Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Pharmacology ; 108(2): 204-212, 2023.
Article in English | MEDLINE | ID: mdl-36724742

ABSTRACT

The nucleus basalis of Meynert (nBM) is the major source of cholinergic neurons in the basal forebrain, which require nerve growth factor (NGF) for their survival. Melatonin, a pleiotropic hormone, has been shown to exert neuroprotection in several experimental models, but its effect on nBM neurons is not well known. Thus, the aim of this study is to evaluate the effect of melatonin in organotypic brain slices of the nBM. Organotypic nBM slices were incubated for 2 weeks without (control) or with 100 ng/mL NGF, 1 µM melatonin, or a combination of both. Cholinergic neurons were immunohistochemically stained for choline acetyltransferase (ChAT) and subjected to a co-localization study with silent information regulator 1 (SIRT1) and melatonin receptor 1A (MT1A), both potentially involved in melatonin neuroprotection. Counting of ChAT-positive neurons in nBM slices showed that melatonin and NGF significantly increased the number of ChAT-positive neurons compared to the control in a dose-dependent manner (1-10 µM). In co-treatment with NGF, melatonin did not potentiate the maximal NGF-mediated effect. Immunohistochemical analysis proved that cholinergic nBM neurons co-localized with SIRT1 and MT1A receptor. Our data show that melatonin improves the survival of cholinergic nBM neurons and confirm that they express SIRT1 and MT1A.


Subject(s)
Basal Nucleus of Meynert , Melatonin , Melatonin/pharmacology , Nerve Growth Factor/pharmacology , Sirtuin 1/metabolism , Cholinergic Neurons/metabolism , Brain/metabolism , Cholinergic Agents/metabolism , Cholinergic Agents/pharmacology
2.
Pharmacology ; 106(7-8): 390-399, 2021.
Article in English | MEDLINE | ID: mdl-33979803

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is a severe neurodegenerative disorder of the brain characterized by degeneration of cholinergic neurons which is directly linked to cognitive decline. Nerve growth factor (NGF) is the most potent protective factor for cholinergic neurons, additionally the NMDA antagonist memantine blocks glutamate-mediated excitotoxic activity. Quinidine is an inhibitor of organic cation transporter 2 (OCT2). OCT2 is located on cholinergic neurons and plays a role in presynaptic reuptake and recycling of acetylcholine in the brain. We hypothesize that quinidine can modulate the protective effects of NGF and memantine on cholinergic neurons in organotypic brain slices of the nucleus basalis of Meynert (nBM). METHODS: Organotypic brain slices of nBM were incubated with 100 ng/mL NGF, 10 µM memantine, 10 µM quinidine, and combinations of these treatments for 2 weeks. Cholinergic neurons were immunohistochemically stained for choline acetyltransferase (ChAT). RESULTS: Our data show that NGF as well as memantine counteracted the cell death of cholinergic nBM neurons. Quinidine alone had no toxic effect on cholinergic neurons but inhibited the protective effect of NGF and memantine when applied simultaneously. DISCUSSION/CONCLUSION: Our data provide evidence that quinidine modulates the survival of cholinergic nBM neurons via OCT2.


Subject(s)
Memantine/pharmacology , Nerve Growth Factor/pharmacology , Neuroprotective Agents/pharmacology , Quinidine/pharmacology , Acetylcholine/metabolism , Animals , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Brain/drug effects , Brain/metabolism , Cell Survival/drug effects , Cholinergic Neurons , Mice , Mice, Inbred C57BL , Organic Cation Transporter 2/antagonists & inhibitors , Tissue Culture Techniques
3.
Adv Exp Med Biol ; 1331: 193-202, 2021.
Article in English | MEDLINE | ID: mdl-34453299

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative disorder of the brain characterized by extracellular beta-amyloid plaques, intraneuronal tau inclusions, vascular impairment, inflammation, neurodegeneration, and memory loss. Acetylcholine is the most important neurotransmitter for memory, and cholinergic neurons selectively degenerate in AD, and a loss of acetylcholine directly correlates with cognitive decline. Nerve growth factor (NGF) is the most potent growth factor to support the survival of these cholinergic neurons. Thus, researchers are interested to deliver NGF directly into the brain to the cholinergic neurons. As the brain is isolated by the blood-brain barrier, the large protein NGF cannot easily pass into the brain, and peripheral administration of NGF also causes severe side effects. Blood cells may represent a potent therapeutic strategy to deliver NGF into the brain. Monocytes can be isolated and loaded with NGF and may transmigrate into the brain. As monocytes are precursors of microglia, they may differentiate and release NGF but also phagocyte and eliminate toxic plaques. Platelets are small anuclear cells and become rapidly activated during vascular lesions, and they may migrate to lesion sites and repair blood vessels and also eliminate toxic beta-amyloid depositions in vessels. In order to guarantee a stable and slow release, the use of biomaterials is of interest, especially collagen hydrogels that may be useful to protect these transmigrating blood cells. In this review, I summarize advantages and challenges of using transmigrating cells to deliver NGF directly into the brain.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Collagen , Humans , Hydrogels , Monocytes , Nerve Growth Factor
4.
Exp Brain Res ; 238(11): 2521-2529, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32862235

ABSTRACT

Vessel damage is a general pathological process in many neurodegenerative disorders, as well as spinal cord injury, stroke, or trauma. Biomaterials can present novel tools to repair and regenerate damaged vessels. The aim of the present study is to test collagen hydrogels loaded with different angiogenic factors to study vessel repair in organotypic brain slice cultures. In the experimental set up I, we made a cut on the organotypic brain slice and tested re-growth of laminin + vessels. In the experimental set up II, we cultured two half brain slices with a gap with a collagen hydrogel placed in between to study endothelial cell migration. In the experimental set up I, we showed that the number of vessels crossing the cut was tendencially increased with the addition of fibroblast growth factor-2 (FGF-2), vascular endothelial growth factor, or platelet-derived growth factor-BB compared to the control group. In the experimental set up II, we demonstrated that a collagen hydrogel loaded with FGF-2 resulted in a significantly increased number of migrated laminin + cells in the gap between the slices compared to the control hydrogel. Co-administration of several growth factors did not further potentiate the effects. Taken together, we show that organotypic brain slices are good models to study brain vessels and FGF-2 is a potent angiogenic factor for endothelial cell proliferation and migration. Our results provide evidence that the collagen hydrogels can be used as an extracellular matrix for the vascular endothelial cells.


Subject(s)
Brain , Animals , Collagen , Endothelial Cells , Fibroblast Growth Factor 2/pharmacology , Hydrogels , Mice , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A
5.
Brain Behav Immun ; 78: 52-64, 2019 05.
Article in English | MEDLINE | ID: mdl-30664922

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative brain disorder and the most common form of dementia coming along with cerebral amyloid angiopathy (CAA) in more than 70% of all cases. However, CAA occurs also in pure form without AD pathology. Vascular life style risk factors such as obesity, hypertension, hypercholesterolemia, diabetes, stress or an old age play an important role in the progression of CAA. So far, no animal model for sporadic CAA has been reported, thus the aim of the present study was to create and characterize a new mouse model for sporadic CAA by treatment with different vascular risk factors. Healthy C57BL6 mice were treated with lifestyle vascular risk factors for 35 or 56 weeks: lipopolysaccharide, social stress, streptozotozin, high cholesterol diet and copper in the drinking water. Four behavioral tests (black-white box, classical maze, cheeseboard maze and plus-maze discriminative avoidance task) showed impaired learning, memory and executive functions as well as anxiety with increased age. The treated animals exhibited increased plasma levels of cortisol, insulin, interleukin-1ß, glucose and cholesterol, confirming the effectiveness of the treatment. Confocal microscopy analysis displayed severe vessel damage already after 35 weeks of treatment. IgG positive staining points to a severe blood-brain barrier (BBB) disruption and furthermore, cerebral bleedings were observed in a much higher amount in the treatment group. Importantly, inclusions of beta-amyloid in the vessels indicated the development of CAA, but no deposition of beta-amyloid plaques and tau pathology in the brains were seen. Taken together, we characterized a novel sporadic CAA mouse model, which offers a strategy to study the progression of the disease and therapeutic and diagnostic interventions.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/pathology , Cerebral Amyloid Angiopathy/etiology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Hemorrhage/pathology , Diabetes Mellitus , Disease Models, Animal , Disease Progression , Female , Humans , Hypercholesterolemia , Hypertension , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity , Plaque, Amyloid , Risk Factors , Stress, Psychological
6.
Int Psychogeriatr ; 30(1): 147-152, 2018 01.
Article in English | MEDLINE | ID: mdl-28816110

ABSTRACT

Alzheimer´s disease (AD) is a severe neurodegenerative brain disorder characterized by beta-amyloid plaques, Tau pathology, inflammation, neurodegeneration, and cerebrovascular dysfunction. Besides that, alterations in monocytes and platelets have been reported in the blood of Alzheimer patients. In the present study, we measured circulating levels of platelet-monocyte aggregates in EDTA blood of cognitively healthy participants and 40 AD patients, and examined their changes induced by stimulation with beta-amyloid peptides. We measured CD14, CD62P, and CD42a using fluorescence-activated cell scanning (FACS) analysis. Our data show that the levels of circulating monocyte-platelet aggregates were not different between healthy controls and AD patients. However, incubation with beta-amyloid-40, -42, and pyroglutamate-beta-amyloid increased the platelet-monocyte aggregation in healthy subjects, but not AD patients. Our data conclude that the interaction between monocytes and platelets is not altered in whole blood of AD patients, but their sensitivity toward beta-amyloid peptides is decreased. There might be a critical link between the interaction of platelets and monocytes in AD, which has to be explored in further studies.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Blood Platelets/metabolism , Monocytes/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Case-Control Studies , Cell Aggregation , Humans , Platelet Aggregation/physiology
7.
Pharmacology ; 100(5-6): 243-245, 2017.
Article in English | MEDLINE | ID: mdl-28743115

ABSTRACT

Clinical diagnosis of Creutzfeldt-Jakob disease (CJD) can be supported by the analysis of Tau and 14-3-3 in the cerebrospinal fluid (CSF). In this short report, we report about a retrospective analysis performed on 2,296 routinely collected CSF samples, and 44 samples with a ratio of phosphoTau181/Tau <0.075 were selected. Analysis was performed with a novel 14-3-3 gamma CircuLex Elisa. We show that control levels were around 6,000 AU/mL and samples from Alzheimer patients were not different from those collected from healthy controls. Four cases of verified CJD had 14-3-3 CSF levels of >100,000 AU/mL, while 10 out of 12 suspected CJD samples with 14-3-3 CSF levels between 50,000-100,000 AU/mL were CJD positive. All samples with 14-3-3 levels between 15,000 and 50,000 AU/mL were not CJD cases but disorders with complex neuropathology. In conclusion, our data suggests that in CSF samples with a phospho-Tau-181/Tau ratio <0.075 CSF levels of 14-3-3 should be analyzed. Our data suggests a very high risk for CJD with 14-3-3 levels above 100,000 AU/mL and a probable diagnosis of CJD based on laboratory parameters above 50,000 AU/mL.


Subject(s)
14-3-3 Proteins/cerebrospinal fluid , Cerebrospinal Fluid/metabolism , Creutzfeldt-Jakob Syndrome/cerebrospinal fluid , Creutzfeldt-Jakob Syndrome/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Creutzfeldt-Jakob Syndrome/diagnosis , Diagnosis, Differential , Humans , Middle Aged , Phosphorylation/physiology , Retrospective Studies , tau Proteins/cerebrospinal fluid
8.
RNA ; 20(12): 1929-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25344396

ABSTRACT

We have generated a novel, neuro-specific ncRNA microarray, covering 1472 ncRNA species, to investigate their expression in different mouse models for central nervous system diseases. Thereby, we analyzed ncRNA expression in two mouse models with impaired calcium channel activity, implicated in Epilepsy or Parkinson's disease, respectively, as well as in a mouse model mimicking pathophysiological aspects of Alzheimer's disease. We identified well over a hundred differentially expressed ncRNAs, either from known classes of ncRNAs, such as miRNAs or snoRNAs or which represented entirely novel ncRNA species. Several differentially expressed ncRNAs in the calcium channel mouse models were assigned as miRNAs and target genes involved in calcium signaling, thus suggesting feedback regulation of miRNAs by calcium signaling. In the Alzheimer mouse model, we identified two snoRNAs, whose expression was deregulated prior to amyloid plaque formation. Interestingly, the presence of snoRNAs could be detected in cerebral spine fluid samples in humans, thus potentially serving as early diagnostic markers for Alzheimer's disease. In addition to known ncRNAs species, we also identified 63 differentially expressed, entirely novel ncRNA candidates, located in intronic or intergenic regions of the mouse genome, genomic locations, which previously have been shown to harbor the majority of functional ncRNAs.


Subject(s)
Alzheimer Disease/genetics , Epilepsy/genetics , MicroRNAs/biosynthesis , Parkinson Disease/genetics , RNA, Untranslated/biosynthesis , Alzheimer Disease/pathology , Animals , Calcium Channels/genetics , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Models, Animal , Epilepsy/pathology , Gene Expression Regulation , Genome , Humans , Mice , MicroRNAs/genetics , Organ Specificity , Parkinson Disease/pathology , RNA, Untranslated/genetics , Tissue Array Analysis
9.
Neurobiol Learn Mem ; 136: 86-96, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27670619

ABSTRACT

Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is mainly characterized by beta-amyloid (Aß) plaque deposition, Tau pathology and dysfunction of the cholinergic system causing memory impairment. The aim of the present study was to examine (1) anxiety and cognition, (2) Aß plaque deposition and (3) degeneration of cholinergic neurons in the nucleus basalis of Meynert (nbM) and cortical cholinergic innervation in an Alzheimer mouse model (APP_SweDI; overexpressing amyloid precursor protein (APP) with the Swedish K670N/M671L, Dutch E693Q, and Iowa D694N mutations). Our results show that 12-month-old APP_SweDI mice were more anxious and had more memory impairment. A large number of Aß plaques were already visible at the age of 6 months and increased with age. A significant decrease in cholinergic neurons was seen in the transgenic mouse model in comparison to the wild-type mice, identified by immunohistochemistry against choline acetyltransferase (ChAT) and p75 neurotrophin receptor as well as by in situ hybridization. Moreover, a significant decrease in cortical cholinergic fiber density was found in the transgenic mice as compared to the wild-type. In the cerebral cortex of APP_SweDI mice, swollen cholinergic varicosities were seen in the vicinity of Aß plaques. In conclusion, the present study shows that in an AD mouse model (APP_SweDI mice) a high Aß plaque load in the cortex causes damage to cholinergic axons in the cortex, followed by subsequent retrograde-induced cell death of cholinergic neurons and some forms of compensatory processes. This degeneration was accompanied by enhanced anxiety and impaired cognition.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor/metabolism , Anxiety/physiopathology , Basal Nucleus of Meynert , Behavior, Animal/physiology , Cerebral Cortex , Cholinergic Neurons/pathology , Disease Models, Animal , Plaque, Amyloid/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Pharmacology ; 98(5-6): 251-260, 2016.
Article in English | MEDLINE | ID: mdl-27463512

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative disorder of the brain, characterized by extracellular beta-amyloid (Aß) plaques, intracellular tau pathology, neurodegeneration and inflammation. There is clear evidence that the blood-brain barrier is damaged in AD and that vessel function is impaired. Alpha-smooth muscle actin (αSMA) is a prominent protein expressed on brain vessels, especially in cells located closer to the arteriole end of the capillaries, which possibly influences the blood vessel contraction. The aim of the present study was to observe αSMA protein and mRNA expression in isolated brain vessel extracts and cortex in an Alzheimer mouse model with strong Aß plaque deposition. Our data revealed a prominent expression of αSMA protein in isolated brain vessel extracts of AD mice by Western blot analysis. Immunostaining showed that these vessels were associated with Aß plaques. Quantitative real-time PCR analysis confirmed this increase at the mRNA expression level and showed a significant increase of transforming growth factor beta-1 mRNA expression in AD mice. In situ hybridization demonstrated a strong expression pattern of αSMA mRNA in the whole cortex and hippocampus. In conclusion, our data provide evidence that αSMA protein and mRNA are enhanced in vessels in an AD mouse model, possibly counteracting vessel malfunction in AD.


Subject(s)
Actins/biosynthesis , Alzheimer Disease/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , RNA, Messenger/biosynthesis , Animals , Blood-Brain Barrier/metabolism , Cerebrovascular Circulation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
J Neurol Neurosurg Psychiatry ; 86(1): 79-86, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24741064

ABSTRACT

BACKGROUND: Recent evidence suggests axonal injury after aneurysmal subarachnoid haemorrhage (aSAH). The microtubule-associated protein, tau, has been shown to be elevated in the cerebrospinal fluid after aSAH, however, brain extracellular tau levels and their relation to long-term neurological and cognitive outcomes have not been investigated. METHODS: Serial cerebral microdialysis (CMD) samples were collected from 22 consecutive aSAH patients with multimodal neuromonitoring to determine CMD-total-tau by ELISA. CMD-total-tau was analysed considering other brain metabolic parameters, brain tissue oxygen tension (PbtO2), and functional and neuropsychological outcome at 12 months. All outcome models were analysed using generalised estimating equations with an autoregressive working correlation matrix to account for multiple measurements of brain extracellular proteins per subject. RESULTS: CMD-total-tau levels positively correlated with brain extracellular fluid levels of lactate (r=0.40, p<0.001), glutamate (r=0.45, p<0.001), pyruvate (r=0.26, p<0.001), and the lactate-pyruvate ratio (r=0.26, p<0.001), and were higher in episodes of hypoxic (PbtO2<20 mm Hg) brain extracellular lactate elevation (>4 mmol/L) (p<0.01). More importantly, high CMD-total-tau levels were associated with poor functional outcome (modified Rankin Scale ≥4) 12 months after aSAH even after adjusting for disease severity and age (p=0.001). A similar association was found with 3/5 neuropsychological tests indicative of impairments in cognition, psychomotor speed, visual conceptualisation and frontal executive functions at 1 year after aSAH (p<0.01). CONCLUSIONS: These results suggest that CMD-total tau may be an important biomarker for predicting long-term outcome in patients with severe aSAH. The value of axonal injury needs further confirmation in a larger patient cohort, preferably combined with advanced imaging techniques.


Subject(s)
Brain/metabolism , Cognition Disorders/metabolism , Intracranial Aneurysm/metabolism , Intracranial Aneurysm/psychology , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/psychology , tau Proteins/metabolism , Aged , Biomarkers/metabolism , Brain/pathology , Cognition Disorders/complications , Cognition Disorders/pathology , Female , Follow-Up Studies , Glutamic Acid , Humans , Intracranial Aneurysm/complications , Lactic Acid/metabolism , Male , Microdialysis , Middle Aged , Neuropsychological Tests , Outcome Assessment, Health Care , Oxygen/metabolism , Pyruvic Acid/metabolism , Subarachnoid Hemorrhage/complications
12.
Biogerontology ; 16(4): 543-58, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25947203

ABSTRACT

Alzheimer's disease (AD) is a severe neurodegenerative disorder characterized mainly by accumulation of amyloid-ß plaques and neurofibrillary tangles, synaptic and neuronal loss. Blood platelets contain the neurotransmitter serotonin and amyloid-precursor protein (APP), and may thus be useful as a peripheral biomarker for AD. The aim of the present study was to functionally characterize platelets by FACS, to examine alterations in APP expression and secretion, and to measure serotonin levels in hypercholesterolemia mice with AD-like pathology and in two AD mouse models, the triple transgenic AD model (3xTg) and the APP overexpressing AD model with the Swedish-Dutch-Iowa mutations (APP_SweDI). These data are supplemented with epidermal growth factor (EGF) levels and compared with changes observed in platelets of patients with AD. We observed decreased platelet APP isoforms in 3xTg mice and patients with AD when analysed by means of Western blot. In patients, a significant increase of APP levels was observed when assessed by ELISA. Secreted APPß proved to be altered amongst all three animal models of AD at different time points and in human patients with AD. Serotonin levels were only reduced in 7 and 14 month old 3xTg mice. Moreover, we found significantly lower EGF levels in human AD patients and could thereby reproduce previous findings. Taken together, our data confirm that platelets are dysfunctional in AD, however, results from AD animal models do not coincide in all aspects, and markedly differ when compared to AD patients. We support previous data that APP, as well as EGF, could become putative biomarkers for diagnosing AD in human platelets.


Subject(s)
Alzheimer Disease/blood , Amyloid beta-Protein Precursor/blood , Blood Platelets/metabolism , Hyperlipidemias/blood , Serotonin/blood , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Apoptosis , Biomarkers/blood , Blood Platelets/pathology , Case-Control Studies , Cross-Sectional Studies , Disease Models, Animal , Epidermal Growth Factor/blood , Female , Genotype , Humans , Hyperlipidemias/pathology , Male , Mice, Transgenic , Phenotype , Species Specificity , Time Factors
13.
Crit Care ; 19: 75, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25887441

ABSTRACT

INTRODUCTION: There is a substantial amount of evidence from animal models that early brain injury (EBI) may play an important role for secondary brain injury after aneurysmal subarachnoid hemorrhage (aSAH). Cerebral microdialysis (CMD) allows online measurement of brain metabolites, including the pro-inflammatory cytokine interleukin-6 (IL-6) and matrix metalloproteinase-9 (MMP-9), which is indicative for disruption of the blood-brain barrier. METHODS: Twenty-six consecutive poor-grade aSAH patients with multimodal neuromonitoring were analyzed for brain hemodynamic and metabolic changes, including CMD-IL-6 and CMD-MMP-9 levels. Statistical analysis was performed by using a generalized estimating equation with an autoregressive function. RESULTS: The baseline cerebral metabolic profile revealed brain metabolic distress and an excitatory response which improved over the following 5 days (P <0.001). Brain tissue hypoxia (brain tissue oxygen tension of less than 20 mm Hg) was common (more than 60% of patients) in the first 24 hours of neuromonitoring and improved thereafter (P <0.05). Baseline CMD-IL-6 and CMD-MMP-9 levels were elevated in all patients (median = 4,059 pg/mL, interquartile range (IQR) = 1,316 to 12,456 pg/mL and median = 851 pg/mL, IQR = 98 to 25,860 pg/mL) and significantly decreased over days (P <0.05). A higher pro-inflammatory response was associated with the development of delayed cerebral ischemia (P = 0.04), whereas admission disease severity and early brain tissue hypoxia were associated with higher CMD-MMP-9 levels (P <0.03). Brain metabolic distress and increased IL-6 levels were associated with poor functional outcome (modified Rankin Scale of more than 3, P ≤0.01). All models were adjusted for probe location, aneurysm securing procedure, and disease severity as appropriate. CONCLUSIONS: Multimodal neuromonitoring techniques allow insight into pathophysiologic changes in the early phase after aSAH. The results may be used as endpoints for future interventions targeting EBI in poor-grade aSAH patients.


Subject(s)
Monitoring, Physiologic/methods , Subarachnoid Hemorrhage/metabolism , Aged , Blood-Brain Barrier/metabolism , Brain Ischemia/etiology , Critical Illness , Female , Humans , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Microdialysis/methods , Middle Aged , Neuroimaging , Oxygen/metabolism , Subarachnoid Hemorrhage/pathology , Subarachnoid Hemorrhage/physiopathology , Treatment Outcome
14.
Mol Cell Neurosci ; 63: 83-95, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25447943

ABSTRACT

Several studies have shown that elevated plasma cholesterol levels (i.e. hypercholesterolemia) serve as a risk factor for late-onset Alzheimer's disease (AD). However, it remains unclear how hypercholesterolemia may contribute to the onset and progression of AD pathology. In order to determine the role of hypercholesterolemia at various stages of AD, we evaluated the effects of high cholesterol diet (5% cholesterol) in wild-type (WT; C57BL6) and triple-transgenic AD (3xTg-AD; Psen1, APPSwe, tauB301L) mice at 7, 14, and 20 months. The transgenic APP-Swedish/Dutch/Iowa AD mouse model (APPSwDI) was used as a control since these animals are more pathologically-accelerated and are known to exhibit extensive plaque deposition and cerebral amyloid angiopathy. Here, we describe the effects of high cholesterol diet on: (1) cognitive function and stress, (2) AD-associated pathologies, (3) neuroinflammation, (4) blood­brain barrier disruption and ventricle size, and (5) vascular dysfunction. Our data show that high dietary cholesterol increases weight, slightly impairs cognitive function, promotes glial cell activation and complement-related pathways, enhances the infiltration of blood-derived proteins and alters vascular integrity, however, it does not induce AD-related pathologies. While normal-fed 3xTg-AD mice display a typical AD-like pathology in addition to severe cognitive impairment and neuroinflammation at 20 months of age, vascular alterations are less pronounced. No microbleedings were seen by MRI, however, the ventricle size was enlarged. Triple-transgenic AD mice, on the other hand, fed a high cholesterol diet do not survive past 14 months of age. Our data indicates that cholesterol does not markedly potentiate AD-related pathology, nor does it cause significant impairments in cognition. However, it appears that high cholesterol diet markedly increases stress-related plasma corticosterone levels as well as some vessel pathologies. Together, our findings represent the first demonstration of prolonged high cholesterol diet and the examination of its effects at various stages of cerebrovascular- and AD-related disease.


Subject(s)
Alzheimer Disease/pathology , Blood-Brain Barrier/drug effects , Cerebral Ventricles/drug effects , Cholesterol/pharmacology , Hypercholesterolemia/pathology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cerebral Ventricles/blood supply , Cholesterol/administration & dosage , Cognition , Corticosterone/blood , Diet, High-Fat/adverse effects , Hypercholesterolemia/etiology , Mice , Mice, Inbred C57BL , Neuroglia/drug effects , Presenilin-1/genetics , tau Proteins/genetics
15.
J Neurosci Res ; 92(3): 298-306, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24323796

ABSTRACT

The recruitment of monocytes into the brain has been implicated in Alzheimer's disease and recent studies have indicated that monocytes can reduce amyloid plaque burden. Our previous investigations have shown that hypercholesterolemic rats develop cognitive, cholinergic, and blood-brain barrier dysfunction, but do not develop amyloid plaques. This study was designed to evaluate the effects of repeated intravenous (i.v.) infusion (via the dorsal penile vein) of primary monocytes on cognition, the cholinergic system, and cortical cytokine levels in hypercholesterolemia Brown-Norway rats. In addition, we also transduced the monocytes with nerve growth factor (NGF) to evaluate whether these cells could be used to deliver a neuroprotective agent to the brain. Our results indicate that repeated i.v. infused monocytes migrate into the brains of hypercholesterolemic rats; however, this migration does not translate into marked effects on learning. Animals receiving NGF-loaded monocytes demonstrate slightly improved learning and significantly elevated cholinergic neuron staining compared to treatment with monocytes alone. Furthermore, our data indicate that repeated infusion of monocytes does not lead to elevated cytokine secretion, indicating that no inflammatory response is induced. This study provides an experimental attempt to evaluate the effects of blood-derived primary monocytes in hypercholesterolemia rats.


Subject(s)
Basal Nucleus of Meynert/pathology , Cholinergic Neurons/drug effects , Hypercholesterolemia/pathology , Hypercholesterolemia/therapy , Monocytes/metabolism , Nerve Growth Factors/therapeutic use , Administration, Intravenous , Analysis of Variance , Animals , Cell Movement/drug effects , Cell Movement/physiology , Choline O-Acetyltransferase/metabolism , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hypercholesterolemia/physiopathology , Male , Maze Learning/drug effects , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nerve Growth Factor , Nerve Growth Factors/metabolism , Rats
16.
Platelets ; 25(1): 16-22, 2014.
Article in English | MEDLINE | ID: mdl-23402285

ABSTRACT

It is known that oxidative stress leads to amyloid precursor protein (APP) dysregulation in platelets. Ethanol (EtOH) is a vascular risk factor and induces oxidative stress. The aim of the present study was thus to investigate whether EtOH affects APP processing in rat and human platelets. Platelets were exposed to 50 mM EtOH with and without 2 mM calcium-chloride (CaCl2) for 20 or 180 minutes at 37°C. Platelet aggregation, serotonin release and APP isoforms 130 and 106/110 kDa were analyzed. As a control, 100 mM H2O2 was tested in rat platelets. Our data show that EtOH alone did not affect any of the analyzed parameters, whereas CaCl2 significantly increased aggregation of rat and human platelets. In addition, CaCl2 alone enhanced serotonin release in rat platelets. EtOH counteracted CaCl2-induced aggregation and serotonin release. In the presence of CaCl2, EtOH reduced the 130 kDa APP isoform in rat and human platelets. In conclusion, this study shows that in the presence of CaCl2, EtOH affects the platelet function and APP processing in rat and human platelets.


Subject(s)
Amyloid beta-Protein Precursor/blood , Blood Platelets/drug effects , Ethanol/pharmacology , Serotonin/blood , Animals , Blood Platelets/metabolism , Humans , Oxidative Stress , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Rats
17.
Pharmacology ; 93(1-2): 24-31, 2014.
Article in English | MEDLINE | ID: mdl-24435206

ABSTRACT

BACKGROUND: It is well established that reactive astrocytes express L-type calcium channels (LTCC), but their functional role is completely unknown. We have recently shown that reactive astrocytes highly express the CaV1.2 α1-subunit around ß-amyloid (Aß) plaques in an Alzheimer mouse model. The aim of the present study was to explore whether Aß peptides may regulate the mRNA expression of all LTCC subunits in primary mouse astrocytes in culture. METHODS: Confluent primary astrocytes were incubated with 10 µg/ml of human or murine Aß or the toxic fragment Aß25-35 for 3 days or for 3 weeks. The LTCC subunits were determined by quantitative RT-PCR. RESULTS: Our data show that murine Aß42 slightly but significantly increased CaV1.2 and CaV1.3 expression when incubated for 3 days. This acute treatment with murine Aß enhanced ß2 and ß3 mRNA levels but decreased α2δ-2 mRNA expression. When astrocytes were incubated for 3 weeks, the levels of CaV1.2 α1 were significantly decreased by the murine Aß and the toxic fragment. As a control, the protein kinase C-ε activator DCP-LA displayed a decrease in CaV2.1 expression. CONCLUSION: In conclusion, our data show that Aß can differentially regulate LTCC expression in primary mouse astrocytes depending on incubation time.


Subject(s)
Amyloid beta-Peptides/pharmacology , Astrocytes/drug effects , Calcium Channels, L-Type/genetics , Animals , Astrocytes/metabolism , Calcium Channels, L-Type/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Female , Glial Fibrillary Acidic Protein , Male , Mice , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism
18.
Front Biosci (Landmark Ed) ; 29(6): 232, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38940051

ABSTRACT

BACKGROUND: Alzheimer's disease is characterized by extracellular beta-amyloid plaques, intraneuronal tau neurofibrillary tangles and excessive neurodegeneration. The mechanisms of neuron degeneration and the potential of these neurons to form new nerve fibers for compensation remain elusive. The present study aimed to evaluate the impact of beta-amyloid and tau on new formations of nerve fibers from mouse organotypic brain slices connected to collagen-based microcontact prints. METHODS: Organotypic brain slices of postnatal day 8-10 wild-type mice were connected to established collagen-based microcontact prints loaded with polyornithine to enhance nerve fiber outgrowth. Human beta-amyloid(42) or P301S mutated aggregated tau was co-loaded to the prints. Nerve fibers were immunohistochemically stained with neurofilament antibodies. The physiological activity of outgrown neurites was tested with neurotracer MiniRuby, voltage-sensitive dye FluoVolt, and calcium-sensitive dye Rhod-4. RESULTS: Immunohistochemical staining revealed newly formed nerve fibers extending along the prints derived from the brain slices. While collagen-only microcontact prints stimulated nerve fiber growth, those loaded with polyornithine significantly enhanced nerve fiber outgrowth. Beta-amyloid(42) significantly increased the neurofilament-positive nerve fibers, while tau had only a weak effect. MiniRuby crystals, retrogradely transported along these newly formed nerve fibers, reached the hippocampus, while FluoVolt and Rhod-4 monitored electrical activity in newly formed nerve fibers. CONCLUSIONS: Our data provide evidence that intact nerve fibers can form along collagen-based microcontact prints from mouse brain slices. The Alzheimer's peptide beta-amyloid(42) stimulates this growth, hinting at a neuroprotective function when physiologically active. This "brain-on-chip" model may offer a platform for screening bioactive factors or testing drug effects on nerve fiber growth.


Subject(s)
Amyloid beta-Peptides , Brain , Nerve Fibers , Animals , Amyloid beta-Peptides/metabolism , Mice , Nerve Fibers/metabolism , Nerve Fibers/drug effects , Nerve Fibers/physiology , Brain/drug effects , Brain/metabolism , tau Proteins/metabolism , Humans , Immunohistochemistry , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/pathology , Mice, Inbred C57BL
19.
Biomolecules ; 14(2)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38397402

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by amyloid-beta (Aß) plaques and tau neurofibrillary tangles (NFT). Modelling aspects of AD is challenging due to its complex multifactorial etiology and pathology. The present study aims to establish a cost-effective and rapid method to model the two primary pathologies in organotypic brain slices. Coronal hippocampal brain slices (150 µm) were generated from postnatal (day 8-10) C57BL6 wild-type mice and cultured for 9 weeks. Collagen hydrogels containing either an empty load or a mixture of human Aß42 and P301S aggregated tau were applied to the slices. The media was further supplemented with various intracellular pathway modulators or heavy metals to augment the appearance of Aß plaques and tau NFTs, as assessed by immunohistochemistry. Immunoreactivity for Aß and tau was significantly increased in the ventral areas in slices with a mixture of human Aß42 and P301S aggregated tau compared to slices with empty hydrogels. Aß plaque- and tau NFT-like pathologies could be induced independently in slices. Heavy metals (aluminum, lead, cadmium) potently augmented Aß plaque-like pathology, which developed intracellularly prior to cell death. Intracellular pathway modulators (scopolamine, wortmannin, MHY1485) significantly boosted tau NFT-like pathologies. A combination of nanomolar concentrations of scopolamine, wortmannin, MHY1485, lead, and cadmium in the media strongly increased Aß plaque- and tau NFT-like immunoreactivity in ventral areas compared to the slices with non-supplemented media. The results highlight that we could harness the potential of the collagen hydrogel-based spreading of human Aß42 and P301S aggregated tau, along with pharmacological manipulation, to produce pathologies relevant to AD. The results offer a novel ex vivo organotypic slice model to investigate AD pathologies with potential applications for screening drugs or therapies in the future.


Subject(s)
Alzheimer Disease , Mice , Animals , Humans , Alzheimer Disease/metabolism , tau Proteins/metabolism , Cadmium/metabolism , Wortmannin/metabolism , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Brain/metabolism , Plaque, Amyloid/complications , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Collagen/metabolism , Hydrogels/metabolism , Scopolamine Derivatives/metabolism
20.
J Neurosci Res ; 91(1): 83-94, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22996751

ABSTRACT

ß-Amyloid (Aß) is a small peptide that plays a potent role in synaptic plasticity as well as forms amyloid plaques in Alzheimer's disease (AD). Recent studies suggest that Aß deposition is deleterious not only in AD, but also in Parkinson's disease (PD) and depression. This Aß effect is associated with inflammatory processes. However, further evaluation is needed to understand how Aß and inflammation interact and contribute to the regulation of the cholinergic, serotonergic, and dopaminergic neuronal populations. The aim of the present study was to investigate the effects of Aß(1-42) on cholinergic neurons of the nucleus basalis of Meynert (which degenerate in AD), on serotonergic neurons of the dorsal raphe nucleus (which play a role in depression), and on dopaminergic neurons of the ventral mesencephalon (which degenerate in PD) in rat organotypic brain slices. Furthermore, we investigated whether anti-inflammatory drugs (celecoxib, citalopram, cyclooxygenase-2 inhibitor, ibuprofen, indomethacin, piclamilast) modulate or counteract Aß-induced effects. Two-week-old organotypic brain slices of the nucleus basalis of Meynert, dorsal raphe nucleus, and ventral mesencephalon were incubated with 50 ng/ml Aß(1-42) with or without anti-inflammatory agents for 3 days. Our results reveal that Aß significantly decreased the number of choline acetyltransferase-positive cholinergic, tryptophan hydroxylase-positive serotonergic, and tyrosine hydroxylase-positive dopaminergic neurons and that anti-inflammatory drugs partially counteracted the Aß-induced neuronal decline. This decline was not due to apoptotic processes (as evaluated by TUNEL, propidium iodide, caspase), oxidative stress (as measured by nitrite, catalase, or superoxide dismutase-2), or inflammation, but was most likely caused by a downregulation of these key enzymes.


Subject(s)
Amyloid beta-Peptides/toxicity , Anti-Inflammatory Agents/pharmacology , Cholinergic Neurons/drug effects , Dopaminergic Neurons/drug effects , Serotonergic Neurons/drug effects , Animals , Apoptosis/drug effects , Blotting, Western , Brain/drug effects , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , In Situ Nick-End Labeling , Nerve Degeneration/chemically induced , Nerve Degeneration/prevention & control , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL