Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 11(10)2020 10 06.
Article in English | MEDLINE | ID: mdl-33036232

ABSTRACT

Lentiviral-mediated integration of a CFTR transgene cassette into airway basal cells is a strategy being considered for cystic fibrosis (CF) cell-based therapies. However, CFTR expression is highly regulated in differentiated airway cell types and a subset of intermediate basal cells destined to differentiate. Since basal stem cells typically do not express CFTR, suppressing the CFTR expression from the lentiviral vector in airway basal cells may be beneficial for maintaining their proliferative capacity and multipotency. We identified miR-106b as highly expressed in proliferating airway basal cells and extinguished in differentiated columnar cells. Herein, we developed lentiviral vectors with the miR-106b-target sequence (miRT) to both study miR-106b regulation during basal cell differentiation and detarget CFTR expression in basal cells. Given that miR-106b is expressed in the 293T cells used for viral production, obstacles of viral genome integrity and titers were overcome by creating a 293T-B2 cell line that inducibly expresses the RNAi suppressor B2 protein from flock house virus. While miR-106b vectors effectively detargeted reporter gene expression in proliferating basal cells and following differentiation in the air-liquid interface and organoid cultures, the CFTR-miRT vector produced significantly less CFTR-mediated current than the non-miR-targeted CFTR vector following transduction and differentiation of CF basal cells. These findings suggest that miR-106b is expressed in certain airway cell types that contribute to the majority of CFTR anion transport in airway epithelium.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Respiratory Mucosa/metabolism , Stem Cells/metabolism , Cell Differentiation , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Genetic Vectors , HEK293 Cells , Humans , Lentivirus/genetics
2.
Sci Transl Med ; 11(485)2019 03 27.
Article in English | MEDLINE | ID: mdl-30918114

ABSTRACT

Cystic fibrosis (CF) is a multiorgan disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). In patients with CF, abnormalities initiate in several organs before birth. However, the long-term impact of these in utero pathologies on disease pathophysiology is unclear. To address this issue, we generated ferrets harboring a VX-770 (ivacaftor)-responsive CFTR G551D mutation. In utero VX-770 administration provided partial protection from developmental pathologies in the pancreas, intestine, and male reproductive tract. Homozygous CFTR G551D/G551D animals showed the greatest VX-770-mediated protection from these pathologies. Sustained postnatal VX-770 administration led to improved pancreatic exocrine function, glucose tolerance, growth and survival, and to reduced mucus accumulation and bacterial infections in the lung. VX-770 withdrawal at any age reestablished disease, with the most rapid onset of morbidity occurring when withdrawal was initiated during the first 2 weeks after birth. The results suggest that CFTR is important for establishing organ function early in life. Moreover, this ferret model provides proof of concept for in utero pharmacologic correction of genetic disease and offers opportunities for understanding CF pathogenesis and improving treatment.


Subject(s)
Aminophenols/administration & dosage , Chloride Channel Agonists/administration & dosage , Cystic Fibrosis/drug therapy , Quinolones/administration & dosage , Animals , Animals, Genetically Modified , Animals, Newborn , Blood Glucose/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Disease Progression , Female , Ferrets , Gene Knock-In Techniques , Genitalia, Male/abnormalities , Genitalia, Male/drug effects , Gestational Age , Humans , Male , Mutation , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/pathology , Pancreas, Exocrine/physiopathology , Pregnancy , Respiratory Tract Infections/etiology , Respiratory Tract Infections/prevention & control , Translational Research, Biomedical
3.
Cell Rep ; 26(7): 1951-1964.e8, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30759402

ABSTRACT

Toolsets available for in-depth analysis of scRNA-seq datasets by biologists with little informatics experience is limited. Here, we describe an informatics tool (PyMINEr) that fully automates cell type identification, cell type-specific pathway analyses, graph theory-based analysis of gene regulation, and detection of autocrine-paracrine signaling networks in silico. We applied PyMINEr to interrogate human pancreatic islet scRNA-seq datasets and discovered several features of co-expression graphs, including concordance of scRNA-seq-graph structure with both protein-protein interactions and 3D genomic architecture, association of high-connectivity and low-expression genes with cell type enrichment, and potential for the graph structure to clarify potential etiologies of enigmatic disease-associated variants. We further created a consensus co-expression network and autocrine-paracrine signaling networks within and across islet cell types from seven datasets. PyMINEr correctly identified changes in BMP-WNT signaling associated with cystic fibrosis pancreatic acinar cell loss. This proof-of-principle study demonstrates that the PyMINEr framework will be a valuable resource for scRNA-seq analyses.


Subject(s)
RNA, Small Cytoplasmic/genetics , Sequence Analysis, RNA/methods , Autocrine Communication , Humans , Paracrine Communication
4.
Endocrinology ; 158(10): 3325-3338, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28977592

ABSTRACT

Although ß-cell dysfunction in cystic fibrosis (CF) leads to diabetes, the mechanism by which the cystic fibrosis transmembrane conductance regulator (CFTR) channel influences islet insulin secretion remains debated. We investigated the CFTR-dependent islet-autonomous mechanisms affecting insulin secretion by using islets isolated from CFTR knockout ferrets. Total insulin content was lower in CF as compared with wild-type (WT) islets. Furthermore, glucose-stimulated insulin secretion (GSIS) was impaired in perifused neonatal CF islets, with reduced first, second, and amplifying phase secretion. Interestingly, CF islets compensated for reduced insulin content under static low-glucose conditions by secreting a larger fraction of islet insulin than WT islets, probably because of elevated SLC2A1 transcripts, increased basal inhibition of adenosine triphosphate-sensitive potassium channels (K-ATP), and elevated basal intracellular Ca2+. Interleukin (IL)-6 secretion by CF islets was higher relative to WT, and IL-6 treatment of WT ferret islets produced a CF-like phenotype with reduced islet insulin content and elevated percentage insulin secretion in low glucose. CF islets exhibited altered expression of INS, CELA3B, and several ß-cell maturation and proliferation genes. Pharmacologic inhibition of CFTR reduced GSIS by WT ferret and human islets but similarly reduced insulin secretion and intracellular Ca2+ in CFTR knockout ferret islets, indicating that the mechanism of action is not through CFTR. Single-molecule fluorescent in situ hybridization, on isolated ferret and human islets and ferret pancreas, demonstrated that CFTR RNA colocalized within KRT7+ ductal cells but not endocrine cells. These results suggest that CFTR affects ß-cell function via a paracrine mechanism involving proinflammatory factors secreted from islet-associated exocrine-derived cell types.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Insulin-Secreting Cells/physiology , Insulin/metabolism , Animals , Animals, Newborn , Calcium/analysis , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Ferrets/genetics , Gene Knockout Techniques , Glucose/pharmacology , Humans , In Situ Hybridization, Fluorescence , Insulin/analysis , Insulin Secretion , Interleukin-6/metabolism , Interleukin-6/pharmacology , Islets of Langerhans/chemistry , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , KATP Channels/antagonists & inhibitors , Male , RNA/analysis
5.
Biores Open Access ; 2(3): 217-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23741634

ABSTRACT

Patient-specific cell replacement therapy is fast becoming the future of medicine, requiring safe, effective methods for reprogramming a patient's own cells. Previously, we showed that a single transient transfection with a plasmid encoding Oct4 was sufficient to reprogram human skin keratinocytes (HSKs), and that this transfection resulted in a decrease in global DNA methylation. In more recent work we showed that decreasing global DNA methylation using the U.S. Food and Drug Administration-approved cancer treatment drug decitabine was sufficient to induce expression of endogenous Oct4. Here we report that a single treatment with decitabine, followed by 5 days in a defined neuronal transformation medium, then 7 days in a neuronal maintenance medium is sufficient to convert HSKs into cells that change their morphology substantially, gain expression of neuronal markers, and lose expression of keratinocyte markers. Within 1 week of treatment the cells express mRNA for ß3-tubulin and doublecortin, and at the end of 2 weeks express mRNA for NeuN, FOXP2, and NCAM1. Additionally, at the end of this protocol, neurofilament-1, nestin, synapsin, FOXP2, and GluR1 proteins are detectable by immunostaining. Thus, we demonstrate a simple method that begins the process for producing cells for cell replacement therapies without using exogenously introduced DNA.

6.
J Dermatol ; 39(7): 617-24, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22486352

ABSTRACT

Previously, we showed that transient transfection with OCT4 not only produced high expression of Oct4 in skin keratinocytes, but also caused a generalized demethylation of keratinocyte DNA. We hypothesized that DNA demethylation alone might allow expression of endogenous OCT4. Here, we report that treatment with the cancer drug decitabine results in generalized DNA demethylation in skin keratinocytes, and by 48 h after treatment, 96% of keratinocytes show expression of the endogenous Oct4 protein and the OCT4 repressor mir-145. This is true for keratinocytes only, as skin fibroblasts treated similarly show no OCT4 or mir-145 expression. Decitabine-treated keratinocytes also show increased mir-302c and proliferation similar to other Oct4(+) cells. Treatment with doxorubicin, another cancer drug, induces expression of mir-145 only in cells that already express OCT4, suggesting that Oct4 regulates its own repressor. Co-treatment with decitabine and doxorubicin results first in increased OCT4 and mir-145, then a decrease in both, suggesting that OCT4 and mir-145 regulate each other. The novel strategy presented here provides a regulatable system to produce Oct4(+) cells for transformation studies and provides a unique method to study the effects of endogenous Oct4 in cancer cells and the surrounding somatic cells.


Subject(s)
Azacitidine/analogs & derivatives , Doxorubicin/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , MicroRNAs/metabolism , Octamer Transcription Factor-3/metabolism , Antineoplastic Agents/pharmacology , Azacitidine/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , DNA Methylation , Decitabine , Gene Expression Regulation/drug effects , Humans , Keratinocytes/cytology , MicroRNAs/genetics , Octamer Transcription Factor-3/genetics , Skin/cytology , Skin/drug effects , Skin/metabolism
7.
Trans Am Clin Climatol Assoc ; 122: 217-28, 2011.
Article in English | MEDLINE | ID: mdl-21686228

ABSTRACT

The lung's epithelial surface is at the same time vitally exchanging gas with the environment and acting as a barrier that protects the organism from the environment. We hypothesized that activation of epithelial-cell G-protein-coupled receptors for immune-defense molecules would temporarily interrupt cadherin-dependent cell-cell adhesion of epithelial cells and thereby focally and temporarily compromise the epithelial barrier to facilitate delivery of other immune molecules and cells to challenged sites. Activation of type 1 histamine or type 2 PAR receptors on the basolateral surface of primary airway epithelial cells or L-cells expressing E-cadherin interrupted cadherin adhesion and caused approximately a 50% decrease in the epithelial barrier for 2-3 minutes. Given basic biochemical observations of others, we further hypothesized that activation of the receptors altered the barrier by phosphorylating tyrosines on an essential cadherin-complex component, beta-catenin. Y-F mutations in beta-catenin completely blocked the effects of activating the same receptors on cadherin-dependent adhesion and on the epithelial barrier. Hence, G-protein-coupled receptors responding to immune-defense molecules temporarily and focally interrupt the lung epithelial barrier by compromising cadherin-based adhesion.


Subject(s)
Cadherins/metabolism , Cell Adhesion , Epithelial Cells/metabolism , Immunity, Mucosal , Receptor, PAR-2/metabolism , Receptors, Histamine H1/metabolism , Respiratory Mucosa/metabolism , Signal Transduction , Animals , Antigens, CD , Cell Line , Dogs , Electric Impedance , Epithelial Cells/drug effects , Epithelial Cells/immunology , Humans , Immunity, Mucosal/drug effects , Kidney/immunology , Kidney/metabolism , Mutation , Oligopeptides/pharmacology , Permeability , Phosphorylation , Receptor, PAR-2/agonists , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Time Factors , Transfection , Tyrosine , beta Catenin/genetics , beta Catenin/metabolism
8.
J Invest Dermatol ; 129(11): 2541-3, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19826444

ABSTRACT

It has been assumed that the slow rate of healing in aging epidermis is due to slowing of the epidermal stem cell proliferative rate. In this issue, Charruyer et al. report that this may not be the case. Using a long-term repopulating model, they demonstrate that epidermal stem cell kinetics are maintained. Instead, it is the compensatory action of the transit-amplifying (TA) cells that changes in aging skin and thus bears responsibility for slowed wound healing.


Subject(s)
Epidermal Cells , Epidermis/physiology , Skin Aging/pathology , Skin Aging/physiology , Animals , Cell Division/physiology , Humans , Keratinocytes/cytology , Keratinocytes/physiology , Stem Cells/cytology , Stem Cells/physiology
9.
Am J Physiol Lung Cell Mol Physiol ; 294(3): L442-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18083766

ABSTRACT

Activation of the type 1 histamine (H1) or the type 2 protease-activated (PAR-2) G protein-coupled receptors interrupts E-cadherin adhesion and decreases the transepithelial resistance (TER) of epithelium. Several reports suggest that cadherin adhesive function depends on the association of cadherin with beta-catenin and that this association is regulated by phosphorylation of tyrosines in beta-catenin. We tested the hypothesis that loss of cadherin adhesion and compromise of TER on activation of the H1 or PAR-2 receptor is due to phosphorylation of tyrosines in beta-catenin. L cells were stably transfected to express E-cadherin (L-E-cad cells) and H1 (L-H1-E-cad cells). L cells and Madin-Darby canine kidney (MDCK) cells constitutively express PAR-2. Stably transfected L-E-cad, L-H1-E-cad, and MDCK cells were also stably transfected with FLAG-tagged wild-type (WT) or mutant beta-catenin, converting tyrosine 142, 489, or 654 to the nonphosphorylatable mimetic, phenylalanine (WT, Y142F, Y489F, or Y654F). Activation of H1 or PAR-2 interrupted adhesion to an immobilized E-cadherin-Fc fusion protein of L-H1-E-cad, L-E-cad, and MDCK cells expressing WT or Y142F beta-catenin but did not interrupt adhesion of L-H1-E-cad, L-E-cad, and MDCK cells expressing the Y489F or Y654F mutant beta-catenins. PAR-2 activation decreased the TER of monolayers of MDCK cells expressing WT or Y142F beta-catenin 40-45%. However, PAR-2 activation did not decrease the TER of monolayers of MDCK cells expressing Y489F or Y654F beta-catenin. The protein tyrosine phosphatase PTP1B binds to the cadherin cytoplasmic domain and dephosphorylates beta-catenin. Inhibition of PTP1B interrupted adhesion to E-cadherin-Fc of MDCK cells expressing WT beta-catenin but did not affect the adhesion of MDCK cells expressing Y489F or Y654F beta-catenin. Similarly, inhibition of PTP1B compromised the TER of MDCK cells expressing WT beta-catenin but did not affect the TER of MDCK cells expressing Y489F or Y654F beta-catenin. We conclude that phosphorylation of tyrosines 489 and 654 in beta-catenin is a necessary step in the process by which G protein-coupled H1 and PAR-2 receptors interrupt E-cadherin adhesion. We also conclude that activation of PAR-2 has no effect on the TER without first interrupting E-cadherin adhesion.


Subject(s)
Cadherins/physiology , Receptor, PAR-2/physiology , Receptors, Histamine H1/physiology , Tyrosine/physiology , beta Catenin/genetics , Amino Acid Substitution , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Dogs , Electric Impedance , Epithelial Cells/drug effects , Epithelial Cells/physiology , Histamine/pharmacology , Humans , L Cells , Mice , Mutation , Oligopeptides/pharmacology , Phenylalanine/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptor, PAR-2/agonists , Transfection , Tyrosine/genetics
10.
J Immunol ; 178(10): 6395-403, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17475869

ABSTRACT

Respiratory pathogens and toxins often assault the lung from the airway lumen. Airway epithelia may initiate and amplify inflammation in response to these attacks, but under certain conditions confinement of inflammation to the airway lumen may be beneficial to the host. Accordingly, we hypothesized that airway epithelial polarity allows different responses to basolateral vs apical stimuli that may modulate inflammation. Using primary human airway epithelial cells differentiated at an air-liquid interface in culture, we found that responses to several cytokines required basolateral mediator application. In contrast, responses to Haemophilus influenzae occurred after either basolateral or apical interaction with airway epithelia. Experiments focused on IFN-gamma receptor polarity confirmed its predominant basolateral location in cultured airway epithelia as well as in normal human airway tissue. Furthermore, physical and pharmacologic disruption of barrier function in airway epithelia allowed responses to apical application of IFN-gamma and other cytokines. These in vitro studies directly correlated with experiments in mice in which an airway epithelial response to IFN-gamma injected into the airway lumen was seen only after disruption of barrier function. The results indicate that airway epithelia with intact barrier function restrict inflammatory responses by limitation of cell activation through requiring interaction of selected mediators with the basolateral surface. However, loss of barrier integrity allows epithelial responses to these mediators if located in the airway lumen to amplify airway defenses.


Subject(s)
Cell Membrane Permeability/immunology , Membrane Proteins/physiology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Animals , Bacteria/immunology , Bacteria/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Decanoic Acids/toxicity , Humans , Interferons/physiology , Interleukin-4/physiology , Mice , Mice, Inbred C57BL , Receptors, Cytokine/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Tumor Necrosis Factor-alpha/physiology
11.
Am J Physiol Lung Cell Mol Physiol ; 291(4): L628-35, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16714334

ABSTRACT

The airway epithelium is an important barrier between the environment and subepithelial tissues. The epithelium is also divided into functionally restricted apical and basolateral domains, and this restriction is dependent on the elements of the barrier. The protease-activated receptor-2 (PAR2) receptor is expressed in airway epithelium, and its activation initiates multiple effects including enhanced airway inflammation and reactivity. We hypothesized that activation of PAR2 would interrupt E-cadherin adhesion and compromise the airway epithelial barrier. The PAR2-activating peptide (PAR2-AP, SLIGRL) caused an immediate approximately 50% decrease in the transepithelial resistance of primary human airway epithelium that persisted for 6-10 min. The decrease in resistance was accompanied by an increase in mannitol flux across the epithelium and occurred in cystic fibrosis transmembrane conductance receptor (CFTR) epithelium pretreated with amiloride to block Na and Cl conductances, confirming that the decrease in resistance represented an increase in paracellular conductance. In parallel experiments, activation of PAR2 interrupted the adhesion of E-cadherin-expressing L cells and of primary airway epithelial cells to an immobilized E-cadherin extracellular domain, confirming the hypothesis that activation of PAR2 interrupts E-cadherin adhesion. Selective interruption of E-cadherin adhesion with antibody to E-cadherin decreased the transepithelial resistance of primary airway epithelium by >80%. Pretreatment of airway epithelium or the E-cadherin-expressing L cells with the long-acting beta-agonist salmeterol prevented PAR2 activation from interrupting E-cadherin adhesion and compromising the airway epithelial barrier. Activation of PAR2 interrupts E-cadherin adhesion and compromises the airway epithelial barrier.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Albuterol/analogs & derivatives , Bronchi/drug effects , Bronchi/physiology , Cadherins/metabolism , Receptor, PAR-2/physiology , Albuterol/pharmacology , Bronchi/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Electric Impedance , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , Humans , In Vitro Techniques , Permeability/drug effects , Receptor, PAR-2/metabolism , Receptors, Histamine H1/metabolism , Receptors, Histamine H1/physiology , Salmeterol Xinafoate , Thionucleotides/pharmacology
12.
Am J Physiol Lung Cell Mol Physiol ; 287(4): L816-23, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15220112

ABSTRACT

Histamine is an important agent of innate immunity, transiently increasing the flux of immune-competent molecules from the vascular space to the tissues and then allowing rapid restoration of the integrity of the endothelial barrier. In previous work we found that histamine alters the endothelial barrier by disrupting cell-cell adhesion and identified VE-cadherin as an essential participant in this process. The previous work did not determine whether histamine directly interrupted VE-cadherin adhesion, whether the effects of histamine were selective for cadherin adhesion, or whether capacitive calcium flux across the cell membrane was necessary for the effects of histamine on cell-cell adhesion. In the current work we found that histamine directly interrupts adhesion of L cells expressing the type 1 histamine (H1) receptor and VE-cadherin to a VE-cadherin-Fc fusion protein. In contrast, integrin-mediated adhesion to fibronectin of the same L cells expressing the H1 receptor was not affected by histamine, demonstrating that the effects of histamine are selective for cadherin adhesion. Some of the effects of many edemagenic agonists on endothelium are dependent on the capacitive flux of calcium across the endothelial cell membrane. Blocking capacitive calcium flux with LaCl3 did not prevent histamine from interrupting VE-cadherin adhesion of transfected L cells, nor did it prevent histamine from interrupting cell-cell adhesion of human umbilical vein endothelial cells. These data support the contentions that histamine directly and selectively interrupts cadherin adhesion and this effect on cadherin adhesion is independent of capacitive calcium flux.


Subject(s)
Cadherins/physiology , Calcium/physiology , Endothelium, Vascular/physiology , Histamine/pharmacology , Animals , Antigens, CD , Base Sequence , Cadherins/genetics , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cells, Cultured , DNA Primers , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Humans , Kinetics , L Cells , Mice , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Umbilical Veins
14.
Am J Physiol Lung Cell Mol Physiol ; 282(6): L1330-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12003790

ABSTRACT

Histamine increases microvascular permeability by creating small transitory (100-400 nm) gaps between adjacent endothelial cells at sites of vascular endothelial (VE)-cadherin-based adhesion. We examined the effects of histamine on the proteins within the VE-cadherin-based adherens junction in primary human umbilical vein endothelial cells. VE-cadherin is linked not only by beta- and alpha-catenin to cortical actin but also by gamma-catenin to the intermediate filament vimentin. In mature human umbilical vein cultures, the VE-cadherin immunoprecipitate contained equivalent amounts of alpha- and beta-catenin, 130% as much beta- as gamma-catenin, and 50% as much actin as vimentin. Within 60 s, histamine decreased the fraction of VE-cadherin in the insoluble portion of the cell lysate by 35 +/- 1.5%. At the same time, histamine decreased the amount of vimentin that immunoprecipitated with VE-cadherin by 50 +/- 6%. Histamine did not affect the amount of actin or the amount of alpha-, beta-, or gamma-catenin that immunoprecipitated with VE-cadherin. Within 60 s, histamine simulated a doubling in the phosphorylation of VE-cadherin and beta- and gamma-catenin. The VE-cadherin immunoprecipitate contained kinase activity that phosphorylated VE-cadherin and gamma-catenin in vitro.


Subject(s)
Adherens Junctions/metabolism , Endothelium, Vascular/metabolism , Histamine/pharmacology , Membrane Proteins/metabolism , Trans-Activators , Vimentin/metabolism , Actins/metabolism , Adherens Junctions/drug effects , Antigens, CD , Cadherins/metabolism , Cells, Cultured , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Desmoplakins , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Phosphorylation/drug effects , Precipitin Tests , alpha Catenin , beta Catenin , gamma Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...