Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Cell Mol Biol ; 60(6): 687-694, 2019 06.
Article in English | MEDLINE | ID: mdl-30571139

ABSTRACT

Mucus overproduction is a major contributor to morbidity and mortality in asthma. Mucus overproduction is induced by orchestrated actions of multiple factors that include inflammatory cytokines and γ-aminobutyric acid (GABA). GABA is produced only by pulmonary neuroendocrine cells (PNECs) in the mouse lung. Recent studies in a neonatal mouse model of allergic inflammation have shown that PNECs play an essential role in mucus overproduction by GABA hypersecretion. Whether PNECs mediate dysregulated GABA signaling for mucus overproduction in asthma is unknown. In this study, we characterized the cellular source of GABA in the lungs of nonhuman primates and humans and assessed GABA secretion and signaling in primate disease models. We found that like in mice, PNECs were the major source of GABA in primate lungs. In addition, an infant nonhuman primate model of asthma exhibited an increase in GABA secretion. Furthermore, subjects with asthma had elevated levels of expression of a subset of GABA type α (GABAα) and type ß (GABAß) receptors in airway epithelium compared with those of healthy control subjects. Last, employing a normal human bronchial epithelial cell model of preinduced mucus overproduction, we showed pharmaceutical blockade of GABAα and GABAß receptor signaling reversed the effect of IL-13 on MUC5AC gene expression and goblet cell proliferation. Together, our data demonstrate an evolutionarily conserved intraepithelial GABA signaling that, in concert with IL-13, plays an essential role in mucus overproduction. Our findings may offer new strategies to ameliorate mucus overproduction in patients with asthma by targeting PNEC secretion and GABA signaling.


Subject(s)
Goblet Cells/pathology , Lung/pathology , Neuroendocrine Cells/metabolism , gamma-Aminobutyric Acid/metabolism , Acute Lung Injury/pathology , Animals , Asthma/pathology , Bronchi/pathology , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Hyperplasia , Interleukin-13/metabolism , Macaca mulatta , Mucus/metabolism , Receptors, GABA/metabolism , Signal Transduction
2.
FASEB J ; 31(9): 4117-4128, 2017 09.
Article in English | MEDLINE | ID: mdl-28566470

ABSTRACT

Pulmonary neuroendocrine cells (PNECs) are the only innervated airway epithelial cells. To what extent neural innervation regulates PNEC secretion and function is unknown. Here, we discover that neurotrophin 4 (NT4) plays an essential role in mucus overproduction after early life allergen exposure by orchestrating PNEC innervation and secretion of GABA. We found that PNECs were the only cellular source of GABA in airways. In addition, PNECs expressed NT4 as a target-derived mechanism underlying PNEC innervation during development. Early life allergen exposure elevated the level of NT4 and caused PNEC hyperinnervation and nodose neuron hyperactivity. Associated with aberrant PNEC innervation, the authors discovered that GABA hypersecretion was required for the induction of mucin Muc5ac expression. In contrast, NT4-/- mice were protected from allergen-induced mucus overproduction and changes along the nerve-PNEC axis without any defects in inflammation. Last, GABA installation restored mucus overproduction in NT4-/- mice after early life allergen exposure. Together, our findings provide the first evidence for NT4-dependent neural regulation of PNEC secretion of GABA in a neonatal disease model. Targeting the nerve-PNEC axis may be a valid treatment strategy for mucus overproduction in airway diseases, such as childhood asthma.-Barrios, J., Patel, K. R., Aven, L., Achey, R., Minns, M. S., Lee, Y., Trinkaus-Randall, V. E., Ai, X. Early life allergen-induced mucus overproduction requires augmented neural stimulation of pulmonary neuroendocrine cell secretion.


Subject(s)
Allergens/immunology , Gene Expression Regulation/immunology , Hypersensitivity/metabolism , Mucus/metabolism , Neuroendocrine Cells/metabolism , Ovalbumin/immunology , Animals , Calcium , Mice, Inbred C57BL , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
3.
Sci Rep ; 5: 13865, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26353920

ABSTRACT

We engineered a transdermal neuromodulation approach that targets peripheral (cranial and spinal) nerves and utilizes their afferent pathways as signaling conduits to influence brain function. We investigated the effects of this transdermal electrical neurosignaling (TEN) method on sympathetic physiology under different experimental conditions. The TEN method involved delivering high-frequency pulsed electrical currents to ophthalmic and maxillary divisions of the right trigeminal nerve and cervical spinal nerve afferents. Under resting conditions, TEN significantly suppressed basal sympathetic tone compared to sham as indicated by functional infrared thermography of facial temperatures. In a different experiment, subjects treated with TEN reported significantly lower levels of tension and anxiety on the Profile of Mood States scale compared to sham. In a third experiment when subjects were experimentally stressed TEN produced a significant suppression of heart rate variability, galvanic skin conductance, and salivary α-amylase levels compared to sham. Collectively these observations demonstrate TEN can dampen basal sympathetic tone and attenuate sympathetic activity in response to acute stress induction. Our physiological and biochemical observations are consistent with the hypothesis that TEN modulates noradrenergic signaling to suppress sympathetic activity. We conclude that dampening sympathetic activity in such a manner represents a promising approach to managing daily stress.


Subject(s)
Stress, Physiological , Stress, Psychological , Sympathetic Nervous System/physiology , Affect , Biomarkers , Cognition , Electric Stimulation , Female , Heart Rate , Humans , Hydrocortisone/metabolism , Male , Psychomotor Performance , Reaction Time , Saliva/metabolism , Skin/innervation , Skin Physiological Phenomena , Thermography , alpha-Amylases/metabolism
4.
FASEB J ; 28(2): 897-907, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24221086

ABSTRACT

Children who are exposed to environmental respiratory insults often develop asthma that persists into adulthood. In this study, we used a neonatal mouse model of ovalbumin (OVA)-induced allergic airway inflammation to understand the long-term effects of early childhood insults on airway structure and function. We showed that OVA sensitization and challenge in early life led to a 2-fold increase in airway smooth muscle (ASM) innervation (P<0.05) and persistent airway hyperreactivity (AHR). In contrast, OVA exposure in adult life elicited short-term AHR without affecting innervation levels. We found that postnatal ASM innervation required neurotrophin (NT)-4 signaling through the TrkB receptor and that early-life OVA exposure significantly elevated NT4 levels and TrkB signaling by 5- and 2-fold, respectively, to increase innervation. Notably, blockade of NT4/TrkB signaling in OVA-exposed pups prevented both acute and persistent AHR without affecting baseline airway function or inflammation. Furthermore, biophysical assays using lung slices and isolated cells demonstrated that NT4 was necessary for hyperreactivity of ASM induced by early-life OVA exposure. Together, our findings show that the NT4/TrkB-dependent increase in innervation plays a critical role in the alteration of the ASM phenotype during postnatal growth, thereby linking early-life allergen exposure to persistent airway dysfunction.


Subject(s)
Muscle, Smooth/metabolism , Nerve Growth Factors/metabolism , Receptor, trkB/metabolism , Allergens/immunology , Animals , Asthma , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Mice , Microscopy, Confocal , Muscle, Smooth/drug effects , Nerve Growth Factors/genetics , Ovalbumin/immunology , Receptor, trkB/genetics
5.
PLoS One ; 8(9): e74469, 2013.
Article in English | MEDLINE | ID: mdl-24040256

ABSTRACT

Phenotypes of lung smooth muscle cells in health and disease are poorly characterized. This is due, in part, to a lack of methodologies that allow for the independent and direct isolation of bronchial smooth muscle cells (BSMCs) and vascular smooth muscle cells (VSMCs) from the lung. In this paper, we describe the development of a bi-fluorescent mouse that permits purification of these two cell populations by cell sorting. By subjecting this mouse to an acute allergen based-model of airway inflammation that exhibits many features of asthma, we utilized this tool to characterize the phenotype of so-called asthmatic BSMCs. First, we examined the biophysical properties of single BSMCs from allergen sensitized mice and found increases in basal tone and cell size that were sustained ex vivo. We then generated for the first time, a comprehensive characterization of the global gene expression changes in BSMCs isolated from the bi-fluorescent mice with allergic airway inflammation. Using statistical methods and pathway analysis, we identified a number of differentially expressed mRNAs in BSMCs from allergen sensitized mice that code for key candidate proteins underlying changes in matrix formation, contractility, and immune responses. Ultimately, this tool will provide direction and guidance for the logical development of new markers and approaches for studying human lung smooth muscle.


Subject(s)
Asthma/genetics , Bronchi/metabolism , Bronchial Hyperreactivity/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Proteome/immunology , Allergens/immunology , Animals , Asthma/immunology , Asthma/pathology , Bronchi/immunology , Bronchi/pathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Cell Size , Disease Models, Animal , Fluorescence , Gene Expression , Gene Expression Profiling , Humans , Immunization , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Ovalbumin/immunology , Proteome/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , Single-Cell Analysis
6.
Organogenesis ; 9(3): 194-8, 2013.
Article in English | MEDLINE | ID: mdl-23974176

ABSTRACT

During embryogenesis, the development of the respiratory tract is closely associated with the formation of an extensive neuronal network. While the topic of respiratory innervation is not new, and similar articles were published previously, recent studies using animal models and genetic tools are breathing new life into the field. In this review, we focus on signaling mechanisms that underlie innervation of the embryonic respiratory tract.


Subject(s)
Embryonic Development , Respiratory System , Signal Transduction , Animals , Brain-Derived Neurotrophic Factor , Humans , Mice , Nerve Growth Factors , Neurons , Rats , Respiratory System/anatomy & histology , Respiratory System/embryology , Respiratory System/innervation
7.
J Neurosci ; 31(43): 15407-15, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22031887

ABSTRACT

Dysfunctional neural control of airway smooth muscle (ASM) is involved in inflammatory diseases, such as asthma. However, neurogenesis in the lung is poorly understood. This study uses mouse models to investigate developmental mechanisms of ASM innervation, a process that is highly coordinated with ASM formation during lung branching morphogenesis. We show that brain-derived neurotrophic factor (BDNF) is an essential ASM-derived signal for innervation. Although BDNF mRNA expression is temporally dissociated with ASM formation and innervation, BDNF protein is coordinately produced through post-transcriptional suppression by miR-206. Using a combination of chemical and genetic approaches to modulate sonic hedgehog (Shh) signaling, a pathway essential for lung branching and ASM formation, we show that Shh signaling blocks miR-206 expression, which in turn increases BDNF protein expression. Together, our work uncovers a functional cascade that involves Shh, miR-206 and BDNF to coordinate ASM formation and innervation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hedgehog Proteins/metabolism , MicroRNAs/metabolism , Muscle, Smooth/physiology , Respiratory System/anatomy & histology , Respiratory System/innervation , Age Factors , Animals , Brain-Derived Neurotrophic Factor/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Hedgehog Proteins/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Muscle, Smooth/innervation , RNA, Messenger/metabolism , Respiratory System/embryology , Signal Transduction/genetics , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...