Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Adv ; 8(13): eabm9718, 2022 04.
Article in English | MEDLINE | ID: mdl-35363522

ABSTRACT

Cystic fibrosis (CF) is characterized by abnormal transepithelial ion transport. However, a description of CF lung disease pathophysiology unifying superficial epithelial and submucosal gland (SMG) dysfunctions has remained elusive. We hypothesized that biophysical abnormalities associated with CF mucus hyperconcentration provide a unifying mechanism. Studies of the anion secretion-inhibited pig airway model of CF revealed elevated SMG mucus concentrations, osmotic pressures, and SMG mucus accumulation. Human airway studies revealed hyperconcentrated CF SMG mucus with raised osmotic pressures and cohesive forces predicted to limit SMG mucus secretion/release. Using proline-rich protein 4 (PRR4) as a biomarker of SMG secretion, CF sputum proteomics analyses revealed markedly lower PRR4 levels compared to healthy and bronchiectasis controls, consistent with a failure of CF SMGs to secrete mucus onto airway surfaces. Raised mucus osmotic/cohesive forces, reflecting mucus hyperconcentration, provide a unifying mechanism that describes disease-initiating mucus accumulation on airway surfaces and in SMGs of the CF lung.


Subject(s)
Cystic Fibrosis , Animals , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Mucus/metabolism , Respiratory System/metabolism , Sputum/metabolism , Swine
2.
Pediatr Pulmonol ; 55(1): 130-135, 2020 01.
Article in English | MEDLINE | ID: mdl-31549486

ABSTRACT

BACKGROUND: The diagnosis of primary ciliary dyskinesia (PCD) is difficult and requires a combination of clinical features, nasal nitric oxide testing, cilia ultrastructural analysis by electron microscopy (EM), and genetics. A recently described cytoplasmic ultrastructural change termed "ciliary inclusions" was reported to be diagnostic of PCD; however, no supporting evidence of PCD was provided. In this study, we sought to confirm, or refute, the diagnosis of PCD in subjects with "ciliary inclusions" on EM. METHODS: Six subjects from five families with previous lab reports of "ciliary inclusions" on EMs of ciliated cells were identified and evaluated at a Genetic Disorders of Mucociliary Clearance Consortium site. We performed a detailed clinical history, nasal nitric oxide measurement, genetic testing including whole-exome sequencing (WES), and when possible, repeat ciliary EM study. RESULTS: Only one of six subjects had multiple and persistent clinical features congruent with PCD. No subject had situs inversus. Only one of six subjects had a very low nasal nitric oxide level. No "ciliary inclusions" were found in three subjects who had a repeat ciliary EM, and ciliary axonemal ultrastructures were normal. Genetic testing, including WES, was negative for PCD-causing genes, and for pathogenic variants in gene pathways that might cause "ciliary inclusions," such as ciliary biogenesis. CONCLUSION: "Ciliary Inclusions", in isolation, are not sufficient to diagnosis PCD. If seen, additional studies should be done to pursue an accurate diagnosis.


Subject(s)
Cilia/ultrastructure , Ciliary Motility Disorders/diagnosis , Child, Preschool , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/metabolism , Female , Genetic Testing , Humans , Infant , Male , Nasal Mucosa/metabolism , Nitric Oxide/metabolism , Exome Sequencing
3.
Am J Respir Cell Mol Biol ; 61(3): 312-321, 2019 09.
Article in English | MEDLINE | ID: mdl-30896965

ABSTRACT

Primary ciliary dyskinesia (PCD) is a genetically and phenotypically heterogeneous disease caused by mutations in over 40 different genes. Individuals with PCD caused by mutations in RSPH1 (radial spoke head 1 homolog) have been reported to have a milder phenotype than other individuals with PCD, as evidenced by a lower incidence of neonatal respiratory distress, higher nasal nitric oxide concentrations, and better lung function. To better understand genotype-phenotype relationships in PCD, we have characterized a mutant mouse model with a deletion of Rsph1. Approximately 50% of cilia from Rsph1-/- cells appeared normal by transmission EM, whereas the remaining cilia revealed a range of defects, primarily transpositions or a missing central pair. Ciliary beat frequency in Rsph1-/- cells was significantly lower than in control cells (20.2 ± 0.8 vs. 25.0 ± 0.9 Hz), and the cilia exhibited an aberrant rotational waveform. Young Rsph1-/- animals demonstrated a low rate of mucociliary clearance in the nasopharynx that was reduced to zero by about 1 month of age. Rsph1-/- animals accumulated mucus in the nasal cavity but had a lower bacterial burden than animals with a deletion of dynein axonemal intermediate chain 1 (Dnaic1-/-). Thus, Rsph1-/- mice display a PCD phenotype similar to but less severe than that observed in Dnaic1-/- mice, similar to what has been observed in humans. The results suggest that some individuals with PCD may not have a complete loss of mucociliary clearance and further suggest that early diagnosis and intervention may be important to maintain this low amount of clearance.


Subject(s)
DNA-Binding Proteins/genetics , Kartagener Syndrome/genetics , Mucociliary Clearance/genetics , Phenotype , Animals , Axoneme/genetics , Cilia/genetics , Humans , Mice , Mutation/genetics , Sequence Deletion/genetics
4.
Am J Respir Cell Mol Biol ; 59(3): 383-396, 2018 09.
Article in English | MEDLINE | ID: mdl-29579396

ABSTRACT

Understanding how expression of airway secretory mucins MUC5B and MUC5AC is regulated in health and disease is important to elucidating the pathogenesis of mucoobstructive respiratory diseases. The transcription factor SPDEF (sterile α-motif pointed domain epithelial specific transcription factor) is a key regulator of MUC5AC, but its role in regulating MUC5B in health and in mucoobstructive lung diseases is unknown. Characterization of Spdef-deficient mice upper and lower airways demonstrated region-specific, Spdef-dependent regulation of basal Muc5b expression. Neonatal Spdef-deficient mice exhibited reductions in BAL Muc5ac and Muc5b. Adult Spdef-deficient mice partially phenocopied Muc5b-deficient mice as they exhibited reduced Muc5b in nasopharyngeal and airway epithelia but not in olfactory Bowman glands, 75% incidence of nasopharyngeal hair/mucus plugs, and mild bacterial otitis media, without defective mucociliary clearance in the nasopharynx. In contrast, tracheal mucociliary clearance was reduced in Spdef-deficient mice in the absence of lung disease. To evaluate the role of Spdef in the development and persistence of Muc5b-predominant mucoobstructive lung disease, Spdef-deficient mice were crossed with Scnn1b-transgenic (Scnn1b-Tg) mice, which exhibit airway surface dehydration-induced airway mucus obstruction and inflammation. Spdef-deficient Scnn1b-Tg mice exhibited reduced Muc5ac, but not Muc5b, expression and BAL content. Airway mucus obstruction was not decreased in Spdef-deficient Scnn1b-Tg mice, consistent with Muc5b-dominant Scnn1b disease, but increased airway neutrophilia was observed compared with Spdef-sufficient Scnn1b-Tg mice. Collectively, these results indicate that Spdef regulates baseline Muc5b expression in respiratory epithelia but does not contribute to Muc5b regulation in a mouse model of Muc5b-predominant mucus obstruction caused by airway dehydration.


Subject(s)
Lung Diseases/metabolism , Mucin-5B/metabolism , Mucociliary Clearance/physiology , Proto-Oncogene Proteins c-ets/genetics , Animals , Epithelial Sodium Channels/genetics , Lung Diseases/genetics , Mice, Transgenic , Mucin-5B/genetics
5.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L318-L331, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29074490

ABSTRACT

The epithelial Na+ channel (ENaC) regulates airway surface hydration. In mouse airways, ENaC is composed of three subunits, α, ß, and γ, which are differentially expressed (α > ß > γ). Airway-targeted overexpression of the ß subunit results in Na+ hyperabsorption, causing airway surface dehydration, hyperconcentrated mucus with delayed clearance, lung inflammation, and perinatal mortality. Notably, mice overexpressing the α- or γ-subunit do not exhibit airway Na+ hyperabsorption or lung pathology. To test whether overexpression of multiple ENaC subunits produced Na+ transport and disease severity exceeding that of ßENaC-Tg mice, we generated double (αß, αγ, ßγ) and triple (αßγ) transgenic mice and characterized their lung phenotypes. Double αγENaC-Tg mice were indistinguishable from WT littermates. In contrast, double ßγENaC-Tg mice exhibited airway Na+ absorption greater than that of ßENaC-Tg mice, which was paralleled by worse survival, decreased mucociliary clearance, and more severe lung pathology. Double αßENaC-Tg mice exhibited Na+ transport rates comparable to those of ßENaC-Tg littermates. However, αßENaC-Tg mice had poorer survival and developed severe parenchymal consolidation. In situ hybridization (RNAscope) analysis revealed both alveolar and airway αENaC-Tg overexpression. Triple αßγENaC-Tg mice were born in Mendelian proportions but died within the first day of life, and the small sample size prevented analyses of cause(s) of death. Cumulatively, these results indicate that overexpression of ßENaC is rate limiting for generation of pathological airway surface dehydration. Notably, airway co-overexpression of ß- and γENaC had additive effects on Na+ transport and disease severity, suggesting dose dependency of these two variables.


Subject(s)
Epithelial Sodium Channels/metabolism , Lung Diseases/pathology , Pneumonia/pathology , Respiratory Mucosa/pathology , Animals , Epithelial Sodium Channels/genetics , Lung Diseases/etiology , Lung Diseases/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Phenotype , Pneumonia/etiology , Pneumonia/metabolism , Respiratory Mucosa/metabolism , Signal Transduction
6.
Am J Pathol ; 188(1): 95-110, 2018 01.
Article in English | MEDLINE | ID: mdl-29107074

ABSTRACT

Human subjects with pseudohypoaldosteronism-1 because of loss-of-function mutations in epithelial sodium channel (ENaC) subunits exhibit meibomian gland (MG) dysfunction. A conditional ßENaC MG knockout (KO) mouse model was generated to elucidate the pathogenesis of absent ENaC function in the MG and associated ocular surface disease. ßENaC MG KO mice exhibited a striking age-dependent, female-predominant MG dysfunction phenotype, with white toothpaste-like secretions observed obstructing MG orifices at 7 weeks of age. There were compensatory increases in tear production but higher tear sodium and indexes of mucin concentration in ßENaC MG KO mice. Histologically, MG acinar atrophy was observed with ductal enlargement and ductal epithelial hyperstratification. Inflammatory cell infiltration was observed in both MG and conjunctiva of ßENaC MG KO mice. In older ßENaC MG KO mice (5 to 11 months), significant ocular surface pathologies were noted, including corneal opacification, ulceration, neovascularization, and ectasia. Inflammation in MG and conjunctiva was confirmed by increased cytokine gene and protein expression and positive Ly-6B.2 immunostaining. Cell proliferation assays revealed lower proliferation rates of MG cells derived from ßENaC MG KO than control mice, suggesting that ßENaC plays a role in cell renewal of mouse MG. Loss of ßENaC function resulted in MG disease and severe ocular surface damage that phenocopied aspects of human pseudohypoaldosteronism-1 MG disease and was sex dependent.


Subject(s)
Epithelial Sodium Channels/genetics , Meibomian Glands/metabolism , Pseudohypoaldosteronism/genetics , Tears/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Epithelial Sodium Channels/metabolism , Female , Male , Mice , Mice, Knockout , Phenotype , Pseudohypoaldosteronism/metabolism , Sex Factors
7.
Invest Ophthalmol Vis Sci ; 57(4): 2328-43, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27127933

ABSTRACT

PURPOSE: We establish novel primary rat meibomian gland (MG) cell culture systems and explore the ion transport activities of the rat MG. METHODS: Freshly excised rat MG tissues were characterized as follows: (1) mRNA expression of selected epithelial ion channels/transporters were measured by RT-PCR, (2) localization of epithelial sodium channel (ENaC) mRNAs was performed by in situ hybridization, and (3) protein expression and localization of ßENaC, the Na+/K+/Cl- cotransporter (NKCC), and the Na+/K+ ATPase were evaluated by immunofluorescence. Primary isolated rat MG cells were cocultured with 3T3 feeder cells and a Rho-associated kinase (ROCK) inhibitor (Y-27632) for expansion. Passaged rat MG cells were cultured as planar sheets under air-liquid interface (ALI) conditions for gene expression and electrophysiologic studies. Passaged rat MG cells also were cultured in matrigel matrices to form spheroids, which were examined ultrastructurally by transmission electron microscopy (TEM) and functionally using swelling assays. RESULTS: Expression of multiple ion channel/transporter genes was detected in rat MG tissues. ß-ENaC mRNA and protein were localized more to MG peripheral acinar cells than central acinar cells or ductular epithelial cells. Electrophysiologic studies of rat MG cell planar cultures demonstrated functional sodium, chloride, and potassium channels, and cotransporters activities. Transmission electron microscopic analyses of rat MG spheroids revealed highly differentiated MG cells with abundant lysosomal lamellar bodies. Rat MG spheroids culture-based measurements demonstrated active volume regulation by ion channels. CONCLUSIONS: This study demonstrates the presence and function of ion channels and volume transport by rat MG. Two novel primary MG cell culture models that may be useful for MG research were established.


Subject(s)
Meibomian Glands/metabolism , 3T3 Cells/physiology , Amides/pharmacology , Animals , Cells, Cultured , Coculture Techniques , Fluorescent Antibody Technique , In Situ Hybridization , Ion Channels/physiology , Ion Transport/physiology , Male , Meibomian Glands/cytology , Meibomian Glands/physiology , Mice , Microscopy, Electron, Transmission , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Sodium-Potassium-Chloride Symporters/physiology , Sodium-Potassium-Exchanging ATPase/physiology , rho-Associated Kinases/antagonists & inhibitors
8.
Am J Respir Cell Mol Biol ; 54(2): 210-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26121027

ABSTRACT

Resident immune cells (e.g., macrophages [MΦs]) and airway mucus clearance both contribute to a healthy lung environment. To investigate interactions between pulmonary MΦ function and defective mucus clearance, a genetic model of lysozyme M (LysM) promoter-mediated MΦ depletion was generated, characterized, and crossed with the sodium channel ß subunit transgenic (Scnn1b-Tg) mouse model of defective mucus clearance. Diphtheria toxin A-mediated depletion of LysM(+) pulmonary MΦs in wild-type mice with normal mucus clearance resulted in lethal pneumonia in 24% of neonates. The pneumonias were dominated by Pasteurella pneumotropica and accompanied by emaciation, neutrophilic inflammation, and elevated Th1 cytokines. The incidence of emaciation and pneumonia reached 51% when LysM(+) MΦ depletion was superimposed on the airway mucus clearance defect of Scnn1b-Tg mice. In LysM(+) MΦ-depleted Scnn1b-Tg mice, pneumonias were associated with a broader spectrum of bacterial species and a significant reduction in airway mucus plugging. Bacterial burden (CFUs) was comparable between Scnn1b-Tg and nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice. However, the nonpneumonic LysM(+) MΦ-depleted Scnn1b-Tg mice exhibited increased airway inflammation, the presence of neutrophilic infiltration, and increased levels of inflammatory cytokines in bronchoalveolar lavage fluid compared with Scnn1b-Tg mice. Collectively, these data identify key MΦ-mucus clearance interactions with respect to both infectious and inflammatory components of muco-obstructive lung disease.


Subject(s)
Lung/immunology , Macrophages/immunology , Mucociliary Clearance , Pasteurella Infections/immunology , Pasteurella pneumotropica/immunology , Pneumonia, Bacterial/immunology , Animals , Animals, Newborn , Cytokines/immunology , Cytokines/metabolism , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Disease Models, Animal , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Genetic Predisposition to Disease , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lung/metabolism , Lung/microbiology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/genetics , Pasteurella Infections/genetics , Pasteurella Infections/metabolism , Pasteurella Infections/microbiology , Pasteurella pneumotropica/pathogenicity , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phenotype , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Promoter Regions, Genetic
9.
Ultrastruct Pathol ; 38(4): 248-55, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23957500

ABSTRACT

Abstract Diagnosis of primary ciliary dyskinesia (PCD) by identification of dynein arm loss in transmission electron microscopy (TEM) images can be confounded by high background noise due to random electron-dense material within the ciliary matrix, leading to diagnostic uncertainty even for experienced morphologists. The authors developed a novel image analysis tool to average the axonemal peripheral microtubular doublets, thereby increasing microtubular signal and reducing random background noise. In a randomized, double-blinded study that compared two experienced morphologists and three different diagnostic approaches, they found that use of this tool led to improvement in diagnostic TEM test performance.


Subject(s)
Axonemal Dyneins/ultrastructure , Image Interpretation, Computer-Assisted/methods , Kartagener Syndrome/diagnosis , Microscopy, Electron, Transmission/methods , Double-Blind Method , Humans , Reproducibility of Results
10.
Am J Hum Genet ; 93(2): 336-45, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23891469

ABSTRACT

Defects of motile cilia cause primary ciliary dyskinesia (PCD), characterized by recurrent respiratory infections and male infertility. Using whole-exome resequencing and high-throughput mutation analysis, we identified recessive biallelic mutations in ZMYND10 in 14 families and mutations in the recently identified LRRC6 in 13 families. We show that ZMYND10 and LRRC6 interact and that certain ZMYND10 and LRRC6 mutations abrogate the interaction between the LRRC6 CS domain and the ZMYND10 C-terminal domain. Additionally, ZMYND10 and LRRC6 colocalize with the centriole markers SAS6 and PCM1. Mutations in ZMYND10 result in the absence of the axonemal protein components DNAH5 and DNALI1 from respiratory cilia. Animal models support the association between ZMYND10 and human PCD, given that zmynd10 knockdown in zebrafish caused ciliary paralysis leading to cystic kidneys and otolith defects and that knockdown in Xenopus interfered with ciliogenesis. Our findings suggest that a cytoplasmic protein complex containing ZMYND10 and LRRC6 is necessary for motile ciliary function.


Subject(s)
Cilia/genetics , Kartagener Syndrome/genetics , Proteins/genetics , Respiratory System/metabolism , Tumor Suppressor Proteins/genetics , Animals , Autoantigens/genetics , Autoantigens/metabolism , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cilia/metabolism , Cilia/pathology , Cytoskeletal Proteins , Exome , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Kartagener Syndrome/metabolism , Kartagener Syndrome/pathology , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Pedigree , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Rats , Respiratory System/pathology , Tumor Suppressor Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zebrafish/genetics , Zebrafish/metabolism
11.
Eukaryot Cell ; 11(8): 966-77, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22562470

ABSTRACT

Ras is a highly conserved GTPase protein that is essential for proper polarized morphogenesis of filamentous fungi. Localization of Ras proteins to the plasma membrane and endomembranes through posttranslational addition of farnesyl and palmitoyl residues is an important mechanism through which cells provide specificity to Ras signal output. Although the Aspergillus fumigatus RasA protein is known to be a major regulator of growth and development, the membrane distribution of RasA during polarized morphogenesis and the role of properly localized Ras signaling in virulence of a pathogenic mold remain unknown. Here we demonstrate that Aspergillus fumigatus RasA localizes primarily to the plasma membrane of actively growing hyphae. We show that treatment with the palmitoylation inhibitor 2-bromopalmitate disrupts normal RasA plasma membrane association and decreases hyphal growth. Targeted mutations of the highly conserved RasA palmitoylation motif also mislocalized RasA from the plasma membrane and led to severe hyphal abnormalities, cell wall structural changes, and reduced virulence in murine invasive aspergillosis. Finally, we provide evidence that proper RasA localization is independent of the Ras palmitoyltransferase homolog, encoded by erfB, but requires the palmitoyltransferase complex subunit, encoded by erfD. Our results demonstrate that plasma membrane-associated RasA is critical for polarized morphogenesis, cell wall stability, and virulence in A. fumigatus.


Subject(s)
Aspergillus fumigatus/metabolism , Fungal Proteins/metabolism , Morphogenesis , ras Proteins/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Aspergillus fumigatus/genetics , Aspergillus fumigatus/growth & development , Aspergillus fumigatus/pathogenicity , Cell Membrane/metabolism , Fungal Proteins/genetics , Hyphae/genetics , Hyphae/growth & development , Hyphae/metabolism , Lipoylation , Molecular Sequence Data , Morphogenesis/genetics , Mutation , Palmitates/pharmacology , Protein Processing, Post-Translational , Protein Transport , Serine C-Palmitoyltransferase/antagonists & inhibitors , Serine C-Palmitoyltransferase/genetics , Serine C-Palmitoyltransferase/metabolism , Virulence/genetics , ras Proteins/genetics
12.
Thorax ; 67(5): 433-41, 2012 May.
Article in English | MEDLINE | ID: mdl-22184204

ABSTRACT

RATIONALE: Primary ciliary dyskinesia (PCD) is an autosomal recessive, genetically heterogeneous disorder characterised by oto-sino-pulmonary disease and situs abnormalities (Kartagener syndrome) due to abnormal structure and/or function of cilia. Most patients currently recognised to have PCD have ultrastructural defects of cilia; however, some patients have clinical manifestations of PCD and low levels of nasal nitric oxide, but normal ultrastructure, including a few patients with biallelic mutations in dynein axonemal heavy chain 11 (DNAH11). OBJECTIVES: To test further for mutant DNAH11 as a cause of PCD, DNAH11 was sequenced in patients with a PCD clinical phenotype, but no known genetic aetiology. METHODS: 82 exons and intron/exon junctions in DNAH11 were sequenced in 163 unrelated patients with a clinical phenotype of PCD, including those with normal ciliary ultrastructure (n=58), defects in outer and/or inner dynein arms (n=76), radial spoke/central pair defects (n=6), and 23 without definitive ultrastructural results, but who had situs inversus (n=17), or bronchiectasis and/or low nasal nitric oxide (n=6). Additionally, DNAH11 was sequenced in 13 subjects with isolated situs abnormalities to see if mutant DNAH11 could cause situs defects without respiratory disease. RESULTS: Of the 58 unrelated patients with PCD with normal ultrastructure, 13 (22%) had two (biallelic) mutations in DNAH11; and two patients without ultrastructural analysis had biallelic mutations. All mutations were novel and private. None of the patients with dynein arm or radial spoke/central pair defects, or isolated situs abnormalities, had mutations in DNAH11. Of the 35 identified mutant alleles, 24 (69%) were nonsense, insertion/deletion or loss-of-function splice-site mutations. CONCLUSIONS: Mutations in DNAH11 are a common cause of PCD in patients without ciliary ultrastructural defects; thus, genetic analysis can be used to ascertain the diagnosis of PCD in this challenging group of patients.


Subject(s)
Axonemal Dyneins/genetics , Cilia/ultrastructure , Ciliary Motility Disorders/genetics , Mutation , Adolescent , Adult , Child , Child, Preschool , Ciliary Motility Disorders/diagnosis , Ciliary Motility Disorders/pathology , Female , Genotype , Humans , Infant , Male , Pedigree , Phenotype , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
13.
Mol Microbiol ; 82(5): 1235-59, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22066998

ABSTRACT

Calcineurin, a heterodimer composed of the catalytic (CnaA) and regulatory (CnaB) subunits, plays key roles in growth, virulence and stress responses of fungi. To investigate the contribution of CnaA and CnaB to hyphal growth and septation, ΔcnaB and ΔcnaAΔcnaB strains of Aspergillus fumigatus were constructed. CnaA colocalizes to the contractile actin ring early during septation and remains at the centre of the mature septum. While CnaB's septal localization is CnaA-dependent, CnaA's septal localization is CnaB-independent, but CnaB is required for CnaA's function at the septum. Catalytic null mutations in CnaA caused stunted growth despite septal localization of the calcineurin complex, indicating the requirement of calcineurin activity at the septum. Compared to the ΔcnaA and ΔcnaB strains, the ΔcnaAΔcnaB strain displayed more defective growth and aberrant septation. While three Ca(2+) -binding motifs in CnaB were sufficient for its association with CnaA at the septum, the amino-terminal arginine-rich domains (16-RRRR-19 and 44-RLRKR-48) are dispensable for septal localization, yet required for complete functionality. Mutation of the 51-KLDK-54 motif in CnaB causes its mislocalization from the septum to the nucleus, suggesting it is a nuclear export signal sequence. These findings confirm a cooperative role for the calcineurin complex in regulating hyphal growth and septation.


Subject(s)
Aspergillus fumigatus/enzymology , Aspergillus fumigatus/growth & development , Calcineurin/metabolism , Hyphae/enzymology , Hyphae/growth & development , Actins/metabolism , Amino Acid Sequence , Aspergillus fumigatus/genetics , Calcineurin/genetics , Calcium/metabolism , Cytoplasm/chemistry , Fungal Proteins/metabolism , Gene Deletion , Hyphae/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism
14.
J Biol Chem ; 284(15): 9781-7, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19208631

ABSTRACT

Synaptotagmin 2 (Syt2) functions as a low affinity, fast exocytic Ca(2+) sensor in neurons, where it is activated by Ca(2+) influx through voltage-gated channels. Targeted insertion of lacZ into the mouse syt2 locus reveals expression in mucin-secreting goblet cells of the airways. In these cells, rapid Ca(2+) entry from the extracellular medium does not contribute significantly to stimulated secretion (Davis, C. W., and Dickey, B. F. (2008) Annu. Rev. Physiol. 70, 487-512). Nonetheless, Syt2(-/-) mice show a severe defect in acute agonist-stimulated airway mucin secretion, and Syt2(+/-) mice show a partial defect. In contrast to Munc13-2(-/-) mice (Zhu, Y., Ehre, C., Abdullah, L. H., Sheehan, J. K., Roy, M., Evans, C. M., Dickey, B. F., and Davis, C. W. (2008) J. Physiol. (Lond.) 586, 1977-1992), Syt2(-/-) mice show no spontaneous mucin accumulation, consistent with the inhibitory action of Syt2 at resting cytoplasmic Ca(2+) in neurons. In human airway goblet cells, inositol trisphosphate receptors are found in rough endoplasmic reticulum that closely invests apical mucin granules, consistent with the known dependence of exocytic Ca(2+) signaling on intracellular stores in these cells. Hence, Syt2 can serve as an exocytic sensor for diverse Ca(2+) signaling systems, and its levels are limiting for stimulated secretory function in airway goblet cells.


Subject(s)
Calcium Signaling , Endoplasmic Reticulum/metabolism , Exocytosis , Gene Expression Regulation , Goblet Cells/metabolism , Synaptotagmin II/physiology , Animals , Calcium/metabolism , Cytoplasm/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucins/metabolism , Neurons/metabolism , Synaptotagmin II/metabolism
15.
Am J Physiol Cell Physiol ; 293(2): C574-83, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17428842

ABSTRACT

In normal nasal epithelium, the olfactory receptor neurons (ORNs) are continuously replaced through the differentiation of progenitor cells. The olfactory epithelium (OE) of the cystic fibrosis (CF) mouse appears normal at birth, yet by 6 mo of age, a marked dysmorphology of sustentacular cells and a dramatic reduction in olfactory receptor neurons are evident. Electroolfactograms revealed that the odor-evoked response in 30-day-old CF mice was reduced approximately 45%; in older CF mice, a approximately 70% reduction was observed compared with the wild type (WT) response. Consistent with studies of CF airway epithelia, Ussing chamber studies of OE isolated from CF mice showed a lack of forskolin-stimulated Cl(-) secretion and an approximately 12-fold increase in amiloride-sensitive sodium absorption compared with WT mice. We hypothesize that the marked hyperabsorption of Na(+), most likely by olfactory sustentacular cells, leads to desiccation of the surface layer in which the sensory cilia reside, followed by degeneration of the ORNs. The CF mouse thus provides a novel model to examine the mechanisms of disease-associated loss of olfactory function.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Receptors, Odorant/metabolism , Smell , Acetophenones/pharmacology , Adenylyl Cyclases/metabolism , Aging/pathology , Aldehydes/pharmacology , Amiloride/pharmacology , Animals , Chlorides/metabolism , Colforsin/pharmacology , Cyclic AMP/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activators/pharmacology , Mice , Mice, Inbred CFTR , Microscopy, Electron, Scanning , Odorants , Olfactory Mucosa/drug effects , Olfactory Mucosa/physiopathology , Olfactory Mucosa/ultrastructure , Olfactory Receptor Neurons/drug effects , Olfactory Receptor Neurons/physiopathology , Olfactory Receptor Neurons/ultrastructure , Pentanols/pharmacology , RNA, Messenger/metabolism , Receptors, Odorant/drug effects , Smell/drug effects , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Time Factors , Tissue Culture Techniques
17.
Am J Physiol Lung Cell Mol Physiol ; 286(4): L631-42, 2004 Apr.
Article in English | MEDLINE | ID: mdl-12959927

ABSTRACT

Airway epithelial stem cells are not well characterized. To examine clonal growth potential, we diluted single, viable B6.129S7-Gtrosa26 (Rosa26) mouse tracheal epithelial cells that constitutively express -galactosidase into non-Rosa26 cells in an air-liquid interface cell culture model; 1.7% of the cells formed colonies of varying size, and, on average, 0.1% of the cells formed large colonies. Thus only a small subset of cells displayed progenitorial capacity suggestive of stem or early transient amplifying cells. Prior studies identified cells with high keratin 5 (K5) promoter activity in specific niches in the mouse trachea and these cells corresponded to the location of bromodeoxyuridine label-retaining cells, thought to be stem cells (Borthwick DW, Shahbazian M, Todd KQ, Dorin JR, and Randell SH, Am J Respir Cell Mol Biol: 24: 662-670, 2001). To explore the hypothesis that stem cells were present in the K5-expressing compartment, we created transgenic mice in which enhanced green fluorescent protein (EGFP) was driven by the K5 promoter. These mice expressed EGFP in most basal cells of the body including a subset of tracheal basal cells apparently located in positions similar to previously identified stem cell niches. Flow cytometrically purified EGFP-positive cells had an overall colony-forming efficiency 4.5-fold greater than EGFP-negative cells, but the ability to generate large colonies was 12-fold greater. Thus adult mouse tracheal epithelial cells with progenitorial capacity sufficient to generate large colonies reside in the basal cell compartment. These studies are a first step toward purification and characterization of airway epithelial stem cells.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Trachea/cytology , Age Factors , Animals , Cell Division , Cell Lineage , Cell Separation , Female , Flow Cytometry , Green Fluorescent Proteins , In Vitro Techniques , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Stem Cells/cytology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...