Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
EMBO J ; 36(13): 1837-1853, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28559417

ABSTRACT

Genetic variants in the triggering receptor expressed on myeloid cells 2 (TREM2) increase the risk for several neurodegenerative diseases including Alzheimer's disease and frontotemporal dementia (FTD). Homozygous TREM2 missense mutations, such as p.T66M, lead to the FTD-like syndrome, but how they cause pathology is unknown. Using CRISPR/Cas9 genome editing, we generated a knock-in mouse model for the disease-associated Trem2 p.T66M mutation. Consistent with a loss-of-function mutation, we observe an intracellular accumulation of immature mutant Trem2 and reduced generation of soluble Trem2 similar to patients with the homozygous p.T66M mutation. Trem2 p.T66M knock-in mice show delayed resolution of inflammation upon in vivo lipopolysaccharide stimulation and cultured macrophages display significantly reduced phagocytic activity. Immunohistochemistry together with in vivo TSPO small animal positron emission tomography (µPET) demonstrates an age-dependent reduction in microglial activity. Surprisingly, perfusion magnetic resonance imaging and FDG-µPET imaging reveal a significant reduction in cerebral blood flow and brain glucose metabolism. Thus, we demonstrate that a TREM2 loss-of-function mutation causes brain-wide metabolic alterations pointing toward a possible function of microglia in regulating brain glucose metabolism.


Subject(s)
Brain/pathology , Frontotemporal Dementia/pathology , Glucose/metabolism , Membrane Glycoproteins/genetics , Microglia/physiology , Mutation, Missense , Perfusion , Receptors, Immunologic/genetics , Animals , Disease Models, Animal , Gene Knock-In Techniques , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Mice , Mutant Proteins/genetics , Positron-Emission Tomography
2.
Ann Neurol ; 86(4): 561-571, 2019 10.
Article in English | MEDLINE | ID: mdl-31359452

ABSTRACT

OBJECTIVE: Clinical trials targeting ß-amyloid peptides (Aß) for Alzheimer disease (AD) failed for arguable reasons that include selecting the wrong stages of AD pathophysiology or Aß being the wrong target. Targeting Aß to prevent cerebral amyloid angiopathy (CAA) has not been rigorously followed, although the causal role of Aß for CAA and related hemorrhages is undisputed. CAA occurs with normal aging and to various degrees in AD, where its impact and treatment is confounded by the presence of parenchymal Aß deposition. METHODS: APPDutch mice develop CAA in the absence of parenchymal amyloid, mimicking hereditary cerebral hemorrhage with amyloidosis Dutch type (HCHWA-D). Mice were treated with a ß-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitor. We used 3-dimensional ultramicroscopy and immunoassays for visualizing CAA and assessing Aß in cerebrospinal fluid (CSF) and brain. RESULTS: CAA onset in mice was at 22 to 24 months, first in frontal leptomeningeal and superficial cortical vessels followed by vessels penetrating the cortical layers. CSF Aß increased with aging followed by a decrease of both Aß40 and Aß42 upon CAA onset, supporting the idea that combined reduction of CSF Aß40 and Aß42 is a specific biomarker for vascular amyloid. BACE1 inhibitor treatment starting at CAA onset and continuing for 4 months revealed a 90% Aß reduction in CSF and largely prevented CAA progression and associated pathologies. INTERPRETATION: This is the first study showing that Aß reduction at early disease time points largely prevents CAA in the absence of parenchymal amyloid. Our observation provides a preclinical basis for Aß-reducing treatments in patients at risk of CAA and in presymptomatic HCHWA-D. ANN NEUROL 2019;86:561-571.


Subject(s)
Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cerebral Amyloid Angiopathy/drug therapy , Disease Progression , Picolinic Acids/therapeutic use , Thiazines/therapeutic use , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Biomarkers/cerebrospinal fluid , Biomarkers/metabolism , Brain/blood supply , Female , Humans , Mice , Mice, Transgenic , Peptide Fragments/cerebrospinal fluid , Picolinic Acids/pharmacology , Thiazines/pharmacology
3.
Neuroimage ; 112: 70-85, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25724758

ABSTRACT

Pharmacological magnetic resonance imaging (phMRI) of the brain has become a widely used tool in both preclinical and clinical drug research. One of its challenges is to condense the observed complex drug-induced brain-activation patterns into semantically meaningful metrics that can then serve as a basis for informed decision making. To aid interpretation of spatially distributed activation patterns, we propose here a set of multivariate metrics termed "domain gauges", which have been calibrated based on different classes of marketed or validated reference drugs. Each class represents a particular "domain" of interest, i.e., a specific therapeutic indication or mode of action. The drug class is empirically characterized by the unique activation pattern it evokes in the brain-the "domain profile". A domain gauge provides, for any tested intervention, a "classifier" as a measure of response strength with respect to the domain in question, and a "differentiator" as a measure of deviation from the domain profile, both along with error ranges. Capitalizing on our in-house database with an unprecedented wealth of standardized perfusion-based phMRI data obtained from rats subjected to various validated treatments, we exemplarily focused on 3 domains based on therapeutic indications: an antipsychotic, an antidepressant and an anxiolytic domain. The domain profiles identified as part of the gauge definition process, as well as the outputs of the gauges when applied to both reference and validation data, were evaluated for their reconcilability with prior biological knowledge and for their performance in drug characterization. The domain profiles provided quantitative activation patterns with high biological plausibility. The antipsychotic profile, for instance, comprised key areas (e.g., cingulate cortex, nucleus accumbens, ventral tegmental area, substantia nigra) which are believed to be strongly involved in mediating an antipsychotic effect, and which are in line with network-level dysfunctions observed in schizophrenia. The domain gauges plausibly positioned the vast majority of the pharmacological and even non-pharmacological treatments. The results also suggest the segregation of sub-domains based on, e.g., the mode of action. Upon judicious selection of domains and careful calibration of the gauges, our approach represents a valuable analytical tool for biological interpretation and decision making in drug discovery.


Subject(s)
Brain/drug effects , Magnetic Resonance Imaging/methods , Psychotropic Drugs/pharmacology , Algorithms , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Discriminant Analysis , Male , Multivariate Analysis , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Reproducibility of Results
4.
Neuroimage ; 74: 326-36, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23454471

ABSTRACT

Imaging methods that enable the investigation of functional networks both in human and animal brain provide important insights into mechanisms underlying pathologies including psychiatric disorders. Since the serotonergic receptor 1A (5-HT(1A)-R) has been strongly implicated in the pathophysiology of depressive and anxiety disorders, as well as in the action of antidepressant drugs, we investigated brain connectivity related to the 5-HT(1A)-R system by use of pharmacological functional magnetic resonance imaging in mice. We characterized functional connectivity elicited by activation of 5-HT(1A)-R and investigated how pharmacological and genetic manipulations of its function may modulate the evoked connectivity. Functional connectivity elicited by administration of the 5-HT(1A)-R agonist 8-OH-DPAT can be described by networks characterized by small-world attributes with nodes displaying highly concerted response patterns. Circuits identified comprised the brain structures known to be involved in stress-related disorders (e.g. prefrontal cortex, amygdala and hippocampus). The results also highlight the dorsomedial thalamus, a structure associated with fear processing, as a hub of the 5-HT(1A)-R functional network. Administration of a specific 5-HT(1A)-R antagonist or use of heterozygous 5-HT(1A)-R knockout mice significantly reduced functional connectivity elicited by 8-OH-DPAT. Whole brain functional connectivity analysis constitutes an attractive tool to characterize impairments in neurotransmission and the efficacy of pharmacological treatment in a comprehensive manner.


Subject(s)
Brain Mapping/methods , Brain/metabolism , Neural Pathways/physiology , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Brain/drug effects , Magnetic Resonance Imaging , Male , Mental Disorders/metabolism , Mice , Mice, Inbred C57BL , Neural Pathways/drug effects , Serotonin Receptor Agonists/pharmacology
5.
Brain ; 135(Pt 11): 3265-81, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23169918

ABSTRACT

Whereas large injuries to the brain lead to considerable irreversible functional impairments, smaller strokes or traumatic lesions are often associated with good recovery. This recovery occurs spontaneously, and there is ample evidence from preclinical studies to suggest that adjacent undamaged areas (also known as peri-infarct regions) of the cortex 'take over' control of the disrupted functions. In rodents, sprouting of axons and dendrites has been observed in this region following stroke, while reduced inhibition from horizontal or callosal connections, or plastic changes in subcortical connections, could also occur. The exact mechanisms underlying functional recovery after small- to medium-sized strokes remain undetermined but are of utmost importance for understanding the human situation and for designing effective treatments and rehabilitation strategies. In the present study, we selectively destroyed large parts of the forelimb motor and premotor cortex of adult rats with an ischaemic injury. A behavioural test requiring highly skilled, cortically controlled forelimb movements showed that some animals recovered well from this lesion whereas others did not. To investigate the reasons behind these differences, we used anterograde and retrograde tracing techniques and intracortical microstimulation. Retrograde tracing from the cervical spinal cord showed a correlation between the number of cervically projecting corticospinal neurons present in the hindlimb sensory-motor cortex and good behavioural recovery. Anterograde tracing from the hindlimb sensory-motor cortex also showed a positive correlation between the degree of functional recovery and the sprouting of neurons from this region into the cervical spinal cord. Finally, intracortical microstimulation confirmed the positive correlation between rewiring of the hindlimb sensory-motor cortex and the degree of forelimb motor recovery. In conclusion, these experiments suggest that following stroke to the forelimb motor cortex, cells in the hindlimb sensory-motor area reorganize and become functionally connected to the cervical spinal cord. These new connections, probably in collaboration with surviving forelimb neurons and more complex indirect connections via the brainstem, play an important role for the recovery of cortically controlled behaviours like skilled forelimb reaching.


Subject(s)
Forelimb/physiopathology , Hindlimb/physiopathology , Motor Cortex/physiology , Pyramidal Tracts/physiology , Recovery of Function/physiology , Stroke/physiopathology , Animals , Disease Models, Animal , Electric Stimulation/methods , Endothelin-1 , Female , Magnetic Resonance Imaging/methods , Motor Cortex/pathology , Motor Cortex/physiopathology , Motor Skills/physiology , Nerve Regeneration/physiology , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques/methods , Neuroimaging/methods , Pyramidal Tracts/pathology , Rats , Rats, Long-Evans , Spinal Cord/pathology , Stroke/pathology
6.
J Neurosci ; 31(3): 1069-80, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21248132

ABSTRACT

Loss of oligodendrocytes is a feature of many demyelinating diseases including multiple sclerosis. Here, we have established and characterized a novel model of genetically induced adult oligodendrocyte death. Specific primary loss of adult oligodendrocytes leads to a well defined and highly reproducible course of disease development that can be followed longitudinally by magnetic resonance imaging. Histological and ultrastructural analyses revealed progressive myelin vacuolation, in parallel to disease development that includes motor deficits, tremor, and ataxia. Myelin damage and clearance were associated with induction of oligodendrocyte precursor cell proliferation, albeit with some regional differences. Remyelination was present in the mildly affected corpus callosum. Consequences of acutely induced cell death of adult oligodendrocytes included secondary axonal damage. Microglia were activated in affected areas but without significant influx of B-cells, T-helper cells, or T-cytotoxic cells. Analysis of the model on a RAG-1 (recombination activating gene-1)-deficient background, lacking functional lymphocytes, did not change the observed disease and pathology compared with immune-competent mice. We conclude that this model provides the opportunity to study the consequences of adult oligodendrocyte death in the absence of primary axonal injury and reactive cells of the adaptive immune system. Our results indicate that if the blood-brain barrier is not disrupted, myelin debris is not removed efficiently, remyelination is impaired, and axonal integrity is compromised, likely as the result of myelin detachment. This model will allow the evaluation of strategies aimed at improving remyelination to foster axon protection.


Subject(s)
Axons/pathology , Cell Death/genetics , Corpus Callosum/pathology , Myelin Sheath/pathology , Oligodendroglia/pathology , Animals , Axons/metabolism , Cell Count , Corpus Callosum/metabolism , Disease Progression , Fluorescent Antibody Technique , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Microscopy, Electron , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Rotarod Performance Test
7.
Neuroimage ; 59(2): 1028-36, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-21945466

ABSTRACT

Two general pathological processes contribute to multiple sclerosis (MS): acute inflammation and degeneration. While magnetic resonance imaging (MRI) is highly sensitive in detecting abnormalities related to acute inflammation both clinically and in animal models of experimental autoimmune encephalomyelitis (EAE), the correlation of these readouts with acute and future disabilities has been found rather weak. This illustrates the need for imaging techniques addressing neurodegenerative processes associated with MS. In the present work we evaluated the sensitivity of different MRI techniques (T(2) mapping, macrophage tracking based on labeling cells in vivo by ultrasmall particles of iron oxide (USPIO), diffusion tensor imaging (DTI) and magnetization transfer imaging (MTI)) to detect histopathological changes in a novel animal model making use of intrinsic, temporally and spatially controlled triggering of oligodendrocyte cell death. This mouse model allows studying the MRI signature associated to neurodegenerative processes of MS in the absence of adaptive inflammatory components that appear to be foremost in the EAE models. Our results revealed pronounced T(2) hyperintensities in brain stem and cerebellar structures, which we attribute to structural alteration of white matter by pronounced vacuolation. Brain areas were found devoid of significant macrophage infiltration in line with the absence of a peripheral inflammatory response. The significant decrease in diffusion anisotropy derived from DTI measures in these structures is mainly caused by a pronounced decrease in diffusivity parallel to the fiber indicative of axonal damage. Triggering of oligodendrocyte ablation did not translate into a significant increase in radial diffusivity. Only minor decreases in MT ratio have been observed, which is attributed to inefficient removal of myelin debris.


Subject(s)
Brain/pathology , Disease Models, Animal , Magnetic Resonance Imaging/methods , Multiple Sclerosis/pathology , Oligodendroglia/pathology , Animals , Apoptosis , Cell Tracking/methods , Humans , Mice , Mice, Transgenic , Reproducibility of Results , Sensitivity and Specificity
8.
BMJ Open ; 12(12): e067159, 2022 12 30.
Article in English | MEDLINE | ID: mdl-36585141

ABSTRACT

INTRODUCTION: The growing worldwide prevalence of Alzheimer's disease (AD) and the lack of effective treatments pose a dire medical challenge. Sleep disruption is also prevalent in the ageing population and is increasingly recognised as a risk factor and an early sign of AD. The ALFASleep project aims to characterise sleep with subjective and objective measurements in cognitively unimpaired middle/late middle-aged adults at increased risk of AD who are phenotyped with fluid and neuroimaging AD biomarkers. This will contribute to a better understanding of the pathophysiological mechanisms linking sleep with AD, thereby paving the way for the development of non-invasive biomarkers and preventive strategies targeting sleep. METHODS AND ANALYSIS: We will invite 200 participants enrolled in the ALFA+ (for ALzheimer and FAmilies) prospective observational study to join the ALFASleep study. ALFA+ participants are cognitively unimpaired middle-aged/late middle-aged adults who are followed up every 3 years with a comprehensive set of evaluations including neuropsychological tests, blood and cerebrospinal fluid (CSF) sampling, and MRI and positron emission tomography acquisition. ALFASleep participants will be additionally characterised with actigraphy and CSF-orexin-A measurements, and a subset (n=90) will undergo overnight polysomnography. We will test associations of sleep measurements and CSF-orexin-A with fluid biomarkers of AD and glial activation, neuroimaging outcomes and cognitive performance. In case we found any associations, we will test whether changes in AD and/or glial activation markers mediate the association between sleep and neuroimaging or cognitive outcomes and whether sleep mediates associations between CSF-orexin-A and AD biomarkers. ETHICS AND DISSEMINATION: The ALFASleep study protocol has been approved by the independent Ethics Committee Parc de Salut Mar, Barcelona (2018/8207/I). All participants have signed a written informed consent before their inclusion (approved by the same ethics committee). Study findings will be presented at national and international conferences and submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER: NCT04932473.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Humans , Middle Aged , Alzheimer Disease/diagnosis , Biomarkers , Cognition/physiology , Cognitive Dysfunction/diagnosis , Observational Studies as Topic , Orexins/cerebrospinal fluid , Sleep Quality
9.
NMR Biomed ; 24(4): 439-46, 2011 May.
Article in English | MEDLINE | ID: mdl-22945293

ABSTRACT

Functional MRI (fMRI) based on the blood oxygen level-dependent (BOLD) contrast is widely used in preclinical neuroscience. The small dimensions of rodent brain place high demands on spatial resolution, and hence on the sensitivity of the fMRI experiment. This work investigates the performance of a 400-MHz cryogenic quadrature transceive radiofrequency probe (CryoProbe) with respect to the enhancement of the BOLD sensitivity. For this purpose, BOLD fMRI experiments were performed in mice during electrical forepaw stimulation using the CryoProbe and a conventional room temperature surface coil of comparable dimensions. Image signal-to-noise ratio (SNR) and temporal SNR were evaluated as quality measures for individual images and for fMRI time series of images, resulting in gains (mean ± standard deviation) with factors of 3.1 ± 0.7 and 1.8 ± 1.0 when comparing the CryoProbe and room temperature coil. The CryoProbe thermal shield temperature did not affect the noise characteristics, with temporal noise levels being 63 ± 16% of the corresponding room temperature value. However, a significant effect on BOLD amplitudes was found, which was attributed to temperature-dependent baseline cerebral blood volumes. Defined local thermal conditions were found to be a critical parameter for achieving an optimal and reproducible fMRI signal. In summary, the CryoProbe represents an attractive alternative for the enhancement of image SNR, temporal SNR and BOLD sensitivity in mouse fMRI experiments.


Subject(s)
Cold Temperature , Forelimb/physiology , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Oxygen/blood , Radio Waves , Somatosensory Cortex/physiology , Animals , Body Temperature/physiology , Brain Mapping , Electric Stimulation , Female , Mice , Mice, Inbred C57BL , Scalp/physiology , Spin Labels
10.
NMR Biomed ; 24(10): 1295-301, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22223365

ABSTRACT

Insulin resistance is a central feature of type II diabetes and is associated with alterations in skeletal muscle lipid metabolism, which manifest themselves, in part, in increased intramyocellular lipid (IMCL) accumulation. The objective of this study was to assess noninvasively the levels of IMCL longitudinally in the tibialis anterior muscle of Lep(ob) /Lep(ob) (ob/ob) mice, a genetic model of obesity and mild diabetes, and Lep(ob) /+ (ob/+) heterozygous control animals, using (1) H MRS at 9.4 T. The use of a cryogenic surface coil transceiver leads to significant increases in sensitivity. Method implementation included the assessment of the reproducibility and spatial heterogeneity of the IMCL signal and the determination of T(2) relaxation times, as IMCL levels were expressed relative to the total creatine signal, and therefore the signal ratios had to be corrected for differences in T(2) relaxation. IMCL levels were found to be significantly higher in ob/ob mice relative to ob/+ heterozygous control mice that do not develop disease. An increase in IMCL levels was observed for ob/ob mice until weeks 16/17; after this time point, IMCL levels decreased again, reaching final levels that were slightly higher than the initial values. These noninvasively detected alterations in skeletal muscle lipid metabolism in ob/ob mice were accompanied by a transient increase in plasma insulin concentrations. This study indicates that IMCL may be reliably assessed in mouse tibialis anterior muscle using a cryogenic surface coil, implying that (1) H MRS at 9.4 T represents a useful technology for the noninvasive measurement of changes in lipid metabolism in the skeletal muscle that accompany obesity.


Subject(s)
Cold Temperature , Lipid Metabolism , Magnetic Resonance Spectroscopy/instrumentation , Magnetic Resonance Spectroscopy/methods , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Aging/metabolism , Animals , Body Weight , Creatine/metabolism , Feasibility Studies , Insulin/metabolism , Male , Mice , Mice, Obese , Spin Labels , Surface Properties
11.
Neurobiol Dis ; 40(1): 284-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20600914

ABSTRACT

Deposition of beta-amyloid along cerebral vessels is found in most patients suffering from Alzheimer's disease. The effects of cerebral amyloid angiopathy (CAA) on the function of cerebral blood vessels were analyzed applying cerebral blood volume (CBV)-based fMRI to transgenic arcA beta mice. In a cortical brain region of interest (ROI), displaying high CAA, arcA beta mice older than 16 months showed reduced response to the vasodilatory substance acetazolamide compared to age-matched wild-type animals, both with regard to rate (vascular reactivity) and extent of vasodilation (maximal vasodilation). In a subcortical ROI, displaying little CAA, no genotype-specific decrease was observed, but maximal vasodilation decreased with age in arcA beta and wild-types. These findings indicate that vascular beta-amyloid deposits reduce the capacity of cerebral blood vessels to dilate upon demand, supporting the hypothesis that vascular beta-amyloid contributes to hypoperfusion and neurological deficits observed in AD and CAA. High diagnostic accuracy of the combined readouts in detecting vascular dysfunction in arcA beta mice was found.


Subject(s)
Acetazolamide , Aging/genetics , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Cerebral Amyloid Angiopathy/genetics , Cerebral Arteries/metabolism , Mice, Transgenic , Vasodilator Agents , Aging/drug effects , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/biosynthesis , Animals , Cerebral Amyloid Angiopathy/diagnosis , Cerebral Amyloid Angiopathy/metabolism , Cerebral Arteries/drug effects , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/genetics , Disease Models, Animal , Genetic Markers , Humans , Mice , Predictive Value of Tests
12.
J Med Chem ; 63(4): 1511-1525, 2020 02 27.
Article in English | MEDLINE | ID: mdl-31951127

ABSTRACT

We recently reported the discovery of a potent, selective, and brain-penetrant V1a receptor antagonist, which was not suitable for full development. Nevertheless, this compound was found to improve surrogates of social behavior in adults with autism spectrum disorder in an exploratory proof-of-mechanism study. Here we describe scaffold hopping that gave rise to triazolobenzodiazepines with improved pharmacokinetic properties. The key to balancing potency and selectivity while minimizing P-gp mediated efflux was fine-tuning of hydrogen bond acceptor basicity. Ascertaining a V1a antagonist specific brain activity pattern by pharmacological magnetic resonance imaging in the rat played a seminal role in guiding optimization efforts, culminating in the discovery of balovaptan (RG7314, RO5285119) 1. In a 12-week clinical phase 2 study in adults with autism spectrum disorder balovaptan demonstrated improvements in Vineland-II Adaptive Behavior Scales, a secondary end point comprising communication, socialization, and daily living skills. Balovaptan entered phase 3 clinical development in August 2018.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/therapeutic use , Autism Spectrum Disorder/drug therapy , Benzodiazepines/therapeutic use , Pyridines/therapeutic use , Receptors, Vasopressin/metabolism , Triazoles/therapeutic use , Adolescent , Adult , Animals , Antidiuretic Hormone Receptor Antagonists/chemical synthesis , Antidiuretic Hormone Receptor Antagonists/pharmacokinetics , Autism Spectrum Disorder/metabolism , Benzodiazepines/chemical synthesis , Benzodiazepines/pharmacokinetics , Brain/metabolism , Child , Clinical Trials as Topic , Drug Discovery , Female , Humans , Male , Mammals , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Triazoles/chemical synthesis , Triazoles/pharmacokinetics
13.
Neuroimage ; 44(4): 1345-54, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19015037

ABSTRACT

The accuracy at which changes in cortical functional topology can be assessed by functional MRI (fMRI) depends on the quality of the reference coordinate system used for comparison of data sets obtained in different imaging sessions. Current procedures comprise an overlay of activation clusters on registered high-resolution anatomical images. Yet, fMRI images are frequently distorted due to susceptibility artifacts, which are prominent in rodent studies due to the small dimensions involved and high magnetic field strengths used. Therefore, a procedure for co-registration of activation maps has been developed based on anatomical landmarks defined on fMR echo planar images (EPI) themselves. Validation studies in control rats revealed that the centers of activated areas in somatosensory cortex S1, evoked through sensory forepaw stimulation fell within an area of 1 x 1 mm(2) in agreement with known electrophysiological coordinates. The technique was applied to detect changes in activation patterns in rats following smaller unilateral spinal cord injuries (SCI) in their cervical segments (C3/C4) 12 weeks after lesion. Despite of an almost complete behavioral recovery, fMRI responses remained altered in SCI animals with both significantly reduced fMRI signal amplitude and reduced latency to reach the peak response. Moreover, in SCI animals the activated S1 area corresponding to the contralesional forepaw was significantly enlarged and the center-of-mass for the ipsilesional paw was shifted rostrally. The mapping technique described combined with the temporal analysis of the BOLD response enabled a noninvasive quantitative characterization of cortical functional reorganization following SCI in rats.


Subject(s)
Magnetic Resonance Imaging/methods , Neuronal Plasticity , Somatosensory Cortex/physiopathology , Spinal Cord Injuries/diagnosis , Spinal Cord Injuries/physiopathology , Subtraction Technique , Adaptation, Physiological , Animals , Male , Rats , Rats, Inbred Lew
14.
Eur J Neurosci ; 30(10): 1860-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19912336

ABSTRACT

Multimodal non-invasive neuroimaging in rodents constitutes an attractive tool for studying neurobiological processes in vivo. At present, imaging studies of brain anatomy and function as well as the investigation of structure-function relationships belong to the standard repertoire of neuroscientists. Molecular imaging adds a new perspective. The mapping of the receptor distribution and receptor occupancy can nowadays be complemented by specific readouts of receptor function either by visualizing the activity of signaling pathways or mapping the physiological consequences of receptor stimulation. Molecular information is obtained through the use of imaging probes that combine a target-specific ligand with a reporter moiety that generates a signal that can be detected from outside the body. For imaging probes targeting the central nervous system, penetration of the intact blood-brain barrier constitutes a major hurdle. Molecular imaging generates specific information and therefore has a large potential for disease phenotyping (diagnostics), therapy development and monitoring of treatment response. Molecular imaging is still in its infancy and major developments in imaging technology, probe design and data analysis are required in order to make an impact. Rodent molecular neuroimaging will play an important role in the development of these tools.


Subject(s)
Brain Mapping , Brain/anatomy & histology , Brain/metabolism , Gene Expression/physiology , Molecular Imaging/methods , Receptors, Cell Surface/metabolism , Animals , Brain/diagnostic imaging , Image Processing, Computer-Assisted/methods , Ligands , Magnetic Resonance Imaging/methods , Mice , Positron-Emission Tomography , Radiopharmaceuticals , Rodentia , Signal Transduction/physiology
15.
Bioconjug Chem ; 20(10): 1940-9, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19803478

ABSTRACT

Different imaging modalities can provide complementary information on biological processes at the cellular or molecular level in vitro and in vivo. However, specific molecular probes suitable for a comparison of different imaging modalities are often not readily accessible because their preparation is usually accomplished by individually developed and optimized syntheses. Herein, we present a general, modular synthetic approach that provides access to multiple probes derived from a single precursor by application of the same, efficient functionalization strategy, the Cu(I)-catalyzed cycloaddition of terminal alkynes and azides (click chemistry). To demonstrate the viability and efficiency of this approach, folic acid (FA) was selected as a targeting vector because the preparation of FA-based imaging probes used for SPECT, PET, MRI, and NIRF by reported synthetic strategies is usually difficult to achieve and often results in low overall yields. We prepared a versatile γ-azido-FA precursor as well as a set of alkyne functionalized probes and precursors including ligand systems suitable for the chelation of various (radio)metals, an NIR dye and (18)F- and (19)F-derivatives, which enabled the parallel development of new FA-imaging probes. The Cu(I)-mediated coupling of the alkynes with the γ-azido-FA precursor was accomplished in high yields and with minimal use of protective groups. The various probes were fully characterized spectroscopically as well as in vitro and in vivo. In vitro, all new FA-derivatives exhibited high affinity toward the folic acid receptor (FR) and/or were specifically internalized into FR-overexpressing KB cells. In vivo experiments with nude mice showed that all probes (except the MRI probes which have not been tested yet) accumulated specifically in FR-positive organs and human KB-cell xenografts. However, in vivo imaging revealed significant differences between the various FA-derivatives with respect to unspecific, off-target localization. In general, the comparison of different probes proved the superiority of the more hydrophilic, radiometal-based imaging agents, a result which will guide future efforts for the development of FA-based imaging probes and therapeutic agents. In addition, the strategy presented herein should be readily applicable to other molecules of interest for imaging and therapeutic purposes and thus represents a valuable alternative to other synthetic approaches.


Subject(s)
Chelating Agents/chemistry , Chelating Agents/metabolism , Click Chemistry , Folate Receptors, GPI-Anchored/chemistry , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Molecular Imaging/methods , Molecular Probes , Animals , Click Chemistry/methods , Humans , KB Cells , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Probes/chemical synthesis , Neoplasm Transplantation/diagnostic imaging , Radionuclide Imaging , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
16.
Cell Rep ; 20(8): 1867-1880, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28834750

ABSTRACT

The NMDA receptor (NMDAR) antagonist ketamine elicits a long-lasting antidepressant response in patients with treatment-resistant depression. Understanding how antagonism of NMDARs alters synapse and circuit function is pivotal to developing circuit-based therapies for depression. Using virally induced gene deletion, ex vivo optogenetic-assisted circuit analysis, and in vivo chemogenetics and fMRI, we assessed the role of NMDARs in the medial prefrontal cortex (mPFC) in controlling depression-related behavior in mice. We demonstrate that post-developmental genetic deletion of the NMDAR subunit GluN2B from pyramidal neurons in the mPFC enhances connectivity between the mPFC and limbic thalamus, but not the ventral hippocampus, and reduces depression-like behavior. Using intersectional chemogenetics, we show that activation of this thalamocortical circuit is sufficient to elicit a decrease in despair-like behavior. Our findings reveal that GluN2B exerts input-specific control of pyramidal neuron innervation and identify a medial dorsal thalamus (MDT)→mPFC circuit that controls depression-like behavior.


Subject(s)
Cerebral Cortex/metabolism , Depression/genetics , Synapses/metabolism , Thalamus/metabolism , Animals , Depression/pathology , Mice
17.
Sci Rep ; 6: 24523, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27080031

ABSTRACT

Functional magnetic resonance imaging (fMRI) has revolutionized neuroscience by opening a unique window that allows neurocircuitry function and pathological alterations to be probed non-invasively across brain disorders. Here we report a novel sustainable anesthesia procedure for small animal neuroimaging that overcomes shortcomings of anesthetics commonly used in rodent fMRI. The significantly improved preservation of cerebrovascular dynamics enhances sensitivity to neural activity changes for which it serves as a proxy in fMRI readouts. Excellent cross-species/strain applicability provides coherence among preclinical findings and is expected to improve translation to clinical fMRI investigations. The novel anesthesia procedure based on the GABAergic anesthetic etomidate was extensively validated in fMRI studies conducted in a range of genetically engineered rodent models of autism and strains commonly used for transgenic manipulations. Etomidate proved effective, yielded long-term stable physiology with basal cerebral blood flow of ~0.5 ml/g/min and full recovery. Cerebrovascular responsiveness of up to 180% was maintained as demonstrated with perfusion- and BOLD-based fMRI upon hypercapnic, pharmacological and sensory stimulation. Hence, etomidate lends itself as an anesthetic-of-choice for translational neuroimaging studies across rodent models of brain disorders.


Subject(s)
Anesthesia , Magnetic Resonance Imaging , Neuroimaging , Anesthetics, Inhalation/pharmacology , Animals , Brain/blood supply , Brain/drug effects , Brain/physiology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Etomidate/pharmacology , Isoflurane/pharmacology , Medetomidine/pharmacology , Mice , Rats , Species Specificity
18.
J Neurosci ; 23(23): 8231-6, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-12967984

ABSTRACT

Quantitative functional magnetic resonance imaging was applied to characterize brain function in amyloid precursor protein 23 (APP23) transgenic mice, which reproduce the neuropathological alterations associated with Alzheimer's disease. Electrical stimulation of the paw led to cerebral blood volume increases in the contralateral somatosensory cortex. In APP23 mice this hemodynamic response decreased with increasing age of the animal and with increasing stimulus amplitude as compared with wild-type animals. The age-dependent dysfunction in APP23 mice may be attributed in part to a compromised cerebrovascular reactivity. Quantitative functional brain mapping that uses standardized sensory inputs should allow for assessment of disease progression and therapy response (e.g., passive immunization against beta-amyloid) in patients also.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Somatosensory Disorders/physiopathology , Age Factors , Alzheimer Disease/complications , Amyloid beta-Protein Precursor/genetics , Animals , Blood Flow Velocity , Blood Gas Monitoring, Transcutaneous , Cerebrovascular Circulation , Disease Models, Animal , Disease Progression , Electric Stimulation , Hindlimb/innervation , Hindlimb/physiopathology , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Mutation , Respiration, Artificial , Somatosensory Cortex/blood supply , Somatosensory Cortex/physiopathology , Somatosensory Disorders/complications
19.
J Neurosci ; 23(24): 8453-9, 2003 Sep 17.
Article in English | MEDLINE | ID: mdl-13679413

ABSTRACT

Neuropathological changes associated with Alzheimer's disease (AD) such as amyloidplaques, cerebral amyloid angiopathy, and related pathologies are reproduced in APP23 transgenic mice overexpressing amyloid precursor protein (APP) with the Swedish mutation. Magnetic resonance angiography (MRA) was applied to probe, in vivo, the cerebral arterial hemodynamics of these mice. Flow voids were detected at the internal carotid artery of 11-month-old APP23 mice. At the age of 20 months, additional flow disturbances were observed in large arteries at the circle of Willis. Vascular corrosion casts obtained from the same mice revealed that vessel elimination, deformation, or both had taken place at the sites where flow voids were detected by MRA. The detailed three-dimensional architecture of the vasculature visible in the casts assisted the identification of smaller vessels most likely formed as substitution or anastomosis within the circle of Willis. Angiograms and corrosion casts from nontransgenic, age-matched mice manifested no major abnormalities in the cerebrovascular arterial flow pattern. Because no transgene overexpression has been found in the cerebrovasculature of APP23 mice and no deposits of amyloid-beta (Abeta) were observed in large arteries in the region of the circle of Willis, the present results suggest that soluble Abeta may exert deleterious effects on the vasculature. Our findings support the idea that cerebral circulatory abnormalities evolving progressively could contribute to AD pathogenesis. The study also shows the power of MRA to identify changes of vascular function in genetically engineered mice. MRA as a noninvasive technique could be applied to test new therapeutic concepts in animal models of AD and in humans.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Brain/blood supply , Brain/physiopathology , Cerebrovascular Circulation , Age Factors , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Blood Flow Velocity , Brain/pathology , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Corrosion Casting , Disease Models, Animal , Disease Progression , Hemodynamics , Humans , Magnetic Resonance Angiography , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
J Neurosci ; 22(16): 7218-24, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12177216

ABSTRACT

APP23 transgenic mice overexpressing amyloid precursor protein (APP751) reproduce neuropathological changes associated with Alzheimer's disease such as high levels of amyloid plaques, cerebral amyloid angiopathy, and associated vascular pathologies. Functional magnetic resonance imaging (fMRI) was applied to characterize brain functionality in these mice through global pharmacological stimulation. The cerebral hemodynamic response to infusion of the GABA(A) antagonist bicuculline was significantly reduced in aged APP23 mice compared with age-matched wild-type littermates. This is in part attributable to a compromised cerebrovascular reactivity, as revealed by the reduced responsiveness to vasodilatory stimulation by acetazolamide. The study shows that fMRI is a sensitive tool to phenotype genetically engineered animals modeling neuropathologies.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/biosynthesis , Brain/blood supply , Brain/physiopathology , Hemodynamics , Acetazolamide/pharmacology , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Bicuculline/pharmacology , Body Weight , Brain/drug effects , Carbonic Anhydrase Inhibitors/pharmacology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance/genetics , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Hemodynamics/drug effects , Hemodynamics/genetics , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL