Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1865(2): 364-378, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29108912

ABSTRACT

Autophagy and apoptosis are two major interconnected host cell responses to viral infection, including influenza A virus (IAV). Thus, delineating these events could facilitate the development of better treatment options and provide an effective anti-viral strategy for controlling IAV infection. We used A549 cells and mouse embryonic fibroblasts (MEF) to study the role of virus-induced autophagy and apoptosis, the cross-talk between both pathways, and their relation to IAV infection [ATCC strain A/Puerto Rico/8/34(H1N1) (hereafter; PR8)]. PR8-infected and mock-infected cells were analyzed by immunoblotting, immunofluorescence confocal microscopy, electron microscopy and flow cytometry (FACS). We found that PR8 infection simultaneously induced autophagy and apoptosis in A549 cells. Autophagy was associated with Bax and Bak activation, intrinsic caspase cleavage and subsequent PARP-1 and BID cleavage. Both Bax knockout (KO) and Bax/Bak double knockout MEFs displayed inhibition of virus-induced cytopathology and cell death and diminished virus-mediated caspase activation, suggesting that virus-induced apoptosis is Bax/Bak-dependent. Biochemical inhibition of autophagy induction with 3-methyladenine blocked both virus replication and apoptosis pathways. These effects were replicated using autophagy-refractory Atg3 KO and Atg5 KO cells. Taken together, our data indicate that PR8 infection simultaneously induces autophagy and Bax/caspase-dependent apoptosis, with autophagy playing a role to support PR8 replication, in part, by modulating virus-induced apoptosis.


Subject(s)
Apoptosis , Autophagy , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/metabolism , Virus Replication/physiology , A549 Cells , Animals , Humans , Influenza, Human/genetics , Influenza, Human/pathology , Mice , Mice, Knockout
2.
BMC Pulm Med ; 19(1): 223, 2019 Nov 27.
Article in English | MEDLINE | ID: mdl-31771541

ABSTRACT

BACKGROUND: The CAnadian REgistry for Pulmonary Fibrosis (CARE-PF) is a multi-center, prospective registry designed to study the natural history of fibrotic interstitial lung disease (ILD) in adults. The aim of this cross-sectional sub-study was to describe the baseline characteristics, risk factors, and comorbidities of patients enrolled in CARE-PF to date. METHODS: Patients completed study questionnaires and clinical measurements at enrollment and each follow-up visit. Environmental exposures were assessed by patient self-report and comorbidities by the Charlson Comorbidity Index (CCI). Baseline characteristics, exposures, and comorbidities were described for the overall study population and for incident cases, and were compared across ILD subtypes. RESULTS: The full cohort included 1285 patients with ILD (961 incident cases (74.8%)). Diagnoses included connective tissue disease-associated ILD (33.3%), idiopathic pulmonary fibrosis (IPF) (24.7%), unclassifiable ILD (22.3%), chronic hypersensitivity pneumonitis (HP) (7.5%), sarcoidosis (3.2%), non-IPF idiopathic interstitial pneumonias (3.0%, including idiopathic nonspecific interstitial pneumonia (NSIP) in 0.9%), and other ILDs (6.0%). Patient-reported exposures were most frequent amongst chronic HP, but common across all ILD subtypes. The CCI was ≤2 in 81% of patients, with a narrow distribution and range of values. CONCLUSIONS: CTD-ILD, IPF, and unclassifiable ILD made up 80% of ILD diagnoses at ILD referral centers in Canada, while idiopathic NSIP was rare when adhering to recommended diagnostic criteria. CCI had a very narrow distribution across our cohort suggesting it may be a poor discriminator in assessing the impact of comorbidities on patients with ILD.


Subject(s)
Alveolitis, Extrinsic Allergic/epidemiology , Environmental Exposure , Idiopathic Pulmonary Fibrosis/epidemiology , Lung Diseases, Interstitial/epidemiology , Registries , Adult , Aged , Canada/epidemiology , Comorbidity , Connective Tissue Diseases/complications , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Risk Factors , Severity of Illness Index , Surveys and Questionnaires
3.
Thorax ; 72(1): 74-82, 2017 01.
Article in English | MEDLINE | ID: mdl-27325752

ABSTRACT

BACKGROUND: Asthma affects 300 million people worldwide. In asthma, the major cause of morbidity and mortality is acute airway narrowing, due to airway smooth muscle (ASM) hypercontraction, associated with airway remodelling. However, little is known about the transcriptional differences between healthy and asthmatic ASM cells. OBJECTIVES: To investigate the transcriptional differences between asthmatic and healthy airway smooth muscle cells (ASMC) in culture and investigate the identified targets using in vitro and ex vivo techniques. METHODS: Human asthmatic and healthy ASMC grown in culture were run on Affymetrix_Hugene_1.0_ST microarrays. Identified candidates were confirmed by PCR, and immunohistochemistry. Functional analysis was conducted using in vitro ASMC proliferation, attachment and contraction assays and ex vivo contraction of mouse airways. RESULTS: We suggest a novel role for latrophilin (LPHN) receptors, finding increased expression on ASMC from asthmatics, compared with non-asthmatics in vivo and in vitro, suggesting a role in mediating airway function. A single nucleotide polymorphism in LPHN1 was associated with asthma and with increased LPHN1 expression in lung tissue. When activated, LPHNs regulated ASMC adhesion and proliferation in vitro, and promoted contraction of mouse airways and ASMC. CONCLUSIONS: Given the need for novel inhibitors of airway remodelling and bronchodilators in asthma, the LPHN family may represent promising novel targets for future dual therapeutic intervention.


Subject(s)
Asthma/genetics , Asthma/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Acetylcholine/pharmacology , Animals , Case-Control Studies , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Humans , Male , Membrane Glycoproteins , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/physiology , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Respiratory System/cytology , Spider Venoms/pharmacology , Transcription, Genetic
4.
Paediatr Respir Rev ; 21: 19-26, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27665512

ABSTRACT

Diabetes is an increasingly common complication of pregnancy. In parallel with this trend, a rise in chronic lung disease in children has been observed in recent decades. While several adverse health outcomes associated with exposure to diabetes in utero have been documented in epidemiological and experimental studies, few have examined the impact of diabetes in pregnancy on offspring lung health and respiratory disease. We provide a comprehensive overview of current literature on this topic, finding suggestive evidence that exposure to diabetes in utero may have adverse effects on lung development. Delayed lung maturation and increased risk of respiratory distress syndrome have been consistently observed among infants born to mothers with diabetes and these findings are also observed in some rodent models of diabetes in pregnancy. Further research is needed to confirm and characterize epidemiologic observations that diabetes in pregnancy may predispose offspring to childhood wheezing illness and asthma. Parallel translational studies in human pregnancy cohorts and experimental models are needed to explore the role of fetal programming and other potential biological mechanisms in this context.


Subject(s)
Diabetes, Gestational/epidemiology , Lung/embryology , Pregnancy in Diabetics/epidemiology , Prenatal Exposure Delayed Effects/epidemiology , Respiratory Tract Diseases/epidemiology , Animals , Asthma/epidemiology , Asthma/metabolism , Bronchopulmonary Dysplasia/epidemiology , Bronchopulmonary Dysplasia/metabolism , Child , Diabetes, Gestational/metabolism , Disease Models, Animal , Female , Hernias, Diaphragmatic, Congenital/epidemiology , Hernias, Diaphragmatic, Congenital/metabolism , Humans , Infant, Newborn , Pregnancy , Pregnancy in Diabetics/metabolism , Prenatal Exposure Delayed Effects/metabolism , Respiratory Distress Syndrome, Newborn/epidemiology , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Sounds , Respiratory Tract Diseases/metabolism
5.
Clin Exp Allergy ; 46(7): 945-56, 2016 07.
Article in English | MEDLINE | ID: mdl-27079765

ABSTRACT

BACKGROUND: Chronic inflammation, typified by increased expression of IL-17A, together with airway and parenchymal remodelling are features of chronic lung diseases. Emerging evidence suggests that phenotypic heterogeneity of repair and inflammatory capacities of fibroblasts may contribute to the differential structural changes observed in different regions of the lung. OBJECTIVE: To investigate phenotypic differences in parenchymal and bronchial fibroblasts, either in terms of inflammation and remodelling or the ability of these fibroblasts to respond to IL-17A. METHODS: Four groups of primary fibroblasts were used: normal human bronchial fibroblast (NHBF), normal human parenchymal fibroblast (NHPF), COPD human bronchial fibroblast (CHBF) and COPD human parenchymal fibroblast (CHPF). Cytokine and extracellular matrix (ECM) expression were measured at baseline and after stimulation with IL-17A. Actinomycin D was used to measure cytokine mRNA stability. RESULTS: At baseline, we observed higher protein production of IL-6 in NHPF than NHBF, but higher levels of IL-8 and GRO-α in NHBF. IL-17A induced a higher expression of GRO-α (CXCL1) and IL-6 in NHPF than in NHBF, and a higher level of IL-8 expression in NHBF. IL-17A treatment decreased the mRNA stability of IL-6 in NHBF when compared with NHPF. CHPF expressed higher protein levels of fibronectin, collagen-I and collagen-III than CHBF, NHBF and NHPF. IL-17A increased fibronectin and collagen-III protein only in NHPF and collagen-III protein production in CHBF and CHPF. CONCLUSIONS AND CLINICAL RELEVANCE: These findings provide insight into the inflammatory and remodelling processes that may be related to the phenotypic heterogeneity of fibroblasts from airway and parenchymal regions and in their response to IL-17A.


Subject(s)
Bronchi/metabolism , Fibroblasts/metabolism , Inflammation/etiology , Inflammation/metabolism , Interleukin-17/metabolism , Parenchymal Tissue/metabolism , Bronchi/cytology , Cytokines/genetics , Cytokines/metabolism , Extracellular Matrix , Fibroblasts/drug effects , Gene Expression , Humans , Interleukin-17/pharmacology , Parenchymal Tissue/cytology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology
6.
Clin Exp Allergy ; 42(1): 85-94, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22092970

ABSTRACT

BACKGROUND: Structural cells are an important reservoir of chemokines that coordinate the influx of various immune cells to the lungs of asthmatics. Airway smooth muscle cells (ASMC) are an important source of these chemokines. CCL15 is a recently described chemo-attractant for neutrophils, eosinophils, monocytes and lymphocytes. OBJECTIVE: To determine the production and the regulation of CCL15 by ASMC and to investigate its production in asthmatic airways. METHODS: Human ASMC were obtained from main bronchial airway segments of patients with mild, moderate and severe asthma. To induce chemokine production, cells were incubated with IL-4, IL-13, TNF-α or IFN-γ in presence or absence of dexamethasone, mithramycin A (SP-1 inhibitor) or the IKK-2 inhibitor, AS602868. CCL15 mRNA expression was evaluated by real-time PCR. Immunoreactive CCL15 was detected by immuno-fluorescence and CCL15 protein concentration in the supernatant was measured using ELISA. RESULTS: CCL15 is constitutively expressed in human ASMC and is strongly up-regulated by TNF-α. This up-regulation is inhibited by dexamethasone, mithramycin A and AS602868. TNF-α-induced CCL15 levels can be synergistically enhanced by the presence of IFN-γ, at both the transcriptional and translation level. This synergism is NF-κB-dependent. Asthmatic biopsies demonstrated higher expression of CCL15 compared with non-asthmatic controls. CONCLUSION AND CLINICAL RELEVANCE: Our results show that ASMC are a potent source of CCL15 in the airways and may directly participate in the recruitment of inflammatory cells to asthmatic airways. Targeting the production of CCL15 by ASMC might reduce the inflammatory response within the airways of asthmatic patients.


Subject(s)
Asthma/physiopathology , Bronchi/cytology , Chemokines, CC/metabolism , Macrophage Inflammatory Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Up-Regulation , Adult , Asthma/immunology , Biopsy , Chemokines, CC/drug effects , Chemokines, CC/genetics , Chemokines, CC/immunology , Female , Humans , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Macrophage Inflammatory Proteins/drug effects , Macrophage Inflammatory Proteins/genetics , Macrophage Inflammatory Proteins/immunology , Male , Middle Aged , Myocytes, Smooth Muscle/immunology , Real-Time Polymerase Chain Reaction , Severity of Illness Index , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
7.
Front Pharmacol ; 13: 991072, 2022.
Article in English | MEDLINE | ID: mdl-36386177

ABSTRACT

Airway cholinergic nerves play a key role in airway physiology and disease. In asthma and other diseases of the respiratory tract, airway cholinergic neurons undergo plasticity and contribute to airway hyperresponsiveness and mucus secretion. We currently lack human in vitro models for airway cholinergic neurons. Here, we aimed to develop a human in vitro model for peripheral cholinergic neurons using human pluripotent stem cell (hPSC) technology. hPSCs were differentiated towards vagal neural crest precursors and subsequently directed towards functional airway cholinergic neurons using the neurotrophin brain-derived neurotrophic factor (BDNF). Cholinergic neurons were characterized by ChAT and VAChT expression, and responded to chemical stimulation with changes in Ca2+ mobilization. To culture these cells, allowing axonal separation from the neuronal cell bodies, a two-compartment PDMS microfluidic chip was subsequently fabricated. The two compartments were connected via microchannels to enable axonal outgrowth. On-chip cell culture did not compromise phenotypical characteristics of the cells compared to standard culture plates. When the hPSC-derived peripheral cholinergic neurons were cultured in the chip, axonal outgrowth was visible, while the somal bodies of the neurons were confined to their compartment. Neurons formed contacts with airway smooth muscle cells cultured in the axonal compartment. The microfluidic chip developed in this study represents a human in vitro platform to model neuro-effector interactions in the airways that may be used for mechanistic studies into neuroplasticity in asthma and other lung diseases.

8.
Eur Respir J ; 38(4): 841-50, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21565918

ABSTRACT

Inhibition of Rho-associated coiled-coil forming kinases (ROCKs) reduces allergic airway responses in mice. The purpose of this study was to determine the roles of the two ROCK isoforms, ROCK1 and ROCK2, in these responses. Wildtype (WT) mice and heterozygous ROCK1 and ROCK2 knockout mice (ROCK1(+/-) and ROCK2(+/-), respectively) were sensitised and challenged with ovalbumin. ROCK expression and activation were assessed by western blotting. Airway responsiveness was measured by forced oscillation. Bronchoalveolar lavage was performed and the lungs were fixed for histological assessment. Compared with WT mice, ROCK1 and ROCK2 expression were 50% lower in lungs of ROCK1(+/-) and ROCK2(+/-) mice, respectively, without changes in the other isoform. In WT lungs, ROCK activation increased after ovalbumin challenge and was sustained for several hours. This activation was reduced in ROCK1(+/-) and ROCK2(+/-) lungs. Airway responsiveness was comparable in WT, ROCK1(+/-), and ROCK2(+/-) mice challenged with PBS. Ovalbumin challenge caused airway hyperresponsiveness in WT, but not ROCK1(+/-) or ROCK2(+/-) mice. Lavage eosinophils and goblet cell hyperplasia were significantly reduced in ovalbumin-challenged ROCK1(+/-) and ROCK2(+/-) versus WT mice. Ovalbumin-induced changes in lavage interleukin-13, interleukin-5 and lymphocytes were also reduced in ROCK1(+/-) mice. In conclusion, both ROCK1 and ROCK2 are important in regulating allergic airway responses.


Subject(s)
Bronchial Hyperreactivity/immunology , Hypersensitivity/immunology , rho-Associated Kinases/immunology , Animals , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Eosinophils/immunology , Eosinophils/pathology , Female , Gene Expression/immunology , Goblet Cells/immunology , Goblet Cells/pathology , Hypersensitivity/genetics , Hypersensitivity/pathology , Interleukin-13/immunology , Interleukin-5/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Respiratory Mechanics/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , rho-Associated Kinases/genetics
9.
J Med Genet ; 46(8): 497-510, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19505876

ABSTRACT

The inactivation of programmed cell death has profound effects not only on the development but also on the overall integrity of multicellular organisms. Beside developmental abnormalities, it may lead to tumorigenesis, autoimmunity, and other serious health problems. Deregulated apoptosis may also be the leading cause of cancer therapy chemoresistance. Caspase family of cysteinyl-proteases plays the key role in the initiation and execution of programmed cell death. This review gives an overview of the role of caspases, their natural modulators like IAPs, FLIPs, and Smac/Diablo in apoptosis and upon inactivation, and also in cancer development. Besides describing the basic mechanisms governing programmed cell death, a large part of this review is dedicated to previous studies that were focused on screening tumours for mutations within caspase genes as well as their regulators. The last part of this review discusses several emerging treatments that involve modulation of caspases and their regulators. Thus, we also highlight caspase cascade modulating experimental anticancer drugs like cFLIP-antagonist CDDO-Me; cIAP1 antagonists OSU-03012 and ME-BS; and XIAP small molecule antagonists 1396-11, 1396-12, 1396-28, triptolide, AEG35156, survivin/Hsp90 antagonist shephedrin, and some of the direct activators of procaspase-3.


Subject(s)
Apoptosis/genetics , Caspases/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Animals , Caspase Inhibitors , Humans , Models, Biological , Neoplasms/enzymology
10.
Eur Respir J ; 34(6): 1436-43, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19460789

ABSTRACT

Acetylcholine is the primary parasympathetic neurotransmitter in the airways and is known to cause bronchoconstriction and mucus secretion. Recent findings suggest that acetylcholine also regulates aspects of remodelling and inflammation through its action on muscarinic receptors. In the present study, we aimed to determine the effects of muscarinic receptor stimulation on cytokine production by human airway smooth muscle cells (primary and immortalised cell lines). The muscarinic receptor agonists carbachol and methacholine both induced modest effects on basal interleukin (IL)-8 and -6 secretion, whereas the secretion of RANTES, eotaxin, vascular endothelial growth factor-A and monocyte chemoattractant protein-1 was not affected. Secretion of IL-8 and -6 was only observed in immortalised airway smooth muscle cells that express muscarinic M3 receptors. In these cells, methacholine also significantly augmented IL-8 secretion in combination with cigarette smoke extract in a synergistic manner, whereas synergistic effects on IL-6 secretion were not significant. Muscarinic M3 receptors were the primary subtype involved in augmenting cigarette smoke extract-induced IL-8 secretion, as only tiotropium bromide and muscarinic M3 receptor subtype selective antagonists abrogated the effects of methacholine. Collectively, these results indicate that muscarinic M3 receptor stimulation augments cigarette smoke extract-induced cytokine production by airway smooth muscle. This interaction could be of importance in patients with chronic obstructive pulmonary disease.


Subject(s)
Interleukin-8/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Muscarinic M3/metabolism , Smoking/adverse effects , Acetylcholine/metabolism , Bronchi/metabolism , Cells, Cultured , Chemokine CCL5/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation , Interleukin-6/metabolism , Methacholine Chloride/pharmacology , Neurotransmitter Agents/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology
11.
Sci Rep ; 8(1): 805, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29339735

ABSTRACT

Asthma is an obstructive respiratory disease characterised by chronic inflammation with airway hyperresponsiveness. In asthmatic airways, there is an increase in airway smooth muscle (ASM) cell bulk, which differs from non-asthmatic ASM in characteristics. This study aimed to assess the usefulness of hTERT immortalisation of human ASM cells as a research tool. Specifically we compared proliferative capacity, inflammatory mediator release and extracellular matrix (ECM) production in hTERT immortalised and parent primary ASM cells from asthmatic and non-asthmatic donors. Our studies revealed no significant differences in proliferation, IL-6 and eotaxin-1 production, or CTGF synthesis between donor-matched parent and hTERT immortalised ASM cell lines. However, deposition of ECM proteins fibronectin and fibulin-1 was significantly lower in immortalised ASM cells compared to corresponding primary cells. Notably, previously reported differences in proliferation and inflammatory mediator release between asthmatic and non-asthmatic ASM cells were retained, but excessive ECM protein deposition in asthmatic ASM cells was lost in hTERT ASM cells. This study shows that hTERT immortalised ASM cells mirror primary ASM cells in proliferation and inflammatory profile characteristics. Moreover, we demonstrate both strengths and weaknesses of this immortalised cell model as a representation of primary ASM cells for future asthma pathophysiological research.


Subject(s)
Myocytes, Smooth Muscle/physiology , Phenotype , Respiratory System/cytology , Telomerase/metabolism , Asthma/pathology , Cell Proliferation , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Immunologic Factors/metabolism , Telomerase/genetics
12.
Curr Drug Targets ; 7(5): 525-40, 2006 May.
Article in English | MEDLINE | ID: mdl-16719764

ABSTRACT

Asthma incidence has climbed markedly in the past two decades despite an increased use of medications that suppress airway inflammation and repress contraction of smooth muscle that encircles the airways. Asthmatics exhibit episodes of airway inflammation that potentiates reversible airway smooth muscle spasm. A hallmark diagnostic symptom of asthma is airway hyperresponsiveness to inhaled non-allergic stimuli, such as methacholine, that directly induce airway smooth muscle contraction. Inhaled gluccocorticoids are used for first-line prevention of airway inflammation, and are frequently combined with inhaled beta2-adrenoceptor agonists that can effectively relax airway smooth muscle and restore airway conductance. Leukotriene receptor antagonists and anti-cholinergics can also be used in many patients to ensure optimal control of symptoms. With increasing disease duration irreversible airway restriction develops from inflammation-driven fibro-proliferative airway remodeling that includes increased deposition of extracellular matrix, the accumulation of airway smooth muscle, and increased numbers of myofibroblasts. Mature airway smooth muscle cells are phenotypically plastic, enabling them to subserve contractile, proliferative, migratory and secretory functional responses that contribute to airway remodeling and persistent hyperresponsiveness. This review assesses current understanding of acute and chronic effects of common anti-asthma medications on the diverse phenotype and functional characteristics of airway smooth muscle cells. Furthermore, we describe the significance of these effects in the treatment of asthma symptoms and pathogenesis.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Bronchi/physiology , Myocytes, Smooth Muscle/physiology , Trachea/physiology , Adrenergic beta-Agonists/pharmacology , Bronchi/drug effects , Bronchial Hyperreactivity/physiopathology , Cell Proliferation/drug effects , Cholinergic Antagonists/pharmacology , Gene Expression Regulation , Glucocorticoids/pharmacology , Humans , Muscle Contraction , Myocytes, Smooth Muscle/drug effects , Phenotype , Trachea/drug effects
13.
J Appl Physiol (1985) ; 98(2): 489-97, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15465883

ABSTRACT

We hypothesized that differences in actin filament length could influence force fluctuation-induced relengthening (FFIR) of contracted airway smooth muscle and tested this hypothesis as follows. One-hundred micromolar ACh-stimulated canine tracheal smooth muscle (TSM) strips set at optimal reference length (Lref) were allowed to shorten against 32% maximal isometric force (Fmax) steady preload, after which force oscillations of +/-16% Fmax were superimposed. Strips relengthened during force oscillations. We measured hysteresivity and calculated FFIR as the difference between muscle length before and after 20-min imposed force oscillations. Strips were relaxed by ACh removal and treated for 1 h with 30 nM latrunculin B (sequesters G-actin and promotes depolymerization) or 500 nM jasplakinolide (stabilizes actin filaments and opposes depolymerization). A second isotonic contraction protocol was then performed; FFIR and hysteresivity were again measured. Latrunculin B increased FFIR by 92.2 +/- 27.6% Lref and hysteresivity by 31.8 +/- 13.5% vs. pretreatment values. In contrast, jasplakinolide had little influence on relengthening by itself; neither FFIR nor hysteresivity was significantly affected. However, when jasplakinolide-treated tissues were then incubated with latrunculin B in the continued presence of jasplakinolide for 1 more h and a third contraction protocol performed, latrunculin B no longer substantially enhanced TSM relengthening. In TSM treated with latrunculin B + jasplakinolide, FFIR increased by only 3.03 +/- 5.2% Lref and hysteresivity by 4.14 +/- 4.9% compared with its first (pre-jasplakinolide or latrunculin B) value. These results suggest that actin filament length, in part, determines the relengthening of contracted airway smooth muscle.


Subject(s)
Acetylcholine/pharmacology , Actin Cytoskeleton/physiology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Depsipeptides/pharmacology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Thiazoles/pharmacology , Trachea/physiology , Animals , Dogs , Dose-Response Relationship, Drug , In Vitro Techniques , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Stress, Mechanical , Thiazolidines , Trachea/drug effects
14.
Cell Death Dis ; 6: e1696, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25789971

ABSTRACT

Transforming growth factor-ß(1) (TGF-ß(1)) is an important regulator of fibrogenesis in heart disease. In many other cellular systems, TGF-ß(1) may also induce autophagy, but a link between its fibrogenic and autophagic effects is unknown. Thus we tested whether or not TGF-ß(1)-induced autophagy has a regulatory function on fibrosis in human atrial myofibroblasts (hATMyofbs). Primary hATMyofbs were treated with TGF-ß(1) to assess for fibrogenic and autophagic responses. Using immunoblotting, immunofluorescence and transmission electron microscopic analyses, we found that TGF-ß(1) promoted collagen type Iα2 and fibronectin synthesis in hATMyofbs and that this was paralleled by an increase in autophagic activation in these cells. Pharmacological inhibition of autophagy by bafilomycin-A1 and 3-methyladenine decreased the fibrotic response in hATMyofb cells. ATG7 knockdown in hATMyofbs and ATG5 knockout (mouse embryonic fibroblast) fibroblasts decreased the fibrotic effect of TGF-ß(1) in experimental versus control cells. Furthermore, using a coronary artery ligation model of myocardial infarction in rats, we observed increases in the levels of protein markers of fibrosis, autophagy and Smad2 phosphorylation in whole scar tissue lysates. Immunohistochemistry for LC3ß indicated the localization of punctate LC3ß with vimentin (a mesenchymal-derived cell marker), ED-A fibronectin and phosphorylated Smad2. These results support the hypothesis that TGF-ß(1)-induced autophagy is required for the fibrogenic response in hATMyofbs.


Subject(s)
Autophagy/genetics , Fibrosis/genetics , Heart Atria/metabolism , Myofibroblasts/metabolism , Transforming Growth Factor beta1/biosynthesis , Adenine/administration & dosage , Adenine/analogs & derivatives , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Proliferation/drug effects , Collagen Type I/metabolism , Fibronectins/biosynthesis , Fibrosis/pathology , Heart Atria/pathology , Humans , Macrolides/administration & dosage , Mice , Microtubule-Associated Proteins/genetics , Myofibroblasts/pathology , Primary Cell Culture , Rats , Signal Transduction/drug effects , Smad2 Protein/biosynthesis , Smad2 Protein/genetics , Transforming Growth Factor beta1/genetics
15.
Cell Death Dis ; 6: e1944, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26512955

ABSTRACT

Exposure to metabolic disease during fetal development alters cellular differentiation and perturbs metabolic homeostasis, but the underlying molecular regulators of this phenomenon in muscle cells are not completely understood. To address this, we undertook a computational approach to identify cooperating partners of the myocyte enhancer factor-2 (MEF2) family of transcription factors, known regulators of muscle differentiation and metabolic function. We demonstrate that MEF2 and the serum response factor (SRF) collaboratively regulate the expression of numerous muscle-specific genes, including microRNA-133a (miR-133a). Using tandem mass spectrometry techniques, we identify a conserved phosphorylation motif within the MEF2 and SRF Mcm1 Agamous Deficiens SRF (MADS)-box that regulates miR-133a expression and mitochondrial function in response to a lipotoxic signal. Furthermore, reconstitution of MEF2 function by expression of a neutralizing mutation in this identified phosphorylation motif restores miR-133a expression and mitochondrial membrane potential during lipotoxicity. Mechanistically, we demonstrate that miR-133a regulates mitochondrial function through translational inhibition of a mitophagy and cell death modulating protein, called Nix. Finally, we show that rodents exposed to gestational diabetes during fetal development display muscle diacylglycerol accumulation, concurrent with insulin resistance, reduced miR-133a, and elevated Nix expression, as young adult rats. Given the diverse roles of miR-133a and Nix in regulating mitochondrial function, and proliferation in certain cancers, dysregulation of this genetic pathway may have broad implications involving insulin resistance, cardiovascular disease, and cancer biology.


Subject(s)
Cell Differentiation/genetics , MEF2 Transcription Factors/chemistry , Mitochondria/physiology , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Serum Response Factor/chemistry , Amino Acid Motifs , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Diabetes, Gestational , Female , Gene Expression Regulation , Humans , MEF2 Transcription Factors/metabolism , MEF2 Transcription Factors/physiology , Membrane Potential, Mitochondrial/genetics , MicroRNAs/metabolism , Mitochondria/genetics , Muscle Fibers, Skeletal/cytology , Mutagenesis, Site-Directed , Myocytes, Cardiac/cytology , Myocytes, Smooth Muscle/cytology , Phosphorylation , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Serum Response Factor/metabolism , Serum Response Factor/physiology , Tandem Mass Spectrometry
16.
J Appl Physiol (1985) ; 90(1): 358-68, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11133929

ABSTRACT

Morphological, functional, molecular and cell biology studies have revealed a striking multifunctional nature of individual smooth muscle cells (SMC). SMCs manifest phenotypic plasticity in response to changes in environment and functional requirements, acquiring a range of structural and functional properties bounded by two extremes, called "synthetic" and "contractile." Each phenotypic state is characterized by expression of a unique set of structural, contractile, and receptor proteins and isoforms that correlate with differing patterns of gene expression. Recent studies have identified signaling pathways and transcription factors (e.g., RhoA GTPase/ROCK, also known as Rho kinase, and serum response factor) that regulate the transcriptional activities of genes encoding proteins associated with the contractile apparatus. Mechanical plasticity of contractile-state smooth muscle further extends SMC functional diversity. This may also be regulated, in part, by the RhoA GTPase/ROCK pathway, via reorganization of cytoskeletal and contractile proteins. Future studies that define transcriptional and posttranscriptional mechanisms of SMC plasticity are necessary to fully understand the role of SMC in the pathogenesis and morbidity of human diseases of the airways, vasculature, and gastrointestinal tract.


Subject(s)
Muscle, Skeletal/physiology , Muscle, Smooth/physiology , Papillary Muscles/physiology , Animals , Genetic Markers , Humans , Muscle Contraction/physiology , Muscle, Skeletal/cytology , Phenotype , Transcription, Genetic/physiology
17.
J Appl Physiol (1985) ; 75(5): 2013-21, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8307854

ABSTRACT

To investigate heterogeneity of airway smooth muscle response, we studied strips of large and small branches from third- to sixth-generation bronchi obtained from ragweed antigen-sensitized and control dogs. The response to electrical field stimulation and carbamylcholine chloride was greater in strips from larger branches of the same generation when expressed as "tissue stress" (force per unit cross-sectional area of the whole tissue), whereas no difference emerged with use of the more appropriate "smooth muscle stress" (force per unit cross-sectional area of the muscle tissue). The response to histamine was significantly higher in small branches than in large ones, and histamine sensitivity [mean effective concentration (EC50)] was 7.79 x 10(-6) [geometric standard error of the mean (GSEM) 1.20] and 1.49 x 10(-5) M (GSEM 1.14), respectively (P < 0.01). Strips from control and sensitized animals at each site and strips from different generations did not show any significant difference. When we clustered our preparations according to dimensions, the response to histamine was significantly higher in small bronchi than in large ones and histamine EC50 was 8.95 x 10(-6) (GSEM 1.17) and 1.57 x 10(-5) M (GSEM 1.18), respectively (P < 0.05). We conclude that evaluation of muscle response in different tissues requires appropriate normalization. Furthermore, classification into generations is inadequate to study bronchial responsiveness, inasmuch as major differences originate from airway size.


Subject(s)
Bronchi/physiology , Muscle, Smooth/physiology , Animals , Animals, Newborn/physiology , Bronchi/anatomy & histology , Bronchi/cytology , Carbachol/pharmacology , Dogs , Electric Stimulation , Histamine/pharmacology , Histocytochemistry , In Vitro Techniques , Muscle Contraction/physiology , Muscle, Smooth/anatomy & histology , Paraffin Embedding , Pollen/immunology
18.
J Appl Physiol (1985) ; 72(1): 39-45, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1537740

ABSTRACT

Although we have reported that tracheal smooth muscle from sensitized dogs shows altered mechanical properties, we did not know, because of technical difficulties with the preparation, whether similar changes occur in the properties of sensitized central bronchial smooth muscle (BSM), the site at which the acute asthmatic response is believed to develop. We have now succeeded in developing a cartilage-free BSM preparation that retains optimal mechanical properties. Such strips were obtained from mongrel dogs that had been sensitized to ragweed pollen. Controls were littermates injected with adjuvant alone. Length-tension relationships were obtained for both control and sensitized BSM strips (CBSM and SBSM, respectively). The maximal active stresses were the same (P greater than 0.05) when normalized to muscle fraction in total tissue cross-sectional area [6.2 +/- 0.6 x 10(4) and 5.9 +/- 0.6 x 10(4) (SE) for SBSM and CBSM, respectively]. This suggests that optimal tension is an insensitive indicator of bronchial hyperresponsiveness and that isotonic studies might be more revealing. The maximal shortening velocity (Vo) for SBSM at 2 s [0.35 +/- 0.017 (SE) lo/s, where lo signifies optimal muscle length], in the course of a 10-s contraction, was significantly greater (P less than 0.05) than Vo measured for CBSM (0.27 +/- 0.015 lo/s). However, Vo did not differ at the 8-s point of contraction. The sensitized group demonstrated a statistically significantly greater maximal shortening capacity (0.67 +/- 0.04 lo) than the control group (0.51 +/- 0.04 lo). At 2 s of contraction, 80% of maximal SBSM shortening had been completed and was significantly greater than for CBSM.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Asthma/physiopathology , Bronchial Hyperreactivity/physiopathology , Muscle, Smooth/physiopathology , Animals , Asthma/pathology , Biomechanical Phenomena , Bronchial Hyperreactivity/pathology , Disease Models, Animal , Dogs , Female , In Vitro Techniques , Isometric Contraction/physiology , Isotonic Contraction/physiology , Male , Muscle, Smooth/pathology
19.
J Appl Physiol (1985) ; 89(5): 1985-90, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053353

ABSTRACT

SM22 is a 201-amino acid actin-binding protein expressed at high levels in smooth muscle cells. It has structural homology to calponin, but how SM22 binds to actin remains unknown. We performed site-directed mutagenesis to generate a series of NH(2)-terminal histidine (His)-tagged mutants of human SM22 in Escherichia coli and used these to analyze the functional importance of potential actin binding domains. Purified full-length recombinant SM22 bound to actin in vitro, as demonstrated by cosedimentation assay. Binding did not vary with calcium concentration. The COOH-terminal domain of SM22 is required for actin affinity, because COOH terminally truncated mutants [SM22-(1-186) and SM22-(1-166)] exhibited markedly reduced cosedimentation with actin, and no actin binding of SM22-(1-151) could be detected. Internal deletion of a putative actin binding site (154-KKAQEHKR-161) partially prevented actin binding, as did point mutation to neutralize either or both pairs of positively charged residues at the ends of this region (KK154LL and/or KR160LL). Internal deletion of amino acids 170-180 or 170-186 also partially or almost completely inhibited actin cosedimentation, respectively. Of the three consensus protein kinase C or casein kinase II phosphorylation sites in SM22, only Ser-181 was readily phosphorylated by protein kinase C in vitro, and such phosphorylation greatly decreased actin binding. Substitution of Ser-181 to aspartic acid (to mimic serine phosphorylation) also reduced actin binding. Immunostains of transiently transfected airway myocytes revealed that full-length NH(2)-terminal FLAG-tagged SM22 colocalizes with actin filaments, whereas FLAG-SM22-(1-151) does not. These data confirm that SM22 binds to actin in vitro and in vivo and, for the first time, demonstrate that multiple regions within the COOH-terminal domain are required for full actin affinity.


Subject(s)
Actins/metabolism , Microfilament Proteins , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Smooth/chemistry , Actins/analysis , Animals , Asthma/metabolism , Calcium/pharmacology , Humans , Muscle Proteins/chemistry , Muscle, Smooth/enzymology , Mutagenesis/physiology , Phosphorylation , Protein Binding/drug effects , Protein Binding/physiology , Protein Kinase C/metabolism , Protein Structure, Tertiary , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Trachea/chemistry , Trachea/enzymology
20.
Comp Biochem Physiol B Biochem Mol Biol ; 119(3): 415-24, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9734326

ABSTRACT

The major part of research dealing with the biophysical and biochemical properties of airway smooth muscle is based on the assumption that the cells constituting the tissue are homogenous. For striated muscle this has been shown untenable. In recent years almost every property of vascular smooth muscle has been also demonstrated to be heterogeneous. This realization has been late in arriving on the airway smooth muscle research scene. Our own studies have shown that mechanical properties are, in quantitative terms, heterogeneously distributed down the airways and that contractility, for example, in extrapulmonary and intrapulmonary airways differs markedly. Another indication of heterogeneity is derived from studies of the biochemical properties of airway smooth muscle cells (ASMCs) in culture. Dramatic changes in phenotype expression were found with days in culture. Just after isolation from the tissue, the cells were of contractile type and contained mature isoforms of contractile, regulatory and cytoskeletal proteins. After the fourth day in culture the cellular phenotype changed such that contractile filaments diminished rapidly with smooth muscle isoforms being replaced by non-muscle isoforms. The cell assumed secretory or synthetic properties and commenced proliferating rapidly. It is possible that similar changes in phenotype could occur in vivo in cells undergoing hypertrophy or hyperplasia. Thus, a thickened medial layer of the type seen in the walls of airways from asthmatic airways is not necessarily one endowed with increased contractility and, in fact, the latter may be subnormal. Finally, using the so-called motility assay, we studied the velocity of translation of actin filaments by myosin molecules obtained from antigen-sensitized and control airway smooth muscle. We found no change in maximum velocity of actin translation. This was under conditions where the myosin light chain (MLC) was fully phosphorylated. However, in these tissues we found heterogeneity in myosin light chain kinase (MLCK) content which, we inferred, accounted for the difference in shortening velocity between control and sensitized muscle strips in vitro.


Subject(s)
Cytoskeletal Proteins/genetics , Gene Expression Regulation , Muscle, Smooth/physiology , Respiratory Physiological Phenomena , Respiratory Tract Diseases/physiopathology , Animals , Biomarkers , Cells, Cultured , Cytoskeletal Proteins/biosynthesis , Dogs , Humans , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , Reference Values , Respiratory System/metabolism , Respiratory System/physiopathology , Respiratory Tract Diseases/genetics , Trachea
SELECTION OF CITATIONS
SEARCH DETAIL