Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
bioRxiv ; 2023 May 05.
Article in English | MEDLINE | ID: mdl-37205335

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disorder affecting brain and spinal cord motor neurons. Mutations in the copper/zinc superoxide dismutase gene ( SOD1 ) are associated with ∼20% of inherited and 1-2% of sporadic ALS cases. Much has been learned from mice expressing transgenic copies of mutant SOD1, which typically involve high-level transgene expression, thereby differing from ALS patients expressing one mutant gene copy. To generate a model that more closely represents patient gene expression, we created a knock-in point mutation (G85R, a human ALS-causing mutation) in the endogenous mouse Sod1 gene, leading to mutant SOD1 G85R protein expression. Heterozygous Sod1 G85R mutant mice resemble wild type, whereas homozygous mutants have reduced body weight and lifespan, a mild neurodegenerative phenotype, and express very low mutant SOD1 protein levels with no detectable SOD1 activity. Homozygous mutants exhibit partial neuromuscular junction denervation at 3-4 months of age. Spinal cord motor neuron transcriptome analyses of homozygous Sod1 G85R mice revealed up-regulation of cholesterol synthesis pathway genes compared to wild type. Transcriptome and phenotypic features of these mice are similar to Sod1 knock-out mice, suggesting the Sod1 G85R phenotype is largely driven by loss of SOD1 function. By contrast, cholesterol synthesis genes are down-regulated in severely affected human TgSOD1 G93A transgenic mice at 4 months. Our analyses implicate dysregulation of cholesterol or related lipid pathway genes in ALS pathogenesis. The Sod1 G85R knock-in mouse is a useful ALS model to examine the importance of SOD1 activity in control of cholesterol homeostasis and motor neuron survival. SIGNIFICANCE STATEMENT: Amyotrophic lateral sclerosis is a devastating disease involving the progressive loss of motor neurons and motor function for which there is currently no cure. Understanding biological mechanisms leading to motor neuron death is critical for developing new treatments. Using a new knock-in mutant mouse model carrying a Sod1 mutation that causes ALS in patients, and in the mouse, causes a limited neurodegenerative phenotype similar to Sod1 loss-of-function, we show that cholesterol synthesis pathway genes are up-regulated in mutant motor neurons, whereas the same genes are down-regulated in transgenic SOD1 mice with a severe phenotype. Our data implicate dysregulation of cholesterol or other related lipid genes in ALS pathogenesis and provide new insights that could contribute to strategies for disease intervention.

2.
J Neurophysiol ; 126(2): 427-439, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34191630

ABSTRACT

Mammalian circadian (24 h) rhythms are timed by the pattern of spontaneous action potential firing in the suprachiasmatic nucleus (SCN). This oscillation in firing is produced through circadian regulation of several membrane currents, including large-conductance Ca2+- and voltage-activated K+ (BK) and L-type Ca2+ channel (LTCC) currents. During the day steady-state BK currents depend mostly on LTCCs for activation, whereas at night they depend predominantly on ryanodine receptors (RyRs). However, the contribution of these Ca2+ channels to BK channel activation during action potential firing has not been thoroughly investigated. In this study, we used a pharmacological approach to determine that both LTCCs and RyRs contribute to the baseline membrane potential of SCN action potential waveforms, as well as action potential-evoked BK current, during the day and night, respectively. Since the baseline membrane potential is a major determinant of circadian firing rate, we focused on the LTCCs contributing to low voltage activation of BK channels during the subthreshold phase. For these experiments, two LTCC subtypes found in SCN (CaV1.2 and CaV1.3) were coexpressed with BK channels in heterologous cells, where their differential contributions could be separately measured. CaV1.3 channels produced currents that were shifted to more hyperpolarized potentials compared with CaV1.2, resulting in increased subthreshold Ca2+ and BK currents during an action potential command. These results show that although multiple Ca2+ sources in SCN can contribute to the activation of BK current during an action potential, specific BK-CaV1.3 partnerships may optimize the subthreshold BK current activation that is critical for firing rate regulation.NEW & NOTEWORTHY BK K+ channels are important regulators of firing. Although Ca2+ channels are required for their activation in excitable cells, it is not well understood how BK channels activate using these Ca2+ sources during an action potential. This study demonstrates the differences in BK current activated by CaV1.2 and CaV1.3 channels in clock neurons and heterologous cells. The results define how specific ion channel partnerships can be engaged during distinct phases of the action potential.


Subject(s)
Action Potentials , Calcium Channels, L-Type/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology
3.
Neuroscience ; 396: A3-A20, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30594291

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease that has significant overlap with frontotemporal dementia (FTD). Mutations in specific genes have been identified that can cause and/or predispose patients to ALS. However, the clinical variability seen in ALS patients suggests that additional genes impact pathology, susceptibility, severity, and/or progression of the disease. To identify molecular pathways involved in ALS, we undertook a meta-analysis of published genetic modifiers both in patients and in model organisms, and undertook bioinformatic pathway analysis. From 72 published studies, we generated a list of 946 genes whose perturbation (1) impacted ALS in patient populations, (2) altered defects in laboratory models, or (3) modified defects caused by ALS gene ortholog loss of function. Herein, these are all called modifier genes. We found 727 modifier genes that encode proteins with human orthologs. Of these, 43 modifier genes were identified as modifiers of more than one ALS gene/model, consistent with the hypothesis that shared genes and pathways may underlie ALS. Further, we used a gene ontology-based bioinformatic analysis to identify pathways and associated genes that may be important in ALS. To our knowledge this is the first comprehensive survey of ALS modifier genes. This work suggests that shared molecular mechanisms may underlie pathology caused by different ALS disease genes. Surprisingly, few ALS modifier genes have been tested in more than one disease model. Understanding genes that modify ALS-associated defects will help to elucidate the molecular pathways that underlie ALS and provide additional targets for therapeutic intervention.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Genes, Modifier/genetics , Signal Transduction/genetics , Animals , Computational Biology , Genetic Predisposition to Disease/genetics , Humans
4.
J Gen Physiol ; 150(2): 259-275, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29237755

ABSTRACT

Large conductance K+ (BK) channels are expressed widely in neurons, where their activation is regulated by membrane depolarization and intracellular Ca2+ (Ca2+i). To enable this regulation, BK channels functionally couple to both voltage-gated Ca2+ channels (VGCCs) and channels mediating Ca2+ release from intracellular stores. However, the relationship between BK channels and their specific Ca2+ source for particular patterns of excitability is not well understood. In neurons within the suprachiasmatic nucleus (SCN)-the brain's circadian clock-BK current, VGCC current, and Ca2+i are diurnally regulated, but paradoxically, BK current is greatest at night when VGCC current and Ca2+i are reduced. Here, to determine whether diurnal regulation of Ca2+ is relevant for BK channel activation, we combine pharmacology with day and night patch-clamp recordings in acute slices of SCN. We find that activation of BK current depends primarily on three types of channels but that the relative contribution changes between day and night. BK current can be abrogated with nimodipine during the day but not at night, establishing that L-type Ca2+ channels (LTCCs) are the primary daytime Ca2+ source for BK activation. In contrast, dantrolene causes a significant decrease in BK current at night, suggesting that nighttime BK activation is driven by ryanodine receptor (RyR)-mediated Ca2+i release. The N- and P/Q-type Ca2+ channel blocker ω-conotoxin MVIIC causes a smaller reduction of BK current that does not differ between day and night. Finally, inhibition of LTCCs, but not RyRs, eliminates BK inactivation, but the BK ß2 subunit was not required for activation of BK current by LTCCs. These data reveal a dynamic coupling strategy between BK channels and their Ca2+ sources in the SCN, contributing to diurnal regulation of SCN excitability.


Subject(s)
Calcium Channels/metabolism , Circadian Clocks , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Calcium/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/physiology
5.
Nat Commun ; 7: 10837, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26940770

ABSTRACT

Inactivation is an intrinsic property of several voltage-dependent ion channels, closing the conduction pathway during membrane depolarization and dynamically regulating neuronal activity. BK K(+) channels undergo N-type inactivation via their ß2 subunit, but the physiological significance is not clear. Here, we report that inactivating BK currents predominate during the day in the suprachiasmatic nucleus, the brain's intrinsic clock circuit, reducing steady-state current levels. At night inactivation is diminished, resulting in larger BK currents. Loss of ß2 eliminates inactivation, abolishing the diurnal variation in both BK current magnitude and SCN firing, and disrupting behavioural rhythmicity. Selective restoration of inactivation via the ß2 N-terminal 'ball-and-chain' domain rescues BK current levels and firing rate, unexpectedly contributing to the subthreshold membrane properties that shift SCN neurons into the daytime 'upstate'. Our study reveals the clock employs inactivation gating as a biophysical switch to set the diurnal variation in suprachiasmatic nucleus excitability that underlies circadian rhythm.


Subject(s)
Brain/physiology , Circadian Clocks/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Electrophysiological Phenomena , Female , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/metabolism , Male , Mice , Mice, Knockout , Neurons/physiology , Patch-Clamp Techniques , Suprachiasmatic Nucleus/cytology
6.
Physiol Rep ; 3(11)2015 Nov.
Article in English | MEDLINE | ID: mdl-26537348

ABSTRACT

BK large conductance calcium-activated K(+) channels (KC a1.1) are expressed widely across many tissues, contributing to systemic regulation of cardiovascular, neurological, and other specialized physiological functions. The pore-forming α subunit is encoded by the Kcnma1 gene, originally named mSlo1 in mouse and slowpoke in Drosophila. Global deletion in mouse (Kcnma1(-/-)) produces a plethora of defects in neuron and muscle excitability, as well as other phenotypes related to channel function in nonexcitable cells. While homozygous null mice are viable, the ubiquitous loss of BK function has complicated the interpretation of phenotypes involving the interaction of multiple cell types which independently express BK channels. Here, we report the generation of a targeted allele for conditional inactivation of Kcnma1 using the Cre-loxP system (Kcnma1(fl)-tdTomato). Cre-mediated recombination generates a null allele, and BK currents were not detectable in neurons and muscle cells from Nestin-Cre; Kcnma1(fl/fl) and SM22α-Cre; Kcnma1(fl/fl) mice, respectively. tdTomato expression was detected in Cre-expressing tissues, but not in Cre-negative controls. These data demonstrate the utility of Kcnma1(fl)-tdTomato for conditional deletion of the BK channel, facilitating the understanding of tissue-specific contributions to physiological function in vivo.

7.
J Gen Physiol ; 142(6): 585-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24277602

ABSTRACT

BK Ca(2+)-activated K(+) currents exhibit diverse properties across tissues. The functional variation in voltage- and Ca(2+)-dependent gating underlying this diversity arises from multiple mechanisms, including alternate splicing of Kcnma1, the gene encoding the pore-forming (α) subunit of the BK channel, phosphorylation of α subunits, and inclusion of ß subunits in channel complexes. To address the interplay of these mechanisms in the regulation of BK currents, two native splice variants, BK0 and BKSRKR, were cloned from a tissue that exhibits dynamic daily expression of BK channel, the central circadian pacemaker in the suprachiasmatic nucleus (SCN) of mouse hypothalamus. The BK0 and BKSRKR variants differed by the inclusion of a four-amino acid alternate exon at splice site 1 (SRKR), which showed increased expression during the day. The functional properties of the variants were investigated in HEK293 cells using standard voltage-clamp protocols. Compared with BK0, BKSRKR currents had a significantly right-shifted conductance-voltage (G-V) relationship across a range of Ca(2+) concentrations, slower activation, and faster deactivation. These effects were dependent on the phosphorylation state of S642, a serine residue within the constitutive exon immediately preceding the SRKR insert. Coexpression of the neuronal ß4 subunit slowed gating kinetics and shifted the G-V relationship in a Ca(2+)-dependent manner, enhancing the functional differences between the variants. Next, using native action potential (AP) command waveforms recorded from SCN to elicit BK currents, we found that these splice variant differences persist under dynamic activation conditions in physiological ionic concentrations. AP-induced currents from BKSRKR channels were significantly reduced compared with BK0, an effect that was maintained with coexpression of the ß4 subunit but abolished by the mutation of S642. These results demonstrate a novel mechanism for reducing BK current activation under reconstituted physiological conditions, and further suggest that S642 is selectively phosphorylated in the presence of SRKR.


Subject(s)
Exons , Ion Channel Gating , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Neurons/metabolism , Serine/metabolism , Action Potentials , Alternative Splicing , Animals , Calcium/metabolism , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Mice , Neurons/physiology , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Serine/genetics , Suprachiasmatic Nucleus/cytology
8.
Am J Physiol Cell Physiol ; 304(4): C299-311, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23174562

ABSTRACT

In mammals, almost all aspects of circadian rhythmicity are attributed to activity in a discrete neural circuit of the hypothalamus, the suprachiasmatic nucleus (SCN). A 24-h rhythm in spontaneous firing is the fundamental neural intermediary to circadian behavior, but the ionic mechanisms that pattern circuit rhythmicity, and the integrated impact on behavior, are not well studied. Here, we demonstrate that daily modulation of a major component of the nighttime-phased suppressive K(+) current, encoded by the BK Ca(2+)-activated K(+) current channel (K(Ca)1.1 or Kcnma1), is a critical arbiter of circadian rhythmicity in the SCN circuit. Aberrant induction of BK current during the day in transgenic mice using a Per1 promoter (Tg-BK(R207Q)) reduced SCN firing or silenced neurons, decreasing the circadian amplitude of the ensemble circuit rhythm. Changes in cellular and circuit excitability in Tg-BK(R207Q) SCNs were correlated with elongated behavioral active periods and enhanced responses to phase-shifting stimuli. Unexpectedly, despite the severe reduction in circuit amplitude, circadian behavioral amplitudes in Tg-BK(R207Q) mice were relatively normal. These data demonstrate that downregulation of the BK current during the day is essential for the high amplitude neural activity pattern in the SCN that restricts locomotor activity to the appropriate phase and maintains the clock's robustness against perturbation. However, a residually rhythmic subset prevails over the ensemble circuit to drive the fundamental circadian behavioral rhythm.


Subject(s)
Circadian Rhythm , Gene Expression , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Suprachiasmatic Nucleus/physiology , Action Potentials , Amino Acid Substitution , Animals , Behavior, Animal/physiology , Circadian Clocks , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/physiology , Mice , Mice, Transgenic , Motor Activity , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/metabolism , Tissue Culture Techniques
9.
Biol Psychiatry ; 70(12): 1122-6, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21798517

ABSTRACT

BACKGROUND: Schizophrenia is a chronic and devastating brain disorder characterized by hallucinations and delusions, symptoms reflecting impaired reality testing. Although animal models have captured negative symptoms and cognitive deficits associated with schizophrenia, none have addressed these defining, positive symptoms. METHODS: Here we tested the performance of adults given neonatal ventral hippocampal lesions (NVHL), a neurodevelopmental model of schizophrenia, in two taste aversion procedures. RESULTS: Normal and NVHL rats formed aversions to a palatable food when the food was directly paired with nausea, but only NVHL rats formed a food aversion when the cue predicting that food was paired with nausea. The failure of NVHL rats to discriminate fully real from imagined food parallels the failure of people with schizophrenia to differentiate internal thoughts and beliefs from reality. CONCLUSIONS: These results further validate the NVHL model of schizophrenia and provide a means to assess impaired reality testing in variety of animal models.


Subject(s)
Avoidance Learning/physiology , Discrimination, Psychological/physiology , Hippocampus/physiology , Reality Testing , Schizophrenia/physiopathology , Animals , Animals, Newborn/surgery , Association Learning/physiology , Conditioning, Classical/physiology , Disease Models, Animal , Hallucinations/physiopathology , Hippocampus/physiopathology , Male , Rats , Rats, Long-Evans , Schizophrenic Psychology , Taste
SELECTION OF CITATIONS
SEARCH DETAIL
...