Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Brain Res ; 1836: 148938, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38615924

ABSTRACT

Prepulse inhibition (PPI) of the auditory startle response, a key measure of sensorimotor gating, diminishes with age and is impaired in various neurological conditions. While PPI deficits are often associated with cognitive impairments, their reversal is routinely used in experimental systems for antipsychotic drug screening. Yet, the cellular and circuit-level mechanisms of PPI remain unclear, even under non-pathological conditions. We recently showed that brainstem neurons located in the caudal pontine reticular nucleus (PnC) expressing the glycine transporter type 2 (GlyT2±) receive inputs from the central nucleus of the amygdala (CeA) and contribute to PPI but via an uncharted pathway. Here, using tract-tracing, immunohistochemistry and in vitro optogenetic manipulations coupled to field electrophysiological recordings, we reveal the neuroanatomical distribution of GlyT2± PnC neurons and PnC-projecting CeA glutamatergic neurons and we provide mechanistic insights on how these glutamatergic inputs suppress auditory neurotransmission in PnC sections. Additionally, in vivo experiments using GlyT2-Cre mice confirm that optogenetic activation of GlyT2± PnC neurons enhances PPI and is sufficient to induce PPI in young mice, emphasizing their role. However, in older mice, PPI decline is not further influenced by inhibiting GlyT2± neurons. This study highlights the importance of GlyT2± PnC neurons in PPI and underscores their diminished activity in age-related PPI decline.


Subject(s)
Brain Stem , Glycine Plasma Membrane Transport Proteins , Glycine , Neurons , Prepulse Inhibition , Reflex, Startle , Animals , Prepulse Inhibition/physiology , Neurons/physiology , Neurons/metabolism , Reflex, Startle/physiology , Mice , Brain Stem/physiology , Brain Stem/metabolism , Glycine Plasma Membrane Transport Proteins/metabolism , Male , Glycine/metabolism , Optogenetics , Mice, Transgenic , Mice, Inbred C57BL , Synaptic Transmission/physiology , Central Amygdaloid Nucleus/physiology , Central Amygdaloid Nucleus/metabolism
2.
Am J Physiol Heart Circ Physiol ; 325(5): H1088-H1098, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37712922

ABSTRACT

Cigarette smoke exposure is a well-known risk factor for developing numerous chronic health conditions, including pulmonary disease and cardiometabolic disorders. However, the cellular mechanisms mediating the toxicity of cigarette smoke in extrapulmonary tissues are still poorly understood. Therefore, the purpose of this study was to characterize the acute dose-dependent toxicity of cigarette smoke on mitochondrial metabolism by determining the susceptibility and sensitivity of mitochondrial respiration from murine skeletal (gastrocnemius and soleus) and cardiac muscles, as well as the aorta to cigarette smoke concentrate (CSC). In all tissues, exposure to CSC inhibited tissue-specific respiration capacity, measured by high-resolution respirometry, according to a biphasic pattern. With a break point of 451 ± 235 µg/mL, the aorta was the least susceptible to CSC-induced mitochondrial respiration inhibition compared with the gastrocnemius (151 ± 109 µg/mL; P = 0.008, d = 2.3), soleus (211 ± 107 µg/mL; P = 0.112; d = 1.7), and heart (94 ± 51 µg/mL; P < 0.001; d = 2.6) suggesting an intrinsic resistance of the vascular smooth muscle mitochondria to cigarette smoke toxicity. In contrast, the cardiac muscle was the most susceptible and sensitive to the effects of CSC, demonstrating the greatest decline in tissue-specific respiration with increasing CSC concentration (P < 0.001, except the soleus). However, when normalized to citrate synthase activity to account for differences in mitochondrial content, cardiac fibers' sensitivity to cigarette smoke inhibition was no longer significantly different from both fast-twitch gastrocnemius and slow-twitch soleus muscle fibers, thus suggesting similar mitochondrial phenotypes. Collectively, these findings established the acute dose-dependent toxicity of cigarette smoke on oxidative phosphorylation in permeabilized tissues involved in the development of smoke-related cardiometabolic diseases.NEW & NOTEWORTHY Despite numerous investigations into the mechanisms underlying cigarette smoke-induced mitochondrial dysfunction, no studies have investigated the tissue-specific mitochondrial toxicity to cigarette smoke. We demonstrate that, while aorta is least sensitive and susceptible to cigarette smoke-induced toxicity, the degree of cigarette smoke-induced toxicity in striated muscle depends on the tissue-specific mitochondrial content. We conclude that while the mitochondrial content influences cigarette smoke-induced toxicity in striated muscles, aorta is intrinsically protected against cigarette smoke-induced mitochondrial toxicity.


Subject(s)
Cardiovascular Diseases , Cigarette Smoking , Mice , Humans , Animals , Oxidative Phosphorylation , Muscle, Skeletal/metabolism , Cell Respiration/physiology
3.
BMC Biol ; 19(1): 116, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34082731

ABSTRACT

BACKGROUND: Sensorimotor gating is a fundamental pre-attentive process that is defined as the inhibition of a motor response by a sensory event. Sensorimotor gating, commonly measured using the prepulse inhibition (PPI) of the auditory startle reflex task, is impaired in patients suffering from various neurological and psychiatric disorders. PPI deficits are a hallmark of schizophrenia, and they are often associated with attention and other cognitive impairments. Although the reversal of PPI deficits in animal models is widely used in pre-clinical research for antipsychotic drug screening, the neurotransmitter systems and synaptic mechanisms underlying PPI are still not resolved, even under physiological conditions. Recent evidence ruled out the longstanding hypothesis that PPI is mediated by midbrain cholinergic inputs to the caudal pontine reticular nucleus (PnC). Instead, glutamatergic, glycinergic, and GABAergic inhibitory mechanisms are now suggested to be crucial for PPI, at the PnC level. Since amygdalar dysfunctions alter PPI and are common to pathologies displaying sensorimotor gating deficits, the present study was designed to test that direct projections to the PnC originating from the amygdala contribute to PPI. RESULTS: Using wild type and transgenic mice expressing eGFP under the control of the glycine transporter type 2 promoter (GlyT2-eGFP mice), we first employed tract-tracing, morphological reconstructions, and immunohistochemical analyses to demonstrate that the central nucleus of the amygdala (CeA) sends glutamatergic inputs lateroventrally to PnC neurons, including GlyT2+ cells. Then, we showed the contribution of the CeA-PnC excitatory synapses to PPI in vivo by demonstrating that optogenetic inhibition of this connection decreases PPI, and optogenetic activation induces partial PPI. Finally, in GlyT2-Cre mice, whole-cell recordings of GlyT2+ PnC neurons in vitro paired with optogenetic stimulation of CeA fibers, as well as photo-inhibition of GlyT2+ PnC neurons in vivo, allowed us to implicate GlyT2+ neurons in the PPI pathway. CONCLUSIONS: Our results uncover a feedforward inhibitory mechanism within the brainstem startle circuit by which amygdalar glutamatergic inputs and GlyT2+ PnC neurons contribute to PPI. We are providing new insights to the clinically relevant theoretical construct of PPI, which is disrupted in various neuropsychiatric and neurological diseases.


Subject(s)
Prepulse Inhibition , Reflex, Startle , Acoustic Stimulation , Amygdala , Animals , Carcinoembryonic Antigen , Humans , Mice
4.
Antioxidants (Basel) ; 8(10)2019 Sep 25.
Article in English | MEDLINE | ID: mdl-31557847

ABSTRACT

Creosote bush (Larrea tridentata; LT) leaves extracts were tested for their potential efficacy to mitigate cellular oxidative stress on human SH-SY5Y cells. Here, the differential nuclear staining assay, a bioimager system, and flow cytometric protocols, concurrently with several specific chemicals, were used to measure the percentage of cell viability and several facets implicated in the cytoprotective mechanism of LT extracts. Initially, three LT extracts, prepared with different solvents, ethanol, ethanol:water (e/w), and water, were tested for their capacity to rescue the viability of cells undergoing aggressive H2O2-induced oxidative stress. Results indicate that the LT extract prepared with a mixture of ethanol:water (LT-e/w; 60:40% v/v) displayed the most effective cytoprotection rescue activity. Interestingly, by investigating the LT-e/w mechanism of action, it was found that LT-e/w extract decreases the levels of H2O2-provoked reactive oxidative species (ROS) accumulation, mitochondrial depolarization, phosphatidylserine externalization, caspase-3/7 activation, and poly (ADP-ribose) polymerase (PARP) cleavage significantly, which are hallmarks of apoptosis. Thus, out of the three LT extracts tested, our findings highlight that the LT-e/w extract was the most effective protective reagent on SH-SY5Y cells undergoing oxidative stress in vitro, functioning as a natural anti-apoptotic extract. These findings warrant further LT-e/w extract examination in a holistic context.

5.
Brain Behav ; 9(8): e01307, 2019 08.
Article in English | MEDLINE | ID: mdl-31268249

ABSTRACT

INTRODUCTION: Lead (Pb) exposure yielding blood lead levels (BLL) as low as 2 µg/dl in children is an international problem. More common in US low-income neighborhoods, childhood Pb exposure can cause behavioral and cognitive deficits, including working memory impairments, which can persist into adulthood. So far, studies characterized short-term effects of high Pb exposure on neuronal structure and function. However, long-term consequences of early chronic Pb exposure on neuronal activity are poorly documented. METHODS: Here, we exposed male and female mice (PND [postnatal day] 0 to PND 28) to one of three Pb treatments: 0 ppm (sodium-treated water, control), 30 ppm (low dose), and 330 ppm (high dose) lead acetate. Once the male and female mice were 9-12 months old, extracellular field recordings on hippocampal slices were performed. RESULTS: We show that at CA3 to CA1 synapses, synaptic transmission was decreased and neuronal fiber activity was increased in males exposed to lowest level Pb. In contrast, both synaptic transmission and neuronal fiber activity were increased in females exposed to high Pb. The ventral hippocampus-medial prefrontal cortex (vHPC-mPFC) synapses are crucial for working memory in rodents. The lowest level Pb decreased vHPC-mPFC synaptic transmission, whereas high Pb decreased short-term synaptic depression. CONCLUSIONS: Overall, we show for the first time that early exposure to either high or lowest level Pb has long-term consequences on different synaptic properties of at least two hippocampal synapses. Such consequences of early Pb exposure might worsen the cognitive decline observed in aging men and women. Our results suggest that additional efforts should focus on the consequences of early Pb exposure especially in at-risk communities.


Subject(s)
Environmental Exposure/adverse effects , Lead/toxicity , Neurons/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Age Factors , Animals , Female , Hippocampus/drug effects , Male , Memory, Short-Term/drug effects , Mice , Optogenetics , Time
6.
Molecules ; 23(7)2018 Jul 23.
Article in English | MEDLINE | ID: mdl-30041415

ABSTRACT

Oxidative stress has been linked to neurodegenerative diseases such as Huntington's, Parkinson's, Alzheimer's and amyotrophic lateral sclerosis diseases. Larrea tridentata (LT) also known as Creosote Bush is an evergreen shrub found in the Chihuahuan desert which has been used medicinally by Native American tribes in southwestern North America and the Amerindians of South America. However, studies of the antioxidant capacity of the crude extract of LT towards the discovery of novel molecular therapies bearing antioxidants and drug-like properties are lacking. In this study, we assessed the antioxidant properties of Larrea tridentata, collected specifically from the Chihuahuan desert in the region of El Paso del Norte, TX, USA. LT phytochemicals were obtained from three different extracts (ethanol; ethanol: water (60:40) and water). Then the extracts were evaluated in eight different assays (DPPH, ABTS, superoxide; FRAP activity, nitric oxide, phenolic content, UV visible absorption and cytotoxicity in non-cancerous HS27 cells). The three extracts were not affecting the HS27 cells at concentrations up to 120 µg/mL. Among the three extracts, we found that the mixture of ethanol: water (60:40) LT extract has the most efficient antioxidant properties (IC50 (DPPH at 30 min) = 111.7 ± 3.8 µg/mL; IC50 (ABTS) = 8.49 ± 2.28 µg/mL; IC50 (superoxide) = 0.43 ± 0.17 µg/mL; IC50 (NO) = 230.4 ± 130.4 µg/mL; and the highest phenolic content was estimated to 212.46 ± 7.05 mg GAE/L). In addition, there was a strong correlation between phenolic content and the free-radical scavenging activity assays. HPLC-MS study identified nine compounds from the LT-ethanol: water extract including Justicidin B and Beta peltain have been previously reported as secondary metabolites of Larrea tridentata.


Subject(s)
Antioxidants/pharmacology , Larrea/chemistry , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Antioxidants/chemistry , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Flavonoids/chemistry , Free Radical Scavengers/chemistry , Free Radical Scavengers/pharmacology , Humans , Mass Spectrometry , Molecular Structure , Nitric Oxide/metabolism , Phenols/chemistry , Phytochemicals/chemistry , Phytochemicals/pharmacology , Plant Extracts/chemistry , Plant Leaves
7.
Proc Natl Acad Sci U S A ; 114(30): E6127-E6136, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696314

ABSTRACT

Identification of protective loss-of-function (LoF) mutations holds great promise for devising novel therapeutic interventions, although it faces challenges due to the scarcity of protective LoF alleles in the human genome. Exploiting the detailed mechanistic characterization of animal models of validated disease mutations offers an alternative. Here, we provide insights into protective-variant biology based on our characterization of a model of the 22q11.2 deletion, a strong genetic risk factor for schizophrenia (SCZ). Postnatal brain up-regulation of Mirta22/Emc10, an inhibitor of neuronal maturation, represents the major transcriptional effect of the 22q11.2-associated microRNA dysregulation. Here, we demonstrate that mice in which the Df(16)A deficiency is combined with a LoF Mirta22 allele show rescue of key SCZ-related deficits, namely prepulse inhibition decrease, working memory impairment, and social memory deficits, as well as synaptic and structural plasticity abnormalities in the prefrontal cortex. Additional analysis of homozygous Mirta22 knockout mice, in which no alteration is observed in the above-mentioned SCZ-related phenotypes, highlights the deleterious effects of Mirta22 up-regulation. Our results support a causal link between dysregulation of a miRNA target and SCZ-related deficits and provide key insights into beneficial LoF mutations and potential new treatments.


Subject(s)
Loss of Function Mutation , Membrane Proteins/genetics , Schizophrenia/genetics , Animals , Female , Genetic Predisposition to Disease , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/genetics , Phenotype , Schizophrenia/physiopathology
8.
J Neurosci ; 37(15): 4158-4180, 2017 04 12.
Article in English | MEDLINE | ID: mdl-28283561

ABSTRACT

Using a genetic mouse model that faithfully recapitulates a DISC1 genetic alteration strongly associated with schizophrenia and other psychiatric disorders, we examined the impact of this mutation within the prefrontal cortex. Although cortical layering, cytoarchitecture, and proteome were found to be largely unaffected, electrophysiological examination of the mPFC revealed both neuronal hyperexcitability and alterations in short-term synaptic plasticity consistent with enhanced neurotransmitter release. Increased excitability of layer II/III pyramidal neurons was accompanied by consistent reductions in voltage-activated potassium currents near the action potential threshold as well as by enhanced recruitment of inputs arising from superficial layers to layer V. We further observed reductions in both the paired-pulse ratios and the enhanced short-term depression of layer V synapses arising from superficial layers consistent with enhanced neurotransmitter release at these synapses. Recordings from layer II/III pyramidal neurons revealed action potential widening that could account for enhanced neurotransmitter release. Significantly, we found that reduced functional expression of the voltage-dependent potassium channel subunit Kv1.1 substantially contributes to both the excitability and short-term plasticity alterations that we observed. The underlying dysregulation of Kv1.1 expression was attributable to cAMP elevations in the PFC secondary to reduced phosphodiesterase 4 activity present in Disc1 deficiency and was rescued by pharmacological blockade of adenylate cyclase. Our results demonstrate a potentially devastating impact of Disc1 deficiency on neural circuit function, partly due to Kv1.1 dysregulation that leads to a dual dysfunction consisting of enhanced neuronal excitability and altered short-term synaptic plasticity.SIGNIFICANCE STATEMENT Schizophrenia is a profoundly disabling psychiatric illness with a devastating impact not only upon the afflicted but also upon their families and the broader society. Although the underlying causes of schizophrenia remain poorly understood, a growing body of studies has identified and strongly implicated various specific risk genes in schizophrenia pathogenesis. Here, using a genetic mouse model, we explored the impact of one of the most highly penetrant schizophrenia risk genes, DISC1, upon the medial prefrontal cortex, the region believed to be most prominently dysfunctional in schizophrenia. We found substantial derangements in both neuronal excitability and short-term synaptic plasticity-parameters that critically govern neural circuit information processing-suggesting that similar changes may critically, and more broadly, underlie the neural computational dysfunction prototypical of schizophrenia.


Subject(s)
Action Potentials/physiology , Disease Models, Animal , Neuronal Plasticity/physiology , Neurons/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Prefrontal Cortex/physiopathology , Pregnancy , Schizophrenia/genetics , Schizophrenia/physiopathology
9.
Protein J ; 34(5): 349-58, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26385697

ABSTRACT

Endoplasmic reticulum (ER) proteins including protein disulfide isomerase (PDI) are playing crucial roles in maintaining appropriate protein folding. Under nitrosative stress, an excess of nitric oxide (NO) radical species induced the S-nitrosylation of PDI cysteines which eliminate its isomerase and oxidoreductase capabilities. In addition, the S-nitrosylation-PDI complex is the cause of aggregation especially of the α-synuclein (α-syn) protein (accumulation of Lewy-body aggregates). We recently identified a potent antioxidant small molecule, Ferrostatin-1 (Fer-1), that was able to inhibit a non-apoptotic cell death named ferroptosis. Ferroptosis cell death involved the generation of oxidative stress particularly lipid peroxide. In this work, we reported the neuroprotective role of ferrostatin-1 under rotenone-induced oxidative stress in dopaminergic neuroblastoma cells (SH-SY5Y). We first synthesized the Fer-1 and confirmed that it is not toxic toward the SH-SY5Y cells at concentrations up to 12.5 µM. Second, we showed that Fer-1 compound quenched the commercially available stable radical, the 2,2-diphenyl-1-picrylhydrazyl (DPPH), in non-cellular assay at 82 %. Third, Fer-1 inhibited the ROS/RNS generated under rotenone insult in SH-SY5Y cells. Fourth, we revealed the effective role of Fer-1 in ER stress mediated activation of apoptotic pathway. Finally, we reported that Fer-1 mitigated rotenone-induced α-syn aggregation.


Subject(s)
Cyclohexylamines/pharmacology , Dopamine/metabolism , Neuroblastoma/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Phenylenediamines/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Neuroblastoma/physiopathology , Neurons/cytology , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Rotenone/toxicity
10.
Neuron ; 86(3): 680-95, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25913858

ABSTRACT

22q11.2 deletion carriers show specific cognitive deficits, and ∼30% of them develop schizophrenia. One of the disrupted genes is ZDHHC8, which encodes for a palmitoyltransferase. We show that Zdhhc8-deficient mice have reduced palmitoylation of proteins that regulate axonal growth and branching. Analysis of axonal projections of pyramidal neurons from both Zdhhc8-deficient and Df(16)A(+/-) mice, which model the 22q11.2 deletion, revealed deficits in axonal growth and terminal arborization, which can be prevented by reintroduction of active ZDHHC8 protein. Impaired terminal arborization is accompanied by a reduction in the strength of synaptic connections and altered functional connectivity and working memory. The effect of ZDHHC8 is mediated in part via Cdc42-dependent modulation of Akt/Gsk3ß signaling at the tip of the axon and can be reversed by pharmacologically decreasing Gsk3ß activity during postnatal brain development. Our findings provide valuable mechanistic insights into the cognitive and psychiatric symptoms associated with a schizophrenia-predisposing mutation.


Subject(s)
Alzheimer Disease/pathology , Axons/pathology , Brain/pathology , DiGeorge Syndrome/pathology , Acyltransferases/deficiency , Acyltransferases/genetics , Age Factors , Alzheimer Disease/genetics , Animals , Animals, Newborn , Brain/embryology , Brain/metabolism , Channelrhodopsins , DiGeorge Syndrome/genetics , Disease Models, Animal , Embryo, Mammalian , Excitatory Postsynaptic Potentials/genetics , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins , Humans , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Transgenic , Neural Pathways/embryology , Neural Pathways/growth & development , Neural Pathways/physiology , Neurons/pathology , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Signal Transduction/genetics , Synapsins/metabolism
11.
J Neurosci ; 33(37): 14825-39, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027283

ABSTRACT

We used a mouse model of the schizophrenia-predisposing 22q11.2 microdeletion to evaluate how this genetic lesion affects cortical neural circuits at the synaptic, cellular, and molecular levels. Guided by cognitive deficits, we demonstrated that mutant mice display robust deficits in high-frequency synaptic transmission and short-term plasticity (synaptic depression and potentiation), as well as alterations in long-term plasticity and dendritic spine stability. Apart from previously reported reduction in dendritic complexity of layer 5 pyramidal neurons, altered synaptic plasticity occurs in the context of relatively circumscribed and often subtle cytoarchitectural changes in neuronal density and inhibitory neuron numbers. We confirmed the pronounced DiGeorge critical region 8 (Dgcr8)-dependent deficits in primary micro-RNA processing and identified additional changes in gene expression and RNA splicing that may underlie the effects of this mutation. Reduction in Dgcr8 levels appears to be a major driver of altered short-term synaptic plasticity in prefrontal cortex and working memory but not of long-term plasticity and cytoarchitecture. Our findings inform the cortical synaptic and neuronal mechanisms of working memory impairment in the context of psychiatric disorders. They also provide insight into the link between micro-RNA dysregulation and genetic liability to schizophrenia and cognitive dysfunction.


Subject(s)
DiGeorge Syndrome/pathology , Long-Term Potentiation/genetics , Long-Term Synaptic Depression/genetics , Neurons/physiology , Prefrontal Cortex/pathology , Animals , Cognition Disorders/etiology , Cognition Disorders/genetics , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , DiGeorge Syndrome/complications , DiGeorge Syndrome/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Phosphopyruvate Hydratase/metabolism , Proteins/genetics , RNA-Binding Proteins , Recognition, Psychology/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
12.
J Gen Physiol ; 140(4): 403-19, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23008434

ABSTRACT

Experiments were performed to characterize the properties of the intrinsic Ca(2+) buffers in the sarcoplasmic reticulum (SR) of cut fibers from frog twitch muscle. The concentrations of total and free calcium ions within the SR ([Ca(T)](SR) and [Ca(2+)](SR)) were measured, respectively, with the EGTA/phenol red method and tetramethylmurexide (a low affinity Ca(2+) indicator). Results indicate SR Ca(2+) buffering was consistent with a single cooperative-binding component or a combination of a cooperative-binding component and a linear binding component accounting for 20% or less of the bound Ca(2+). Under the assumption of a single cooperative-binding component, the most likely resting values of [Ca(2+)](SR) and [Ca(T)](SR) are 0.67 and 17.1 mM, respectively, and the dissociation constant, Hill coefficient, and concentration of the Ca-binding sites are 0.78 mM, 3.0, and 44 mM, respectively. This information can be used to calculate a variable proportional to the Ca(2+) permeability of the SR, namely d[Ca(T)](SR)/dt ÷ [Ca(2+)](SR) (denoted release permeability), in experiments in which only [Ca(T)](SR) or [Ca(2+)](SR) is measured. In response to a voltage-clamp step to -20 mV at 15°C, the release permeability reaches an early peak followed by a rapid decline to a quasi-steady level that lasts ~50 ms, followed by a slower decline during which the release permeability decreases by at least threefold. During the quasi-steady level of release, the release amplitude is 3.3-fold greater than expected from voltage activation alone, a result consistent with the recruitment by Ca-induced Ca(2+) release of 2.3 SR Ca(2+) release channels neighboring each channel activated by its associated voltage sensor. Release permeability at -60 mV increases as [Ca(T)](SR) decreases from its resting physiological level to ~0.1 of this level. This result argues against a release termination mechanism proposed in mammalian muscle fibers in which a luminal sensor of [Ca(2+)](SR) inhibits release when [Ca(T)](SR) declines to a low level.


Subject(s)
Calcium Signaling , Calcium/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Buffers , Calsequestrin/metabolism , Hydrogen-Ion Concentration , Membrane Potentials , Muscle Fibers, Fast-Twitch/physiology , Rana temporaria
13.
Mol Cell Neurosci ; 47(4): 293-305, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21635953

ABSTRACT

22q11.2 chromosomal deletions are recurrent copy number mutations that increase the risk of schizophrenia around thirty-fold. Deletion of the orthologous chromosomal region in mice offers an opportunity to characterize changes to neuronal structure and function that may account for the development of this disease. The hippocampus has been implicated in schizophrenia pathogenesis, is reduced in volume in 22q11.2 deletion carriers and displays altered neuronal structure in a mouse model of the mutation (Df(16)A(+/-) mice). Here we investigate hippocampal CA1 physiology, hippocampal-dependent spatial memory and novelty-induced hippocampal activation in Df(16)A(+/-) mice. We found normal spatial reference memory (as assayed by the Morris water maze test) as well as modest but potentially important deficits in physiology. In particular, a reduction in the level of inhibition of CA1 pyramidal neurons was observed, implying a decrease in interneuron activity. Additionally, deficits in LTP were observed using certain induction protocols. Induction of c-Fos expression by exploration of a novel environment suggested a relative sparing of CA1 and dentate gyrus function but showed a robust decrease in the number of activated CA3 pyramidal neurons in Df(16)A(+/-) mice. Overall, experiments performed in this 22q11.2 deletion model demonstrated deficits of various degrees across different regions of the hippocampus, which together may contribute to the increased risk of developing schizophrenia.


Subject(s)
Chromosome Deletion , Hippocampus/physiology , Models, Animal , Action Potentials/physiology , Animals , Chromosomes, Human, Pair 22 , Humans , Interneurons/metabolism , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , Risk Factors , Schizophrenia/genetics
14.
Proc Natl Acad Sci U S A ; 108(11): 4447-52, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21368174

ABSTRACT

Individuals with 22q11.2 microdeletions have cognitive and behavioral impairments and the highest known genetic risk for developing schizophrenia. One gene disrupted by the 22q11.2 microdeletion is DGCR8, a component of the "microprocessor" complex that is essential for microRNA production, resulting in abnormal processing of specific brain miRNAs and working memory deficits. Here, we determine the effect of Dgcr8 deficiency on the structure and function of cortical circuits by assessing their laminar organization, as well as the neuronal morphology, and intrinsic and synaptic properties of layer 5 pyramidal neurons in the prefrontal cortex of Dgcr8(+/-) mutant mice. We found that heterozygous Dgcr8 mutant mice have slightly fewer cortical layer 2/4 neurons and that the basal dendrites of layer 5 pyramidal neurons have slightly smaller spines. In addition to the modest structural changes, field potential and whole-cell electrophysiological recordings performed in layer 5 of the prefrontal cortex revealed greater short-term synaptic depression during brief stimulation trains applied at 50 Hz to superficial cortical layers. This finding was accompanied by a decrease in the initial phase of synaptic potentiation. Our results identify altered short-term plasticity as a neural substrate underlying the cognitive dysfunction and the increased risk for schizophrenia associated with the 22q11.2 microdeletions.


Subject(s)
Gene Deletion , Neuronal Plasticity/physiology , Prefrontal Cortex/physiopathology , Proteins/metabolism , Animals , CA1 Region, Hippocampal/physiopathology , CA3 Region, Hippocampal/physiopathology , Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , Dendritic Spines/metabolism , Dendritic Spines/pathology , Excitatory Postsynaptic Potentials/physiology , Mice , Prefrontal Cortex/pathology , RNA-Binding Proteins , Synapses/metabolism , Time Factors
15.
Int J Dev Neurosci ; 29(3): 259-81, 2011 May.
Article in English | MEDLINE | ID: mdl-20920576

ABSTRACT

Over the last fifteen years it has become established that 22q11.2 deletion syndrome (22q11DS) is a true genetic risk factor for schizophrenia. Carriers of deletions in chromosome 22q11.2 develop schizophrenia at rate of 25-30% and such deletions account for as many as 1-2% of cases of sporadic schizophrenia in the general population. Access to a relatively homogeneous population of individuals that suffer from schizophrenia as the result of a shared etiological factor and the potential to generate etiologically valid mouse models provides an immense opportunity to better understand the pathobiology of this disease. In this review we survey the clinical literature associated with the 22q11.2 microdeletions with a focus on neuroanatomical changes. Then, we highlight results from work modeling this structural mutation in animals. The key biological pathways disrupted by the mutation are discussed and how these changes impact the structure and function of neural circuits is described.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 22/genetics , Genetic Predisposition to Disease , Mental Disorders/genetics , Mental Disorders/pathology , Animals , Brain/abnormalities , Brain/physiology , Brain/physiopathology , Catechol O-Methyltransferase/genetics , Catechol O-Methyltransferase/metabolism , Disease Models, Animal , Epistasis, Genetic , Humans , Induced Pluripotent Stem Cells/physiology , MicroRNAs/metabolism , Models, Genetic , Proline Oxidase/genetics , Proline Oxidase/metabolism , Schizophrenia/genetics , Syndrome
16.
J Neurosci ; 30(2): 523-33, 2010 Jan 13.
Article in English | MEDLINE | ID: mdl-20071515

ABSTRACT

A unilateral activation of the mesencephalic locomotor region (MLR) produces symmetrical bilateral locomotion in all vertebrate species tested to date. How this occurs remains unresolved. This study examined the possibility that the symmetry occurred at the level of the inputs from the MLR to reticulospinal (RS) cells. In lamprey semi-intact preparations, we recorded intracellular responses of pairs of large, homologous RS cells on both sides to stimulation of the MLR on one side. The synaptic responses on both sides were very similar in shape, amplitude, and threshold intensity. Increasing MLR stimulation intensity produced a symmetrical increase in the magnitude of the responses on both sides. Ca(2+) imaging confirmed the bilateral activation of smaller-sized RS cells as well. In a high-divalent cation solution, the synaptic responses of homologous RS cells persisted and exhibited a constant latency during high-frequency stimulation. Moreover, during gradual replacement of normal Ringer's solution with a Ca(2+)-free solution, the magnitude of responses showed a gradual reduction with a similar time course in the homologous RS cells. These results support the idea that the MLR projects monosynaptically to RS cells on both sides with symmetrical inputs. During locomotion of the semi-intact preparation, the discharge pattern was also very similar in homologous bilateral RS cells. Anatomical experiments confirmed the presence of MLR neurons projecting ipsilaterally to the reticular formation intermingled with neurons projecting contralaterally. We conclude that the bilaterally symmetrical MLR inputs to RS cells are likely contributors to generating symmetrical locomotor activity.


Subject(s)
Brain Stem/physiology , Excitatory Postsynaptic Potentials/physiology , Functional Laterality/physiology , Locomotion/physiology , Animals , Biophysics , Brain Stem/cytology , Calcium/metabolism , Electric Stimulation/methods , Electromyography/methods , Fluorescent Dyes/metabolism , In Vitro Techniques , Larva , Neurons/classification , Neurons/physiology , Petromyzon
17.
J Neurosci ; 29(4): 1140-51, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19176823

ABSTRACT

Sensory stimulation elicits sustained depolarizations in lamprey reticulospinal (RS) cells for which intrinsic properties were shown to play a crucial role. The depolarizations last up to minutes, and we tested whether the intrinsic properties required the cooperation of synaptic inputs to maintain RS cells depolarized for such long periods of time. Ascending spinal inputs to RS cells were reversibly blocked by applying xylocaine over the rostral spinal cord segments. The duration of the sustained depolarizations was markedly reduced. The membrane potential oscillations in tune with locomotor activity that were present under control condition were also abolished. The contribution of excitatory glutamatergic inputs was then assessed by applying CNQX and AP-5 over one of two simultaneously recorded homologous RS cells on each side of the brainstem. The level of sensory-evoked depolarization decreased significantly in the cell exposed to the antagonists compared with the other RS cell monitored as a control. In contrast, local application of glycine only produced a transient membrane potential hyperpolarization with a marked reduction in the amplitude of membrane potential oscillations. Locally applied strychnine did not change the duration of the sustained depolarizations, suggesting that mechanisms other than glycinergic inhibition are involved in ending the sustained depolarizations in RS cells. It is concluded that excitatory glutamatergic inputs, including ascending spinal feedback, cooperate with intrinsic properties of RS cells to maintain the cells depolarized for prolonged periods, sustaining long bouts of escape swimming.


Subject(s)
Membrane Potentials/physiology , Neurons/physiology , Reticular Formation/cytology , Spinal Cord/physiology , Synapses/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Anesthetics, Local/pharmacology , Animals , Electric Stimulation/methods , Electromyography , Evoked Potentials/drug effects , Evoked Potentials/physiology , Excitatory Amino Acid Antagonists/pharmacology , Glycine/pharmacology , Glycine Agents/pharmacology , In Vitro Techniques , Larva , Lidocaine/pharmacology , Membrane Potentials/drug effects , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Patch-Clamp Techniques/methods , Petromyzon , Strychnine/pharmacology , Synapses/drug effects , Time Factors , Trigeminal Nerve/physiology , Valine/analogs & derivatives , Valine/pharmacology
18.
Brain Res Rev ; 57(1): 172-82, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17916380

ABSTRACT

The spinal circuitry underlying the generation of basic locomotor synergies has been described in substantial detail in lampreys and the cellular mechanisms have been identified. The initiation of locomotion, on the other hand, relies on supraspinal networks and the cellular mechanisms involved are only beginning to be understood. This review examines some of the findings relative to the neural mechanisms involved in the initiation of locomotion of lampreys. Locomotion can be elicited by sensory stimulation or by internal cues associated with fundamental needs of the animal such as food seeking, exploration, and mating. We have described mechanisms by which escape swimming is elicited in lampreys in response to mechanical skin stimulation. A rather simple neural connectivity is involved, including sensory and relay neurons, as well as the brainstem rhombencephalic reticulospinal cells, which act as command neurons. We have shown that reticulospinal cells have intrinsic membrane properties that allow them to transform a short duration sensory input into a long-lasting excitatory command that activates the spinal locomotor networks. These mechanisms constitute an important feature for the activation of escape swimming. Other sensory inputs can also elicit locomotion in lampreys. For instance, we have recently shown that olfactory signals evoke sustained depolarizations in reticulospinal neurons and chemical activation of the olfactory bulbs with local injections of glutamate induces fictive locomotion. The mechanisms by which internal cues initiate locomotion are less understood. Our research has focused on one particular locomotor center in the brainstem, the mesencephalic locomotor region (MLR). The MLR is believed to channel inputs from many brain regions to generate goal-directed locomotion. It activates reticulospinal cells to elicit locomotor output in a graded fashion contrary to escape locomotor bouts, which are all-or-none. MLR inputs to reticulospinal cells use both glutamatergic and cholinergic transmission; nicotinic receptors on reticulospinal cells are involved. MLR excitatory inputs to reticulospinal cells in the middle (MRRN) are larger than those in the posterior rhombencephalic reticular nucleus (PRRN). Moreover at low stimulation strength, reticulospinal cells in the MRRN are activated first, whereas those in the PRRN require stronger stimulation strengths. The output from the MLR on one side activates reticulospinal neurons on both sides in a highly symmetrical fashion. This could account for the symmetrical bilateral locomotor output evoked during unilateral stimulation of the MLR in all animal species tested to date. Interestingly, muscarinic receptor activation reduces sensory inputs to reticulospinal neurons and, under natural conditions, the activation of MLR cholinergic neurons will likely reduce sensory inflow. Moreover, exposing the brainstem to muscarinic agonists generates sustained recurring depolarizations in reticulospinal neurons through pre-reticular effects. Cells in the caudal half of the rhombencephalon appear to be involved and we propose that the activation of these muscarinoceptive cells could provide additional excitation to reticulospinal cells when the MLR is activated under natural conditions. One important question relates to sources of inputs to the MLR. We found that substance P excites the MLR, whereas GABA inputs tonically maintain the MLR inhibited and removal of this inhibition initiates locomotion. Other locomotor centers exist such as a region in the ventral thalamus projecting directly to reticulospinal cells. This region, referred to as the diencephalic locomotor region, receives inputs from several areas in the forebrain and is likely important for goal-directed locomotion. In summary, this review focuses on the most recent findings relative to initiation of lamprey locomotion in response to sensory and internal cues in lampreys.


Subject(s)
Lampreys/physiology , Locomotion/physiology , Animals , Brain/anatomy & histology , Brain/physiology , Nervous System Physiological Phenomena , Neural Pathways/cytology , Neural Pathways/physiology , Neurons/physiology , Sensation/physiology
20.
J Physiol ; 581(Pt 1): 319-67, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17331996

ABSTRACT

Calsequestrin is a large-capacity Ca-binding protein located in the terminal cisternae of sarcoplasmic reticulum (SR) suggesting a role as a buffer of the concentration of free Ca in the SR ([Ca2+](SR)) serving to maintain the driving force for SR Ca2+ release. Essentially all of the functional studies on calsequestrin to date have been carried out on purified calsequestrin or on disrupted muscle preparations such as terminal cisternae vesicles. To obtain information about calsequestrin's properties during physiological SR Ca2+ release, experiments were carried out on frog cut skeletal muscle fibres using two optical methods. One - the EGTA-phenol red method - monitored the content of total Ca in the SR ([Ca(T)](SR)) and the other used the low affinity Ca indicator tetramethylmurexide (TMX) to monitor the concentration of free Ca in the SR. Both methods relied on a large concentration of the Ca buffer EGTA (20 mM), in the latter case to greatly reduce the increase in myoplasmic [Ca2+] caused by SR Ca2+ release thereby almost eliminating the myoplasmic component of the TMX signal. By releasing almost all of the SR Ca, these optical signals provided information about [Ca(T)](SR) versus [Ca2+](SR) as [Ca2+](SR) varied from its resting level ([Ca2+](SR,R)) to near zero. Since almost all of the Ca in the SR is bound to calsequestrin, this information closely resembles the binding curve of the Ca-calsequestrin reaction. Calcium binding to calsequestrin was found to be cooperative (estimated Hill coefficient = 2.95) and to have a very high capacity (at the start of Ca2+ release, 23 times more Ca was estimated to initiate from calsequestrin as opposed to the pool of free Ca in the SR). The latter result contrasts with an earlier report that only approximately 25% of released Ca2+ comes from calsequestrin and approximately 75% comes from the free pool. The value of [Ca2+](SR,R) was close to the K(D) for calsequestrin, which has a value near 1 mm in in vitro studies. Other evidence indicates that [Ca2+](SR,R) is near 1 mM in cut fibres. These results along with the known rapid kinetics of the Ca-calsequestrin binding reaction indicate that calsequestrin's properties are optimized to buffer [Ca2+](SR) during rapid, physiological SR Ca2+ release. Although the results do not entirely rule out a more active role in the excitation-contraction coupling process, they do indicate that passive buffering of [Ca2+](SR) is a very important function of calsequestrin.


Subject(s)
Calcium/metabolism , Calsequestrin/physiology , Muscle, Skeletal/metabolism , Sarcoplasmic Reticulum/metabolism , Action Potentials/physiology , Animals , Electric Stimulation , Models, Theoretical , Murexide/analogs & derivatives , Muscle, Skeletal/innervation , Patch-Clamp Techniques , Phenolsulfonphthalein , Rana temporaria , Signal Transduction/physiology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...