Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Physiol Genomics ; 55(11): 487-503, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37602394

ABSTRACT

Chronic hypercapnia (CH) is a hallmark of respiratory-related diseases, and the level of hypercapnia can acutely or progressively become more severe. Previously, we have shown time-dependent adaptations in steady-state physiology during mild (arterial Pco2 ∼55 mmHg) and moderate (∼60 mmHg) CH in adult goats, including transient (mild CH) or sustained (moderate CH) suppression of acute chemosensitivity suggesting limitations in adaptive respiratory control mechanisms as the level of CH increases. Changes in specific markers of glutamate receptor plasticity, interleukin-1ß, and serotonergic modulation within key nodes of cardiorespiratory control do not fully account for the physiological adaptations to CH. Here, we used an unbiased approach (bulk tissue RNA sequencing) to test the hypothesis that mild or moderate CH elicits distinct gene expression profiles in important brain stem regions of cardiorespiratory control, which may explain the contrasting responses to CH. Gene expression profiles from the brain regions validated the accuracy of tissue biopsy methodology. Differential gene expression analyses revealed greater effects of CH on brain stem sites compared with the medial prefrontal cortex. Mild CH elicited an upregulation of predominantly immune-related genes and predicted activation of immune-related pathways and functions. In contrast, moderate CH broadly led to downregulation of genes and predicted inactivation of cellular pathways related to the immune response and vascular function. These data suggest that mild CH leads to a steady-state activation of neuroinflammatory pathways within the brain stem, whereas moderate CH drives the opposite response. Transcriptional shifts in immune-related functions may underlie the cardiorespiratory network's capability to respond to acute, more severe hypercapnia when in a state of progressively increased CH.NEW & NOTEWORTHY Mild chronic hypercapnia (CH) broadly upregulated immune-related genes and a predicted activation of biological pathways related to immune cell activity and the overall immune response. In contrast, moderate CH primarily downregulated genes related to major histocompatibility complex signaling and vasculature function that led to a predicted inactivation of pathways involving the immune response and vascular endothelial function. The severity-dependent effect on immune responses suggests that neuroinflammation has an important role in CH and may be important in the maintenance of proper ventilatory responses to acute and chronic hypercapnia.


Subject(s)
Hypercapnia , Transcriptome , Humans , Hypercapnia/genetics , Hypercapnia/metabolism , Hypercapnia/pathology , Transcriptome/genetics , Brain/metabolism , Gene Expression Profiling , Immunity
2.
Function (Oxf) ; 4(5): zqad026, 2023.
Article in English | MEDLINE | ID: mdl-37575478

ABSTRACT

Rationale: Acute intermittent hypoxia (AIH) shows promise for enhancing motor recovery in chronic spinal cord injuries and neurodegenerative diseases. However, human trials of AIH have reported significant variability in individual responses. Objectives: Identify individual factors (eg, genetics, age, and sex) that determine response magnitude of healthy adults to an optimized AIH protocol, acute intermittent hypercapnic-hypoxia (AIHH). Methods: In 17 healthy individuals (age = 27 ± 5 yr), associations between individual factors and changes in the magnitude of AIHH (15, 1-min O2 = 9.5%, CO2 = 5% episodes) induced changes in diaphragm motor-evoked potential (MEP) amplitude and inspiratory mouth occlusion pressures (P0.1) were evaluated. Single nucleotide polymorphisms (SNPs) in genes linked with mechanisms of AIH induced phrenic motor plasticity (BDNF, HTR2A, TPH2, MAOA, NTRK2) and neuronal plasticity (apolipoprotein E, APOE) were tested. Variations in AIHH induced plasticity with age and sex were also analyzed. Additional experiments in humanized (h)ApoE knock-in rats were performed to test causality. Results: AIHH-induced changes in diaphragm MEP amplitudes were lower in individuals heterozygous for APOE4 (i.e., APOE3/4) compared to individuals with other APOE genotypes (P = 0.048) and the other tested SNPs. Males exhibited a greater diaphragm MEP enhancement versus females, regardless of age (P = 0.004). Additionally, age was inversely related with change in P0.1 (P = 0.007). In hApoE4 knock-in rats, AIHH-induced phrenic motor plasticity was significantly lower than hApoE3 controls (P < 0.05). Conclusions: APOE4 genotype, sex, and age are important biological determinants of AIHH-induced respiratory motor plasticity in healthy adults. Addition to Knowledge Base: AIH is a novel rehabilitation strategy to induce functional recovery of respiratory and non-respiratory motor systems in people with chronic spinal cord injury and/or neurodegenerative disease. Figure 5 Since most AIH trials report considerable inter-individual variability in AIH outcomes, we investigated factors that potentially undermine the response to an optimized AIH protocol, AIHH, in healthy humans. We demonstrate that genetics (particularly the lipid transporter, APOE), age and sex are important biological determinants of AIHH-induced respiratory motor plasticity.


Subject(s)
Apolipoprotein E4 , Hypercapnia , Hypoxia , Neurodegenerative Diseases , Spinal Cord Injuries , Adult , Animals , Female , Humans , Male , Rats , Young Adult , Apolipoprotein E4/genetics , Hypercapnia/genetics , Hypoxia/genetics , Neuronal Plasticity/genetics , Rats, Sprague-Dawley
3.
Elife ; 112022 11 17.
Article in English | MEDLINE | ID: mdl-36394266

ABSTRACT

Mutations in the transcription factor Phox2b cause congenital central hypoventilation syndrome (CCHS). The syndrome is characterized by hypoventilation and inability to regulate breathing to maintain adequate O2 and CO2 levels. The mechanism by which CCHS impact respiratory control is incompletely understood, and even less is known about the impact of the non-polyalanine repeat expansion mutations (NPARM) form. Our goal was to investigate the extent by which NPARM Phox2b mutation affect (a) respiratory rhythm; (b) ventilatory responses to hypercapnia (HCVR) and hypoxia (HVR); and (c) number of chemosensitive neurons in mice. We used a transgenic mouse line carrying a conditional Phox2bΔ8 mutation (same found in humans with NPARM CCHS). We crossed them with Atoh1cre mice to introduce mutation in regions involved with respiratory function and central chemoreflex control. Ventilation was measured by plethysmograph during neonatal and adult life. In room air, mutation in neonates and adult did not greatly impact basal ventilation. However, Phox2bΔ8, Atoh1cre increased breath irregularity in adults. The HVR and HCVR were impaired in neonates. The HVR, but not HCVR, was still partially compromised in adults. The mutation reduced the number of Phox2b+/TH--expressing neurons as well as the number of fos-activated cells within the ventral parafacial region (also named retrotrapezoid nucleus [RTN] region) induced by hypercapnia. Our data indicates that Phox2bΔ8 mutation in Atoh1-expressing cells impaired RTN neurons, as well as chemoreflex under hypoxia and hypercapnia specially early in life. This study provided new evidence for mechanisms related to NPARM form of CCHS neuropathology.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Homeodomain Proteins , Hypercapnia , Sleep Apnea, Central , Animals , Humans , Mice , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypercapnia/genetics , Hypoxia/genetics , Mice, Transgenic , Mutation , Sleep Apnea, Central/genetics , Homeodomain Proteins/genetics
4.
Circ Res ; 125(10): 907-920, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31451088

ABSTRACT

RATIONALE: Precise regulation of cerebral blood flow is critical for normal brain function. Insufficient cerebral blood flow contributes to brain dysfunction and neurodegeneration. Carbon dioxide (CO2), via effects on local acidosis, is one of the most potent regulators of cerebral blood flow. Although a role for nitric oxide in intermediate signaling has been implicated, mechanisms that initiate CO2-induced vasodilation remain unclear. OBJECTIVE: Acid-sensing ion channel-1A (ASIC1A) is a proton-gated cation channel that is activated by extracellular acidosis. Based on work that implicated ASIC1A in the amygdala and bed nucleus of the stria terminalis in CO2-evoked and acid-evoked behaviors, we hypothesized that ASIC1A might also mediate microvascular responses to CO2. METHODS AND RESULTS: To test this hypothesis, we genetically and pharmacologically manipulated ASIC1A and assessed effects on CO2-induced dilation of cerebral arterioles in vivo. Effects of inhalation of 5% or 10% CO2 on arteriolar diameter were greatly attenuated in mice with global deficiency in ASIC1A (Asic1a-/-) or by local treatment with the ASIC inhibitor, psalmotoxin. Vasodilator effects of acetylcholine, which acts via endothelial nitric oxide synthase were unaffected, suggesting a nonvascular source of nitric oxide may be key for CO2 responses. Thus, we tested whether neurons may be the cell type through which ASIC1A influences microvessels. Using mice in which Asic1a was specifically disrupted in neurons, we found effects of CO2 on arteriolar diameter were also attenuated. CONCLUSIONS: Together, these data are consistent with a model wherein activation of ASIC1A, particularly in neurons, is critical for CO2-induced nitric oxide production and vasodilation. With these findings, ASIC1A emerges as major regulator of microvascular tone.


Subject(s)
Acid Sensing Ion Channels/deficiency , Cerebrovascular Circulation/physiology , Hypercapnia/metabolism , Vasodilation/physiology , Acid Sensing Ion Channels/genetics , Animals , Carbon Dioxide/pharmacology , Cerebrovascular Circulation/drug effects , Hypercapnia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide/metabolism , Vasodilation/drug effects
5.
Orphanet J Rare Dis ; 14(1): 137, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31196221

ABSTRACT

INTRODUCTION: Mucopolysaccharidosis (MPS) IVA or Morquio A syndrome is an autosomal recessive lysosomal storage disorder (LSD) caused by deficiency of the N-acetylgalactosamine-6-sulfatase (GALNS) enzyme, which impairs lysosomal degradation of keratan sulphate and chondroitin-6-sulphate. The multiple clinical manifestations of MPS IVA present numerous challenges for management and necessitate the need for individualised treatment. Although treatment guidelines are available, the methodology used to develop this guidance has come under increased scrutiny. This programme was conducted to provide evidence-based, expert-agreed recommendations to optimise management of MPS IVA. METHODS: Twenty six international healthcare professionals across multiple disciplines, with expertise in managing MPS IVA, and three patient advocates formed the Steering Committee (SC) and contributed to the development of this guidance. Representatives from six Patient Advocacy Groups (PAGs) were interviewed to gain insights on patient perspectives. A modified-Delphi methodology was used to demonstrate consensus among a wider group of healthcare professionals with experience managing patients with MPS IVA and the manuscript was evaluated against the validated Appraisal of Guidelines for Research and Evaluation (AGREE II) instrument by three independent reviewers. RESULTS: A total of 87 guidance statements were developed covering five domains: (1) general management principles; (2) recommended routine monitoring and assessments; (3) disease-modifying interventions (enzyme replacement therapy [ERT] and haematopoietic stem cell transplantation [HSCT]); (4) interventions to support respiratory and sleep disorders; (5) anaesthetics and surgical interventions (including spinal, limb, ophthalmic, cardio-thoracic and ear-nose-throat [ENT] surgeries). Consensus was reached on all statements after two rounds of voting. The overall guideline AGREE II assessment score obtained for the development of the guidance was 5.3/7 (where 1 represents the lowest quality and 7 represents the highest quality of guidance). CONCLUSION: This manuscript provides evidence- and consensus-based recommendations for the management of patients with MPS IVA and is for use by healthcare professionals that manage the holistic care of patients with the intention to improve clinical- and patient-reported outcomes and enhance patient quality of life. It is recognised that the guidance provided represents a point in time and further research is required to address current knowledge and evidence gaps.


Subject(s)
Chondroitinsulfatases/metabolism , Mucopolysaccharidosis IV/metabolism , Chondroitinsulfatases/genetics , Enzyme Replacement Therapy/methods , Female , Humans , Hypercapnia/genetics , Hypercapnia/metabolism , Male
6.
Neuromuscul Disord ; 29(3): 198-212, 2019 03.
Article in English | MEDLINE | ID: mdl-30765255

ABSTRACT

Myotonic dystrophy type 1 (DM1) is one of the most common muscular dystrophies in adults. This review summarises the current literature regarding the natural history of respiratory dysfunction in DM1, the role of central respiratory drive and peripheral respiratory muscle involvement and its significance in respiratory function, and investigates the relationship between genetics (CTG repeat length) and respiratory dysfunction. The review included all articles that reported spirometry on 10 or more myotonic dystrophy patients. The final review included 55 articles between 1964 and 2017. The major conclusions of this review were (1) confirmation of the current consensus that respiratory dysfunction, predominantly a restrictive ventilatory pattern, is common in myotonic dystrophy and is associated with alveolar hypoventilation, chronic hypercapnia, and sleep disturbance in the form of sleep apnoea and sleep related disordered breathing; (2) contrary to commonly held belief, there is no consensus in the literature regarding the relationship between CTG repeat length and severity of respiratory dysfunction and a relationship has not been established; (3) the natural history and time-course of respiratory functional decline is very poorly understood in the current literature; (4) there is a consensus that there is a significant involvement of central respiratory drive in this alveolar hypoventilation however the current literature does not identify the mechanism for this.


Subject(s)
Hypercapnia/physiopathology , Myotonic Dystrophy/physiopathology , Respiration Disorders/physiopathology , Respiratory Muscles/physiopathology , Sleep Wake Disorders/physiopathology , Humans , Hypercapnia/complications , Hypercapnia/genetics , Myotonic Dystrophy/genetics , Respiration Disorders/complications , Sleep Wake Disorders/genetics , Trinucleotide Repeat Expansion/genetics
7.
Sci Transl Med ; 10(457)2018 09 05.
Article in English | MEDLINE | ID: mdl-30185650

ABSTRACT

The elevation of carbon dioxide (CO2) in tissues and the bloodstream (hypercapnia) occurs in patients with severe lung diseases, including chronic obstructive pulmonary disease (COPD). Whereas hypercapnia has been recognized as a marker of COPD severity, a role for hypercapnia in disease pathogenesis remains unclear. We provide evidence that CO2 acts as a signaling molecule in mouse and human airway smooth muscle cells. High CO2 activated calcium-calpain signaling and consequent smooth muscle cell contraction in mouse airway smooth muscle cells. The signaling was mediated by caspase-7-induced down-regulation of the microRNA-133a (miR-133a) and consequent up-regulation of Ras homolog family member A and myosin light-chain phosphorylation. Exposure of wild-type, but not caspase-7-null, mice to hypercapnia increased airway contraction and resistance. Deletion of the Caspase-7 gene prevented hypercapnia-induced airway contractility, which was restored by lentiviral transfection of a miR-133a antagonist. In a cohort of patients with severe COPD, hypercapnic patients had higher airway resistance, which improved after correction of hypercapnia. Our data suggest a specific molecular mechanism by which the development of hypercapnia may drive COPD pathogenesis and progression.


Subject(s)
Caspase 7/metabolism , Hypercapnia/metabolism , Hypercapnia/physiopathology , Muscle Contraction , Muscle, Smooth/physiopathology , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Acetylcholine/pharmacology , Aged , Aged, 80 and over , Airway Resistance , Animals , Calcium/metabolism , Calpain/metabolism , Carbon Dioxide , Chronic Disease , Down-Regulation/drug effects , Enzyme Activation/drug effects , Female , Humans , Hypercapnia/genetics , MEF2 Transcription Factors/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology
8.
Neuron ; 96(5): 1153-1167.e5, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29103805

ABSTRACT

The precise neural circuitry that mediates arousal during sleep apnea is not known. We previously found that glutamatergic neurons in the external lateral parabrachial nucleus (PBel) play a critical role in arousal to elevated CO2 or hypoxia. Because many of the PBel neurons that respond to CO2 express calcitonin gene-related peptide (CGRP), we hypothesized that CGRP may provide a molecular identifier of the CO2 arousal circuit. Here, we report that selective chemogenetic and optogenetic activation of PBelCGRP neurons caused wakefulness, whereas optogenetic inhibition of PBelCGRP neurons prevented arousal to CO2, but not to an acoustic tone or shaking. Optogenetic inhibition of PBelCGRP terminals identified a network of forebrain sites under the control of a PBelCGRP switch that is necessary to arouse animals from hypercapnia. Our findings define a novel cellular target for interventions that may prevent sleep fragmentation and the attendant cardiovascular and cognitive consequences seen in obstructive sleep apnea. VIDEO ABSTRACT.


Subject(s)
Arousal/genetics , Hypercapnia/genetics , Hypercapnia/physiopathology , Sleep/genetics , Acoustic Stimulation , Animals , Calcitonin Gene-Related Peptide/metabolism , Carbon Dioxide/metabolism , Carbon Dioxide/pharmacology , Electroencephalography , Electromyography , Mice , Mice, Inbred C57BL , Nerve Net/physiopathology , Neurons , Optogenetics , Patch-Clamp Techniques , Prosencephalon/physiopathology , Respiration , Sleep Apnea Syndromes/physiopathology
10.
J Biol Chem ; 292(27): 11561-11571, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28507099

ABSTRACT

CO2 is a physiological gas normally produced in the body during aerobic respiration. Hypercapnia (elevated blood pCO2 >≈50 mm Hg) is a feature of several lung pathologies, e.g. chronic obstructive pulmonary disease. Hypercapnia is associated with increased susceptibility to bacterial infections and suppression of inflammatory signaling. The NF-κB pathway has been implicated in these effects; however, the molecular mechanisms underpinning cellular sensitivity of the NF-κB pathway to CO2 are not fully elucidated. Here, we identify several novel CO2-dependent changes in the NF-κB pathway. NF-κB family members p100 and RelB translocate to the nucleus in response to CO2 A cohort of RelB protein-protein interactions (e.g. with Raf-1 and IκBα) are altered by CO2 exposure, although others are maintained (e.g. with p100). RelB is processed by CO2 in a manner dependent on a key C-terminal domain located in its transactivation domain. Loss of the RelB transactivation domain alters NF-κB-dependent transcriptional activity, and loss of p100 alters sensitivity of RelB to CO2 Thus, we provide molecular insight into the CO2 sensitivity of the NF-κB pathway and implicate altered RelB/p100-dependent signaling in the CO2-dependent regulation of inflammatory signaling.


Subject(s)
Carbon Dioxide/immunology , Hypercapnia/immunology , NF-kappa B p52 Subunit/immunology , Signal Transduction/immunology , Transcription Factor RelB/immunology , A549 Cells , Animals , Humans , Hypercapnia/genetics , Hypercapnia/pathology , Mice , NF-kappa B p52 Subunit/genetics , Protein Domains , Signal Transduction/genetics , Transcription Factor RelB/genetics , Transcription, Genetic/genetics , Transcription, Genetic/immunology
11.
Article in English | MEDLINE | ID: mdl-28396262

ABSTRACT

Adaptive capacities, governing the ability of animals to cope with an environmental stressor, have been demonstrated to be strongly dependent upon genetic factors. Two isogenic lines of rainbow trout, previously described for their sensitivity and resilience to an acute confinement challenge, were used in the present study to investigate whether adaptive capacities remain consistent when fish are exposed to a different type of challenge. For this purpose, the effects of a 4-hour hypercapnia (CO2 increase) challenge at concentrations relevant in aquaculture conditions are described for the two isogenic lines. Oxygen consumption, cortisol release, group dispersion and group swimming activity were measured before, during and after the challenge. Sensitivity and resilience for each measure were extracted from temporal responses and analyzed using multivariate statistics. The two fish lines displayed significant differences in their cortisol response, translating differences in the stress axis sensitivity to the stressor. On the contrary, both lines showed, for other measures, similar temporal patterns across the study. Notable within line variability in the stress response was observed, despite identical genome between fish. The results are discussed in the context of animal robustness.


Subject(s)
Adaptation, Psychological , Hypercapnia/metabolism , Oncorhynchus mykiss/metabolism , Animals , Carbon Dioxide/metabolism , Genotype , Hypercapnia/genetics , Oncorhynchus mykiss/genetics , Stress, Physiological/genetics
12.
J Neurosci ; 37(17): 4565-4583, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28363984

ABSTRACT

Current understanding of the contribution of C1 neurons to blood pressure (BP) regulation derives predominantly from experiments performed in anesthetized animals or reduced ex vivo preparations. Here, we use ArchaerhodopsinT3.0 (ArchT) loss-of-function optogenetics to explore BP regulation by C1 neurons in intact, unanesthetized rats. Using a lentivirus that expresses ArchT under the Phox2b-activated promoter PRSx8 (PRSx8-ArchT), ∼65% of transduced neurons were C1 (balance retrotrapezoid nucleus, RTN). Other rats received CaMKII-ArchT3.0 AAV2 (CaMKII-ArchT), which transduced C1 neurons and larger numbers of unidentified glutamatergic and GABAergic cells. Under anesthesia, ArchT photoactivation reduced sympathetic nerve activity and BP and silenced/strongly inhibited most (7/12) putative C1 neurons. In unanesthetized PRSx8-ArchT-treated rats breathing room air, bilateral ArchT photoactivation caused a very small BP reduction that was only slightly larger under hypercapnia (6% FiCO2), but was greatly enhanced during hypoxia (10 and 12% FiO2), after sino-aortic denervation, or during isoflurane anesthesia. The degree of hypotension correlated with percentage of ArchT-transduced C1 neurons. ArchT photoactivation produced similar BP changes in CaMKII-ArchT-treated rats. Photoactivation in PRSX8-ArchT rats reduced breathing frequency (FR), whereas FR increased in CaMKII-ArchT rats. We conclude that the BP drop elicited by ArchT activation resulted from C1 neuron inhibition and was unrelated to breathing changes. C1 neurons have low activity under normoxia, but their activation is important to BP stability during hypoxia or anesthesia and contributes greatly to the hypertension caused by baroreceptor deafferentation. Finally, C1 neurons are marginally activated by hypercapnia and the large breathing stimulation caused by this stimulus has very little impact on resting BP.SIGNIFICANCE STATEMENT C1 neurons are glutamatergic/peptidergic/catecholaminergic neurons located in the medulla oblongata, which may operate as a switchboard for differential, behavior-appropriate activation of selected sympathetic efferents. Based largely on experimentation in anesthetized or reduced preparations, a rostrally located subset of C1 neurons may contribute to both BP stabilization and dysregulation (hypertension). Here, we used Archaerhodopsin-based loss-of-function optogenetics to explore the contribution of these neurons to BP in conscious rats. The results suggest that C1 neurons contribute little to resting BP under normoxia or hypercapnia, C1 neuron discharge is restrained continuously by arterial baroreceptors, and C1 neuron activation is critical to stabilize BP under hypoxia or anesthesia. This optogenetic approach could also be useful to explore the role of C1 neurons during specific behaviors or in hypertensive models.


Subject(s)
Anesthesia , Blood Pressure , Hypercapnia/physiopathology , Hypoxia/physiopathology , Medulla Oblongata/physiopathology , Pressoreceptors , Anesthetics, Inhalation/pharmacology , Animals , Blood Pressure/drug effects , Chemoreceptor Cells , Hypercapnia/genetics , Hypertension/physiopathology , Isoflurane/pharmacology , Male , Neurons , Optogenetics , Rats , Rats, Sprague-Dawley , Transduction, Genetic
13.
PLoS One ; 12(1): e0168930, 2017.
Article in English | MEDLINE | ID: mdl-28045995

ABSTRACT

INTRODUCTION: A previous study has suggested that the Human Leukocyte Antigen (HLA) allele DQB1*06:02 affects hypoxic ventilatory response (HVR) but not hypercapnic ventilatory response (HCVR) in an Asian population. The current study evaluated the relationship in Caucasians and Asians. In addition we assessed whether gender or polymorphisms in genes participating in the control of breathing affect HVR and HCVR. METHODS: A re-breathing system was used to measure HVR and HCVR in 551 young adults (56.8% Caucasians, 30% Asians). HLA-DQB1*06:02 and tagged polymorphisms and coding variants in genes participating in breathing (PHOX2B, GPR4 and TASK2/KCNK5) were analyzed. The associations between HVR/HCVR and HLA-DQB1*06:02, genetic polymorphisms, and gender were evaluated using ANOVA or frequentist association testing with SNPTEST. RESULTS: HVR and gender are strongly correlated. HCVR and gender are not. Mean HVR in women was 0.276±0.168 (liter/minute/%SpO2) compared to 0.429±0.266 (liter/minute/%SpO2) in men, p<0.001 (55.4% higher HVR in men). Women had lower baseline minute ventilation (8.08±2.36 l/m vs. 10.00±3.43l/m, p<0.001), higher SpO2 (98.0±1.3% vs. 96.6±1.7%, p<0.001), and lower EtCO2 (4.65±0.68% vs. 4.82±1.02%, p = 0.025). One hundred and two (18.5%) of the participants had HLA-DQB1*06:02. No association was seen between HLA-DQB1*06:02 and HVR or HCVR. Genetic analysis revealed point wise, uncorrected significant associations between two TASK2/KCNK5 variants (rs2815118 and rs150380866) and HCVR. CONCLUSIONS: This is the largest study to date reporting the relationship between gender and HVR/ HCVR and the first study assessing the association between genetic polymorphisms in humans and HVR/HCVR. The data suggest that gender has a large effect on hypoxic breathing response.


Subject(s)
HLA-DQ beta-Chains/genetics , Hypercapnia/genetics , Hypoxia/genetics , Potassium Channels, Tandem Pore Domain/genetics , Respiration , Adolescent , Adult , Analysis of Variance , Asian People , Female , Genotype , Healthy Volunteers , Homeodomain Proteins/genetics , Humans , Hypercapnia/ethnology , Hypoxia/ethnology , Male , Polymorphism, Genetic , Receptors, G-Protein-Coupled/genetics , Sex Factors , Transcription Factors/genetics , Ventilators, Mechanical , White People , Young Adult
14.
Int J Mol Sci ; 18(2)2017 Jan 27.
Article in English | MEDLINE | ID: mdl-28134810

ABSTRACT

Acidosis commonly exists in the tissue microenvironment of various pathophysiological conditions such as tumors, inflammation, ischemia, metabolic disease, and respiratory disease. For instance, the tumor microenvironment is characterized by acidosis and hypoxia due to tumor heterogeneity, aerobic glycolysis (the "Warburg effect"), and the defective vasculature that cannot efficiently deliver oxygen and nutrients or remove metabolic acid byproduct. How the acidic microenvironment affects the function of blood vessels, however, is not well defined. GPR4 (G protein-coupled receptor 4) is a member of the proton-sensing G protein-coupled receptors and it has high expression in endothelial cells (ECs). We have previously reported that acidosis induces a broad inflammatory response in ECs. Acidosis also increases the expression of several endoplasmic reticulum (ER) stress response genes such as CHOP (C/EBP homologous protein) and ATF3 (activating transcription factor 3). In the current study, we have examined acidosis/GPR4- induced ER stress pathways in human umbilical vein endothelial cells (HUVEC) and other types of ECs. All three arms of the ER stress/unfolded protein response (UPR) pathways were activated by acidosis in ECs as an increased expression of phosphorylated eIF2α (eukaryotic initiation factor 2α), phosphorylated IRE1α (inositol-requiring enzyme 1α), and cleaved ATF6 upon acidic pH treatment was observed. The expression of other downstream mediators of the UPR, such as ATF4, ATF3, and spliced XBP-1 (X box-binding protein 1), was also induced by acidosis. Through genetic and pharmacological approaches to modulate the expression level or activity of GPR4 in HUVEC, we found that GPR4 plays an important role in mediating the ER stress response induced by acidosis. As ER stress/UPR can cause inflammation and cell apoptosis, acidosis/GPR4-induced ER stress pathways in ECs may regulate vascular growth and inflammatory response in the acidic microenvironment.


Subject(s)
Acidosis/metabolism , Endoplasmic Reticulum Stress , Human Umbilical Vein Endothelial Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Acidosis/complications , Acidosis/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Hydrogen-Ion Concentration , Hypercapnia/complications , Hypercapnia/genetics , Models, Biological , Mutant Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/pharmacology , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
15.
Pharmacogenomics ; 18(2): 143-156, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27977335

ABSTRACT

AIM: Fatty acid amide hydrolase (FAAH) degrades anandamide, an endogenous cannabinoid. We hypothesized that FAAH variants will predict risk of morphine-related adverse outcomes due to opioid-endocannabinoid interactions. PATIENTS & METHODS: In 101 postsurgical adolescents receiving morphine analgesia, we prospectively studied ventilatory response to 5% CO2 (HCVR), respiratory depression (RD) and vomiting. Blood was collected for genotyping and morphine pharmacokinetics. RESULTS: We found significant FAAH-morphine interaction for missense (rs324420) and several regulatory variants, with HCVR (p < 0.0001) and vomiting (p = 0.0339). HCVR was more depressed in patients who developed RD compared with those who did not (p = 0.0034), thus FAAH-HCVR association predicts risk of impending RD from morphine use. CONCLUSION: FAAH genotypes predict risk for morphine-related adverse outcomes.


Subject(s)
Amidohydrolases/blood , Amidohydrolases/genetics , Hypercapnia/blood , Hypercapnia/genetics , Morphine/blood , Pulmonary Ventilation/physiology , Adolescent , Analgesics, Opioid/adverse effects , Analgesics, Opioid/blood , Child , Cohort Studies , Female , Humans , Hypercapnia/chemically induced , Linkage Disequilibrium/genetics , Male , Morphine/adverse effects , Polymorphism, Single Nucleotide/genetics , Postoperative Complications/blood , Postoperative Complications/chemically induced , Postoperative Complications/genetics , Prospective Studies , Pulmonary Ventilation/drug effects , Treatment Outcome
16.
Sci Rep ; 6: 26415, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27199002

ABSTRACT

Chronic obstructive pulmonary disease can cause muscle fibre transformation due to chronic intermittent hypoxia-hypercapnia (CIHH). Studies have shown that high expression of Sox6 in muscle could suppress type-I fibres through downregulating the PPARß (peroxisome proliferator-activated receptor ß)/ERRγ (oestrogen-related receptor γ)/microRNA pathway. However, whether this pathway is involved in CIHH-induced muscle fibre transformation is unknown. Electrical stimulation (ES) is an effective approach to ameliorate muscle dysfunction. Here, we explored the effects of ES on CIHH-induced muscle fibre transformation and the microRNA/Sox6 pathway. After CIHH exposure, both the soleus (SOL) and gastrocnemius (GC) muscles showed decreased type-I fibres. The PPARß/ERRγ/mir-499&208b (PEM, for GC) and PPARß/mir-499&208b (PM, for SOL) signalling cascades were suppressed, followed by elevated Sox6 expression. Low frequency electrical stimulation (LFES) activated the PEM/PM pathway and enhanced type-I fibre numbers through suppressing Sox6 in SOL and GC. High frequency electrical stimulation (HFES) promoted type-I fibre expression through activating the PEM pathway in GC. Although PPARß expression and type-I fibres were suppressed in SOL after HFES, no significant change was found in mir-499&208b/Sox6 expression. These results suggest that the microRNA/Sox6 pathway is disturbed after CIHH. Both low and high frequency electrical stimulations induce muscle fibre transformation partly through regulating the microRNA/Sox6 pathway.


Subject(s)
Electric Stimulation Therapy/methods , Hypercapnia/therapy , Hypoxia/therapy , MicroRNAs/genetics , Muscle Fibers, Skeletal/pathology , SOXD Transcription Factors/metabolism , Animals , Disease Models, Animal , Humans , Hypercapnia/genetics , Hypercapnia/metabolism , Hypercapnia/physiopathology , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , PPAR-beta/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Signal Transduction
17.
PLoS One ; 11(3): e0152525, 2016.
Article in English | MEDLINE | ID: mdl-27023369

ABSTRACT

Skeletal muscle dysfunction in chronic obstructive pulmonary disease (COPD) patients is common. Neuromuscular Electrical Stimulation (NMES) is a powerful exercise training that may relieve muscle dysfunction in COPD. This study investigated whether electrical stimulation may have atypical adaptations via activation of miRNA related pathways in counteracting COPD muscle dysfunction. Forty-eight male Sprague-Dawley rats were randomly assigned to 3 groups. With the exception of the rats in the control group, the experimental rats were exposed to chronic intermittent hypoxia-hypercapnia (CIHH) (9∼11%O2,5.5∼6.5%CO2) for 2 or 4 weeks. Electrical stimulation was performed immediately after each CIHH session. Following assessment of the running capacity, biopsy samples were obtained from the gastrocnemius of the rats. The miR-1, miR-133a and miR-133b levels were measured, as well as their related proteins: phosphorylation of Akt (p-AKT), PGC-1alpha (PGC-1α), histone deacetylase 4 (HDAC4) and serum response factor (SRF). Myosin heavy chainIIa (MHCIIa) and myosin heavy chainIIb (MHCIIb) were also measured to assess fiber type changes. After 2 weeks, compared with the controls, only miR-1 and miR-133a were significantly increased (p<0.05) in the exposure group. After 4 weeks, the exposure group exhibited a decreased running distance (p = 0.054) and MHCIIa-to-MHCIIb shift (p<0.05). PGC-1α (p = 0.051), nuclear HDAC4 (p = 0.058), HDAC4, p-AKT, PGC-1α and SRF was also significantly decreased (p<0.05). In contrast, miR-1 and miR-133a were significantly increased (p<0.05). Four weeks of electrical stimulation can partly reversed those changes, and miR-133b exhibited a transient increase after 2 weeks electrical stimulation. Our study indicate miRNAs may have roles in the response of CIHH-impaired muscle to changes during electrical stimulation.


Subject(s)
Hypercapnia/genetics , Hypercapnia/physiopathology , Hypoxia/genetics , Hypoxia/physiopathology , MicroRNAs/metabolism , Muscle, Skeletal/physiopathology , Signal Transduction/genetics , Animals , Cell Nucleus/metabolism , Chronic Disease , Electric Stimulation , Gene Expression Regulation , Hypercapnia/complications , Hypoxia/complications , Male , MicroRNAs/genetics , Muscle, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phenotype , Physical Endurance , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Running , Serum Response Factor/metabolism , Transcription Factors/metabolism
18.
Nature ; 527(7577): 240-4, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26560302

ABSTRACT

Animals have evolved homeostatic responses to changes in oxygen availability that act on different timescales. Although the hypoxia-inducible factor (HIF) transcriptional pathway that controls long-term responses to low oxygen (hypoxia) has been established, the pathway that mediates acute responses to hypoxia in mammals is not well understood. Here we show that the olfactory receptor gene Olfr78 is highly and selectively expressed in oxygen-sensitive glomus cells of the carotid body, a chemosensory organ at the carotid artery bifurcation that monitors blood oxygen and stimulates breathing within seconds when oxygen declines. Olfr78 mutants fail to increase ventilation in hypoxia but respond normally to hypercapnia. Glomus cells are present in normal numbers and appear structurally intact, but hypoxia-induced carotid body activity is diminished. Lactate, a metabolite that rapidly accumulates in hypoxia and induces hyperventilation, activates Olfr78 in heterologous expression experiments, induces calcium transients in glomus cells, and stimulates carotid sinus nerve activity through Olfr78. We propose that, in addition to its role in olfaction, Olfr78 acts as a hypoxia sensor in the breathing circuit by sensing lactate produced when oxygen levels decline.


Subject(s)
Lactic Acid/metabolism , Olfactory Receptor Neurons/metabolism , Oxygen/metabolism , Receptors, Odorant/metabolism , Respiration , Animals , Calcium Signaling , Carotid Body/cytology , Carotid Body/drug effects , Carotid Body/metabolism , Carotid Sinus/innervation , Female , HEK293 Cells , Humans , Hypercapnia/genetics , Hypercapnia/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Lactic Acid/pharmacology , Mice , Oxygen/blood , Receptors, Odorant/deficiency
19.
Rev. patol. respir ; 18(1): 11-22, ene.-mar. 2015. ilus, tab
Article in Spanish | IBECS | ID: ibc-139106

ABSTRACT

El edema agudo de pulmón (EAP), fundamentalmente de origen cardiogénico, supone una importante carga asistencial en las urgencias hospitalarias, así como una importante causa de muerte. Junto a un tratamiento médico óptimo, muchas guías recomiendan el uso de CPAP o ventilación mecánica no invasiva. Aunque los meta-análisis publicados hasta ahora muestran suficiente evidencia para recomendar el uso de dispositivos ventilatorios en el edema agudo de pulmón, existe un ensayo clínico que incluye cerca de 1.000 pacientes en el que no se mostró una clara ventaja de la CPAP o la ventilación mecánica no invasiva con respecto a la oxigenoterapia en el tratamiento de estos pacientes. Esto ha generado cierta controversia en el manejo del edema agudo de pulmón con terapias ventilatorias. Como alternativa existen otros dispositivos no mecánicos, como la CPAP de Boussignac o el oxígeno con alto flujo humidificado, que en estudios iniciales parecen tener resultados similares a la CPAP o la ventilación mecánica no invasiva


Acute pulmonary edema (APE), fundamentally of cardiogenic origin, entails a significant care load in the hospital emergency services and is an important cause of death. Together with optimal medical treatment, many guidelines recommend the use of continuous positive airway pressure (CPAP) or non-invasive mechanical ventilation. Although the meta-analyses published up to date show sufficient evidence to recommend the use of ventilatory devices in acute pulmonary edema, there is a clinical trial including approximately 1000 patients in which no clear advantage of the CPAP or non-invasive mechanical ventilation over oxygen therapy in the treatment of these patients was demonstrated. This has generated some controversy regarding the management of acute pulmonary edema with ventilatory therapies. As an alternative, there are other non-mechanical devices such as Boussignac CPAP or high flow humidified oxygen therapy whose results seem to be similar to CPAP or non-invasive mechanical ventilation in the initial studies


Subject(s)
Female , Humans , Male , Therapeutics/psychology , Therapeutics/standards , Edema/metabolism , Edema/pathology , Shock, Cardiogenic/genetics , Shock, Cardiogenic/metabolism , Pharmacology/instrumentation , Pharmacology/methods , Hypercapnia/metabolism , Hypercapnia/pathology , Therapeutics/instrumentation , Therapeutics/methods , Edema/complications , Edema/diagnosis , Shock, Cardiogenic/diagnosis , Shock, Cardiogenic/physiopathology , Pharmacology/classification , Pharmacology/standards , Hypercapnia/diagnosis , Hypercapnia/genetics
20.
J Physiol ; 593(2): 415-30, 2015 01 15.
Article in English | MEDLINE | ID: mdl-25630262

ABSTRACT

Raphé-derived serotonin (5-HT) and thyrotropin-releasing hormone (TRH) play important roles in fundamental, homeostatic control systems such as breathing and specifically the ventilatory CO2 chemoreflex. Brown Norway (BN) rats exhibit an inherent and severe ventilatory insensitivity to hypercapnia but also exhibit relatively normal ventilation at rest and during other conditions, similar to multiple genetic models of 5-HT system dysfunction in mice. Herein, we tested the hypothesis that the ventilatory insensitivity to hypercapnia in BN rats is due to altered raphé gene expression and the consequent deficiencies in raphé-derived neuromodulators such as TRH. Medullary raphé transcriptome comparisons revealed lower expression of multiple 5-HT neuron-specific genes in BN compared to control Dahl salt-sensitive rats, predictive of reduced central nervous system monoamines by bioinformatics analyses and confirmed by high-performance liquid chromatography measurements. In particular, raphé Trh mRNA and peptide levels were significantly reduced in BN rats, and injections of the stable TRH analogue Taltirelin (TAL) stimulated breathing dose-dependently, with greater effects in BN versus control Sprague-Dawley rats. Importantly, TAL also effectively normalized the ventilatory CO2 chemoreflex in BN rats, but TAL did not affect CO2 sensitivity in control Sprague-Dawley rats. These data establish a molecular basis of the neuromodulatory deficiency in BN rats, and further suggest an important functional role for TRH signalling in the mammalian CO2 chemoreflex.


Subject(s)
Hypercapnia/metabolism , Raphe Nuclei/metabolism , Thyrotropin-Releasing Hormone/metabolism , Transcriptome , Animals , Carbon Dioxide/pharmacology , Hypercapnia/genetics , Neurotransmitter Agents/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raphe Nuclei/drug effects , Rats , Rats, Inbred BN , Rats, Inbred Dahl , Rats, Sprague-Dawley , Reflex , Serotonin/metabolism , Species Specificity , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...