Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Allergy Clin Immunol ; 149(2): 467-479, 2022 02.
Article in English | MEDLINE | ID: mdl-34953791

ABSTRACT

Asthma is classically described as having either a type 2 (T2) eosinophilic phenotype or a non-T2 neutrophilic phenotype. T2 asthma usually responds to classical bronchodilation therapy and corticosteroid treatment. Non-T2 neutrophilic asthma is often more severe. Patients with non-T2 asthma or late-onset T2 asthma show poor response to the currently available anti-inflammatory therapies. These therapeutic failures result in increased morbidity and cost associated with asthma and pose a major health care problem. Recent evidence suggests that some non-T2 asthma is associated with elevated TH17 cell immune responses. TH17 cells producing Il-17A and IL-17F are involved in the neutrophilic inflammation and airway remodeling processes in severe asthma and have been suggested to contribute to the development of subsets of corticosteroid-insensitive asthma. This review explores the pathologic role of TH17 cells in corticosteroid insensitivity of severe asthma and potential targets to treat this endotype of asthma.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Asthma/immunology , Th17 Cells/immunology , Asthma/drug therapy , Cell Differentiation , Humans , Interleukin-17/antagonists & inhibitors , Interleukin-17/physiology , Interleukin-6/antagonists & inhibitors , Neutrophils/immunology , Severity of Illness Index , Th17 Cells/cytology , rho-Associated Kinases/antagonists & inhibitors
2.
Epilepsia ; 61(3): 572-588, 2020 03.
Article in English | MEDLINE | ID: mdl-32030748

ABSTRACT

OBJECTIVE: Immediately preceding sudden unexpected death in epilepsy (SUDEP), patients experienced a final generalized tonic-clonic seizure (GTCS), rapid ventilation, apnea, bradycardia, terminal apnea, and asystole. Whether a progressive pathophysiology develops and increases risk of SUDEP remains unknown. Here, we determined (a) heart rate, respiratory rate, and blood oxygen saturation (SaO2 ) in low-risk and high-risk knockout (KO) mice; and (b) whether blocking receptors for orexin, a cardiorespiratory neuromodulator, influences cardiorespiratory function mice or longevity in high-risk KO mice. METHODS: Heart rate and SaO2 were determined noninvasively with ECGenie and pulse oximetry. Respiration was determined with noninvasive airway mechanics technology. The role of orexin was determined within subject following acute treatment with a dual orexin receptor antagonist (DORA, 100 mg/kg). The number of orexin neurons in the lateral hypothalamus was determined with immunohistochemistry. RESULTS: Intermittent bradycardia was more prevalent in high-risk KO mice, an effect that may be the result of increased parasympathetic drive. High-risk KO mice had more orexin neurons in the lateral hypothalamus. Blocking of orexin receptors differentially influenced heart rate in KO, but not wild-type (WT) mice. When DORA administration increased heart rate, it also decreased heart rate variability, breathing frequency, and/or hypopnea-apnea. Blocking orexin receptors prevented the methacholine (MCh)-induced increase in breathing frequency in KO mice and reduced MCh-induced seizures, via a direct or indirect mechanism. DORA improved oxygen saturation in KO mice with intermittent hypoxia. Daily administration of DORA to high-risk KO mice increased longevity. SIGNIFICANCE: High-risk KO mice have a unique cardiorespiratory phenotype that is characterized by progressive changes in five interdependent endpoints. Blocking of orexin receptors attenuates some of these endpoints and increases longevity, supporting the notion that windows of opportunity for intervention exist in this preclinical SUDEP model.


Subject(s)
Apnea/genetics , Bradycardia/genetics , Epilepsy/genetics , Hypoxia/genetics , Kv1.1 Potassium Channel/genetics , Sudden Unexpected Death in Epilepsy , Animals , Apnea/physiopathology , Bradycardia/physiopathology , Epilepsy/physiopathology , Heart Rate/drug effects , Heart Rate/physiology , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/pathology , Hypoxia/physiopathology , Methacholine Chloride/toxicity , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Orexin Receptor Antagonists/pharmacology , Orexins/metabolism , Oximetry , Oxygen , Parasympathetic Nervous System/physiopathology , Parasympathomimetics/toxicity , Respiratory Rate/drug effects , Seizures/chemically induced
3.
Epilepsia ; 59(2): 345-357, 2018 02.
Article in English | MEDLINE | ID: mdl-29327348

ABSTRACT

OBJECTIVE: Increased breathing rate, apnea, and respiratory failure are associated with sudden unexpected death in epilepsy (SUDEP). We recently demonstrated the progressive nature of epilepsy and mortality in Kcna1-/- mice, a model of temporal lobe epilepsy and SUDEP. Here we tested the hypothesis that respiratory dysfunction progresses with age in Kcna1-/- mice, thereby increasing risk of respiratory failure and sudden death (SD). METHODS: Respiratory parameters were determined in conscious mice at baseline and following increasing doses of methacholine (MCh) using noninvasive airway mechanics (NAM) systems. Kcna1+/+ , Kcna1+/- , and Kcna1-/- littermates were assessed during 3 age ranges when up to ~30%, ~55%, and ~90% of Kcna1-/- mice have succumbed to SUDEP: postnatal day (P) 32-36, P40-46, and P48-56, respectively. Saturated arterial O2 (SaO2 ) was determined with pulse oximetry. Lung and brain tissues were isolated and Kcna1 gene and protein expression were evaluated by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) and Western blot techniques. Airway smooth muscle responsiveness was assessed in isolated trachea exposed to MCh. RESULTS: Kcna1-/- mice experienced an increase in basal respiratory drive, chronic oxygen desaturation, frequent apnea-hypopnea (A-H), an atypical breathing sequence of A-H-tachypnea-A-H, increased tidal volume, and hyperventilation induced by MCh. The MCh-provoked hyperventilation was dramatically attenuated with age. Of interest, only Kcna1-/- mice developed seizures following exposure to MCh. Seizures were provoked by lower concentrations of MCh as Kcna1-/- mice approached SD. MCh-induced seizures experienced by a subset of younger Kcna1-/- mice triggered death. Respiratory parameters of these younger Kcna1-/- mice resembled older near-SD Kcna1-/- mice. Kcna1 gene and protein were not expressed in Kcna1+/+ and Kcna1+/- lungs, and MCh-mediated airway smooth muscle contractions exhibited similar half-maximal effective concentration( EC50 ) in isolated Kcna1+/+ and Kcna1-/- trachea. SIGNIFICANCE: The Kcna1-/- model of SUDEP exhibits progressive respiratory dysfunction, which suggests a potential increased susceptibility for respiratory failure during severe seizures that may result in sudden death.


Subject(s)
Apnea/genetics , Death, Sudden , Epilepsy, Temporal Lobe/physiopathology , Hypoxia/genetics , Kv1.1 Potassium Channel/genetics , Respiratory Insufficiency/genetics , Animals , Apnea/complications , Apnea/metabolism , Bronchoconstrictor Agents/pharmacology , Disease Models, Animal , Disease Progression , Epilepsy , Epilepsy, Temporal Lobe/complications , Gene Expression , Hyperventilation/chemically induced , Hypoxia/complications , Hypoxia/metabolism , Kv1.1 Potassium Channel/metabolism , Methacholine Chloride/pharmacology , Mice , Mice, Knockout , Muscle, Smooth/drug effects , Respiratory Insufficiency/complications , Respiratory Insufficiency/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tachypnea/complications , Tachypnea/genetics , Tachypnea/metabolism , Tidal Volume , Trachea/drug effects
4.
Respir Res ; 17(1): 103, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27549302

ABSTRACT

BACKGROUND: Pirfenidone was recently approved for treatment of idiopathic pulmonary fibrosis. However, the therapeutic dose of pirfenidone is very high, causing side effects that limit its doses and therapeutic effectiveness. Understanding the molecular mechanisms of action of pirfenidone could improve its safety and efficacy. Because activated fibroblasts are critical effector cells associated with the progression of fibrosis, this study investigated the genes that change expression rapidly in response to pirfenidone treatment of pulmonary fibroblasts and explored their contributions to the anti-fibrotic effects of pirfenidone. METHODS: We used the GeneChip microarray to screen for genes that were rapidly up-regulated upon exposure of human lung fibroblast cells to pirfenidone, with confirmation for specific genes by real-time PCR and western blots. Biochemical and functional analyses were used to establish their anti-fibrotic effects in cellular and animal models of pulmonary fibrosis. RESULTS: We identified Regulator of G-protein Signaling 2 (RGS2) as an early pirfenidone-induced gene. Treatment with pirfenidone significantly increased RGS2 mRNA and protein expression in both a human fetal lung fibroblast cell line and primary pulmonary fibroblasts isolated from patients without or with idiopathic pulmonary fibrosis. Pirfenidone treatment or direct overexpression of recombinant RGS2 in human lung fibroblasts inhibited the profibrotic effects of thrombin, whereas loss of RGS2 exacerbated bleomycin-induced pulmonary fibrosis and mortality in mice. Pirfenidone treatment reduced bleomycin-induced pulmonary fibrosis in wild-type but not RGS2 knockout mice. CONCLUSIONS: Endogenous RGS2 exhibits anti-fibrotic functions. Upregulated RGS2 contributes significantly to the anti-fibrotic effects of pirfenidone.


Subject(s)
Fibroblasts/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Pyridones/pharmacology , RGS Proteins/metabolism , Animals , Bleomycin , Calcium Signaling/drug effects , Cell Line , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling/methods , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RGS Proteins/deficiency , RGS Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thrombin/pharmacology , Time Factors , Transfection , Up-Regulation
5.
Am J Respir Cell Mol Biol ; 53(1): 42-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25368964

ABSTRACT

G protein-coupled receptors (GPCRs) are important regulators of cell functions in asthma. We recently reported that regulator of G-protein signaling (RGS) 2, a selective modulator of Gq-coupled GPCRs, is a key regulator of airway hyper-responsiveness (AHR), the pathophysiologic hallmark of asthma. Because RGS2 protein levels in airway cells were significantly lower in patients with asthma compared with patients without asthma, we further investigated the potential pathological importance of RGS2 repression in asthma. The human RGS2 gene maps to chromosome 1q31. We first screened patients with asthma for RGS2 gene promoter single-nucleotide polymorphisms (SNPs) and found significant differences in the distribution of two RGS2 SNPs (A638G, rs2746071 and C395G, rs2746072) between patients with asthma and nonasthmatic subjects. These two SNPs are always associated with each other and have the same higher prevalence in patients with asthma (65%) as compared with nonasthmatic subjects (35%). Point mutations corresponding to these SNPs decrease RGS2 promoter activity by 44%. The importance of RGS2 down-regulation was then determined in an acute IL-13 mouse model of asthma. Intranasal administration of IL-13 in mice also decreased RGS2 expression in lungs by ∼50% and caused AHR. Although naive RGS2 knockout (KO) mice exhibit spontaneous AHR, acute IL-13 exposure further increased AHR in RGS2 KO mice. Loss of RGS2 also significantly enhanced IL-13-induced mouse airway remodeling, including peribronchial smooth muscle thickening and fibrosis, without effects on goblet cell hyperplasia or airway inflammation in mice. Thus, genetic variations and increased inflammatory cytokines can lead to RGS2 repression, which exacerbates AHR and airway remodeling in asthma.


Subject(s)
Asthma/genetics , Asthma/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , RGS Proteins , Airway Remodeling , Animals , Asthma/chemically induced , Asthma/pathology , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/metabolism , Disease Models, Animal , Female , Humans , Interleukin-13/toxicity , Male , Mice , Mice, Knockout , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , RGS Proteins/genetics , RGS Proteins/metabolism
7.
J Allergy Clin Immunol ; 130(4): 968-76.e3, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22704538

ABSTRACT

BACKGROUND: Drugs targeting individual G protein-coupled receptors are used as asthma therapies, but this strategy is limited because of G protein-coupled receptor signal redundancy. Regulator of G protein signaling 2 (RGS2), an intracellular selective inhibitor of multiple bronchoconstrictor receptors, may play a central role in the pathophysiology and treatment of asthma. OBJECTIVE: We defined functions and mechanisms of RGS2 in regulating airway hyperresponsiveness (AHR), the pathophysiologic hallmark of asthma. METHODS: Real-time PCR and Western blot were used to determine changes in RGS2 expression in ovalbumin-sensitized/-challenged mice. We also used immunohistochemistry and real-time PCR to compare RGS2 expression between human asthmatic and control subjects. The AHR of RGS2 knockout mice was assessed by using invasive tracheostomy and unrestrained plethysmography. Effects of loss of RGS2 on mouse airway smooth muscle (ASM) remodeling, contraction, intracellular Ca(2+), and mitogenic signaling were determined in vivo and in vitro. RESULTS: RGS2 was highly expressed in human and murine bronchial epithelium and ASM and was markedly downregulated in lungs of ovalbumin-sensitized/-challenged mice. Lung tissues and blood monocytes from asthma patients expressed significantly lower RGS2 protein (lung) and mRNA (monocytes) than from nonasthma subjects. The extent of reduction of RGS2 on human monocytes correlated with increased AHR. RGS2 knockout caused spontaneous AHR in mice. Loss of RGS2 augmented Ca(2+) mobilization and contraction of ASM cells. Loss of RGS2 also increased ASM mass and stimulated ASM cell growth via extracellular signal-regulated kinase and phosphatidylinositol 3-kinase pathways. CONCLUSION: We identified RGS2 as a potent modulator of AHR and a potential novel therapeutic target for asthma.


Subject(s)
Bronchial Hyperreactivity/etiology , RGS Proteins/immunology , RGS Proteins/physiology , Animals , Calcium/metabolism , Cell Proliferation , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/physiology , Phosphatidylinositol 3-Kinases/physiology , RGS Proteins/antagonists & inhibitors , RGS Proteins/deficiency , RGS Proteins/genetics , Signal Transduction
8.
J Biol Chem ; 286(29): 25813-22, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21636851

ABSTRACT

Aberrant up-regulation of P-Rex1 expression plays important roles in cancer progression and metastasis. The present study investigated the regulatory mechanism underlying P-Rex1 gene expression in prostate cancer cells. We showed that P-Rex1 expression was much higher in metastatic prostate cancer cells than in prostate epithelial cells and non-metastatic prostate cancer cells. Histone deacetylase (HDAC) inhibitors or silence of endogenous HDAC1 and HDAC2 markedly elevated P-Rex1 transcription in non-metastatic prostate cancer cells, whereas overexpression of recombinant HDAC1 in metastatic prostate cancer cells suppressed P-Rex1 expression. HDAC inhibitor trichostatin A (TSA) also significantly increased P-Rex1 promoter activity and caused acetylated histones to accumulate and associate with the P-Rex1 promoter. One Sp1 site, essential for basal promoter activity, was identified as critical for the TSA effect. TSA treatment did not alter the DNA-binding activity of Sp1 toward the P-Rex1 promoter; however, it facilitated the dissociation of the repressive HDAC1 and HDAC2 from the Sp1 binding region. Interestingly, HDAC1 association with Sp1 and with the P-Rex1 promoter were much weaker in metastatic prostate cancer PC-3 cells than in non-metastatic prostate cancer cells, and HDAC inhibitors only had very modest stimulatory effects on P-Rex1 promoter activity and P-Rex1 expression in PC-3 cells. Altogether, our studies demonstrate that HDACs could regulate P-Rex1 gene transcription by interaction with Sp1 and by region-specific changes in histone acetylation within the P-Rex1 promoter. Disassociation of HDACs from Sp1 on the P-Rex1 promoter may contribute to aberrant up-regulation of P-Rex1 in cancer.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Guanine Nucleotide Exchange Factors/genetics , Prostatic Neoplasms/pathology , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Male , Neoplasm Metastasis , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Sp1 Transcription Factor/metabolism , Substrate Specificity , Transcriptional Activation/drug effects , Up-Regulation/genetics
9.
Int J Cancer ; 130(7): 1521-31, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-21500190

ABSTRACT

G-protein-coupled receptor (GPCR)-stimulated androgen-independent activation of androgen receptor (AR) contributes to acquisition of a hormone-refractory phenotype by prostate cancer. We previously reported that regulator of G-protein signaling (RGS) 2, an inhibitor of GPCRs, inhibits androgen-independent AR activation (Cao et al., Oncogene 2006;25:3719-34). Here, we show reduced RGS2 protein expression in human prostate cancer specimens compared to adjacent normal or hyperplastic tissue. Methylation-specific PCR analysis and bisulfite sequencing indicated that methylation of the CpG island in the RGS2 gene promoter correlated with RGS2 downregulation in prostate cancer. In vitro methylation of this promoter suppressed reporter gene expression in transient transfection studies, whereas reversal of this promoter methylation with 5-aza-2'-deoxycytidine (5-Aza-dC) induced RGS2 reexpression in androgen-independent prostate cancer cells and inhibited their growth under androgen-deficient conditions. Interestingly, the inhibitory effect of 5-Aza-dC was significantly reduced by an RGS2-targeted short hairpin RNA, indicating that reexpressed RGS2 contributed to this growth inhibition. Restoration of RGS2 levels by ectopic expression in androgen-independent prostate cancer cells suppressed growth of xenografts in castrated mice. Thus, RGS2 promoter hypermethylation represses its expression and unmasks a latent pathway for AR transactivation in prostate cancer cells. Targeting this reversible process may provide a new strategy for suppressing prostate cancer progression by reestablishing its androgen sensitivity.


Subject(s)
Androgens/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RGS Proteins/genetics , Androgens/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , CpG Islands/drug effects , CpG Islands/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Down-Regulation/drug effects , Down-Regulation/genetics , Epigenomics/methods , Epithelial Cells/drug effects , Epithelial Cells/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Promoter Regions, Genetic/drug effects , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/pathology , RGS Proteins/antagonists & inhibitors , RGS Proteins/metabolism , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Receptors, G-Protein-Coupled
10.
J Pharmacol Exp Ther ; 342(2): 305-11, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22543031

ABSTRACT

We recently reported that phosphoinositide 3-kinase γ (PI3Kγ) directly regulates airway smooth muscle (ASM) contraction by modulating Ca(2+) oscillations. Because ASM contraction plays a critical role in airway hyperresponsiveness (AHR) of asthma, the aim of the present study was to determine whether targeting PI3Kγ in ASM cells could suppress AHR in vitro and in vivo. Intranasal administration into mice of interleukin-13 (IL-13; 10 µg per mouse), a key pathophysiologic cytokine in asthma, induced AHR after 48 h, as assessed by invasive tracheostomy. Intranasal administration of a broad-spectrum PI3K inhibitor or a PI3Kγ-specific inhibitor 1 h before AHR assessment attenuated IL-13 effects. Airway responsiveness to bronchoconstrictor agonists was also examined in precision-cut mouse lung slices pretreated without or with IL-13 for 24 h. Acetylcholine and serotonin dose-response curves indicated that IL-13-treated lung slices had a 40 to 50% larger maximal airway constriction compared with controls. Furthermore, acetylcholine induced a larger initial Ca(2+) transient and increased Ca(2+) oscillations in IL-13-treated primary mouse ASM cells compared with control cells, correlating with increased cell contraction. As expected, PI3Kγ inhibitor treatment attenuated IL-13-augmented airway contractility of lung slices and ASM cell contraction. In both control and IL-13-treated ASM cells, small interfering RNA-mediated knockdown of PI3Kγ by 70% only reduced the initial Ca(2+) transient by 20 to 30% but markedly attenuated Ca(2+) oscillations and contractility of ASM cells by 50 to 60%. This report is the first to demonstrate that PI3Kγ in ASM cells is important for IL-13-induced AHR and that acute treatment with a PI3Kγ inhibitor can ameliorate AHR in a murine model of asthma.


Subject(s)
Bronchial Hyperreactivity/metabolism , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Interleukin-13/immunology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Phosphoinositide-3 Kinase Inhibitors , Acetylcholine/pharmacology , Animals , Asthma/genetics , Asthma/immunology , Asthma/metabolism , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/immunology , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , Interleukin-13/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Contraction/genetics , Muscle Contraction/immunology , Muscle, Smooth/immunology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Serotonin/pharmacology , Trachea/drug effects , Trachea/immunology , Trachea/metabolism
11.
Front Genet ; 13: 901228, 2022.
Article in English | MEDLINE | ID: mdl-36035149

ABSTRACT

Disruptive variants in lysine methyl transferase 5B (KMT5B/SUV4-20H1) have been identified as likely-pathogenic among humans with neurodevelopmental phenotypes including motor deficits (i.e., hypotonia and motor delay). However, the role that this enzyme plays in early motor development is largely unknown. Using a Kmt5b gene trap mouse model, we assessed neuromuscular strength, skeletal muscle weight (i.e., muscle mass), neuromuscular junction (NMJ) structure, and myofiber type, size, and distribution. Tests were performed over developmental time (postnatal days 17 and 44) to represent postnatal versus adult structures in slow- and fast-twitch muscle types. Prior to the onset of puberty, slow-twitch muscle weight was significantly reduced in heterozygous compared to wild-type males but not females. At the young adult stage, we identified decreased neuromuscular strength, decreased skeletal muscle weights (both slow- and fast-twitch), increased NMJ fragmentation (in slow-twitch muscle), and smaller myofibers in both sexes. We conclude that Kmt5b haploinsufficiency results in a skeletal muscle developmental deficit causing reduced muscle mass and body weight.

12.
J Pharmacol Exp Ther ; 333(2): 393-403, 2010 May.
Article in English | MEDLINE | ID: mdl-20110378

ABSTRACT

Signaling through G protein-coupled receptors (GPCRs) promotes breast cancer metastasis. G proteins convey GPCR signals by dissociating into Galpha and Gbetagamma subunits. The aim of the present study was to determine whether blockade of Gbetagamma signaling suppresses breast cancer cell migration and invasion, which are critical components of metastasis. Conditioned media (CM) of NIH-3T3 fibroblasts are widely used as chemoattractants in in vitro cancer metastasis studies. Expression of a Gbetagamma scavenger peptide attenuated NIH-3T3 CM-induced migration and invasion of both metastatic breast cancer MDA-MB-231 and MDA-MB-436 cells by 40 to 50% without effects on cell viability. Migration and invasion of cells in response to NIH-3T3 CM were also blocked by 8-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-1-naph-thalene-carboxylic acid) (M119K), a Gbetagamma inhibitor, with maximum inhibition exceeding 80% and half-maximal inhibitory concentration (IC50) values of 1 to 2 microM. M119K also attenuated Rac-dependent formation of lamellipodia, a key structure required for metastasis. Constitutively active Rac1 rescued Gbetagamma blockade-mediated inhibition of breast cancer cell migration, whereas dominant negative Rac1 inhibited cell migration similar to Gbetagamma blockade. Furthermore, M119K suppressed Gi protein-coupled CXC chemokine receptor 4 (CXCR4)-dependent MDA-MB-231 cell migration by 80% with an IC50 value of 1 microM, whereas tyrosine kinase receptor-dependent cell migration was significantly less inhibited. However, CXCR4-dependent inhibition of adenylyl cyclase, a Gialpha-mediated response in MDA-MB-231 cells, was not blocked by M119K but was blocked by pertussis toxin, which selectively inactivates Gialpha. This report is the first to directly demonstrate the role of Gbetagamma in cancer cell migration and invasion and suggests that targeting Gbetagamma signaling pathways may provide a novel strategy for suppressing breast cancer metastasis.


Subject(s)
Breast Neoplasms/physiopathology , Cell Movement/physiology , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Neoplasm Invasiveness/physiopathology , Adenylyl Cyclases/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclohexanes/pharmacology , Female , Humans , Microscopy, Fluorescence , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/physiopathology , Peptide Fragments/physiology , Pseudopodia/drug effects , Receptors, CXCR4/physiology , Recombinant Proteins , Signal Transduction/drug effects , Signal Transduction/physiology , Xanthenes/pharmacology , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/physiology
13.
J Pharmacol Exp Ther ; 334(3): 703-9, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20501633

ABSTRACT

Phosphoinositide 3-kinase gamma (PI3Kgamma) has been implicated in the pathogenesis of asthma, but its mechanism has been considered indirect, through release of inflammatory cell mediators. Because airway smooth muscle (ASM) contractile hyper-responsiveness plays a critical role in asthma, the aim of the present study was to determine whether PI3Kgamma can directly regulate contractility of ASM. Immunohistochemistry staining indicated expression of PI3Kgamma protein in ASM cells of mouse trachea and lung, which was confirmed by Western blot analysis in isolated mouse tracheal ASM cells. PI3Kgamma inhibitor II inhibited acetylcholine (ACh)-stimulated airway contraction of cultured precision-cut mouse lung slices in a dose-dependent manner with 75% inhibition at 10 muM. In contrast, inhibitors of PI3Kalpha, PI3Kbeta, or PI3Kdelta, at concentrations 40-fold higher than their reported IC(50) values for their primary targets, had no effect. It is noteworthy that airways in lung slices pretreated with PI3Kgamma inhibitor II still exhibited an ACh-induced initial contraction, but the sustained contraction was significantly reduced. Furthermore, the PI3Kgamma-selective inhibitor had a small inhibitory effect on the ACh-stimulated initial Ca(2+) transient in ASM cells of mouse lung slices or isolated mouse ASM cells but significantly attenuated the sustained Ca(2+) oscillations that are critical for sustained airway contraction. This report is the first to show that PI3Kgamma directly controls contractility of airways through regulation of Ca(2+) oscillations in ASM cells. Thus, in addition to effects on airway inflammation, PI3Kgamma inhibitors may also exert direct effects on the airway contraction that contribute to pathologic airway hyper-responsiveness.


Subject(s)
Calcium Signaling/physiology , Muscle, Smooth/physiology , Phosphatidylinositol 3-Kinases/metabolism , Respiratory System/drug effects , Respiratory System/enzymology , Acetylcholine/pharmacology , Animals , Bronchial Hyperreactivity/physiopathology , Calcium Signaling/drug effects , Cell Separation , Class Ib Phosphatidylinositol 3-Kinase , Enzyme Inhibitors/pharmacology , Immunohistochemistry , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Isoenzymes/physiology , Lung/drug effects , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Phosphoinositide-3 Kinase Inhibitors , Trachea/cytology , Trachea/drug effects
14.
Biochem Pharmacol ; 180: 114172, 2020 10.
Article in English | MEDLINE | ID: mdl-32712053

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disease with irreversible loss of lung tissue and function. Myofibroblasts in the lung are key cellular mediators of IPF progression. Transforming growth factor (TGF)-ß1, a major profibrogenic cytokine, induces pulmonary myofibroblast differentiation, and emerging evidence has established the importance of microRNAs (miRs) in the development of IPF. The objective of this study was to define the pro-fibrotic roles and mechanisms of miRs in TGF-ß1-induced pulmonary myofibroblast differentiation. Using RNA sequencing, we identified miR-424 as an important TGF-ß1-induced miR in human lung fibroblasts (HLFs). Quantitative RT-PCR confirmed that miR-424 expression was increased by 2.6-fold in HLFs in response to TGF-ß1 and was 1.7-fold higher in human fibrotic lung tissues as compared to non-fibrotic lung tissues. TGF-ß1-induced upregulation of miR-424 was blocked by the Smad3 inhibitor SIS3, suggesting the involvement of this canonical TGF-ß1 signaling pathway. Transfection of a miR-424 hairpin inhibitor into HLFs reduced TGF-ß1-induced expression of classic myofibroblast differentiation markers including ɑ-smooth muscle actin (ɑ-SMA) and connective tissue growth factor (CTGF), whereas a miR-424 mimic significantly enhanced TGF-ß1-induced myofibroblast differentiation. In addition, TGF-ß1 induced Smad3 phosphorylation in HLFs, and this response was reduced by the miR-424 inhibitor. In silico analysis identified Slit2, a protein that inhibits TGF-ß1 profibrogenic signaling, as a putative target of regulation by miR-424. Slit2 is less highly expressed in human fibrotic lung tissues than in non-fibrotic lung tissues, and knockdown of Slit2 by its siRNA enhanced TGF-ß1-induced HLF differentiation. Overexpression of a miR-424 mimic down-regulated expression of Slit2 but not the Slit2 major receptor ROBO1 in HLFs. Luciferase reporter assays showed that the miR-424 mimic represses Slit2 3' untranslated region (3'-UTR) reporter activity, and mutations at the seeding regions in the 3'-UTR of Slit2 abolish this inhibition. Together, these data demonstrate a pro-fibrotic role of miR-424 in TGF-ß1-induced HLF differentiation. It functions as a positive feed-back regulator of the TGF-ß1 signaling pathway by reducing expression of the negative regulator Slit2. Thus, targeting miR-424 may provide a new therapeutic strategy to prevent myofibroblast differentiation and IPF progression.


Subject(s)
Cell Differentiation/physiology , Intercellular Signaling Peptides and Proteins/biosynthesis , Lung/metabolism , MicroRNAs/biosynthesis , Myofibroblasts/metabolism , Nerve Tissue Proteins/biosynthesis , Transforming Growth Factor beta1/pharmacology , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Gene Expression , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Lung/cytology , Lung/drug effects , MicroRNAs/genetics , Myofibroblasts/drug effects , Nerve Tissue Proteins/genetics
15.
Sci Signal ; 13(659)2020 11 24.
Article in English | MEDLINE | ID: mdl-33234690

ABSTRACT

Overuse of ß2-adrenoceptor agonist bronchodilators evokes receptor desensitization, decreased efficacy, and an increased risk of death in asthma patients. Bronchodilators that do not target ß2-adrenoceptors represent a critical unmet need for asthma management. Here, we characterize the utility of osthole, a coumarin derived from a traditional Chinese medicine, in preclinical models of asthma. In mouse precision-cut lung slices, osthole relaxed preconstricted airways, irrespective of ß2-adrenoceptor desensitization. Osthole administered in murine asthma models attenuated airway hyperresponsiveness, a hallmark of asthma. Osthole inhibited phosphodiesterase 4D (PDE4D) activity to amplify autocrine prostaglandin E2 signaling in airway smooth muscle cells that eventually triggered cAMP/PKA-dependent relaxation of airways. The crystal structure of the PDE4D complexed with osthole revealed that osthole bound to the catalytic site to prevent cAMP binding and hydrolysis. Together, our studies elucidate a specific molecular target and mechanism by which osthole induces airway relaxation. Identification of osthole binding sites on PDE4D will guide further development of bronchodilators that are not subject to tachyphylaxis and would thus avoid ß2-adrenoceptor agonist resistance.


Subject(s)
Asthma , Coumarins , Animals , Asthma/drug therapy , Coumarins/metabolism , Coumarins/therapeutic use , Drugs, Chinese Herbal , Humans , Lung/metabolism , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology
16.
Cell Death Dis ; 10(9): 670, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511493

ABSTRACT

Transforming growth factor (TGF)-ß1, a main profibrogenic cytokine in the progression of idiopathic pulmonary fibrosis (IPF), induces differentiation of pulmonary fibroblasts to myofibroblasts that produce high levels of collagen, leading to concomitantly loss of lung elasticity and function. Recent studies implicate the importance of microRNAs (miRNAs) in IPF but their regulation and individual pathological roles remain largely unknown. We used both RNA sequencing and quantitative RT-PCR strategies to systematically study TGF-ß1-induced alternations of miRNAs in human lung fibroblasts (HFL). Our data show that miR-133a was significantly upregulated by TGF-ß1 in a time- and concentration-dependent manner. Surprisingly, miR-133a inhibits TGF-ß1-induced myofibroblast differentiation whereas miR-133a inhibitor enhances TGF-ß1-induced myofibroblast differentiation. Interestingly, quantitative proteomics analysis indicates that miR-133a attenuates myofibroblast differentiation via targeting multiple components of TGF-ß1 profibrogenic pathways. Western blot analysis confirmed that miR-133a down-regulates TGF-ß1-induced expression of classic myofibroblast differentiation markers such as ɑ-smooth muscle actin (ɑ-SMA), connective tissue growth factor (CTGF) and collagens. miRNA Target Searcher analysis and luciferase reporter assays indicate that TGF-ß receptor 1, CTGF and collagen type 1-alpha1 (Col1a1) are direct targets of miR-133a. More importantly, miR-133a gene transferred into lung tissues ameliorated bleomycin-induced pulmonary fibrosis in mice. Together, our study identified TGF-ß1-induced miR-133a as an anti-fibrotic factor. It functions as a feed-back negative regulator of TGF-ß1 profibrogenic pathways. Thus, manipulations of miR-133a expression may provide a new therapeutic strategy to halt and perhaps even partially reverse the progression of IPF.


Subject(s)
Cell Differentiation/genetics , Idiopathic Pulmonary Fibrosis/metabolism , MicroRNAs/metabolism , Myofibroblasts/metabolism , Transforming Growth Factor beta1/pharmacology , Actins/genetics , Actins/metabolism , Animals , Bleomycin/toxicity , Cell Differentiation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Female , HEK293 Cells , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Myofibroblasts/drug effects , NIH 3T3 Cells , Proteomics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Eur J Pharmacol ; 578(2-3): 349-58, 2008 Jan 14.
Article in English | MEDLINE | ID: mdl-17936747

ABSTRACT

Alpha(1)-Adrenoceptors and extracellular signal-regulated kinases 1 and 2 (ERK1/2) regulate salivary secretion. However, whether alpha(1)-adrenoceptors couple to ERK1/2 activation and the specific alpha(1)-adrenoceptor subtypes involved in salivary glands is unknown. Western blotting of ERK1/2 phosphorylation showed phenylephrine activated ERK1/2 by 2-3-fold in submandibular gland slices and 3-4-fold in submandibular acinar (SMG-C10) cells with an EC(50) of 2.7+/-2 microM. ERK1/2 activation was blocked by either prazosin or HEAT, indicating alpha(1)-adrenoceptors stimulate ERK1/2 in native glands and SMG-C10 cells. Inhibition of [(125)I]HEAT binding by 5-methylurapidil (selective for alpha(1A) over alpha(1B/)alpha(1D)), but not BMY 7378 (selective for alpha(1D) over alpha(1A/)alpha(1B)), was biphasic and best-fit by a two-site binding model with K(i)(H) and K(i)(L) values for 5-methylurapidil of 0.64+/-0.3 and 91+/-7 nM, respectively, in SMG-C10 membranes. From these binding data, we obtained subtype-selective concentrations of 5-methylurapidil to determine the alpha(1)-adrenoceptor subtype/s activating ERK1/2 in SMG-C10 cells. 5-methylurapidil (20 nM) did not affect phenylephrine- or A-61603- (alpha(1A)-selective agonist) induced ERK1/2 activation; whereas, 30 microM chloroethylclonidine (alpha(1B)-selective antagonist) inhibited ERK1/2 activation by phenylephrine, indicating alpha(1B)-adrenoceptors, but not alpha(1A)-adrenoceptors, activate ERK1/2 in submandibular cells. We also examined alpha(1)-adrenoceptor location and dependence on cholesterol-rich microdomains for activating ERK1/2. Sucrose density gradient centrifugation showed 71+/-3% of alpha(1)-adrenoceptor binding sites were in plasma membranes. Cholesterol-disrupting agents filipin and methyl-beta-cyclodextrin inhibited phenylephrine-stimulated ERK1/2. These results show only alpha(1B)-adrenoceptors activate ERK1/2 and suggest subtype-specific ERK1/2 signaling by alpha(1B)-adrenoceptors may be determined by localization to cholesterol-rich microdomains in submandibular cells.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Submandibular Gland/metabolism , Adrenergic alpha-Agonists/pharmacology , Adrenergic alpha-Antagonists/metabolism , Adrenergic alpha-Antagonists/pharmacology , Animals , Binding, Competitive , Cell Line , Cell Membrane/enzymology , Cell Membrane/metabolism , Cholesterol/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , In Vitro Techniques , Male , Membrane Microdomains/enzymology , Membrane Microdomains/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/drug effects , Saliva/metabolism , Signal Transduction/drug effects , Submandibular Gland/cytology , Submandibular Gland/drug effects , Submandibular Gland/enzymology
18.
Sci Rep ; 8(1): 15543, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341388

ABSTRACT

Dysregulation of microRNAs (miRNAs) contributes to epithelial-mesenchymal transition (EMT) of cancer, but the pathological roles of miRNAs in airway EMT of lung diseases remains largely unknown. We performed sequencing and real-time PCR analysis of the miRNA expression profile of human airway epithelial cells undergoing EMT, and revealed miR-133a to be one of the most common up-regulated miRNAs. MiR-133a was previously reported to be persistently up-regulated in airway epithelial cells of smokers. We found that mice exposed to cigarette smoke (CS) showed airway hyper-responsiveness, a typical symptom occurring in CS-related lung diseases, up-regulation of miR-133a and EMT marker protein N-cadherin in airway epithelium. Importantly, miR-133a overexpression induces airway epithelial cells to undergo spontaneous EMT via down-regulation of grainyhead-like 2 (GRHL2), an epithelial specific transcriptional factor. Loss of GRHL2 causes down-regulation of epithelial splicing regulatory protein 1 (ESRP1), a central coordinator of alternative splicing processes that are critical in the regulation of EMT. Down-regulation of ESRP1 induces isoform switching of adherens junction-associated protein p120-catenin, and leads to the loss of E-cadherin. Our study is the first to demonstrate that up-regulated miR-133a orchestrates airway EMT via alternative splicing processes, which points to novel therapeutic possibilities for the treatment of CS-related lung disease.


Subject(s)
Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , MicroRNAs/biosynthesis , Up-Regulation , Animals , Cells, Cultured , Environmental Exposure , Gene Expression Profiling , Humans , Mice , RNA Splicing , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Smoke/adverse effects
19.
Reprod Biol Endocrinol ; 5: 41, 2007 Nov 02.
Article in English | MEDLINE | ID: mdl-17980032

ABSTRACT

BACKGROUND: Uterine smooth muscle cells exhibit ionic currents that appear to be important in the control of uterine contractility, but how these currents might produce the changes in contractile activity seen in pregnant myometrium has not been established. There are conflicting reports concerning the role of voltage-gated potassium (Kv) channels and large-conductance, calcium-activated potassium (BK) channels in the regulation of uterine contractility. In this study we provide molecular and functional evidence for a role for Kv channels in the regulation of spontaneous contractile activity in mouse myometrium, and also demonstrate a change in Kv channel regulation of contractility in pregnant mouse myometrium. METHODS: Functional assays which evaluated the effects of channel blockers and various contractile agonists were accomplished by quantifying contractility of isolated uterine smooth muscle obtained from nonpregnant mice as well as mice at various stages of pregnancy. Expression of Kv channel proteins in isolated uterine smooth muscle was evaluated by Western blots. RESULTS: The Kv channel blocker 4-aminopyridine (4-AP) caused contractions in nonpregnant mouse myometrium (EC50 = 54 micromolar, maximal effect at 300 micromolar) but this effect disappeared in pregnant mice; similarly, the Kv4.2/Kv4.3 blocker phrixotoxin-2 caused contractions in nonpregnant, but not pregnant, myometrium. Contractile responses to 4-AP were not dependent upon nerves, as neither tetrodotoxin nor storage of tissues at room temperature significantly altered these responses, nor were responses dependent upon the presence of the endometrium. Spontaneous contractions and contractions in response to 4-AP did not appear to be mediated by BK, as the BK channel-selective blockers iberiotoxin, verruculogen, or tetraethylammonium failed to affect either spontaneous contractions or 4-AP-elicited responses. A number of different Kv channel alpha subunit proteins were found in isolated myometrium from both nonpregnant and term-pregnant mice, and one of these proteins - Kv4.3 - was found to disappear in term-pregnant tissues. CONCLUSION: These findings suggest a role for Kv channels in the regulation of uterine contractility, and that changes in the expression and/or function of specific Kv channels may account for the functional changes seen in pregnant myometrium.


Subject(s)
Potassium Channels, Voltage-Gated/physiology , Pregnancy, Animal , Uterine Contraction/physiology , 4-Aminopyridine/pharmacology , Animals , Dose-Response Relationship, Drug , Female , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channels/physiology , Male , Mice , Mice, Inbred C57BL , Myometrium/drug effects , Potassium Channels, Voltage-Gated/metabolism , Pregnancy , Shal Potassium Channels/metabolism , Shal Potassium Channels/physiology , Uterine Contraction/metabolism
20.
Peptides ; 28(3): 505-14, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17174009

ABSTRACT

Peptidomic analysis of an extract of the skin of the stream brown frog Rana sakuraii Matsui and Matsui, 1990 led to the isolation of a C-terminally alpha-amidated peptide (VR-23; VIGSILGALASGLPTLISWIKNR x NH2) with broad-spectrum antimicrobial activity that shows structural similarity to the bee venom peptide, melittin together with two peptides belonging to the temporin family (temporin-1SKa; FLPVILPVIGKLLNGIL x NH2 and temporin-1SKb; FLPVILPVIGKLLSGIL x NH2), and peptides whose primary structures identified them as belonging to the brevinin-2 (2 peptides) and ranatuerin-2 (1 peptide) families. Using a forward primer that was designed from a conserved region of the 5'-untranslated regions of Rana temporaria preprotemporins in a 3'-RACE procedure, a cDNA clone encoding preprotemporin-1SKa was prepared from R. sakuraii skin total RNA. Further preprotemporin cDNAs encoding temporin-1SKc (AVDLAKIANIAN KVLSSL F x NH2) and temporin-1SKd (FLPMLAKLLSGFL x NH2) were obtained by RT-PCR. Unexpectedly, the 3'-RACE procedure using the same primer led to amplification of a cDNA encoding a preprobradykinin whose signal peptide region was identical to that of preprotemporin-1SKa except for the substitution Ser18-->Asn. R. sakuraii bradykinin ([Arg0,Leu1,Thr6,Trp8] BK) was 28-fold less potent than mammalian BK in effecting B2 receptor-mediated relaxation of mouse trachea and the des[Arg0] derivative was only a weak partial agonist. The evolutionary history of the Japanese brown frogs is incompletely understood but a comparison of the primary structures of the R. sakuraii dermal peptides with those of Tago's brown frog Rana tagoi provides evidence for a close phylogenetic relationship between these species.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Bradykinin/genetics , Ranidae/genetics , 5' Untranslated Regions , Amino Acid Sequence , Amphibian Proteins/chemistry , Amphibian Proteins/genetics , Amphibian Proteins/isolation & purification , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/isolation & purification , Bradykinin/chemistry , Bradykinin/isolation & purification , Bradykinin/pharmacology , Cloning, Molecular , DNA, Complementary/genetics , Female , Gene Expression , In Vitro Techniques , Male , Melitten/chemistry , Melitten/genetics , Mice , Molecular Sequence Data , Muscle Relaxation/drug effects , Peptides/chemistry , Peptides/genetics , Peptides/isolation & purification , Proteins/chemistry , Proteins/genetics , Ranidae/metabolism , Sequence Homology, Amino Acid , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL