Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Br J Anaesth ; 131(6): 1030-1042, 2023 12.
Article in English | MEDLINE | ID: mdl-37714750

ABSTRACT

BACKGROUND: Clinical studies suggest that anaesthesia exposure early in life affects neurobehavioural development. We designed a non-human primate (NHP) study to evaluate cognitive, behavioural, and brain functional and structural alterations after isoflurane exposure during infancy. These NHPs displayed decreased close social behaviour and increased astrogliosis in specific brain regions, most notably in the amygdala. Here we hypothesise that resting-state functional connectivity MRI can detect alterations in connectivity of brain areas that relate to these social behaviours and astrogliosis. METHODS: Imaging was performed in 2-yr-old NHPs under light anaesthesia, after early-in-life (postnatal days 6-12) exposure to 5 h of isoflurane either one or three times, or to room air. Brain images were segmented into 82 regions of interest; the amygdala and the posterior cingulate cortex were chosen for a seed-based resting-state functional connectivity MRI analysis. RESULTS: We found differences between groups in resting-state functional connectivity of the amygdala and the auditory cortices, medial premotor cortex, and posterior cingulate cortex. There were also alterations in resting-state functional connectivity between the posterior cingulate cortex and secondary auditory, polar prefrontal, and temporal cortices, and the anterior insula. Relationships were identified between resting-state functional connectivity alterations and the decrease in close social behaviour and increased astrogliosis. CONCLUSIONS: Early-in-life anaesthesia exposure in NHPs is associated with resting-state functional connectivity alterations of the amygdala and the posterior cingulate cortex with other brain regions, evident at the juvenile age of 2 yr. These changes in resting-state functional connectivity correlate with the decrease in close social behaviour and increased astrogliosis. Using resting-state functional connectivity MRI to study the neuronal underpinnings of early-in-life anaesthesia-induced behavioural alterations could facilitate development of a biomarker for anaesthesia-induced developmental neurotoxicity.


Subject(s)
Isoflurane , Animals , Isoflurane/adverse effects , Gliosis , Brain/diagnostic imaging , Gyrus Cinguli/diagnostic imaging , Magnetic Resonance Imaging/methods , Primates , Brain Mapping/methods , Neural Pathways/diagnostic imaging , Neural Pathways/physiology
2.
Am J Respir Cell Mol Biol ; 67(4): 482-490, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35776523

ABSTRACT

Asthma is a common respiratory disease characterized, in part, by excessive airway smooth muscle (ASM) contraction (airway hyperresponsiveness). Various GABAAR (γ-aminobutyric acid type A receptor) activators, including benzodiazepines, relax ASM. The GABAAR is a ligand-operated Cl- channel best known for its role in inhibitory neurotransmission in the central nervous system. Although ASM cells express GABAARs, affording a seemingly logical site of action, the mechanism(s) by which GABAAR ligands relax ASM remains unclear. PI320, a novel imidazobenzodiazepine designed for tissue selectivity, is a promising asthma drug candidate. Here, we show that PI320 alleviates methacholine (MCh)-induced bronchoconstriction in vivo and relaxes peripheral airways preconstricted with MCh ex vivo using the forced oscillation technique and precision-cut lung slice experiments, respectively. Surprisingly, the peripheral airway relaxation demonstrated in precision-cut lung slices does not appear to be GABAAR-dependent, as it is not inhibited by the GABAAR antagonist picrotoxin or the benzodiazepine antagonist flumazenil. Furthermore, we demonstrate here that PI320 inhibits MCh-induced airway constriction in the absence of external Ca2, suggesting that PI320-mediated relaxation is not mediated by inhibition of Ca2+ influx in ASM. However, PI320 does inhibit MCh-induced intracellular Ca2+ oscillations in peripheral ASM, a key mediator of contraction that is dependent on sarcoplasmic reticulum Ca2+ mobilization. Furthermore, PI320 inhibits peripheral airway constriction induced by experimentally increasing the intracellular concentration of inositol triphosphate (IP3). These novel data suggest that PI320 relaxes murine peripheral airways by inhibiting intracellular Ca2+ mobilization in ASM, likely by inhibiting Ca2+ release through IP3Rs (IP3 receptors).


Subject(s)
Asthma , Calcium , Animals , Asthma/drug therapy , Asthma/metabolism , Calcium/metabolism , Calcium Signaling , Flumazenil/metabolism , Inositol/metabolism , Ligands , Lung/metabolism , Methacholine Chloride/pharmacology , Mice , Muscle Contraction , Muscle, Smooth/metabolism , Picrotoxin/metabolism , gamma-Aminobutyric Acid/metabolism
3.
Br J Anaesth ; 128(4): 602-605, 2022 04.
Article in English | MEDLINE | ID: mdl-35115157

ABSTRACT

Anaesthesia exposure early in life potentially impairs neurobehavioural development. A recent study in the Journal investigated the possibility that progesterone mitigates anaesthesia-induced developmental neurotoxicity in neonatal rats exposed to sevoflurane. The novel findings show that the steroid hormone progesterone protects against development of behavioural alterations caused by sevoflurane. The protective mechanism is proposed to relate to anti-inflammatory properties of progesterone, which brings up important questions regarding the role of inflammation in mediating the neurobehavioural alterations in anaesthesia-induced developmental neurotoxicity. We discuss this mechanism and encourage new research that may clarify the underlying mechanisms of progesterone-induced protection and extend these findings into a translational model.


Subject(s)
Anesthesia , Neurotoxicity Syndromes , Anesthesia/adverse effects , Animals , Humans , Inflammation/chemically induced , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Rats , Sevoflurane/toxicity
4.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L912-L924, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34549600

ABSTRACT

Asthma affects millions of people worldwide and its prevalence is increasing. It is characterized by chronic airway inflammation, airway remodeling, and pathologic bronchoconstriction, and it poses a continuous treatment challenge with very few new therapeutics available. Thus, many asthmatics turn to plant-based complementary products, including ginger, for better symptom control, indicating an unmet need for novel therapies. Previously, we demonstrated that 6-shogaol (6S), the primary bioactive component of ginger, relaxes human airway smooth muscle (hASM) likely by inhibition of phosphodiesterases (PDEs) in the ß-adrenergic (cyclic nucleotide PDEs), and muscarinic (phospholipase C, PLC) receptor pathways. However, oral 6S is extensively metabolized and it is unknown if the resulting metabolites remain bioactive. Here, we screened all the known human metabolites of 6S and several metabolite-based synthetic derivatives to better understand their mechanism of action and structure-function relationships. We demonstrate that several metabolites and metabolite-based synthetic derivatives are able to prevent Gq-coupled stimulation of intracellular calcium [Ca2+]i and inositol trisphosphate (IP3) synthesis by inhibiting PLC, similar to the parent compound 6S. We also show that these compounds prevent recontraction of ASM after ß-agonist relaxation likely by inhibiting PDEs. Furthermore, they potentiate isoproterenol-induced relaxation. Importantly, moving beyond cell-based assays, metabolites also retain the functional ability to relax Gq-coupled-contractions in upper (human) and lower (murine) airways. The current study indicates that, although oral ginger may be metabolized rapidly, it retains physiological activity through its metabolites. Moreover, we are able to use naturally occurring metabolites as inspiration to develop novel therapeutics for brochoconstrictive diseases.


Subject(s)
Calcium/metabolism , Muscle Relaxation/drug effects , Myocytes, Smooth Muscle/drug effects , Zingiber officinale , Animals , Asthma/chemically induced , Asthma/metabolism , Bronchoconstriction/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Zingiber officinale/metabolism , Humans , Isoproterenol/pharmacology , Lung/drug effects , Lung/metabolism , Mice, Inbred C57BL , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism
5.
Br J Anaesth ; 127(3): 447-457, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34266661

ABSTRACT

BACKGROUND: Infant anaesthesia causes acute brain cell apoptosis, and later in life cognitive deficits and behavioural alterations, in non-human primates (NHPs). Various brain injuries and neurodegenerative conditions are characterised by chronic astrocyte activation (astrogliosis). Glial fibrillary acidic protein (GFAP), an astrocyte-specific protein, increases during astrogliosis and remains elevated after an injury. Whether infant anaesthesia is associated with a sustained increase in GFAP is unknown. We hypothesised that GFAP is increased in specific brain areas of NHPs 2 yr after infant anaesthesia, consistent with prior injury. METHODS: Eight 6-day-old NHPs per group were exposed to 5 h isoflurane once (1×) or three times (3×), or to room air as a control (Ctr). Two years after exposure, their brains were assessed for GFAP density changes in the primary visual cortex (V1), perirhinal cortex (PRC), hippocampal subiculum, amygdala, and orbitofrontal cortex (OFC). We also assessed concomitant microglia activation and hippocampal neurogenesis. RESULTS: Compared with controls, GFAP densities in V1 were increased in exposed groups (Ctr: 0.208 [0.085-0.427], 1×: 0.313 [0.108-0.533], 3×: 0.389 [0.262-0.652]), whereas the density of activated microglia was unchanged. In addition, GFAP densities were increased in the 3× group in the PRC and the subiculum, and in both exposure groups in the amygdala, but there was no increase in the OFC. There were no differences in hippocampal neurogenesis among groups. CONCLUSIONS: Two years after infant anaesthesia, NHPs show increased GFAP without concomitant microglia activation in specific brain areas. These long-lasting structural changes in the brain caused by infant anaesthesia exposure may be associated with functional alterations at this age.


Subject(s)
Anesthesia, Inhalation/adverse effects , Anesthetics, Inhalation/toxicity , Brain/drug effects , Gliosis/chemically induced , Isoflurane/toxicity , Microglia/drug effects , Administration, Inhalation , Age Factors , Anesthetics, Inhalation/administration & dosage , Animals , Animals, Newborn , Apoptosis/drug effects , Brain/metabolism , Brain/pathology , Calcium-Binding Proteins/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Gliosis/pathology , Isoflurane/administration & dosage , Macaca mulatta , Male , Microfilament Proteins/metabolism , Microglia/metabolism , Microglia/pathology , Time Factors
6.
Curr Opin Anaesthesiol ; 34(4): 402-408, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34010174

ABSTRACT

PURPOSE OF REVIEW: Long-term behavioural and cognitive impairments after exposure to general anaesthetics during infancy is an intensely investigated and controversial topic. Recent clinical studies with prospective assessments associate exposure with long-term behavioural alterations rather than cognitive impairments. This review aims to provide an understanding of the long-term cognitive impairments and behavioural alterations found in recent animal studies and to summarize latest advances in strategies to protect against anaesthesia-induced developmental neurotoxicity (AIDN). RECENT FINDINGS: Preclinical studies, particularly those in nonhuman primates (NHPs), provide accumulating evidence that anaesthesia exposure during infancy is associated with long-term alterations in behaviour, but cognitive impairments are more controversial. Results from recent studies aiming to find mitigating strategies to reduce AIDN or to identify alternative anaesthetic agents include the co-administration of dexmedetomidine with the anaesthetic drugs or the alternative use of hypnotic neurosteroids without being harmful to the developing brain. SUMMARY: Recent findings in animal studies with translational relevance support the proposed association between early-in-life anaesthesia exposure and long-term alterations in behaviour. Studies aiming to prevent AIDN are promising and need evaluation in the NHP model. The careful design of subsequent translational studies will be critical to advance the field forward towards safer anaesthesia exposure in children.


Subject(s)
Anesthetics, General , Neurotoxicity Syndromes , Animals , Brain , Cognition , Humans , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/prevention & control , Prospective Studies
7.
Br J Anaesth ; 126(2): 486-499, 2021 02.
Article in English | MEDLINE | ID: mdl-33198945

ABSTRACT

BACKGROUND: Clinical studies show that children exposed to anaesthetics for short times at young age perform normally on intelligence tests, but display altered social behaviours. In non-human primates (NHPs), infant anaesthesia exposure for several hours causes neurobehavioural impairments, including delayed motor reflex development and increased anxiety-related behaviours assessed by provoked response testing. However, the effects of anaesthesia on spontaneous social behaviours in juvenile NHPs have not been investigated. We hypothesised that multiple, but not single, 5 h isoflurane exposures in infant NHPs are associated with impairments in specific cognitive domains and altered social behaviours at juvenile age. METHODS: Eight Rhesus macaques per group were anaesthetised for 5 h using isoflurane one (1×) or three (3×) times between postnatal days 6 and 12 or were exposed to room air (control). Cognitive testing, behavioural assessments in the home environment, and provoked response testing were performed during the first 2 yr of life. RESULTS: The cognitive functions tested did not differ amongst groups. However, compared to controls, NHPs in the 3× group showed less close social behaviour (P=0.016), and NHPs in the 1× group displayed increased anxiety-related behaviours (P=0.038) and were more inhibited towards novel objects (P<0.001). CONCLUSIONS: 5 h exposures of NHPs to isoflurane during infancy are associated with decreased close social behaviour after multiple exposures and more anxiety-related behaviours and increased behavioural inhibition after single exposure, but they do not affect the cognitive domains tested. Our findings are consistent with behavioural alterations in social settings reported in clinical studies, which may guide future research.


Subject(s)
Anesthetics, Inhalation/toxicity , Behavior, Animal/drug effects , Brain/drug effects , Cognition/drug effects , Isoflurane/toxicity , Neurotoxicity Syndromes/etiology , Social Behavior , Age Factors , Anesthetics, Inhalation/administration & dosage , Animals , Animals, Newborn , Anxiety/chemically induced , Anxiety/physiopathology , Anxiety/psychology , Brain/physiopathology , Drug Administration Schedule , Exploratory Behavior/drug effects , Female , Isoflurane/administration & dosage , Macaca mulatta , Male , Motor Activity/drug effects , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Reaction Time/drug effects , Time Factors
8.
Neurotoxicology ; 79: 75-83, 2020 07.
Article in English | MEDLINE | ID: mdl-32387222

ABSTRACT

BACKGROUND: Preclinical studies suggest that exposures of infant animals to general anesthetics cause acute neurotoxicity and affect their neurobehavioral development representing a potential risk to human infants undergoing anesthesia. Alternative or mitigating strategies to counteract such adverse effects are desirable. Dexmedetomidine (DEX) is a clinically established sedative with potential neuroprotective properties. DEX ameliorates experimental brain injury as well as neurotoxicity caused by anesthetic doses of sevoflurane (SEVO) or other general anesthetics in infant animals. However, it is unknown whether DEX also is beneficial when given together with lower doses of these drugs. Here we tested the hypothesis that DEX co-administration with a sub-anesthetic dose of SEVO reduces responsiveness to external stimuli while also protecting against SEVO-induced brain cell apoptosis. METHOD: Rats were exposed on postnatal day 7 for 6 h to SEVO 1.1, 1.8, or 2.5% and were given intraperitoneal injections of saline or DEX at different doses (1-25 µg/kg) three times during the exposure. Responsiveness to external stimuli, respiratory rates, and blood gases were assessed. Apoptosis was determined in cortical and subcortical brain areas by activated caspase-3 immunohistochemistry. RESULTS: Rats exposed to SEVO 1.1% alone were sedated but still responsive to external stimuli whereas those exposed to SEVO 1.8% reached complete unresponsiveness. SEVO-induced brain cell apoptosis increased dose-dependently, with SEVO 1.1% causing a small increase in apoptosis above that in controls. Co-administration of DEX at 1 µg/kg did not alter the responsiveness to stimuli nor the apoptosis induced by SEVO 1.1%. In contrast, co-administration of DEX at 5 µg/kg or higher with SEVO 1.1% reduced responsiveness but potentiated apoptosis. CONCLUSIONS: In the neonatal rat model, co-administration of a clinically relevant dose of DEX (1 µg/kg) with a sub-anesthetic dose of SEVO (1.1%) does not affect the neurotoxicity of the anesthetic while co-administration of higher doses of DEX with SEVO 1.1% potentiates it.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/toxicity , Anesthetics, Inhalation/toxicity , Apoptosis/drug effects , Brain/drug effects , Dexmedetomidine/toxicity , Neurotoxicity Syndromes/etiology , Sevoflurane/toxicity , Animals , Animals, Newborn , Brain/pathology , Brain/physiopathology , Dose-Response Relationship, Drug , Drug Synergism , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/physiopathology , Rats, Wistar , Respiratory Rate/drug effects , Sensory Thresholds/drug effects
9.
FASEB J ; 34(6): 7404-7426, 2020 06.
Article in English | MEDLINE | ID: mdl-32307754

ABSTRACT

Fragile X syndrome (FXS) is the leading known inherited intellectual disability and the most common genetic cause of autism. The full mutation results in transcriptional silencing of the Fmr1 gene and loss of fragile X mental retardation protein (FMRP) expression. Defects in neuroenergetic capacity are known to cause a variety of neurodevelopmental disorders. Thus, we explored the integrity of forebrain mitochondria in Fmr1 knockout mice during the peak of synaptogenesis. We found inefficient thermogenic respiration due to futile proton leak in Fmr1 KO mitochondria caused by coenzyme Q (CoQ) deficiency and an open cyclosporine-sensitive channel. Repletion of mitochondrial CoQ within the Fmr1 KO forebrain closed the channel, blocked the pathological proton leak, restored rates of protein synthesis during synaptogenesis, and normalized the key phenotypic features later in life. The findings demonstrate that FMRP deficiency results in inefficient oxidative phosphorylation during the neurodevelopment and suggest that dysfunctional mitochondria may contribute to the FXS phenotype.


Subject(s)
Cell Respiration/physiology , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Mitochondria/metabolism , Mitochondria/pathology , Thermogenesis/physiology , Animals , Autistic Disorder/metabolism , Autistic Disorder/pathology , Disease Models, Animal , Female , Fragile X Mental Retardation Protein/metabolism , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Knockout , Neurogenesis/physiology , Protons
10.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L264-L275, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31800261

ABSTRACT

Duodenogastroesophageal reflux (DGER) is associated with chronic lung disease. Bile acids (BAs) are established markers of DGER aspiration and are important risk factors for reduced post-transplant lung allograft survival by disrupting the organ-specific innate immunity, facilitating airway infection and allograft failure. However, it is unknown whether BAs also affect airway reactivity. We investigated the acute effects of 13 BAs detected in post-lung-transplant surveillance bronchial washings (BW) on airway contraction. We exposed precision-cut slices from human and mouse lungs to BAs and monitored dynamic changes in the cross-sectional luminal area of peripheral airways using video phase-contrast microscopy. We also used guinea pig tracheal rings in organ baths to study BA effects in proximal airway contraction induced by electrical field stimulation. We found that most secondary BAs at low micromolar concentrations strongly and reversibly relaxed smooth muscle and inhibited peripheral airway constriction induced by acetylcholine but not by noncholinergic bronchoconstrictors. Similarly, secondary BAs strongly inhibited cholinergic constrictions in tracheal rings. In contrast, TC-G 1005, a specific agonist of the BA receptor Takeda G protein-coupled receptor 5 (TGR5), did not cause airway relaxation, and Tgr5 deletion in knockout mice did not affect BA-induced relaxation, suggesting that this receptor is not involved. BAs inhibited acetylcholine-induced inositol phosphate synthesis in human airway smooth muscle cells overexpressing the muscarinic M3 receptor. Our results demonstrate that select BAs found in BW of patients with lung transplantation can affect airway reactivity by inhibiting the cholinergic contractile responses of the proximal and peripheral airways, possibly by acting as antagonists of M3 muscarinic receptors.


Subject(s)
Acetylcholine/metabolism , Bile Acids and Salts/pharmacology , Bronchoconstriction/drug effects , Lung/physiopathology , Animals , Bronchoconstrictor Agents/pharmacology , Chenodeoxycholic Acid/pharmacology , Electric Stimulation , Guinea Pigs , Humans , Inositol Phosphates/biosynthesis , Lung/drug effects , Male , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Muscarinic/metabolism , Serotonin/pharmacology , Taurolithocholic Acid/pharmacology , Trachea/drug effects
11.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L82-L93, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30284927

ABSTRACT

Nonvisual opsin (OPN) receptors have recently been implicated in blue light-mediated photorelaxation of smooth muscle in various organs. Since photorelaxation has not yet been demonstrated in airway smooth muscle (ASM) or in human tissues, we questioned whether functional OPN receptors are expressed in mouse and human ASM. mRNA, encoding the OPN 3 receptor, was detected in both human and mouse ASM. To demonstrate the functionality of the OPN receptors, we performed wire myography of ex vivo ASM from mouse and human upper airways. Blue light-mediated relaxation of ACh-preconstricted airways was intensity and wavelength dependent (maximum relaxation at 430-nm blue light) and was inhibited by blockade of the large-conductance calcium-activated potassium channels with iberiotoxin. We further implicated OPN receptors as key mediators in functional photorelaxation by demonstrating increased relaxation in the presence of a G protein receptor kinase 2 inhibitor or an OPN chromophore (9- cis retinal). We corroborated these responses in peripheral airways of murine precision-cut lung slices. This is the first demonstration of photorelaxation in ASM via an OPN receptor-mediated pathway.


Subject(s)
Light , Muscle Relaxation , Myocytes, Smooth Muscle/metabolism , Rod Opsins/metabolism , Trachea/metabolism , Animals , Humans , Mice , Myocytes, Smooth Muscle/cytology , Signal Transduction , Trachea/cytology
12.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L385-L390, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30489155

ABSTRACT

Airway smooth muscle (ASM) cells express GABA A receptors (GABAARs), and previous reports have demonstrated that GABAAR activators relax ASM. However, given the activity of GABAARs in central nervous system inhibitory neurotransmission, concern exists that these activators may lead to undesirable sedation. MIDD0301 is a novel imidazobenzodiazepine and positive allosteric modulator of the GABAAR with limited brain distribution, thus eliminating the potential for sedation. Here, we demonstrate that MIDD0301 relaxes histamine-contracted guinea pig ( P < 0.05, n = 6-9) and human ( P < 0.05, n = 6-10) tracheal smooth muscle ex vivo in organ bath experiments, dilates mouse peripheral airways ex vivo in precision-cut lung-slice experiments ( P < 0.001, n = 16 airways from three mice), and alleviates bronchoconstriction in vivo in mice, as assessed by the forced-oscillation technique ( P < 0.05, n = 6 mice). Only trace concentrations of the compound were detected in the brains of mice after inhalation of nebulized 5 mM MIDD0301. Given its favorable pharmacokinetic properties and demonstrated ability to relax ASM in a number of clinically relevant experimental paradigms, MIDD0301 is a promising drug candidate for bronchoconstrictive diseases, such as asthma.


Subject(s)
Asthma/drug therapy , Blood-Brain Barrier/drug effects , GABA Agents/pharmacology , Receptors, GABA-A/drug effects , Animals , Guinea Pigs , Humans , Ligands , Lung/drug effects , Lung/metabolism , Male , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Receptors, GABA-A/metabolism , Trachea/drug effects , Trachea/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L105-L113, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30407863

ABSTRACT

We have previously reported that mice genetically deficient in the actin binding protein gelsolin exhibit impaired airway smooth muscle (ASM) relaxation. Primary cultured ASM cells from these mice demonstrate enhanced inositol triphosphate (IP3) synthesis and increased intracellular calcium in response to Gq-coupled agonists. We hypothesized that this was due to increased intracellular availability of unbound phosphatidylinositol 4,5-bisphosphate (PIP2), based on the fact that gelsolin contains a short peptide region that binds PIP2, presumably making it a less available substrate. We now questioned whether a peptide that corresponds to the PIP2 binding region of gelsolin could modulate ASM signaling and contraction. The 10 amino acid sequence of the gelsolin peptide within the PIP2-binding region was incubated with primary cultures of human ASM cells, and IP3 synthesis was measured in response to a Gq-coupled agonist. Gelsolin peptide-treated cells generated less IP3 under basal and bradykinin or acetylcholine (Gq-coupled) conditions. Acetylcholine-induced contractile force measured in isolated tracheal rings from mice and human tracheal muscle strips in organ baths was attenuated in the presence of the gelsolin peptide. The gelsolin peptide also attenuated methacholine-induced airway constriction in murine precision-cut lung slices. Furthermore, this peptide fragment delivered to the respiratory system of mice via nebulization attenuated subsequent methacholine-induced increases in airway resistance in vivo. The current study demonstrates that introduction of this small gelsolin peptide into the airway may be a novel therapeutic option in bronchoconstrictive diseases.


Subject(s)
Bronchoconstriction/drug effects , Gelsolin/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/metabolism , Peptides/pharmacology , Trachea/metabolism , Animals , Gelsolin/chemistry , Humans , Male , Mice , Muscle, Smooth/pathology , Peptides/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Trachea/pathology
14.
J Physiol ; 596(13): 2491-2506, 2018 07.
Article in English | MEDLINE | ID: mdl-29790164

ABSTRACT

KEY POINTS: We investigated the excitation-contraction coupling mechanisms in small pulmonary veins (SPVs) in rat precision-cut lung slices. We found that SPVs contract strongly and reversibly in response to extracellular ATP and other vasoconstrictors, including angiotensin-II and endothelin-1. ATP-induced vasoconstriction in SPVs was associated with the stimulation of purinergic P2Y2 receptors in vascular smooth muscle cell, activation of phospholipase C-ß and the generation of intracellular Ca2+ oscillations mediated by cyclic Ca2+ release events via the inositol 1,4,5-trisphosphate receptor. Active constriction of SPVs may play an important role in the development of pulmonary hypertension and pulmonary oedema. ABSTRACT: The small pulmonary veins (SPVs) may play a role in the development of pulmonary hypertension and pulmonary oedema via active changes in SPV diameter, mediated by vascular smooth muscle cell (VSMC) contraction. However, the excitation-contraction coupling mechanisms during vasoconstrictor stimulation remain poorly understood in these veins. We used rat precision-cut lung slices and phase-contrast and confocal microscopy to investigate dynamic changes in SPV cross-sectional luminal area and intracellular Ca2+ signalling in their VSMCs. We found that the SPV (∼150 µm in diameter) contract strongly in response to extracellular ATP and other vasoconstrictors, including angiotensin-II and endothelin-1. ATP-induced SPV contraction was fast, concentration-dependent, completely reversible upon ATP washout, and inhibited by purinergic receptor antagonists suramin and AR-C118925 but not by MRS2179. Immunofluorescence showed purinergic P2Y2 receptors expressed in SPV VSMCs. ATP-induced SPV contraction was inhibited by phospholipase Cß inhibitor U73122 and accompanied by intracellular Ca2+ oscillations in the VSMCs. These Ca2+ oscillations and SPV contraction were inhibited by the inositol 1,4,5-trisphosphate receptor inhibitor 2-APB but not by ryanodine. The results of the present study suggest that ATP-induced vasoconstriction in SPVs is associated with the activation of purinergic P2Y2 receptors in VSMCs and the generation of Ca2+ oscillations.


Subject(s)
Calcium/physiology , Muscle Contraction , Myocytes, Smooth Muscle/physiology , Pulmonary Veins/physiology , Receptors, Purinergic P2Y2/metabolism , Vasoconstriction , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Cross-Sectional Studies , Excitation Contraction Coupling , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Myocytes, Smooth Muscle/cytology , Phospholipase C beta/metabolism , Pulmonary Veins/cytology , Rats
15.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L747-58, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26773068

ABSTRACT

Enhanced contractility of airway smooth muscle (ASM) is a major pathophysiological characteristic of asthma. Expanding the therapeutic armamentarium beyond ß-agonists that target ASM hypercontractility would substantially improve treatment options. Recent studies have identified naturally occurring phytochemicals as candidates for acute ASM relaxation. Several flavonoids were evaluated for their ability to acutely relax human and murine ASM ex vivo and murine airways in vivo and were evaluated for their ability to inhibit procontractile signaling pathways in human ASM (hASM) cells. Two members of the flavonol subfamily, galangin and fisetin, significantly relaxed acetylcholine-precontracted murine tracheal rings ex vivo (n = 4 and n = 5, respectively, P < 0.001). Galangin and fisetin also relaxed acetylcholine-precontracted hASM strips ex vivo (n = 6-8, P < 0.001). Functional respiratory in vivo murine studies demonstrated that inhaled galangin attenuated the increase in lung resistance induced by inhaled methacholine (n = 6, P < 0.01). Both flavonols, galangin and fisetin, significantly inhibited purified phosphodiesterase-4 (PDE4) (n = 7, P < 0.05; n = 7, P < 0.05, respectively), and PLCß enzymes (n = 6, P < 0.001 and n = 6, P < 0.001, respectively) attenuated procontractile Gq agonists' increase in intracellular calcium (n = 11, P < 0.001), acetylcholine-induced increases in inositol phosphates, and CPI-17 phosphorylation (n = 9, P < 0.01) in hASM cells. The prorelaxant effect retained in these structurally similar flavonols provides a novel pharmacological method for dual inhibition of PLCß and PDE4 and therefore may serve as a potential treatment option for acute ASM constriction.


Subject(s)
Flavonoids/pharmacology , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Phospholipase C beta/antagonists & inhibitors , Animals , Aorta/drug effects , Aorta/physiopathology , Asthma/drug therapy , Bronchoconstriction/drug effects , Calcium Signaling , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Drug Evaluation, Preclinical , Flavonoids/chemistry , Flavonols , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Male , Mice , Muscle Contraction , Muscle, Smooth/physiology , Muscle, Smooth/physiopathology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Phospholipase C beta/physiology
16.
J Physiol ; 591(23): 5999-6015, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24144878

ABSTRACT

Hydrogen sulphide (H2S) is a signalling molecule that appears to regulate diverse cell physiological process in several organs and systems including vascular and airway smooth muscle cell (SMC) contraction. Decreases in endogenous H2S synthesis have been associated with the development of cardiovascular diseases and asthma. Here we investigated the mechanism of airway SMC relaxation induced by H2S in small intrapulmonary airways using mouse lung slices and confocal and phase-contrast video microscopy. Exogenous H2S donor Na2S (100 µm) reversibly inhibited Ca(2+) release and airway contraction evoked by inositol-1,4,5-trisphosphate (InsP3) uncaging in airway SMCs. Similarly, InsP3-evoked Ca(2+) release and contraction was inhibited by endogenous H2S precursor l-cysteine (10 mm) but not by l-serine (10 mm) or either amino acid in the presence of dl-propargylglycine (PPG). Consistent with the inhibition of Ca(2+) release through InsP3 receptors (InsP3Rs), Na2S reversibly inhibited acetylcholine (ACh)-induced Ca(2+) oscillations in airway SMCs. In addition, Na2S, the H2S donor GYY-4137, and l-cysteine caused relaxation of airways pre-contracted with either ACh or 5-hydroxytryptamine (5-HT). Na2S-induced airway relaxation was resistant to a guanylyl cyclase inhibitor (ODQ) and a protein kinase G inhibitor (Rp-8-pCPT-cGMPS). The effects of H2S on InsP3-evoked Ca(2+) release and contraction as well as on the relaxation of agonist-contracted airways were mimicked by the thiol-reducing agent dithiothreitol (DTT, 10 mm) and inhibited by the oxidizing agent diamide (30 µm). These studies indicate that H2S causes airway SMC relaxation by inhibiting Ca(2+) release through InsP3Rs and consequent reduction of agonist-induced Ca(2+) oscillations in SMCs. The results suggest a novel role for endogenously produced H2S that involves the modulation of InsP3-evoked Ca(2+) release - a cell-signalling system of critical importance for many physiological and pathophysiological processes.


Subject(s)
Calcium/physiology , Hydrogen Sulfide/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Lung/drug effects , Muscle, Smooth/drug effects , Animals , In Vitro Techniques , Lung/physiology , Male , Mice , Mice, Inbred C3H , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/physiology
17.
FASEB J ; 27(12): 4975-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23995289

ABSTRACT

Large conductance voltage- and calcium-activated potassium (BK) channels are highly expressed in airway smooth muscle (ASM). Utilizing the ovalbumin (OVA) and house dust mite (HDM) models of asthma in C57BL/6 mice, we demonstrate that systemic administration of the BK channel agonist rottlerin (5 µg/g) during the challenge period reduced methacholine-induced airway hyperreactivity (AHR) in OVA- and HDM-sensitized mice (47% decrease in peak airway resistance in OVA-asthma animals, P<0.01; 54% decrease in HDM-asthma animals, P<0.01) with a 35-40% reduction in inflammatory cells and 20-35% reduction in Th2 cytokines in bronchoalveolar lavage fluid. Intravenous rottlerin (5 µg/g) reduced AHR within 5 min in the OVA-asthma mice by 45% (P<0.01). With the use of an ex vivo lung slice technique, rottlerin relaxed acetylcholine-stimulated murine airway lumen area to 87 ± 4% of the precontracted area (P<0.01 vs. DMSO control). Rottlerin increased BK channel activity in human ASM cells (V50 shifted by 73.5±13.5 and 71.8±14.6 mV in control and asthmatic cells, respectively, both P<0.05 as compared with pretreatment) and reduced the frequency of acetylcholine-induced Ca(2+) oscillations in murine ex vivo lung slices. These findings suggest that rottlerin, with both anti-inflammatory and ASM relaxation properties, may have benefit in treating asthma.


Subject(s)
Acetophenones/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Benzopyrans/therapeutic use , Large-Conductance Calcium-Activated Potassium Channels/agonists , Acetophenones/pharmacology , Action Potentials , Animals , Anti-Inflammatory Agents/pharmacology , Antigens, Dermatophagoides/toxicity , Asthma/chemically induced , Benzopyrans/pharmacology , Calcium Signaling , Cells, Cultured , Female , Humans , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Ovalbumin/toxicity , Trachea/drug effects , Trachea/pathology
18.
PLoS One ; 8(7): e69598, 2013.
Article in English | MEDLINE | ID: mdl-23936056

ABSTRACT

Intracellular Ca(2+) dynamics of airway smooth muscle cells (ASMC) mediate ASMC contraction and proliferation, and thus play a key role in airway hyper-responsiveness (AHR) and remodelling in asthma. We evaluate the importance of store-operated Ca(2+) entry (SOCE) in these Ca(2+) dynamics by constructing a mathematical model of ASMC Ca(2+) signaling based on experimental data from lung slices. The model confirms that SOCE is elicited upon sufficient Ca(2+) depletion of the sarcoplasmic reticulum (SR), while receptor-operated [Ca(2+) entry (ROCE) is inhibited in such conditions. It also shows that SOCE can sustain agonist-induced Ca(2+) oscillations in the absence of other [Ca(2+) influx. SOCE up-regulation may thus contribute to AHR by increasing the Ca(2+) oscillation frequency that in turn regulates ASMC contraction. The model also provides an explanation for the failure of the SERCA pump blocker CPA to clamp the cytosolic Ca(2+) of ASMC in lung slices, by showing that CPA is unable to maintain the SR empty of Ca(2+). This prediction is confirmed by experimental data from mouse lung slices, and strongly suggests that CPA only partially inhibits SERCA in ASMC.


Subject(s)
Calcium Channels/genetics , Lung/metabolism , Models, Theoretical , Myocytes, Smooth Muscle/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Signaling , Gene Expression Regulation , Indoles/pharmacology , Ion Transport/drug effects , Lung/cytology , Lung/drug effects , Mice , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Tissue Culture Techniques
19.
J Gen Physiol ; 141(2): 165-78, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23359281

ABSTRACT

Protein kinase C (PKC) has been implicated in the regulation of smooth muscle cell (SMC) contraction and may contribute to airway hyperresponsiveness. Here, we combined optical and biochemical analyses of mouse lung slices to determine the effects of PKC activation on Ca(2+) signaling, Ca(2+) sensitivity, protein phosphorylation, and contraction in SMCs of small intrapulmonary airways. We found that 10 µM phorbol-12-myristate-13-acetate or 1 µM phorbol 12,13-dibutyrate induced repetitive, unsynchronized, and transient contractions of the SMCs lining the airway lumen. These contractions were associated with low frequency Ca(2+) oscillations in airway SMCs that resulted from Ca(2+) influx through L-type voltage-gated Ca(2+) channels and the subsequent release of Ca(2+) from intracellular stores through ryanodine receptors. Phorbol ester stimulation of lung slices in which SMC intracellular Ca(2+) concentration ([Ca(2+)](i)) was "clamped" at a high concentration induced strong airway contraction, indicating that PKC mediated sensitization of the contractile response to [Ca(2+)](i). This Ca(2+) sensitization was accompanied by phosphorylation of both the PKC-potentiated PP1 inhibitory protein of 17 kD (CPI-17) and the regulatory myosin light chain. Thrombin, like the phorbol esters, induced a strong Ca(2+) sensitization that was inhibited by the PKC inhibitor GF-109203X and also potentiated airway contraction to membrane depolarization with KCl. In conclusion, we suggest that PKC activation in small airways leads to both the generation of Ca(2+) oscillations and strong Ca(2+) sensitization; agents associated with airway inflammation, such as thrombin, may activate this pathway to sensitize airway smooth muscle to agonists that cause membrane depolarization and Ca(2+) entry and induce airway hyperresponsiveness.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Muscle, Smooth/physiology , Myocardial Contraction/physiology , Myocytes, Smooth Muscle/physiology , Protein Kinase C/metabolism , Animals , Enzyme Activation , Mice , Mice, Inbred C3H , Muscle, Smooth/cytology , Organ Culture Techniques
20.
J Smooth Muscle Res ; 49: 112-24, 2013.
Article in English | MEDLINE | ID: mdl-24662476

ABSTRACT

Enhanced airway smooth muscle (ASM) contraction is an important component in the pathophysiology of asthma. We have shown that ligand gated chloride channels modulate ASM contractile tone during the maintenance phase of an induced contraction, however the role of chloride flux in depolarization-induced contraction remains incompletely understood. To better understand the role of chloride flux under these conditions, muscle force (human ASM, guinea pig ASM), peripheral small airway luminal area (rat ASM) and airway smooth muscle plasma membrane electrical potentials (human cultured ASM) were measured. We found ex vivo guinea pig airway rings, human ASM strips and small peripheral airways in rat lungs slices relaxed in response to niflumic acid following depolarization-induced contraction induced by K(+) channel blockade with tetraethylammonium chloride (TEA). In isolated human airway smooth muscle cells TEA induce depolarization as measured by a fluorescent indicator or whole cell patch clamp and this depolarization was reversed by niflumic acid. These findings demonstrate that ASM depolarization induced contraction is dependent on chloride channel activity. Targeting of chloride channels may be a novel approach to relax hypercontractile airway smooth muscle in bronchoconstrictive disorders.


Subject(s)
Bronchoconstriction/drug effects , Chloride Channels/antagonists & inhibitors , Chloride Channels/physiology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Niflumic Acid/pharmacology , Potassium Channel Blockers/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Tetraethylammonium/antagonists & inhibitors , Tetraethylammonium/pharmacology , Trachea/drug effects , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Guinea Pigs , Humans , In Vitro Techniques , Lung/anatomy & histology , Lung/drug effects , Membrane Potentials/drug effects , Muscle, Smooth/cytology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...