Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
1.
Stroke ; 55(6): 1660-1671, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660789

ABSTRACT

BACKGROUND: Activation of the acid-sensing ion channels (ASICs) by tissue acidosis, a common feature of brain ischemia, contributes to ischemic brain injury, while blockade of ASICs results in protection. Cholestane-3ß,5α,6ß-triol (Triol), a major cholesterol metabolite, has been demonstrated as an endogenous neuroprotectant; however, the mechanism underlying its neuroprotective activity remains elusive. In this study, we tested the hypothesis that inhibition of ASICs is a potential mechanism. METHODS: The whole-cell patch-clamp technique was used to examine the effect of Triol on ASICs heterogeneously expressed in Chinese hamster ovary cells and ASICs endogenously expressed in primary cultured mouse cortical neurons. Acid-induced injury of cultured mouse cortical neurons and middle cerebral artery occlusion-induced ischemic brain injury in wild-type and ASIC1 and ASIC2 knockout mice were studied to examine the protective effect of Triol. RESULTS: Triol inhibits ASICs in a subunit-dependent manner. In Chinese hamster ovary cells, it inhibits homomeric ASIC1a and ASIC3 without affecting ASIC1ß and ASIC2a. In cultured mouse cortical neurons, it inhibits homomeric ASIC1a and heteromeric ASIC1a-containing channels. The inhibition is use-dependent but voltage- and pH-independent. Structure-activity relationship analysis suggests that hydroxyls at the 5 and 6 positions of the A/B ring are critical functional groups. Triol alleviates acidosis-mediated injury of cultured mouse cortical neurons and protects against middle cerebral artery occlusion-induced brain injury in an ASIC1a-dependent manner. CONCLUSIONS: Our study identifies Triol as a novel ASIC inhibitor, which may serve as a new pharmacological tool for studying ASICs and may also be developed as a potential drug for treating stroke.


Subject(s)
Acid Sensing Ion Channels , Acidosis , Cricetulus , Mice, Knockout , Animals , Acid Sensing Ion Channels/metabolism , Acid Sensing Ion Channels/genetics , Mice , CHO Cells , Acidosis/metabolism , Acidosis/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Neurons/drug effects , Neurons/metabolism , Cricetinae , Neuroprotective Agents/pharmacology , Cholestanols/pharmacology , Mice, Inbred C57BL , Acid Sensing Ion Channel Blockers/pharmacology , Male , Cells, Cultured
2.
Biomolecules ; 13(3)2023 03 10.
Article in English | MEDLINE | ID: mdl-36979442

ABSTRACT

KB-R7943, an isothiourea derivative, is widely used as a pharmacological inhibitor of reverse sodium-calcium exchanger (NCX). It has been shown to have neuroprotective and analgesic effects in animal models; however, the detailed molecular mechanisms remain elusive. In the current study, we investigated whether KB-R7943 modulates acid-sensing ion channels (ASICs), a group of proton-gated cation channels implicated in the pathophysiology of various neurological disorders, using the whole-cell patch clamp techniques. Our data show that KB-R7943 irreversibly inhibits homomeric ASIC1a channels heterologously expressed in Chinese Hamster Ovary (CHO) cells in a use- and concentration-dependent manner. It also reversibly inhibits homomeric ASIC2a and ASIC3 channels in CHO cells. Both the transient and sustained current components of ASIC3 are inhibited. Furthermore, KB-R7943 inhibits ASICs in primary cultured peripheral and central neurons. It inhibits the ASIC-like currents in mouse dorsal root ganglion (DRG) neurons and the ASIC1a-like currents in mouse cortical neurons. The inhibition of the ASIC1a-like current is use-dependent and unrelated to its effect on NCX since neither of the other two well-characterized NCX inhibitors, including SEA0400 and SN-6, shows an effect on ASIC. Our data also suggest that the isothiourea group, which is lacking in other structurally related analogs that do not affect ASIC1a-like current, may serve as a critical functional group. In summary, we characterize KB-R7943 as a new ASIC inhibitor. It provides a novel pharmacological tool for the investigation of the functions of ASICs and could serve as a lead compound for developing small-molecule drugs for treating ASIC-related disorders.


Subject(s)
Acid Sensing Ion Channels , Sodium-Calcium Exchanger , Cricetinae , Mice , Animals , Cricetulus , Sodium-Calcium Exchanger/genetics , CHO Cells
3.
Biomolecules ; 12(9)2022 09 01.
Article in English | MEDLINE | ID: mdl-36139053

ABSTRACT

In this study, we characterize biophysical changes in NMDA receptor function in response to brief non-injurious ischemic stress (ischemic preconditioning). Electrophysiological studies show NMDA receptor function is reduced following preconditioning in cultured rat cortical neurons. This functional change is not due to changes in the reversal potential of the receptor, but an increase in desensitization. We performed concentration-response analysis of NMDA-evoked currents, and demonstrate that preconditioned neurons show a reduced potency of NMDA to evoke currents, an increase in Mg2+ sensitivity, but no change in glycine sensitivity. Antagonists studies show a reduced inhibition of GluN2B antagonists that have an allosteric mode of action (ifenprodil and R-25-6981), but competitive antagonists at the GluR2A and 2B receptor (NVP-AMM077 and conantokin-G) appear to have similar potency to block currents. Biochemical studies show a reduction in membrane surface GluN2B subunits, and an increased co-immunoprecipitation of GluN2A with GluN2B subunits, suggestive of tri-heteromeric receptor formation. Finally, we show that blocking actin remodeling with jasplakinolide, a mechanism of rapid ischemic tolerance, prevents NMDA receptor functional changes and co-immunoprecipitation of GluN2A and 2B subunits. Together, this study shows that alterations in NMDA receptor function following preconditioning ischemia are associated with neuroprotection in rapid ischemic tolerance.


Subject(s)
N-Methylaspartate , Receptors, N-Methyl-D-Aspartate , Actins , Animals , Glycine/pharmacology , Ischemia , Rats
4.
J Cereb Blood Flow Metab ; 42(8): 1349-1363, 2022 08.
Article in English | MEDLINE | ID: mdl-35301897

ABSTRACT

Extracellular proton concentration is at 40 nM when pH is 7.4. In disease conditions such as brain ischemia, proton concentration can reach µM range. To respond to this increase in extracellular proton concentration, the mammalian brain expresses at least three classes of proton receptors. Acid-sensing ion channels (ASICs) are the main neuronal cationic proton receptor. The proton-activated chloride channel (PAC), which is also known as (aka) acid-sensitive outwardly rectifying anion channel (ASOR; TMEM206), mediates acid-induced chloride currents. Besides proton-activated channels, GPR4, GPR65 (aka TDAG8, T-cell death-associated gene 8), and GPR68 (aka OGR1, ovarian cancer G protein-coupled receptor 1) function as proton-sensitive G protein-coupled receptors (GPCRs). Though earlier studies on these GPCRs mainly focus on peripheral cells, we and others have recently provided evidence for their functional importance in brain injury. Specifically, GPR4 shows strong expression in brain endothelium, GPR65 is present in a fraction of microglia, while GPR68 exhibits predominant expression in brain neurons. Here, to get a better view of brain acid signaling and its contribution to ischemic injury, we will review the recent findings regarding the differential contribution of proton-sensitive GPCRs to cerebrovascular function, neuroinflammation, and neuronal injury following acidosis and brain ischemia.


Subject(s)
Brain Ischemia , Protons , Acid Sensing Ion Channels/metabolism , Animals , Hydrogen-Ion Concentration , Mammals/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
5.
Front Genet ; 12: 737741, 2021.
Article in English | MEDLINE | ID: mdl-35095995

ABSTRACT

Whole transcriptome RNA-sequencing was performed to quantify RNA expression changes in whole blood samples collected from steady state sickle cell anemia (SCA) and control subjects. Pediatric SCA and control subjects were recruited from Atlanta (GA)-based hospital(s) systems and consented for RNA sequencing. RNA sequencing was performed on an Ion Torrent S5 sequencer, using the Ion Total RNA-seq v2 protocol. Data were aligned to the hg19 reference genome and analyzed in the Partek Genomics studio package (v7.0). 223 genes were differentially expressed between SCA and controls (± 1.5 fold change FDR p < 0.001) and 441 genes show differential transcript expression (± 1.5 fold FDR p < 0.001). Differentially expressed RNA are enriched for hemoglobin associated genes and ubiquitin-proteasome pathway genes. Further analysis shows higher gamma globin gene expression in SCA (33-fold HBG1 and 49-fold HBG2, both FDR p < 0.05), which did not correlate with hemoglobin F protein levels. eQTL analysis identified SNPs in novel non-coding RNA RYR2 gene as having a potential regulatory role in HBG1 and HBG2 expression levels. Gene expression correlation identified JHDM1D-AS1(KDM7A-DT), a non-coding RNA associated with angiogenesis, enhanced GATA1 and decreased JAK-STAT signaling to correlate with HBG1 and HBG2 mRNA levels. These data suggest novel regulatory mechanisms for fetal hemoglobin regulation, which may offer innovative therapeutic approaches for SCA.

6.
Stroke ; 51(12): 3690-3700, 2020 12.
Article in English | MEDLINE | ID: mdl-33059544

ABSTRACT

BACKGROUND AND PURPOSE: Brain acidosis is prevalent in stroke and other neurological diseases. Acidosis can have paradoxical injurious and protective effects. The purpose of this study is to determine whether a proton receptor exists in neurons to counteract acidosis-induced injury. METHODS: We analyzed the expression of proton-sensitive GPCRs (G protein-coupled receptors) in the brain, examined acidosis-induced signaling in vitro, and studied neuronal injury using in vitro and in vivo mouse models. RESULTS: GPR68, a proton-sensitive GPCR, was present in both mouse and human brain, and elicited neuroprotection in acidotic and ischemic conditions. GPR68 exhibited wide expression in brain neurons and mediated acidosis-induced PKC (protein kinase C) activation. PKC inhibition exacerbated pH 6-induced neuronal injury in a GPR68-dependent manner. Consistent with its neuroprotective function, GPR68 overexpression alleviated middle cerebral artery occlusion-induced brain injury. CONCLUSIONS: These data expand our knowledge on neuronal acid signaling to include a neuroprotective metabotropic dimension and offer GPR68 as a novel therapeutic target to alleviate neuronal injuries in ischemia and multiple other neurological diseases.


Subject(s)
Acidosis/metabolism , Brain/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neurons/metabolism , Neuroprotection/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Ischemic Stroke/metabolism , Mice , Mice, Knockout , Neuroprotection/physiology , Protein Kinase C/metabolism , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neurosci ; 40(15): 3119-3129, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32144179

ABSTRACT

Mitochondrial fission catalyzed by dynamin-related protein 1 (Drp1) is necessary for mitochondrial biogenesis and maintenance of healthy mitochondria. However, excessive fission has been associated with multiple neurodegenerative disorders, and we recently reported that mice with smaller mitochondria are sensitized to ischemic stroke injury. Although pharmacological Drp1 inhibition has been put forward as neuroprotective, the specificity and mechanism of the inhibitor used is controversial. Here, we provide genetic evidence that Drp1 inhibition is neuroprotective. Drp1 is activated by dephosphorylation of an inhibitory phosphorylation site, Ser637. We identify Bß2, a mitochondria-localized protein phosphatase 2A (PP2A) regulatory subunit, as a neuron-specific Drp1 activator in vivo Bß2 KO mice of both sexes display elongated mitochondria in neurons and are protected from cerebral ischemic injury. Functionally, deletion of Bß2 and maintained Drp1 Ser637 phosphorylation improved mitochondrial respiratory capacity, Ca2+ homeostasis, and attenuated superoxide production in response to ischemia and excitotoxicity in vitro and ex vivo Last, deletion of Bß2 rescued excessive stroke damage associated with dephosphorylation of Drp1 S637 and mitochondrial fission. These results indicate that the state of mitochondrial connectivity and PP2A/Bß2-mediated dephosphorylation of Drp1 play a critical role in determining the severity of cerebral ischemic injury. Therefore, Bß2 may represent a target for prophylactic neuroprotective therapy in populations at high risk of stroke.SIGNIFICANCE STATEMENT With recent advances in clinical practice including mechanical thrombectomy up to 24 h after the ischemic event, there is resurgent interest in neuroprotective stroke therapies. In this study, we demonstrate reduced stroke damage in the brain of mice lacking the Bß2 regulatory subunit of protein phosphatase 2A, which we have shown previously acts as a positive regulator of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1). Importantly, we provide evidence that deletion of Bß2 can rescue excessive ischemic damage in mice lacking the mitochondrial PKA scaffold AKAP1, apparently via opposing effects on Drp1 S637 phosphorylation. These results highlight reversible phosphorylation in bidirectional regulation of Drp1 activity and identify Bß2 as a potential pharmacological target to protect the brain from stroke injury.


Subject(s)
Brain Ischemia/genetics , Brain Ischemia/prevention & control , Dynamins/genetics , Neurons/metabolism , Animals , Calcium/metabolism , Dynamins/metabolism , Female , Homeostasis , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Phosphorylation , Primary Cell Culture , Protein Phosphatase 2/genetics , Stroke/pathology , Stroke/prevention & control , Superoxides/metabolism
8.
Hippocampus ; 30(7): 715-723, 2020 07.
Article in English | MEDLINE | ID: mdl-32057164

ABSTRACT

Prohormone convertase 2 (PC2) is essential for the biosynthesis of many neuropeptides, including several of them in hippocampus. In mouse brain, lacking an enzymatically active PC2 (PC2-null) causes accumulation of many neuropeptides in their precursor or intermediate forms. Little is known about how a PC2-null state may affect the function of the hippocampus. In this study, adult PC2-null mice and their wildtype (WT) littermates were subjected to three analyses to determine possible changes associated with PC2-null at physiological, behavioral, and molecular levels, respectively, under normal and stressed conditions. Electrophysiological recordings of hippocampal slices were performed to measure evoked field-excitatory postsynaptic potentials (EPSP), long-term potentiation (LTP), and paired-pulse facilitation (PPF). Morris water maze (MWM) testing was conducted to examine behavioral changes that are indicative of hippocampal integrity. Quantitative mass spectrometry analysis was used to determine changes in the hippocampal proteome in response to a focal cerebral ischemic insult. We found that there were no significant differences in the threshold of evoked EPSPs between PC2-null and WT animals. However, an increase in LTP in both triggering rate and amplitude was observed in PC2-null mice, suggesting that PC2 may be involved in regulating synaptic strength. The PPF, on the other hand, showed a decrease in PC2-null mice, suggesting a presynaptic mechanism. Consistent with changes in LTP, PC2-null mice displayed decreased latencies in finding the escape platform in the MWM test. Further, after distal focal cerebral ischemia, the hippocampal proteomes incurred changes in both WT and PC2-null mice, with a prominent change in proteins associated with neurotransmission, exocytosis, and transport processes seen in the PC2-null but not WT mice. Taken together, our results suggest that PC2 is involved in regulating hippocampal synaptic plasticity, learning, and memory behaviors, as well as the hippocampal response to stresses originating in other regions of the brain.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Hippocampus/enzymology , Maze Learning/physiology , Proprotein Convertase 2/deficiency , Animals , Brain Ischemia/enzymology , Brain Ischemia/genetics , Female , Male , Mice , Mice, Knockout , Organ Culture Techniques , Proprotein Convertase 2/genetics
9.
Article in English | MEDLINE | ID: mdl-31149323

ABSTRACT

BACKGROUND: Stroke occurs more often and results in more severe brain injury in African Americans than in Caucasians. The former also exhibit different responses to thrombolytic therapy than the latter do. There is an imminent need for stroke biomarkers for African Americans, who have been underrepresented in biomarker research for stroke diagnosis and prognosis. Proteomics offers sources for protein biomarkers that are not available by other Omics approaches. In this pilot study, plasma proteomes of African American stroke patients were analyzed and compared to that of hypertensive, non-stroke controls. METHODS: Plasma samples were prepared from whole blood specimens that were collected from stroke patients admitted to Grady Memorial Hospital in Atlanta, and their age- and sex-matched, hypertensive controls from the outpatient clinic. Samples were pooled according to patient groups and sex. Plasma proteins were analyzed with quantitative mass spectrometry. The identified and quantified proteins were compared between stroke and control patients of each sex. Proteins that showed changes in abundances in stroke patients were further analyzed with the assistance of bioinformatics tools for their known biological functions or potential implications in stroke. RESULTS: A total of 128 annotated proteins were identified. Results of bioinformatic analysis of plasma proteins whose levels were increased in stroke patients showed, as expected, their association with blood coagulation and inflammation processes. Interestingly, a number of proteins showed different or even opposing changes in male and female stroke patients, notably those involved in IL-4 and IL-6 signaling, complement activation, and blood coagulation disorders. For a few proteins that were increased in female but unchanged or decreased in male stroke patients, an association with fibromuscular dysplasia was recognized. CONCLUSION: Plasma proteins that differ in quantities between stroke patients and controls were readily detected using a simple proteomic approach. Sex-dependent changes and changes that have not been reported for African American stroke patients offer potentially novel biomarkers for stroke in this underserved population.

10.
Transl Stroke Res ; 10(6): 664-671, 2019 12.
Article in English | MEDLINE | ID: mdl-30535792

ABSTRACT

We hypothesize that enhancing mitochondrial base excision repair (BER) capability in brain will reduce reperfusion-associated ischemic brain injury. Post-stroke reperfusion was modeled in mice via transient filament occlusion of the middle cerebral artery (60 min) (transient MCAO). Administration of a TAT-modified form of a DNA glycosylase (EndoIII) following reperfusion of the brain reduced resultant brain infarct volume. Protection was dose-dependent, BER enzyme specific, and regionally specific (more effective via the jugular vein). EndoIII is compatible with tissue plasminogen activator (tPA). The time window of a single dose of EndoIII effect is 3 h following reperfusion onset. These data suggest a novel approach to enhance protection of reperfused brain in the setting of revascularization procedures (thrombectomy or thrombolytic therapy) following stroke.


Subject(s)
Brain Ischemia/drug therapy , DNA Repair , DNA, Mitochondrial/genetics , Reperfusion Injury/drug therapy , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Animals , Brain Ischemia/genetics , Brain Ischemia/surgery , DNA Glycosylases/therapeutic use , Disease Models, Animal , Female , Male , Mice , Neuroprotective Agents/therapeutic use , Reperfusion/methods , Reperfusion Injury/genetics , Stroke/surgery , Thrombolytic Therapy
11.
Dose Response ; 16(3): 1559325818784501, 2018.
Article in English | MEDLINE | ID: mdl-30140178

ABSTRACT

Human performance, endurance, and resilience have biological limits that are genetically and epigenetically predetermined but perhaps not yet optimized. There are few systematic, rigorous studies on how to raise these limits and reach the true maxima. Achieving this goal might accelerate translation of the theoretical concepts of conditioning, hormesis, and stress adaptation into technological advancements. In 2017, an Air Force-sponsored conference was held at the University of Massachusetts for discipline experts to display data showing that the amplitude and duration of biological performance might be magnified and to discuss whether there might be harmful consequences of exceeding typical maxima. The charge of the workshop was "to examine and discuss and, if possible, recommend approaches to control and exploit endogenous defense mechanisms to enhance the structure and function of biological tissues." The goal of this white paper is to fulfill and extend this workshop charge. First, a few of the established methods to exploit endogenous defense mechanisms are described, based on workshop presentations. Next, the white paper accomplishes the following goals to provide: (1) synthesis and critical analysis of concepts across some of the published work on endogenous defenses, (2) generation of new ideas on augmenting biological performance and resilience, and (3) specific recommendations for researchers to not only examine a wider range of stimulus doses but to also systematically modify the temporal dimension in stimulus inputs (timing, number, frequency, and duration of exposures) and in measurement outputs (interval until assay end point, and lifespan). Thus, a path forward is proposed for researchers hoping to optimize protocols that support human health and longevity, whether in civilians, soldiers, athletes, or the elderly patients. The long-term goal of these specific recommendations is to accelerate the discovery of practical methods to conquer what were once considered intractable constraints on performance maxima.

12.
PLoS One ; 12(9): e0183113, 2017.
Article in English | MEDLINE | ID: mdl-28863142

ABSTRACT

Mild traumatic brain injury (mTBI) is a complex, neurophysiological condition that can have detrimental outcomes. Yet, to date, no objective method of diagnosis exists. Physical damage to the blood-brain-barrier and normal waste clearance via the lymphatic system may enable the detection of biomarkers of mTBI in peripheral circulation. Here we evaluate the accuracy of whole transcriptome analysis of blood to predict the clinical diagnosis of post-concussion syndrome (PCS) in a military cohort. Sixty patients with clinically diagnosed chronic concussion and controls (no history of concussion) were recruited (retrospective study design). Male patients (46) were split into a training set comprised of 20 long-term concussed (> 6 months and symptomatic) and 12 controls (no documented history of concussion). Models were validated in a testing set (control = 9, concussed = 5). RNA_Seq libraries were prepared from whole blood samples for sequencing using a SOLiD5500XL sequencer and aligned to hg19 reference genome. Patterns of differential exon expression were used for diagnostic modeling using support vector machine classification, and then validated in a second patient cohort. The accuracy of RNA profiles to predict the clinical diagnosis of post-concussion syndrome patients from controls was 86% (sensitivity 80%; specificity 89%). In addition, RNA profiles reveal duration of concussion. This pilot study shows the potential utility of whole transcriptome analysis to establish the clinical diagnosis of chronic concussion syndrome.


Subject(s)
Post-Concussion Syndrome/blood , Post-Concussion Syndrome/diagnosis , RNA/blood , Adult , Brain Injuries/blood , Brain Injuries/diagnosis , DNA, Complementary/metabolism , Female , Gene Expression Profiling , Genomics , Humans , Male , Middle Aged , Military Medicine , Military Personnel , Pilot Projects , Retrospective Studies , Sensitivity and Specificity , Support Vector Machine , Transcriptome
13.
J Cereb Blood Flow Metab ; 37(2): 528-540, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26861816

ABSTRACT

Acidosis in the brain plays a critical role in neuronal injury in neurological diseases, including brain ischemia. One key mediator of acidosis-induced neuronal injury is the acid-sensing ion channels (ASICs). Current literature has focused on ASIC1a when studying acid signaling. The importance of ASIC2, which is also widely expressed in the brain, has not been appreciated. We found here a region-specific effect of ASIC2 on acid-mediated responses. Deleting ASIC2 reduced acid-activated current in cortical and striatal neurons, but had no significant effect in cerebellar granule neurons. In addition, we demonstrated that ASIC2 was important for ASIC1a expression, and that ASIC2a but not 2b facilitated ASIC1a surface trafficking in the brain. Further, we showed that ASIC2 deletion attenuated acidosis/ischemia-induced neuronal injury in organotypic hippocampal slices but had no effect in organotypic cerebellar slices. Consistent with an injurious role of ASIC2, we showed that ASIC2 deletion significantly protected the mouse brain from ischemic damage in vivo. These data suggest a critical region-specific contribution of ASIC2 to neuronal injury and reveal an important functional difference between ASIC2a and 2b in the brain.


Subject(s)
Acid Sensing Ion Channels/metabolism , Brain Ischemia/pathology , Brain/pathology , Neurons/pathology , Acid Sensing Ion Channels/analysis , Acid Sensing Ion Channels/genetics , Acidosis , Animals , Brain/metabolism , Brain Ischemia/genetics , Brain Ischemia/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neurons/metabolism , Neuroprotection
14.
Article in English | MEDLINE | ID: mdl-29348796

ABSTRACT

In the context of seeking novel therapeutic targets for treating ischemic stroke, the preconditioning ischemia-induced brain ischemic tolerance has been used as a model of endogenously operative, broad-based neuroprotective mechanisms. Targeting such mechanisms is considered potentially less prone to adverse side effects, as those seen in many failed clinical trials that focus on single targets using exogenous compounds. Results from previous studies have revealed an overall decrease in potassium channel activity in tolerance development. The objective of this study is to identify ion channel genes that are differentially regulated under different brain ischemic conditions, as a mean to identify those ion channels that are associated with ischemic brain injury and ischemic tolerance. In mice in vivo, transient focal cerebral ischemia was induced by middle cerebral artery occlusion. In cultured neuronal cells in vitro, simulated ischemia was modeled by oxygen-glucose deprivation. For both in vivo and in vitro studies, three principal ischemic conditions were included: ischemic-preconditioned, injured and tolerant, respectively, plus appropriate controls. In these model systems, transcript levels of a panel of 84 neuronal ion channels genes were analyzed with a quantitative real-time PCR mini-array. The results showed that, both in vivo and in vitro, there was a predominant down regulation in neuronal ion channel genes under ischemic-tolerant conditions, and an up regulation in ischemic injury. Similar changes were observed among potassium, sodium and calcium channel genes. A number of regulated genes exhibited opposing changes under ischemic-injured and ischemic-tolerant conditions. This subset of ion channel genes exemplifies potentially novel leads for developing multi-factorial therapeutic targets for treating ischemic stroke.

15.
PLoS One ; 11(12): e0169170, 2016.
Article in English | MEDLINE | ID: mdl-28036405

ABSTRACT

The objective of this study was to evaluate the effects of increasing dietary crude protein (CP) levels and also comparing the effects of static versus oscillating dietary CP on ruminal nutrient digestibility, ruminal fermentation, nitrogen (N) metabolism, and microbial efficiency in beef cattle diets using a dual-flow continuous culture system. Eight fermenters (1,223 ± 21 mL) were used in a replicated 4 x 4 Latin square design with periods lasting 12 d each (8 d for adaptation and 4 d for sampling). Dietary treatments were: 1) 10% CP, 2) 12% CP, 3) 14% CP, and 4) 10 and 14% CP diets oscillating at 48-h intervals. Experimental diets consisted of 50% orchard hay and 50% concentrate. Fermenters were fed 72 g/d and solid and liquid dilution rates were adjusted to 5.5 and 11%/h, respectively. Data were analyzed using the MIXED procedure in SAS with α = 0.05. Apparent and true ruminal digestibilities of dry matter and organic matter were not affected (P > 0.05) by increasing dietary CP, nor by oscillating dietary CP. Total volatile fatty acids concentration and molar proportions of acetate, propionate, butyrate, valerate, iso-butyrate and iso-valerate were not affected (P > 0.05) by increasing or oscillating dietary CP. Ruminal NH3-N concentration increased linearly (P < 0.01) in response to increasing dietary CP. Total N, non-ammonia N, and rumen undegraded protein flows did not differ among treatments or between oscillating dietary CP and static 12% CP. Microbial N and NH3-N flows and microbial efficiency did not differ when comparing oscillating versus static CP (P > 0.05). However, there was a quadratic effect (P < 0.05) for these variables when dietary CP was increased. These results indicate that either ruminal microorganisms do not respond to oscillating CP levels or are capable of coping with 48-h periods of undernourishment.


Subject(s)
Animal Feed/analysis , Bioreactors , Diet/veterinary , Dietary Proteins/analysis , Digestion/physiology , Fermentation , Rumen/metabolism , Ammonia/metabolism , Animal Nutritional Physiological Phenomena/physiology , Animals , Cattle , Dietary Proteins/administration & dosage , Dietary Proteins/chemistry , Dietary Supplements , Fatty Acids/metabolism , Nitrogen/metabolism
16.
Sleep ; 39(11): 2033-2040, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27568798

ABSTRACT

STUDY OBJECTIVES: Episodes of brief limb ischemia (remote preconditioning) in mice induce tolerance to modeled ischemic stroke (focal brain ischemia). Since stroke outcomes are in part dependent on sleep-wake history, we sought to determine if sleep is critical for the neuroprotective effect of limb ischemia. METHODS: EEG/EMG recording electrodes were implanted in mice. After a 24 h baseline recording, limb ischemia was induced by tightening an elastic band around the left quadriceps for 10 minutes followed by 10 minutes of release for two cycles. Two days following remote preconditioning, a second 24 h EEG/EMG recording was completed and was immediately followed by a 60-minute suture occlusion of the middle cerebral artery (modeled ischemic stroke). This experiment was then repeated in a model of circadian and sleep abnormalities (Bmal1 knockout [KO] mice sleep 2 h more than wild-type littermates). Brain infarction was determined by vital dye staining, and sleep was assessed by trained identification of EEG/EMG recordings. RESULTS: Two days after limb ischemia, wild-type mice slept an additional 2.4 h. This additional sleep was primarily comprised of non-rapid eye movement (NREM) sleep during the middle of the light-phase (i.e., naps). Repeating the experiment but preventing increases in sleep after limb ischemia abolished tolerance to ischemic stroke. In Bmal1 knockout mice, remote preconditioning did not increase daily sleep nor provide tolerance to subsequent focal ischemia. CONCLUSIONS: These results suggest that sleep induced by remote preconditioning is both sufficient and necessary for its neuroprotective effects on stroke outcome.


Subject(s)
Brain Ischemia/therapy , Ischemic Preconditioning/methods , Neuroprotection/physiology , Sleep/physiology , Stroke/therapy , Animals , Brain Ischemia/diagnosis , Brain Ischemia/physiopathology , Electroencephalography , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats, Sprague-Dawley , Stroke/diagnosis , Stroke/physiopathology
17.
J Biol Chem ; 291(35): 18370-83, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27402850

ABSTRACT

An important contributor to brain ischemia is known to be extracellular acidosis, which activates acid-sensing ion channels (ASICs), a family of proton-gated sodium channels. Lines of evidence suggest that targeting ASICs may lead to novel therapeutic strategies for stroke. Investigations of the role of ASICs in ischemic brain injury have naturally focused on the role of extracellular pH in ASIC activation. By contrast, intracellular pH (pHi) has received little attention. This is a significant gap in our understanding because the ASIC response to extracellular pH is modulated by pHi, and activation of ASICs by extracellular protons is paradoxically enhanced by intracellular alkalosis. Our previous studies show that acidosis-induced cell injury in in vitro models is attenuated by intracellular acidification. However, whether pHi affects ischemic brain injury in vivo is completely unknown. Furthermore, whereas ASICs in native neurons are composed of different subunits characterized by distinct electrophysiological/pharmacological properties, the subunit-dependent modulation of ASIC activity by pHi has not been investigated. Using a combination of in vitro and in vivo ischemic brain injury models, electrophysiological, biochemical, and molecular biological approaches, we show that the intracellular alkalizing agent quinine potentiates, whereas the intracellular acidifying agent propionate inhibits, oxygen-glucose deprivation-induced cell injury in vitro and brain ischemia-induced infarct volume in vivo Moreover, we find that the potentiation of ASICs by quinine depends on the presence of the ASIC1a, ASIC2a subunits, but not ASIC1b, ASIC3 subunits. Furthermore, we have determined the amino acids in ASIC1a that are involved in the modulation of ASICs by pHi.


Subject(s)
Acid Sensing Ion Channels/metabolism , Brain Ischemia/metabolism , Stroke/metabolism , Animals , Brain Ischemia/physiopathology , Disease Models, Animal , Hydrogen-Ion Concentration , Mice , Propionates/metabolism , Stroke/physiopathology
18.
Ann Clin Transl Neurol ; 3(2): 70-81, 2016 02.
Article in English | MEDLINE | ID: mdl-26900583

ABSTRACT

OBJECTIVE: Molecular diagnostic medicine holds much promise to change point of care treatment. An area where additional diagnostic tools are needed is in acute stroke care, to assist in diagnosis and prognosis. Previous studies using microarray-based gene expression analysis of peripheral blood following stroke suggests this approach may be effective. Next-generation sequencing (NGS) approaches have expanded genomic analysis and are not limited to previously identified genes on a microarray chip. Here, we report on a pilot NGS study to identify gene expression and exon expression patterns for the prediction of stroke diagnosis and prognosis. METHODS: We recruited 28 stroke patients and 28 age- and sex-matched hypertensive controls. RNA was extracted from 3 mL blood samples, and RNA-Seq libraries were assembled and sequenced. RESULTS: Bioinformatical analysis of the aligned RNA data reveal exonic (30%), intronic (36%), and novel RNA components (not currently annotated: 33%). We focused our study on patients with confirmed middle cerebral artery occlusion ischemic stroke (n = 17). On the basis of our observation of differential splicing of gene transcripts, we used all exonic RNA expression rather than gene expression (combined exons) to build prediction models using support vector machine algorithms. Based on model building, these models have a high predicted accuracy rate >90% (spec. 88% sen. 92%). We further stratified outcome based on the improvement in NIHss scores at discharge; based on model building we observe a predicted 100% accuracy rate. INTERPRETATION: NGS-based exon expression analysis approaches have a high potential for patient diagnosis and outcome prediction, with clear utility to aid in clinical patient care.

19.
CNS Neurosci Ther ; 22(6): 468-76, 2016 06.
Article in English | MEDLINE | ID: mdl-26890278

ABSTRACT

BACKGROUND: ASIC1a, the predominant acid-sensing ion channels (ASICs), is implicated in neurological disorders including stroke, traumatic spinal cord injury, and ALS. Potent ASIC1a inhibitors should have promising therapeutic potential for ASIC1a-related diseases. AIMS: We examined the inhibitory effects of a number of amiloride analogs on ASIC1a currents, aimed at understanding the structure-activity relationship and identifying potent ASIC1a inhibitors for stroke intervention. METHODS: Whole-cell patch-clamp techniques and a mouse model of middle cerebral artery occlusion (MCAO)-induced focal ischemia were used. Surflex-Dock was used to dock the analogs into the pocket with default parameters. RESULTS: Amiloride and its analogs inhibit ASIC1a currents expressed in Chinese hamster ovary cells with a potency rank order of benzamil > phenamil > 5-(N,N-dimethyl)amiloride (DMA) > amiloride > 5-(N,N-hexamethylene)amiloride (HMA) ≥ 5-(N-methyl-N-isopropyl)amiloride (MIA) > 5-(N-ethyl-N-isopropyl)amiloride (EIPA). In addition, amiloride and its analogs inhibit ASIC currents in cortical neurons with the same potency rank order. In mice, benzamil and EIPA decreased MCAO-induced infarct volume. Similar to its effect on the ASIC current, benzamil showed a much higher potency than EIPA. CONCLUSION: Addition of a benzyl group to the terminal guanidinyl group resulted in enhanced inhibitory activity on ASIC1a. On the other hand, the bulky groups added to the 5-amino residues slightly decreased the activity. Among the tested amiloride analogs, benzamil is the most potent ASIC1a inhibitor.


Subject(s)
Acid Sensing Ion Channels/physiology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Amiloride/chemistry , Amiloride/therapeutic use , Animals , Biophysics , CHO Cells , Cells, Cultured , Cerebral Cortex/cytology , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Embryo, Mammalian , Infarction, Middle Cerebral Artery/drug therapy , Inhibitory Concentration 50 , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Models, Molecular , Neurons/drug effects , Patch-Clamp Techniques , Transfection
20.
Neurosci Lett ; 625: 16-9, 2016 06 20.
Article in English | MEDLINE | ID: mdl-26739198

ABSTRACT

Mild stress from ischemia, seizure, hypothermia, or infection can produce a transient neuroprotected state in the brain. In the neuroprotected state, the brain responds differently to a severe stress and sustains less injury. At the genomic level, the response of the neuroprotected brain to a severe stress is characterized by widespread differential regulation of genes with diverse functions. This reprogramming of gene expression observed in the neuroprotected brain in response to a stress is consistent with an epigenetic model of regulation mediated by changes in DNA methylation and histone modification. Here, we summarize our evolving understanding of the molecular basis for endogenous neuroprotection and review recent findings that implicate DNA methylation and protein mediators of histone modification as epigenetic regulators of the brain's response to injury.


Subject(s)
Brain Injuries/genetics , Epigenesis, Genetic , Gene Expression , Neuroprotection , Animals , DNA Methylation , Histone Code , Humans , Polycomb-Group Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...