Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Neurosci ; 44(5)2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38050105

ABSTRACT

Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by subtle broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.


Subject(s)
Alzheimer Disease , Amyloidosis , Mice , Humans , Animals , Neurons/physiology , Synapses/physiology , Neuronal Plasticity/physiology
2.
J Neurosci ; 34(10): 3517-22, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24599452

ABSTRACT

During development, experience plays a crucial role in sculpting neuronal connections. Patterned neural activity guides formation of functional neural circuits through the selective stabilization of some synapses and the pruning of others. Activity-regulated factors are fundamental to this process, but their roles in synapse stabilization and maturation is still poorly understood. CPG15, encoded by the activity-regulated gene candidate plasticity gene 15, is a small, glycosylphosphatidylinositol (GPI)-linked, extracellular protein that promotes synapse stabilization. Here we show that global knock-out of cpg15 results in abnormal postnatal development of the excitatory network in visual cortex and an associated disruption in development of visual receptive field properties. In addition, whereas repeated stimulation induced potentiation and depression in wild-type mice, the depression was slower in cpg15 knock-out mice, suggesting impairment in short-term depression-like mechanisms. These findings establish the requirement for cpg15 in activity-dependent development of the visual system and demonstrate the importance of timely excitatory network development for normal visual function.


Subject(s)
Nerve Net/metabolism , Nerve Tissue Proteins/deficiency , Neuronal Plasticity/physiology , Visual Cortex/metabolism , Visual Pathways/metabolism , Animals , Excitatory Postsynaptic Potentials/physiology , Female , GPI-Linked Proteins/deficiency , Male , Mice , Mice, Knockout , Nerve Net/growth & development , Visual Cortex/growth & development , Visual Pathways/growth & development
3.
J Neurosci ; 33(17): 7245-52, 2013 Apr 24.
Article in English | MEDLINE | ID: mdl-23616533

ABSTRACT

The small GTPase Rap1 contributes to fear learning and cortico-amygdala plasticity by inhibiting glutamate release from cortical neurons, but mechanisms of this inhibition remain unknown. Conversely, L-type calcium channels (LTCCs) become involved in glutamate release after fear learning and LTP induction. Here, we show that Rap1 deletion in mouse primary cortical neurons increases synaptic vesicle exocytosis without altering endocytosis or vesicle pool size in an LTCC-dependent manner. We identify Erk1/2 as the downstream effector of Rap1 and show that its inhibition increases plasma membrane expression of LTCCs near presynaptic terminals. We propose that the Rap1 signaling enables plasticity and fear learning by regulating LTCCs at cortico-amygdala synapses.


Subject(s)
Calcium Channels, L-Type/metabolism , Exocytosis/physiology , Neurotransmitter Agents/antagonists & inhibitors , Neurotransmitter Agents/metabolism , Signal Transduction/physiology , rap1 GTP-Binding Proteins/deficiency , Animals , Cells, Cultured , Female , Male , Mice , Neurons/metabolism , rap1 GTP-Binding Proteins/genetics
4.
Opt Express ; 22(18): 21368-81, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25321515

ABSTRACT

Multiphoton excitation fluorescence microscopy is the preferred method for in vivo deep tissue imaging. Many biological applications demand both high imaging speed and the ability to resolve multiple fluorophores. One of the successful methods to improve imaging speed in a highly turbid specimen is multifocal multiphoton microscopy (MMM) based on use of multi-anode photomultiplier tubes (MAPMT). This approach improves imaging speed by using multiple foci for parallelized excitation without sacrificing signal to noise ratio (SNR) due to the scattering of emission photons. In this work, we demonstrate that the MAPMT based MMM can be extended with spectral resolved imaging capability. Instead of generating multiple excitation foci in a 2D grid pattern, a linear array of foci is generated. This leaves one axis of the 2D MAPMT available for spectral dispersion and detection. The spectral-resolved MMM can detect several emission signals simultaneously with high imaging speed optimized for high-throughput, high-contents applications. The new procedure is illustrated using imaging data from the kidney, peripheral nerve regeneration and dendritic morphological data from the brain.


Subject(s)
Microscopy, Fluorescence, Multiphoton/instrumentation , Photons , Fluorescence
5.
Neurosci Lett ; 836: 137874, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38857696

ABSTRACT

Clusterin is a secreted glycoprotein that participates in multiple physiological processes through its chaperon function. In Alzheimer's disease, the brain functions under an increased oxidative stress condition that causes an elevation of protein oxidation, resulting in enhanced pathology. Accordingly, it is important to determine the type of human brain cells that are mostly prone to methionine oxidation in Alzheimer's disease and specifically monitoring the methionine-oxidation levels of clusterin in human and mice brains and its effect on clusterin's function. We analyzed the level of methionine sulfoxide (MetO)-clusterin in these brains, using a combination of immunoprecipitation and Western-blott analyses. Also, we determine the effect of methionine oxidation on clusterin ability to bind beta-amyloid, in vitro, using calorimetric assay. Our results show that human neurons and astrocytes of Alzheimer's disease brains are mostly affected by methionine oxidation. Moreover, MetO-clusterin levels are elevated in postmortem Alzheimer's disease human and mouse brains in comparison to controls. Finally, oxidation of methionine residues of purified clusterin reduced its binding efficiency to beta-amyloid. In conclusion, we suggest that methionine oxidation of brain-clusterin is enhanced in Alzheimer's disease and that this oxidation compromises its chaperon function, leading to exacerbation of beta-amyloid's toxicity in Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Astrocytes , Brain , Clusterin , Methionine , Oxidation-Reduction , Aged , Animals , Humans , Male , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Brain/metabolism , Clusterin/metabolism , Methionine/metabolism , Methionine/analogs & derivatives , Neurons/metabolism , Protein Binding
6.
bioRxiv ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-36712054

ABSTRACT

While Alzheimer's disease (AD) has been extensively studied with a focus on cognitive networks, sensory network dysfunction has received comparatively less attention despite compelling evidence of its significance in both Alzheimer's disease patients and mouse models. We recently found that neurons in the primary visual cortex of an amyloid mouse model exhibit an imbalance of postsynaptic structures favoring neuronal hyperactivity alongside increased c-Fos expression, which regulates plasticity and memory. Here, we investigate aberrant visual network and brain-wide c-Fos expression and functional connectivity patterns, network responses to light deprivation, and visual system presynaptic deficits of a mouse model of Alzheimer's disease. We found that the mouse model of AD exhibits aberrant c-Fos expression and functional connectivity patterns across multiple brain regions, and functional connectivity between brain regions is a significant predictor for aberrant c-Fos expression. We also show that one week of light deprivation increases c-Fos expression across the brain in nonpathological controls but not the AD model, indicating experience-dependent plasticity deficits in multiple brain regions. Using in vivo and ex vivo imaging of presynaptic termini, we found that aberrant visual cortical c-Fos expression is associated with selective loss of excitatory cortical but not inhibitory or subcortical synapses. Our findings reveal novel structural and functional connectivity deficits in the visual network pre-plaque amyloidosis.

7.
J Alzheimers Dis ; 101(1): 111-131, 2024.
Article in English | MEDLINE | ID: mdl-39121131

ABSTRACT

Background: While Alzheimer's disease (AD) has been extensively studied with a focus on cognitive networks, visual network dysfunction has received less attention despite compelling evidence of its significance in AD patients and mouse models. We recently reported c-Fos and synaptic dysregulation in the primary visual cortex of a pre-amyloid plaque AD-model. Objective: We test whether c-Fos expression and presynaptic density/dynamics differ in cortical and subcortical visual areas in an AD-model. We also examine whether aberrant c-Fos expression is inherited through functional connectivity and shaped by light experience. Methods: c-Fos+ cell density, functional connectivity, and their experience-dependent modulation were assessed for visual and whole-brain networks in both sexes of 4-6-month-old J20 (AD-model) and wildtype (WT) mice. Cortical and subcortical differences in presynaptic vulnerability in the AD-model were compared using ex vivo and in vivo imaging. Results: Visual cortical, but not subcortical, networks show aberrant c-Fos expression and impaired experience-dependent modulation. The average functional connectivity of a brain region in WT mice significantly predicts aberrant c-Fos expression, which correlates with impaired experience-dependent modulation in the AD-model. We observed a subtle yet selective weakening of excitatory visual cortical synapses. The size distribution of cortical boutons in the AD-model is downscaled relative to those in WT mice, suggesting a synaptic scaling-like adaptation of bouton size. Conclusions: Visual network structural and functional disruptions are biased toward cortical regions in pre-plaque J20 mice, and the cellular and synaptic dysregulation in the AD-model represents a maladaptive modification of the baseline physiology seen in WT conditions.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Proto-Oncogene Proteins c-fos , Synapses , Animals , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Synapses/pathology , Synapses/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Mice , Male , Female , Visual Cortex/metabolism , Visual Cortex/pathology , Mice, Inbred C57BL
8.
bioRxiv ; 2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37205469

ABSTRACT

Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by the broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.

9.
bioRxiv ; 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-36865208

ABSTRACT

Familiarity creates subjective memory of repeated innocuous experiences, reduces neural and behavioral responsiveness to those experiences, and enhances novelty detection. The neural correlates of the internal model of familiarity and the cellular mechanisms of enhanced novelty detection following multi-day repeated passive experience remain elusive. Using the mouse visual cortex as a model system, we test how the repeated passive experience of a 45° orientation-grating stimulus for multiple days alters spontaneous and non-familiar stimuli evoked neural activity in neurons tuned to familiar or non-familiar stimuli. We found that familiarity elicits stimulus competition such that stimulus selectivity reduces in neurons tuned to the familiar 45° stimulus; it increases in those tuned to the 90° stimulus but does not affect neurons tuned to the orthogonal 135° stimulus. Furthermore, neurons tuned to orientations 45° apart from the familiar stimulus dominate local functional connectivity. Interestingly, responsiveness to natural images, which consists of familiar and non-familiar orientations, increases subtly in neurons that exhibit stimulus competition. We also show the similarity between familiar grating stimulus-evoked and spontaneous activity increases, indicative of an internal model of altered experience.

10.
Sci Rep ; 13(1): 20907, 2023 11 27.
Article in English | MEDLINE | ID: mdl-38017135

ABSTRACT

Familiarity creates subjective memory of repeated innocuous experiences, reduces neural and behavioral responsiveness to those experiences, and enhances novelty detection. The neural correlates of the internal model of familiarity and the cellular mechanisms of enhanced novelty detection following multi-day repeated passive experience remain elusive. Using the mouse visual cortex as a model system, we test how the repeated passive experience of a 45° orientation-grating stimulus for multiple days alters spontaneous and non-familiar stimuli evoked neural activity in neurons tuned to familiar or non-familiar stimuli. We found that familiarity elicits stimulus competition such that stimulus selectivity reduces in neurons tuned to the familiar 45° stimulus; it increases in those tuned to the 90° stimulus but does not affect neurons tuned to the orthogonal 135° stimulus. Furthermore, neurons tuned to orientations 45° apart from the familiar stimulus dominate local functional connectivity. Interestingly, responsiveness to natural images, which consists of familiar and non-familiar orientations, increases subtly in neurons that exhibit stimulus competition. We also show the similarity between familiar grating stimulus-evoked and spontaneous activity increases, indicative of an internal model of altered experience.


Subject(s)
Recognition, Psychology , Visual Cortex , Mice , Animals , Recognition, Psychology/physiology , Neurons/physiology , Photic Stimulation
11.
Cell Rep ; 42(1): 111946, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640331

ABSTRACT

Neuronal hyperactivity induces memory deficits in Alzheimer's disease. However, how hyperactivity disrupts memory is unclear. Using in vivo synaptic imaging in the mouse visual cortex, we show that structural excitatory-inhibitory synapse imbalance in the apical dendrites favors hyperactivity in early amyloidosis. Consistent with this, natural images elicit neuronal hyperactivity in these mice. Compensatory changes that maintain activity homeostasis disrupt functional connectivity and increase population sparseness such that a small fraction of neurons dominates population activity. These properties reduce the selectivity of neural response to natural images and render visual recognition memory vulnerable to interference. Deprivation of non-specific visual experiences improves the neural representation and behavioral expression of visual familiarity. In contrast, in non-pathological conditions, deprivation of non-specific visual experiences induces disinhibition, increases excitability, and disrupts visual familiarity. We show that disrupted familiarity occurs when the fraction of high-responsive neurons and the persistence of neural representation of a memory-associated stimulus are not constrained.


Subject(s)
Alzheimer Disease , Neurons , Mice , Animals , Neurons/metabolism , Dendrites , Alzheimer Disease/metabolism , Homeostasis/physiology , Recognition, Psychology , Amyloidogenic Proteins/metabolism
12.
J Neurosci ; 31(12): 4755-64, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21430174

ABSTRACT

L-type calcium channels play only a minor role in basal neurotransmitter release in brain neurons but contribute significantly after induction of plasticity. Very little is known about mechanisms that enable L-type calcium channel participation in neurotransmitter release. Here, using mouse primary cortical neurons, we found that inhibition of Erk1/2 (extracellular signal-regulated kinases 1 and 2) enhanced synaptic vesicle exocytosis by increasing calcium influx through L-type calcium channels. Furthermore, inhibition of Erk1/2 increased the surface fraction of these channels. These findings indicate a novel inhibitory effect of Erk1/2 on synaptic transmission through L-type calcium channels.


Subject(s)
Calcium Channels, L-Type/physiology , Exocytosis/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Synaptic Vesicles/physiology , Animals , Axons/physiology , Calcium/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/physiology , DNA/genetics , Electric Stimulation , Endocytosis/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Pregnancy , Transfection
13.
Front Cell Neurosci ; 14: 592607, 2020.
Article in English | MEDLINE | ID: mdl-33408613

ABSTRACT

Synapse loss is the strongest correlate for cognitive decline in Alzheimer's disease. The mechanisms underlying synapse loss have been extensively investigated using mouse models expressing genes with human familial Alzheimer's disease mutations. In this review, we summarize how multiphoton in vivo imaging has improved our understanding of synapse loss mechanisms associated with excessive amyloid in the living animal brain. We also discuss evidence obtained from these imaging studies for the role of cell-intrinsic calcium dyshomeostasis and cell-extrinsic activities of microglia, which are the immune cells of the brain, in mediating synapse loss.

14.
Methods Mol Biol ; 557: 165-81, 2009.
Article in English | MEDLINE | ID: mdl-19799182

ABSTRACT

The study of location and intensity of double-strand breaks (DSBs) in mammalian systems is more challenging than in yeast because, unlike yeast, the progression through meiosis is not synchronous and only a small fraction of all testis cells are actually at the stage where DSB formation is initiated. We devised a quantitative approach that is sensitive enough to detect the position of rare DNA strand breaks in mouse germ cell-enriched testicular cell populations. The method can detect DNA breaks at any desired location in the genome but is not specific for DSBs-overhangs, nicks, or gaps with a free 3' OH group are also detected. The method was successfully used to compare testicular cells from mouse strains that possess or lack an active recombination hot spot at the H2-Ea gene. Breaks that were due to meiotic hot spot activity could be distinguished from the background of DNA breaks. This highly sensitive approach could be used to study other biological processes where rare DNA breaks are generated.


Subject(s)
DNA Breaks, Double-Stranded , DNA Mutational Analysis/methods , Germ Cells/metabolism , Testis/metabolism , Animals , Base Sequence , Germ Cells/cytology , Male , Mice , Models, Biological , Polymerase Chain Reaction/methods , Restriction Mapping/methods , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/cytology
15.
Cell Rep ; 28(6): 1584-1595.e5, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390571

ABSTRACT

A key feature of brain plasticity is the experience-dependent selection of optimal connections, implemented by a set of activity-regulated genes that dynamically adjust synapse strength and number. The activity-regulated gene cpg15/neuritin has been previously implicated in stabilization and maturation of excitatory synapses. Here, we combine two-photon microscopy with genetic and sensory manipulations to dissect excitatory synapse formation in vivo and examine the role of activity and CPG15 in dendritic spine formation, PSD95 recruitment, and synapse stabilization. We find that neither visual experience nor CPG15 is required for spine formation. However, PSD95 recruitment to nascent spines and their subsequent stabilization requires both. Further, cell-autonomous CPG15 expression is sufficient to replace experience in facilitating PSD95 recruitment and spine stabilization. CPG15 directly interacts with α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on immature dendritic spines, suggesting a signaling mode for this small extracellular molecule acting as an experience-dependent "selector" for spine stabilization and synapse maturation.


Subject(s)
Dendritic Spines/metabolism , Disks Large Homolog 4 Protein/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials , Female , GPI-Linked Proteins/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Neuronal Plasticity , Receptors, AMPA/metabolism
17.
Genetics ; 171(2): 427-41, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15965249

ABSTRACT

Recombination and microsatellite mutation in humans contribute to disorders including cancer and trinucleotide repeat (TNR) disease. TNR expansions in wild-type yeast may arise by flap ligation during lagging-strand replication. Here we show that overexpression of DNA ligase I (CDC9) increases the rates of TNR expansion, of TNR contraction, and of mitotic recombination. Surprisingly, this effect is observed with catalytically inactive forms of Cdc9p protein, but only if they possess a functional PCNA-binding site. Furthermore, in vitro analysis indicates that the interaction of PCNA with Cdc9p and Rad27p (Fen1) is mutually exclusive. Together our genetic and biochemical analysis suggests that, although DNA ligase I seals DNA nicks during replication, repair, and recombination, higher than normal levels can yield genetic instability by disrupting the normal interplay of PCNA with other proteins such as Fen1.


Subject(s)
DNA Ligases/metabolism , Gene Expression , Genomic Instability/genetics , Proliferating Cell Nuclear Antigen/metabolism , Recombination, Genetic/genetics , Saccharomycetales/genetics , Trinucleotide Repeat Expansion/genetics , Acetyltransferases , Cloning, Molecular , DNA Ligase ATP , DNA Ligases/genetics , DNA Primers , Flap Endonucleases/metabolism , Gene Deletion , Immunoblotting , Membrane Proteins/metabolism , Mutagenesis, Site-Directed , Proliferating Cell Nuclear Antigen/genetics , Saccharomyces cerevisiae Proteins/metabolism
18.
Neuron ; 91(2): 205-7, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27477010

ABSTRACT

In this issue of Neuron, Fossati et al. (2016) report that through its domain structure, SRGAP2A, a Rho-GTPase-activating protein, can co-regulate excitatory and inhibitory synapse development, offering a putative evolutionary genetic mechanism for preserving excitatory/inhibitory balance during speciation.


Subject(s)
GTPase-Activating Proteins/genetics , Neurons/metabolism , Synapses/physiology , Animals , GTPase-Activating Proteins/metabolism , Humans , Mutation/genetics
20.
Neuron ; 89(4): 756-69, 2016 02 17.
Article in English | MEDLINE | ID: mdl-26853302

ABSTRACT

Older concepts of a hard-wired adult brain have been overturned in recent years by in vivo imaging studies revealing synaptic remodeling, now thought to mediate rearrangements in microcircuit connectivity. Using three-color labeling and spectrally resolved two-photon microscopy, we monitor in parallel the daily structural dynamics (assembly or removal) of excitatory and inhibitory postsynaptic sites on the same neurons in mouse visual cortex in vivo. We find that dynamic inhibitory synapses often disappear and reappear again in the same location. The starkest contrast between excitatory and inhibitory synapse dynamics is on dually innervated spines, where inhibitory synapses frequently recur while excitatory synapses are stable. Monocular deprivation, a model of sensory input-dependent plasticity, shortens inhibitory synapse lifetimes and lengthens intervals to recurrence, resulting in a new dynamic state with reduced inhibitory synaptic presence. Reversible structural dynamics indicate a fundamentally new role for inhibitory synaptic remodeling--flexible, input-specific modulation of stable excitatory connections.


Subject(s)
Neural Inhibition/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/ultrastructure , Synapses/physiology , Synaptic Transmission/physiology , Visual Cortex/cytology , Animals , Carrier Proteins/metabolism , Disks Large Homolog 4 Protein , Female , Functional Laterality , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/genetics , Organ Culture Techniques , Pregnancy , Sensory Deprivation , Synapses/ultrastructure , gamma-Aminobutyric Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL