdala. Brain regions showing human APP expression without the concomitant presence of QC (the anterodorsal thalamic nucleus and perifornical nucleus) do not display pE-Abeta plaque formation. However, we also identified brain regions with substantial expression of human APP and QC in the absence of pE-Abeta deposition (the Edinger-Westphal nucleus and locus coeruleus). In these brain regions, the enzymes required to generate N-truncated Abeta peptides as substrates for QC might be lacking. Our observations provide additional evidence for an involvement of QC in AD pathogenesis via QC-catalyzed pE-Abeta formation.


Subject(s)
Alzheimer Disease/metabolism , Aminoacyltransferases/metabolism , Amyloid beta-Peptides/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Alzheimer Disease/genetics , Aminoacyltransferases/genetics , Amyloid beta-Peptides/genetics , Animals , Goats , Humans , Immunohistochemistry , Mice , Models, Animal , Rats
13.
Neurobiol Aging ; 62: 168-179, 2018 02.
Article in English | MEDLINE | ID: mdl-29154037

ABSTRACT

Age and sex are risk factors of Alzheimer's disease (AD). Among the neurotransmitter systems, gamma-aminobutyric acid (GABA) has been implicated in AD pathogenesis but the relevance of sex-specific GABAergic dysfunction during AD progression remains unknown. In the present study, we utilized state-of-the-art high-resolution magic angle spinning nuclear magnetic resonance to systematically monitor the brain region-, age-, and sex-specific modulation of GABA levels in wild-type and Tg2576 mice with amyloid pathology. In addition, we followed the possible role of reactive astrocytes in sex-specific GABA modulation. In female Tg2576 mice, hippocampal GABA levels were significantly elevated, along with higher number of reactive astrocytes and amyloid deposition. The elevated GABA was found to be produced via the monoamine oxidase-B route from putrescine in reactive astrocytes, more substantially in female than male mice, thus suggesting a role of astrocytes in memory impairment and sex-related differences in AD. Our results paint a coherent model of memory impairment in AD and signify that dynamic changes in regional GABA may be at the root of marked sex disparities observed in AD.


Subject(s)
Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Brain/metabolism , Sex Characteristics , gamma-Aminobutyric Acid/metabolism , Alzheimer Disease/pathology , Amyloidogenic Proteins/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/physiology , Disease Models, Animal , Female , Longitudinal Studies , Magnetic Resonance Spectroscopy/methods , Male , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/pathology , Mice, Transgenic , Monoamine Oxidase/metabolism , Putrescine/metabolism , Risk Factors
14.
Chemistry ; 23(62): 15834-15838, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-28857302

ABSTRACT

The morphology, structure, and dynamics of mature amyloid ß (Aß) fibrils formed by the Aß variant, which is truncated at residue 11 and chemically modified by enzymatic pyroglutamate formation (pGlu11 -Aß(11-40)), was studied along with the investigation of the toxicity of these Aß variants to neurons and astrocytes. The fibrils of pGlu11 -Aß (11-40) were more toxic than wildtype Aß (1-40) and the longer pGlu3-Aß (3-40) especially at higher concentration, whereas the overall morphology was quite similar. The secondary structure of pGlu11 -Aß (11-40) fibrils shows the typical two ß-strands connected by a short turn as known for mature fibrils of Aß (1-40) and also pGlu3 -Aß (3-40). Further insights into tertiary contacts exhibit some similarities of pGlu11 -Aß (11-40) fibrils with wildtype Aß (1-40), but also a so far not described contact between Gly25 and Ile31 . This highlights the biological importance of chemical modifications on the molecular structure of Aß.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Pyrrolidonecarboxylic Acid/chemistry , Amyloid beta-Peptides/toxicity , Animals , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Kinetics , Mice , Mice, Inbred C57BL , Microscopy, Electron , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/toxicity , X-Ray Diffraction
15.
Aging Cell ; 15(5): 953-63, 2016 10.
Article in English | MEDLINE | ID: mdl-27470171

ABSTRACT

Alzheimer's disease (AD) is histopathologically characterized by neurodegeneration, the formation of intracellular neurofibrillary tangles and extracellular Aß deposits that derive from proteolytic processing of the amyloid precursor protein (APP). As rodents do not normally develop Aß pathology, various transgenic animal models of AD were designed to overexpress human APP with mutations favouring its amyloidogenic processing. However, these mouse models display tremendous differences in the spatial and temporal appearance of Aß deposits, synaptic dysfunction, neurodegeneration and the manifestation of learning deficits which may be caused by age-related and brain region-specific differences in APP transgene levels. Consequentially, a comparative temporal and regional analysis of the pathological effects of Aß in mouse brains is difficult complicating the validation of therapeutic AD treatment strategies in different mouse models. To date, no antibodies are available that properly discriminate endogenous rodent and transgenic human APP in brains of APP-transgenic animals. Here, we developed and characterized rat monoclonal antibodies by immunohistochemistry and Western blot that detect human but not murine APP in brains of three APP-transgenic mouse and one APP-transgenic rat model. We observed remarkable differences in expression levels and brain region-specific expression of human APP among the investigated transgenic mouse lines. This may explain the differences between APP-transgenic models mentioned above. Furthermore, we provide compelling evidence that our new antibodies specifically detect endogenous human APP in immunocytochemistry, FACS and immunoprecipitation. Hence, we propose these antibodies as standard tool for monitoring expression of endogenous or transfected APP in human cells and APP expression in transgenic animals.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/immunology , Amyloid beta-Protein Precursor/immunology , Antibodies/immunology , Antibody Specificity/immunology , Gene Expression , Transgenes/genetics , Animals , Disease Models, Animal , Humans , Immunohistochemistry , Mice, Inbred C57BL , Mice, Transgenic , Rats , Reproducibility of Results
16.
Alzheimers Dement ; 12(9): 964-976, 2016 09.
Article in English | MEDLINE | ID: mdl-27107518

ABSTRACT

INTRODUCTION: In Alzheimer's disease (AD), pathologic amyloid-beta (Aß) is synaptotoxic and impairs neuronal function at the microscale, influencing brain networks at the macroscale before Aß deposition. The latter can be detected noninvasively, in vivo, using resting-state functional MRI (rsfMRI), a technique used to assess brain functional connectivity (FC). METHODS: RsfMRI was performed longitudinally in TG2576 and PDAPP mice, starting before Aß deposition to determine the earliest FC changes. Additionally, the role of pathologic Aß on early FC alterations was investigated by treating TG2576 mice with the 3D6 anti-Aß-antibody. RESULTS: Both transgenic models showed hypersynchronized FC before Aß deposition and hyposynchronized FC at later stages. Early anti-Aß treatment in TG2576 mice prevented hypersynchronous FC and the associated synaptic impairments and excitatory/inhibitory disbalances. DISCUSSION: Hypersynchrony of FC may be used as a new noninvasive read out of early AD and can be recovered by anti-Aß treatment, encouraging preventive treatment strategies in familial AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/physiopathology , Alzheimer Disease/diagnostic imaging , Animals , Autoantibodies/pharmacology , Brain/diagnostic imaging , Brain Mapping , Cerebrovascular Circulation/physiology , Cortical Synchronization/physiology , Disease Models, Animal , Disease Progression , Longitudinal Studies , Magnetic Resonance Imaging , Mice, Transgenic , Neural Pathways/diagnostic imaging , Neural Pathways/physiopathology , Neuroprotective Agents/pharmacology , Oxygen/blood , Plaque, Amyloid/diagnostic imaging , Plaque, Amyloid/physiopathology , Plaque, Amyloid/prevention & control , Prodromal Symptoms , Rest
17.
Eur Neuropsychopharmacol ; 26(6): 1048-61, 2016 06.
Article in English | MEDLINE | ID: mdl-26996375

ABSTRACT

Prolyl oligopeptidase (PREP) has been implicated in neurodegeneration and neuroinflammation and has been considered a drug target to enhance memory in dementia. However, the true physiological role of PREP is not yet understood. In this paper, we report the phenotyping of a mouse line where the PREP gene has been knocked out. This work indicates that the lack of PREP in mice causes reduced anxiety but also hyperactivity. The cortical volumes of PREP knockout mice were smaller than those of wild type littermates. Additionally, we found increased expression of diazepam binding inhibitor protein in the cortex and of the somatostatin receptor-2 in the hippocampus of PREP knockout mice. Furthermore, immunohistochemistry and tail suspension test revealed lack of response of PREP knockout mice to lipopolysaccharide insult. Further analysis revealed significantly increased levels of polysialylated-neural cell adhesion molecule in PREP deficient mice. These findings might be explained as possible alteration in brain plasticity caused by PREP deficiency, which in turn affect behaviour and brain development.


Subject(s)
Anxiety/genetics , Anxiety/psychology , Behavior, Animal , Neuronal Plasticity/genetics , Serine Endopeptidases/deficiency , Synapses/genetics , Animals , Anxiety/pathology , Body Weight/genetics , Brain/pathology , Cytokines/blood , Hindlimb Suspension , Hyperkinesis/genetics , Hyperkinesis/psychology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Phenotype , Prolyl Oligopeptidases , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/genetics
18.
Eur J Immunol ; 45(12): 3339-50, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26381487

ABSTRACT

Antiretroviral therapy (ART) has yielded major advances in fighting the HIV pandemic by restoring protective immunity. However, a significant proportion of HIV patients co-infected with the opportunistic fungal pathogen Cryptococcus neoformans paradoxically develops a life-threatening immune reconstitution inflammatory syndrome (IRIS) during antiretroviral therapy. Despite several clinical studies, the underlying pathomecha-nisms are poorly understood. Here, we present the first mouse model of cryptococcal IRIS that allows for a detailed analysis of disease development. Lymphocyte-deficient RAG-1(-/-) mice are infected with C. neoformans and 4 weeks later adoptively transferred with purified CD4(+) T cells. Reconstitution of CD4(+) T cells is sufficient to induce a severe inflammatory disease similar to clinical IRIS in C. neoformans-infected RAG-1(-/-) mice of different genetic backgrounds and immunological phenotypes (i.e. C57BL/6 and BALB/c). Multiorgan inflammation is accompanied by a systemic release of distinct proinflammatory cytokines, i.e. IFN-γ, IL-6, and TNF-α. IRIS development is characterized by infection-dependent activation of donor CD4(+) T cells, which are the source of IFN-γ. Interestingly, IFN-γ-mediated effects are not required for disease induction. Taken together, this novel mouse model of cryptococcal IRIS provides a useful tool to verify potential mechanisms of pathogenesis, revealing targets for diagnosis and therapeutic interventions.


Subject(s)
Cryptococcosis/complications , Cryptococcus neoformans , Immune Reconstitution Inflammatory Syndrome/etiology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Disease Models, Animal , Homeodomain Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
19.
J Alzheimers Dis ; 48(4): 1083-94, 2015.
Article in English | MEDLINE | ID: mdl-26402099

ABSTRACT

The pyruvate-derived acetyl-CoA is a principal direct precursor substrate for bulk energy synthesis in the brain. Deficits of pyruvate dehydrogenase in the neocortex are common features of Alzheimer's disease and other age-related encephalopathies in humans. Therefore, amyloid-ß overload in brains of diverse transgenic Alzheimer's disease model animals was investigated as one of neurotoxic compounds responsible for pyruvate dehydrogenase inhibition yielding deficits of cholinergic neurotransmission and cognitive functions. Brains of aged, 14-16-month-old Tg2576 mice contained 0.6 µmol/kg levels of amyloid-ß1 - 42. Activities of pyruvate dehydrogenase complex, choline acetyltransferase, and several enzymes of acetyl-CoA and energy metabolism were found to be unchanged in both forebrain mitochondria and synaptosomes of Tg2576 mice, indicating preservation of structural integrity at least in cholinergic neuronal cells. However, in transgenic brain synaptosomes, pyruvate utilization, mitochondrial levels, and cytoplasmic acetyl-CoA levels, as well as acetylcholine content and its quantal release, were all found to be decreased by 25-40% . On the contrary, activation of pyruvate utilization was detected and no alterations in acetyl-CoA content and citrate or α-ketoglutarate accumulation were observed in transgenic whole brain mitochondria. These data indicate that amyloid-ß evoked deficits in acetyl-CoA are confined to mitochondrial and cytoplasmic compartments of Tg2576 nerve terminals, becoming early primary signals paving the path for further stages of neurodegeneration. On the other hand, acetyl-CoA synthesis in mitochondrial compartments of glial cells seems to be activated despite amyloid-ß accumulated in transgenic brains.


Subject(s)
Acetyl Coenzyme A/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Neuroglia/metabolism , Neurons/metabolism , Acetylcholine/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Citric Acid/metabolism , Cytoplasm/metabolism , Disease Models, Animal , Female , Humans , Ketoglutaric Acids/metabolism , Male , Mice, Transgenic , Mitochondria/metabolism , Mutation , Neuroglia/pathology , Neurons/pathology , Peptide Fragments/metabolism , Pyruvic Acid/metabolism , Synaptosomes/metabolism
20.
Biochim Biophys Acta ; 1852(1): 146-55, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25446989

ABSTRACT

Recently, Aß peptide variants with an N-terminal truncation and pyroglutamate modification were identified and shown to be highly neurotoxic and prone to aggregation. This modification of Aß is catalyzed by glutaminyl cyclase (QC) and pharmacological inhibition of QC diminishes Aß deposition and accompanying gliosis and ameliorates memory impairment in transgenic mouse models of Alzheimer's disease (AD). QC expression was initially described in the hypothalamus, where thyrotropin-releasing hormone (TRH) is one of its physiological substrates. In addition to its hormonal role, a novel neuroprotective function of TRH following excitotoxicity and Aß-mediated neurotoxicity has been reported in the hippocampus. Functionally matching this finding, we recently demonstrated QC expression by hippocampal interneurons in mouse brain. Here, we detected neuronal co-expression of QC and TRH in the hippocampus of young adult wild type mice using double immunofluorescence labeling. This provides evidence for TRH being a physiological QC substrate in hippocampus. Additionally, in neocortex of aged but not of young mice transgenic for amyloid precursor protein an increase of QC mRNA levels was found compared to wild type littermates. This phenomenon was not observed in hippocampus, which is later affected by Aß pathology. However, in hippocampus of transgenic - but not of wild type mice - a correlation between QC and TRH mRNA levels was revealed. This co-regulation of the enzyme QC and its substrate TRH was reflected by a co-induction of both proteins in reactive astrocytes in proximity of Aß deposits. Also, in primary mouse astrocytes a co-induction of QC and TRH was demonstrated upon Aß stimulation.


Subject(s)
Aminoacyltransferases/metabolism , Astrocytes/enzymology , Hippocampus/enzymology , Neurons/enzymology , Thyrotropin-Releasing Hormone/metabolism , Aminoacyltransferases/genetics , Amyloid beta-Peptides/metabolism , Animals , Base Sequence , DNA Primers , Hippocampus/cytology , Mice , Mice, Transgenic , Polymerase Chain Reaction , RNA, Messenger/genetics , Substrate Specificity , Thyrotropin-Releasing Hormone/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...