Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Theranostics ; 11(14): 6644-6667, 2021.
Article in English | MEDLINE | ID: mdl-34093845

ABSTRACT

Mouse models of Alzheimer's disease (AD) are valuable but do not fully recapitulate human AD pathology, such as spontaneous Tau fibril accumulation and neuronal loss, necessitating the development of new AD models. The transgenic (TG) TgF344-AD rat has been reported to develop age-dependent AD features including neuronal loss and neurofibrillary tangles, despite only expressing APP and PSEN1 mutations, suggesting an improved modelling of AD hallmarks. Alterations in neuronal networks as well as learning performance and cognition tasks have been reported in this model, but none have combined a longitudinal, multimodal approach across multiple centres, which mimics the approaches commonly taken in clinical studies. We therefore aimed to further characterise the progression of AD-like pathology and cognition in the TgF344-AD rat from young-adults (6 months (m)) to mid- (12 m) and advanced-stage (18 m, 25 m) of the disease. Methods: TgF344-AD rats and wild-type (WT) littermates were imaged at 6 m, 12 m and 18 m with [18F]DPA-714 (TSPO, neuroinflammation), [18F]Florbetaben (Aß) and [18F]ASEM (α7-nicotinic acetylcholine receptor) and with magnetic resonance spectroscopy (MRS) and with (S)-[18F]THK5117 (Tau) at 15 and 25 m. Behaviour tests were also performed at 6 m, 12 m and 18 m. Immunohistochemistry (CD11b, GFAP, Aß, NeuN, NeuroChrom) and Tau (S)-[18F]THK5117 autoradiography, immunohistochemistry and Western blot were also performed. Results: [18F]DPA-714 positron emission tomography (PET) showed an increase in neuroinflammation in TG vs wildtype animals from 12 m in the hippocampus (+11%), and at the advanced-stage AD in the hippocampus (+12%), the thalamus (+11%) and frontal cortex (+14%). This finding coincided with strong increases in brain microgliosis (CD11b) and astrogliosis (GFAP) at these time-points as assessed by immunohistochemistry. In vivo [18F]ASEM PET revealed an age-dependent increase uptake in the striatum and pallidum/nucleus basalis of Meynert in WT only, similar to that observed with this tracer in humans, resulting in TG being significantly lower than WT by 18 m. In vivo [18F]Florbetaben PET scanning detected Aß accumulation at 18 m, and (S)-[18F]THK5117 PET revealed subsequent Tau accumulation at 25m in hippocampal and cortical regions. Aß plaques were low but detectable by immunohistochemistry from 6 m, increasing further at 12 and 18 m with Tau-positive neurons adjacent to Aß plaques at 18 m. NeuroChrom (a pan neuronal marker) immunohistochemistry revealed a loss of neuronal staining at the Aß plaques locations, while NeuN labelling revealed an age-dependent decrease in hippocampal neuron number in both genotypes. Behavioural assessment using the novel object recognition task revealed that both WT & TgF344-AD animals discriminated the novel from familiar object at 3 m and 6 m of age. However, low levels of exploration observed in both genotypes at later time-points resulted in neither genotype successfully completing the task. Deficits in social interaction were only observed at 3 m in the TgF344-AD animals. By in vivo MRS, we showed a decrease in neuronal marker N-acetyl-aspartate in the hippocampus at 18 m (-18% vs age-matched WT, and -31% vs 6 m TG) and increased Taurine in the cortex of TG (+35% vs age-matched WT, and +55% vs 6 m TG). Conclusions: This multi-centre multi-modal study demonstrates, for the first time, alterations in brain metabolites, cholinergic receptors and neuroinflammation in vivo in this model, validated by robust ex vivo approaches. Our data confirm that, unlike mouse models, the TgF344-AD express Tau pathology that can be detected via PET, albeit later than by ex vivo techniques, and is a useful model to assess and longitudinally monitor early neurotransmission dysfunction and neuroinflammation in AD.


Subject(s)
Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Magnetic Resonance Spectroscopy , Plaque, Amyloid/metabolism , Positron-Emission Tomography , tau Proteins/metabolism , Aging/metabolism , Aging/physiology , Alzheimer Disease/pathology , Animals , Behavior Rating Scale , Cognitive Dysfunction/genetics , Cognitive Dysfunction/physiopathology , Disease Models, Animal , Female , Fluorine Radioisotopes , Frontal Lobe/metabolism , Frontal Lobe/pathology , Gliosis/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Inflammation/metabolism , Locomotion/genetics , Locomotion/physiology , Male , Neurons/metabolism , Neurons/pathology , Rats , Rats, Transgenic , Receptors, Cholinergic/metabolism , Thalamus/metabolism , Thalamus/pathology
3.
Theranostics ; 10(18): 7938-7955, 2020.
Article in English | MEDLINE | ID: mdl-32724451

ABSTRACT

The sigma 1 receptor (S1R) is widely expressed in the CNS and is mainly located on the endoplasmic reticulum. The S1R is involved in the regulation of many neurotransmission systems and, indirectly, in neurodegenerative diseases. The S1R may therefore represent an interesting neuronal biomarker in neurodegenerative diseases such as Parkinson's (PD) or Alzheimer's diseases (AD). Here we present the characterisation of the S1R-specific 18F-labelled tracer 18F-IAM6067 in two animal models and in human brain tissue. Methods: Wistar rats were used for PET-CT imaging (60 min dynamic acquisition) and metabolite analysis (1, 2, 5, 10, 20, 60 min post-injection). To verify in vivo selectivity, haloperidol, BD1047 (S1R ligand), CM398 (S2R ligand) and SB206553 (5HT2B/C antagonist) were administrated for pre-saturation studies. Excitotoxic lesions induced by intra-striatal injection of AMPA were also imaged by 18F-IAM6067 PET-CT to test the sensitivity of the methods in a well-established model of neuronal loss. Tracer brain uptake was also verified by autoradiography in rats and in a mouse model of PD (intrastriatal 6-hydroxydopamine (6-OHDA) unilateral lesion). Finally, human cortical binding was investigated by autoradiography in three groups of subjects (control subjects with Braak ≤2, and AD patients, Braak >2 & ≤4 and Braak >4 stages). Results: We demonstrate that despite rapid peripheral metabolism of 18F-IAM6067, radiolabelled metabolites were hardly detected in brain samples. Brain uptake of 18F-IAM6067 showed differences in S1R anatomical distribution, namely from high to low uptake: pons-raphe, thalamus medio-dorsal, substantia nigra, hypothalamus, cerebellum, cortical areas and striatum. Pre-saturation studies showed 79-90% blockade of the binding in all areas of the brain indicated above except with the 5HT2B/C antagonist SB206553 and S2R ligand CM398 which induced no significant blockade, indicating good specificity of 18F-IAM6067 for S1Rs. No difference between ipsi- and contralateral sides of the brain in the mouse model of PD was detected. AMPA lesion induced a significant 69% decrease in 18F-IAM6067 uptake in the globus pallidus matching the neuronal loss as measured by NeuN, but only a trend to decrease (-16%) in the caudate putamen despite a significant 91% decrease in neuronal count. Moreover, no difference in the human cortical binding was shown between AD groups and controls. Conclusion: This work shows that 18F-IAM6067 is a specific and selective S1R radiotracer. The absence or small changes in S1R detected here in animal models and human tissue warrants further investigations and suggests that S1R might not be the anticipated ideal biomarker for neuronal loss in neurodegenerative diseases such as AD and PD.


Subject(s)
Alzheimer Disease/diagnosis , Brain/diagnostic imaging , Parkinson Disease, Secondary/diagnosis , Radiopharmaceuticals/administration & dosage , Receptors, sigma/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Autoradiography , Brain/pathology , Disease Models, Animal , Female , Fluorine Radioisotopes/administration & dosage , Humans , Male , Middle Aged , Molecular Imaging/methods , Oxidopamine/administration & dosage , Oxidopamine/toxicity , Parkinson Disease, Secondary/etiology , Parkinson Disease, Secondary/pathology , Positron Emission Tomography Computed Tomography/methods , Rats , Rats, Wistar , Sigma-1 Receptor
4.
Neuroimage ; 184: 349-358, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30219292

ABSTRACT

Blood-brain barrier (BBB) breakdown has been hypothesized to play a key role in the onset and progression of Alzheimer's disease (AD). However, the question of whether AD itself contributes to loss of BBB integrity is still uncertain, as many in-vivo studies have failed to detect signs of AD-related BBB breakdown. We hypothesize AD-related BBB damage is subtle, and that these negative results arise from a lack of measurement sensitivity. With the aim of developing a more sensitive measure of BBB breakdown, we have designed a novel MRI scanning protocol to quantify the trans-BBB exchange of endogenous water. Using this method, we detect increased BBB water permeability in a rat model of AD that is associated with reduced expression of the tight junction protein occludin. BBB permeability to MRI contrast agent, assessed using dynamic contrast-enhanced (DCE)-MRI, did not differ between transgenic and wild-type animals and was uncorrelated with occludin expression. Our data supports the occurrence of AD-related BBB breakdown, and indicates that such BBB pathology is subtle and may be undetectable using existing 'tracer leakage' methods. Our validated water-exchange MRI method provides a new powerful tool with which to study BBB damage in-vivo.


Subject(s)
Alzheimer Disease/pathology , Blood-Brain Barrier/pathology , Magnetic Resonance Imaging/methods , Animals , Brain/pathology , Capillary Permeability/physiology , Rats , Rats, Transgenic , Water/analysis
5.
Sci Rep ; 7(1): 4955, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694463

ABSTRACT

Gadolinium (Gd)-stained MRI is based on Gd contrast agent (CA) administration into the brain parenchyma. The strong signal increase induced by Gd CA can be converted into resolution enhancement to record microscopic MR images. Moreover, inhomogeneous distribution of the Gd CA in the brain improves the contrast between different tissues and provides new contrasts in MR images. Gd-stained MRI detects amyloid plaques, one of the microscopic lesions of Alzheimer's disease (AD), in APPSL/PS1M146L mice or in primates. Numerous transgenic mice with various plaque typologies have been developed to mimic cerebral amyloidosis and comparison of plaque detection between animal models and humans with new imaging methods is a recurrent concern. Here, we investigated detection of amyloid plaques by Gd-stained MRI in five mouse models of amyloidosis (APPSL/PS1M146L, APP/PS1dE9, APP23, APPSwDI, and 3xTg) presenting with compact, diffuse and intracellular plaques as well as in post mortem human-AD brains. The brains were then evaluated by histology to investigate the impact of size, compactness, and iron load of amyloid plaques on their detection by MRI. We show that Gd-stained MRI allows detection of compact amyloid plaques as small as 25 µm, independently of their iron load, in mice as well as in human-AD brains.


Subject(s)
Alzheimer Disease/diagnostic imaging , Amyloidosis/diagnostic imaging , Magnetic Resonance Imaging/methods , Plaque, Amyloid/diagnostic imaging , Alzheimer Disease/metabolism , Amyloidosis/metabolism , Animals , Autopsy , Contrast Media/administration & dosage , Disease Models, Animal , Gadolinium/administration & dosage , Humans , Iron/metabolism , Mice , Mice, Transgenic , Plaque, Amyloid/metabolism
6.
Immunol Lett ; 188: 89-95, 2017 08.
Article in English | MEDLINE | ID: mdl-28690185

ABSTRACT

The two most used methods to select camelid single-domain antibody-fragments (VHHs) are: displaying their repertoires on the surface of filamentous bacteriophages (phage display) or linking them to ribosomes (ribosome display). In this study, we compared specific VHHs isolated from two different immune libraries coming from two different alpacas by using these two selection methods. Three anti-GFAP (glial fibrillary acidic protein) VHHs were derived from an immune library obtained by ribosome display after immunization of one alpaca with purified GFAP, a protein expressed by astroglial cells. In parallel, three other anti-GFAP VHHs were derived from an immune library by phage display after immunization of another alpaca with a human brain tissue extract containing GFAP. All the VHHs were closely related and one VHH was found to be strictly identical in both studies. This highlights the selection pressure exerted by the camelid immune system to shape the paratope of an antibody against a defined antigen.


Subject(s)
Cell Surface Display Techniques , Gene Library , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Amino Acid Sequence , Animals , Antibody Affinity/immunology , Glial Fibrillary Acidic Protein/immunology , Humans , Protein Binding/immunology , Single-Domain Antibodies/chemistry , Workflow
7.
MAbs ; 9(6): 1016-1027, 2017.
Article in English | MEDLINE | ID: mdl-28657418

ABSTRACT

Today, molecular imaging of neurodegenerative diseases is mainly based on small molecule probes. Alternatively, antibodies are versatile tools that may be developed as new imaging agents. Indeed, they can be readily obtained to specifically target any antigen of interest and their scaffold can be functionalized. One of the critical issues involved in translating antibody-based probes to the clinic is the design and synthesis of perfectly-defined conjugates. Camelid single-domain antibody-fragments (VHHs) are very small and stable antibodies that are able to diffuse in tissues and potentially cross the blood brain barrier (BBB). Here, we selected a VHH (R3VQ) specifically targeting one of the main lesions of Alzheimer's disease (AD), namely the amyloid-beta (Aß) deposits. It was used as a scaffold for the design of imaging probes for magnetic resonance imaging (MRI) and labeled with the contrastophore gadolinium using either a random or site-specific approach. In contrast to the random strategy, the site-specific conjugation to a single reduced cysteine in the C-terminal part of the R3VQ generates a well-defined bioconjugate in a high yield process. This new imaging probe is able to cross the BBB and label Aß deposits after intravenous injection. Also, it displays improved r1 and r2 relaxivities, up to 30 times higher than a widely used clinical contrast agent, and it allows MRI detection of amyloid deposits in post mortem brain tissue of a mouse model of AD. The ability to produce chemically-defined VHH conjugates that cross the BBB opens the way for future development of tailored imaging probes targeting intracerebral antigens.

8.
J Control Release ; 243: 1-10, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27671875

ABSTRACT

Detection of intracerebral targets with imaging probes is challenging due to the non-permissive nature of blood-brain barrier (BBB). The present work describes two novel single-domain antibodies (VHHs or nanobodies) that specifically recognize extracellular amyloid deposits and intracellular tau neurofibrillary tangles, the two core lesions of Alzheimer's disease (AD). Following intravenous administration in transgenic mouse models of AD, in vivo real-time two-photon microscopy showed gradual extravasation of the VHHs across the BBB, diffusion in the parenchyma and labeling of amyloid deposits and neurofibrillary tangles. Our results demonstrate that VHHs can be used as specific BBB-permeable probes for both extracellular and intracellular brain targets and suggest new avenues for therapeutic and diagnostic applications in neurology.


Subject(s)
Camelids, New World/immunology , Neurofibrillary Tangles/immunology , Plaque, Amyloid/immunology , Single-Domain Antibodies/immunology , Administration, Intravenous , Alzheimer Disease/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Disease Models, Animal , Male , Mice , Mice, Transgenic , Microscopy/methods , Single-Domain Antibodies/metabolism , Tissue Distribution
9.
J Neurosci ; 35(6): 2817-29, 2015 Feb 11.
Article in English | MEDLINE | ID: mdl-25673868

ABSTRACT

Astrocyte reactivity is a hallmark of neurodegenerative diseases (ND), but its effects on disease outcomes remain highly debated. Elucidation of the signaling cascades inducing reactivity in astrocytes during ND would help characterize the function of these cells and identify novel molecular targets to modulate disease progression. The Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway is associated with reactive astrocytes in models of acute injury, but it is unknown whether this pathway is directly responsible for astrocyte reactivity in progressive pathological conditions such as ND. In this study, we examined whether the JAK/STAT3 pathway promotes astrocyte reactivity in several animal models of ND. The JAK/STAT3 pathway was activated in reactive astrocytes in two transgenic mouse models of Alzheimer's disease and in a mouse and a nonhuman primate lentiviral vector-based model of Huntington's disease (HD). To determine whether this cascade was instrumental for astrocyte reactivity, we used a lentiviral vector that specifically targets astrocytes in vivo to overexpress the endogenous inhibitor of the JAK/STAT3 pathway [suppressor of cytokine signaling 3 (SOCS3)]. SOCS3 significantly inhibited this pathway in astrocytes, prevented astrocyte reactivity, and decreased microglial activation in models of both diseases. Inhibition of the JAK/STAT3 pathway within reactive astrocytes also increased the number of huntingtin aggregates, a neuropathological hallmark of HD, but did not influence neuronal death. Our data demonstrate that the JAK/STAT3 pathway is a common mediator of astrocyte reactivity that is highly conserved between disease states, species, and brain regions. This universal signaling cascade represents a potent target to study the role of reactive astrocytes in ND.


Subject(s)
Alzheimer Disease/physiopathology , Astrocytes , Huntington Disease/physiopathology , Janus Kinases , STAT3 Transcription Factor , Signal Transduction , Alzheimer Disease/pathology , Animals , Electron Transport Complex IV/metabolism , Humans , Huntington Disease/pathology , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
10.
Neurobiol Aging ; 34(6): 1599-609, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23352115

ABSTRACT

The present study investigates in aged mice the working memory (WM) enhancing potential of the selective α4ß2* nicotinic receptor agonist S 38232 as compared with the cholinesterase inhibitor donepezil, and their effect on cAMP response element binding protein (CREB) phosphorylation (pCREB) as a marker of neuronal activity. We first showed that aged mice exhibit a WM deficit and an increase of pCREB in the prelimbic cortex (PL) as compared with young mice, whereas no modification appears in the CA1. Further, we showed that systemic administration of S 38232 restored WM in aged mice and alleviated PL CREB overphosphorylation. Donepezil alleviated age-related memory deficits, however, by increasing pCREB in the CA1, while pCREB in PL remained unaffected. Finally, whereas neuronal inhibition by lidocaine infusion in the PL appeared deleterious in young mice, the infusion of Rp-cAMPS (a compound known to inhibit CREB phosphorylation) or S 38232 rescued WM in aged animals. Thus, by targeting the α4ß2*-nicotinic receptor of the PL, S 38232 alleviates PL CREB overphosphorylation and restores WM in aged mice, which opens new pharmacologic perspectives of therapeutic strategy.


Subject(s)
Aging/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Memory Disorders/metabolism , Receptors, Nicotinic/biosynthesis , Aging/drug effects , Animals , Cholinesterase Inhibitors/pharmacology , Cyclic AMP/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Male , Memory Disorders/drug therapy , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Nicotinic Agonists/pharmacology , Nicotinic Agonists/therapeutic use , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
11.
J Neurosci ; 31(46): 16517-28, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090478

ABSTRACT

The multiple memory systems hypothesis posits that different neural circuits function in parallel and may compete for information processing and storage. For example, instrumental conditioning would depend on the striatum, whereas spatial memory may be mediated by a circuit centered on the hippocampus. However, the nature of the task itself is not sufficient to select durably one system over the other. In this study, we investigated the effects of natural and pharmacological rewards on the selection of a particular memory system during learning. We compared the effects of food- or drug-induced activation of the reward system on cue-guided versus spatial learning using a Y-maze discrimination task. Drug-induced reward severely impaired the acquisition of a spatial discrimination task but spared the cued version of the task. Immunohistochemical analysis of the phosphorylated form of the cAMP response element binding (CREB) protein and c-Fos expression induced by behavioral testing revealed that the spatial deficit was associated with a decrease of both markers within the hippocampus and the prefrontal cortex. In contrast, drug reward potentiated the cued learning-induced CREB phosphorylation within the dorsal striatum. Administration of the protein kinase A inhibitor 8-Bromo-adenosine-3',5'-cyclic monophosphorothioate Rp isomer (Rp-cAMPS) into the dorsal striatum before training completely reversed the drug-induced spatial deficit and restored CREB phosphorylation levels within the hippocampus and the prefrontal cortex. Therefore, drug-induced striatal hyperactivity may underlie the declarative memory deficit reported here. This mechanism could represent an important early step toward the development of addictive behaviors by promoting conditioning to the detriment of more flexible forms of memory.


Subject(s)
CREB-Binding Protein/metabolism , Corpus Striatum/metabolism , Cues , Cyclic AMP-Dependent Protein Kinases/metabolism , Reward , Signal Transduction/physiology , Space Perception/physiology , Analysis of Variance , Animals , Behavior, Animal , Brain Mapping , Choice Behavior/drug effects , Corpus Striatum/drug effects , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Discrimination, Psychological/drug effects , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Microinjections/methods , Morphine/administration & dosage , Narcotics/administration & dosage , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Reaction Time/drug effects , Signal Transduction/drug effects , Space Perception/drug effects , Thionucleotides/pharmacology , Ventral Tegmental Area/drug effects
12.
Psychopharmacology (Berl) ; 215(4): 709-20, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21274701

ABSTRACT

RATIONALE: Normal or pathological ageing is characterized by working-memory dysfunction paired with a marked reduction in several neurotransmitters activity. The development of therapeutic strategy centered on the glutamatergic system known to bear a critical role in cognitive functions, is therefore of major importance in the treatment of mild forms of AD or age-related memory dysfunctions. OBJECTIVES: In Experiment 1, we investigated the effects of ageing on spatial working memory measured by sequential alternation (SA). Thus, the decay of alternation rates over a series of trials separated by varying intertrial temporal intervals (ITI, from 5 sec to 180 sec) was studied in mice of different age groups. In Experiment 2, we investigated the memory-enhancing potential of S 18986--a modulator of AMPA receptors--on age-related SA impairments, in comparison with memantine--an antagonist of NMDA receptors--. RESULTS: In Experiment 1, aged mice responded at chance with shorter ITI's and exhibited greater levels of interference in the SA task as compared to young adult mice. In Experiment 2, (1) S 18986 at 0.03 and 0.1 mg/kg reversed the memory deficit in aged mice but did not modify performance in young adult mice; (2) memantine at 10 mg/kg also increased SA rates in aged mice but did not improve performance in young adult mice. CONCLUSION: The SA task is a useful tool to reveal age-induced time-dependent working memory impairments. As compared to memantine, S 18986--a compound targeting AMPA receptors--contributes a valuable therapy in the treatment of age-related cognitive dysfunctions or mild forms of AD.


Subject(s)
Aging/drug effects , Benzothiadiazines/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Memantine/therapeutic use , Memory Disorders/prevention & control , Memory, Short-Term/drug effects , Aging/metabolism , Aging/psychology , Alzheimer Disease/drug therapy , Animals , Behavior, Animal/drug effects , Benzothiadiazines/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Male , Maze Learning/drug effects , Memantine/administration & dosage , Memory Disorders/metabolism , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...