Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Int J STEM Educ ; 9(1): 11, 2022.
Article in English | MEDLINE | ID: mdl-35127335

ABSTRACT

BACKGROUND: Across the globe, there have been significant reforms to improve STEM education at all levels. A significant part of this has been teacher reform. While the responses and resilience of STEM teachers to educational reforms in secondary education have received significant attention, the responses and resilience of STEM teachers in higher education remains understudied. In higher education, educational reforms of academic roles have seen increasing numbers of STEM academics focussed on education. Responses of STEM academics to education reform of the academic role have some parallels with teacher resilience, but there are also potential misalignments within a culture which values and prioritises science disciplinary research. This study examined the responses of STEM academics in higher education to educational reform of the academic role using the theoretical construct of resilience and Bronfenbrenner's socio-ecological model. This was a 2-year case study of 32 academics and senior educational leaders in higher education in STEM. Data collection included semi-structured interviews which were theme coded and inductively analysed. RESULTS: The responses and resilience of STEM academics focussed on education appeared to be dependent on interactions between individual disposition in the microsystem and influences of the exosystem and the external macrosystem. Five major themes emerged about the value and quality, scholarship and expertise, progress and mobility, status and identity and community and culture of STEM academics focussed on education. The exosystem was a significant unidirectional influence on STEM academics where judgements were made concerning academic performance, awards, and promotion. Responses of senior leaders in the exosystem were influenced by the macrosystem and culture of science. Academics focussed on research, rather than education were more valued and more likely to be both financially rewarded and promoted. CONCLUSION: During this pressured decade, where COVID-19 has intensified stress, more attention on the direction and reciprocal relationships in the socio-ecological model of higher education is needed in order for educational reform in higher education STEM to be effective. Resilience of STEM academics to educational reform in higher education is a dynamic quality, and the capacity to "bounce back", learn from challenges, and realise expectations of educational reform will depend on an understanding of resilience and support of Bronfenbrenner's spheres of influence.

2.
Clin Exp Pharmacol Physiol ; 42(10): 1118-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26173747

ABSTRACT

The consumption of a high fat diet (HFD) is associated with proteinuria and altered sodium handling and excretion, which can lead to kidney disease. In the proximal tubule, the Na(+) /H(+) Exchanger 3 (NHE3) is responsible for normal protein reabsorption and the reabsorption of approximately 70% of the renal sodium load. It is the Na(+) /K(+) -ATPase that provides the driving force for the reabsorption of sodium and its exit across the basolateral membrane. This study investigates the effects that consumption of a HFD for 12 weeks has on NHE3 and Na(+) /K(+) -ATPase expression in the kidney. Western blot analysis identified a significant reduction in NHE3 and its modulator, phosphorylated protein kinase B, in renal lysate from obese rats. In the obese rats, a reduction in NHE3 expression in the proximal tubule may impact on the acidification of endosomes which are responsible for albumin uptake, suggesting a key role for the exchanger in protein endocytosis in obesity. Western blot analysis identified a reduction in Na(+) /K(+) -ATPase which could also potentially impact on albumin uptake and sodium reabsorption. This study demonstrates that consumption of a HFD for 12 weeks reduces renal NHE3 and Na(+) /K(+) -ATPase expression, an effect that may contribute to the albuminuria associated with obesity. Furthermore the reduction in these transporters is not likely to contribute to the reduced sodium excretion in obesity. These data highlight a potential link between NHE3 and Na(+) /K(+) -ATPase in the pathophysiological changes in renal protein handling observed in obesity.


Subject(s)
Diet, High-Fat/adverse effects , Gene Expression Regulation, Enzymologic , Kidney/metabolism , Obesity/etiology , Obesity/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Male , Obesity/complications , Obesity/genetics , Phosphoproteins/metabolism , Proteinuria/complications , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Potassium-Exchanging ATPase/genetics
3.
Br J Pharmacol ; 169(6): 1239-51, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23594166

ABSTRACT

BACKGROUND AND PURPOSE: Albuminuria is an important biomarker of renal dysfunction and is a major mediator of renal damage and fibrosis during kidney disease. The mechanisms underlying albumin-induced renal fibrosis remain unclear. There has been significant interest in γ-secretase activity in tubular epithelial cells in recent times; however, its potential role in albumin-induced fibrosis has not been investigated. EXPERIMENTAL APPROACH: The primary aim of this study was to examine the role of γ-secretase in albumin-induced fibrotic effects in proximal tubular cells. The effects of increasing albumin concentrations on fibrosis indicators and mediators in the human HK-2 cell line were examined in the presence and absence of a γ-secretase inhibitor, compound E. KEY RESULTS: Treatment with albumin resulted in a number of pro-fibrotic effects, including up-regulation of fibronectin, TGF-ß1 and the EGF-R. Interestingly, similar effects were observed in response to treatment with the γ-secretase inhibitor, compound E. Co-treatment of cells with albumin and an EGF-R inhibitor, AG-1478, resulted in significant inhibition of the observed pro-fibrotic effects, suggesting a major role for the EGF-R in albumin-induced fibrotic events. Albumin-induced effects on the EGF-R appeared to be mediated through inhibition of γ-secretase activity and were dependent on ERK-MAPK signalling. CONCLUSIONS AND IMPLICATIONS: These results provide novel insights into the mechanisms of albumin-induced fibrotic effects in tubular epithelial cells, suggesting important roles for the γ-secretase and the EGF-R. These results suggest that the proposed use of γ-secretase inhibitors as anti-fibrotic agents requires further investigation.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Endocytosis/drug effects , Enzyme Inhibitors/pharmacology , Kidney Tubules, Proximal/drug effects , Proteolysis/drug effects , Serum Albumin, Bovine/metabolism , Urothelium/drug effects , Amyloid Precursor Protein Secretases/metabolism , Animals , Benzodiazepinones/pharmacology , Cattle , Cell Line , Culture Media, Serum-Free , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Fibronectins/metabolism , Fibrosis , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , MAP Kinase Signaling System/drug effects , Opossums , Quinazolines/pharmacology , Transforming Growth Factor beta1/metabolism , Tyrphostins/pharmacology , Urothelium/metabolism , Urothelium/pathology
4.
Int J Biochem Cell Biol ; 44(5): 815-23, 2012 May.
Article in English | MEDLINE | ID: mdl-22349218

ABSTRACT

Albumin endocytosis in the proximal tubule is mediated by a number of proteins, including the scavenger receptor megalin/cubilin and the PSD-95/Dlg/ZO-1 (PDZ) scaffolds NHERF1 and NHERF2. In addition, in a number of in vitro and in vivo models, the loss of ClC-5 results in a decreased cell surface expression and whole cell level of megalin, suggesting an interaction between these two proteins in vivo. We investigated if ClC-5 and megalin interact directly, and as ClC-5 binds to NHERF2, we investigated if this PDZ scaffold was required for a megalin/ClC-5 complex. GST-pulldown and immunoprecipitation experiments using rat kidney lysate demonstrated an interaction between ClC-5 and megalin, which was mediated by their C-termini. As this interaction may be controlled by a scaffold protein, we characterised any interaction between megalin and NHERF2. Immunoprecipitation experiments indicated that megalin interacts with NHERF2 in vivo, and that this interaction was via an internal NHERF binding domain in the C-terminus of megalin and PDZ2 and the C-terminus of NHERF2. Silencing NHERF2 had no effect on megalin protein levels in the whole cell or plasma membrane. Using siRNA against NHERF2, we demonstrated that NHERF2 was required to facilitate the interaction between megalin and ClC-5. Using fusion proteins, we characterised a protein complex containing ClC-5 and megalin, which is scaffolded by NHERF2, in the absence of any other proteins. Importantly, these observations are the first to describe an interaction between megalin and ClC-5, which is scaffolded by NHERF2 in proximal tubule cells.


Subject(s)
Albumins/metabolism , Antiporters/metabolism , Cytoskeletal Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Organic Anion Transporters/metabolism , Animals , Antiporters/genetics , Binding Sites , Cytoskeletal Proteins/genetics , Endocytosis/physiology , Gene Expression , Immunoprecipitation , Kidney Tubules, Proximal/physiology , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Organic Anion Transporters/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism
5.
Genes Brain Behav ; 6(8): 750-5, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17331106

ABSTRACT

Photosensitive seizures occur most commonly in childhood and adolescence, usually as a manifestation of complex idiopathic generalized epilepsies (IGEs). Molecular mechanisms underlying this condition are yet to be determined because no susceptibility genes have been identified. The NEDD4-2 (Neuronally Expressed Developmentally Downregulated 4) gene encodes a ubiquitin protein ligase proposed to regulate cell surface levels of several ion channels, receptors and transporters involved in regulating neuronal excitability, including voltage-gated sodium channels (VGSCs), the most clinically relevant of the epilepsy genes. The regulation of NEDD4-2 in vivo involves complex interactions with accessory proteins in a cell type specific manner. We screened NEDD4-2 for mutations in a cohort of 253 families with IGEs. We identified three NEDD4-2 missense changes in highly conserved residues; S233L, E271A and H515P in families with photosensitive generalized epilepsy. The NEDD4-2 variants were as effective as wild-type NEDD4-2 in downregulating the VGSC subtype Na(v)1.2 when assessed in the Xenopus oocyte heterologous expression system showing that the direct interaction with the ion channel was not altered by these variants. These data raise the possibility that photosensitive epilepsy may arise from defective interaction of NEDD4-2 with as yet unidentified accessory or target proteins.


Subject(s)
Epilepsy, Generalized/genetics , Epilepsy, Reflex/genetics , Ion Channel Gating/genetics , Ubiquitin-Protein Ligases/genetics , Case-Control Studies , Chromosomes, Human, Pair 18/genetics , Cohort Studies , Endosomal Sorting Complexes Required for Transport , Epilepsy, Generalized/metabolism , Epilepsy, Reflex/metabolism , Female , Genetic Predisposition to Disease , Humans , Ion Channel Gating/physiology , Male , Mutation, Missense , Nedd4 Ubiquitin Protein Ligases , Pedigree , Sequence Deletion , Sequence Homology, Amino Acid , Sodium Channels/metabolism , Xenopus Proteins
6.
Am J Physiol Renal Physiol ; 288(4): F800-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15536170

ABSTRACT

Matrix accumulation in the renal tubulointerstitium is predictive of a progressive decline in renal function. Transforming growth factor-beta(1) (TGF-beta(1)) and, more recently, connective tissue growth factor (CTGF) are recognized to play key roles in mediating the fibrogenic response, independently of the primary renal insult. Further definition of the independent and interrelated effects of CTGF and TGF-beta(1) is critical for the development of effective antifibrotic strategies. CTGF (20 ng/ml) induced fibronectin and collagen IV secretion in primary cultures of human proximal tubule cells (PTC) and cortical fibroblasts (CF) compared with control values (P < 0.005 in all cases). This effect was inhibited by neutralizing antibodies to either TGF-beta or to the TGF-beta type II receptor (TbetaRII). TGF-beta(1) induced a greater increase in fibronectin and collagen IV secretion in both PTC (P < 0.01) and CF (P < 0.01) compared with that observed with CTGF alone. The combination of TGF-beta(1) and CTGF was additive in their effects on both PTC and CF fibronectin and collagen IV secretion. TGF-beta(1) (2 ng/ml) stimulated CTGF mRNA expression within 30 min, which was sustained for up to 24 h, with a consequent increase in CTGF protein (P < 0.05), whereas CTGF had no effect on TGF-beta(1) mRNA or protein expression. TGF-beta(1) (2 ng/ml) induced phosphorylated (p)Smad-2 within 15 min, which was sustained for up to 24 h. CTGF had a delayed effect on increasing pSmad-2 expression, which was evident at 24 h. In conclusion, this study has demonstrated the key dependence of the fibrogenic actions of CTGF on TGF-beta. It has further uniquely demonstrated that CTGF requires TGF-beta, signaling through the TbetaRII in both PTCs and CFs, to exert its fibrogenic response in this in vitro model.


Subject(s)
Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/metabolism , Kidney Diseases/pathology , Transforming Growth Factor beta/metabolism , Antibodies , Cells, Cultured , Collagen Type IV/metabolism , Connective Tissue Growth Factor , DNA-Binding Proteins/metabolism , Fibronectins/metabolism , Fibrosis , Gene Expression/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Kidney/metabolism , Kidney/pathology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Smad2 Protein , Trans-Activators/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta1
7.
Diabet Med ; 21(10): 1102-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15384957

ABSTRACT

BACKGROUND: Statins are known to enhance atherosclerotic plaque stability through influences on extracellular matrix homeostasis. Net matrix production reflects the relative balance of matrix production and degradation through enzymes such as matrix metalloproteinases (MMPs) and their inhibitors, tissue inhibitor of MMP (TIMPs). The effects of statins on endothelial cell production of these parameters following co-exposure with a proatherogenic stimulus such as high glucose are not known. METHODS: Human endothelial cells were exposed for 72 h to 5 mm (control) or 25 mm (high) glucose +/- atorvastatin (1 micromol/l). Extracellular matrix homeostasis was assessed by measuring matrix metalloproteinase (MMP)-2 secretion, tissue inhibitor of MMP (TIMP)-1 and -2 secretion and net collagen IV production. Results were expressed as percentage +/- SEM of control values. RESULTS: Exposure to high glucose increased cellular collagen IV expression to 190.1 +/- 11.7% (P < 0.0001) of control levels. No change in MMP-2 secretion (111.6 +/- 5.2%; P > 0.05) was observed but both TIMP-1 and TIMP-2 expression were increased to 136.3 +/- 6.4% and 144.0 +/- 27.5%, respectively (both P < 0.05). The presence of atorvastatin in high glucose conditions reduced collagen IV expression to 136.1 +/- 20.6%. This was paralleled by increased secretion of MMP-2 to 145.8 +/- 7.8% (P < 0.01), increased TIMP-2 expression to 208.0 +/- 21.3% (P < 0.005 compared with high glucose) but no change in TIMP-1 expression (155.1 +/- 14.6%) compared with high glucose alone. The presence of atorvastatin in control conditions did not affect levels of collagen IV expression (114.5 +/- 13.2%). CONCLUSIONS: Endothelial cell exposure to high glucose was associated with a MMP/TIMP profile that increased extracellular matrix production which was attenuated by concurrent exposure to atorvastatin. Consequently, a mechanism by which the atherosclerotic plaque regression that is observed in patients taking these drugs has been demonstrated.


Subject(s)
Collagen Type IV/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/drug effects , Glucose/pharmacology , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pyrroles/pharmacology , Atorvastatin , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Matrix Metalloproteinase 2/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Umbilical Veins/drug effects , Umbilical Veins/metabolism
8.
Am J Physiol Renal Physiol ; 287(2): F268-73, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15113747

ABSTRACT

Postprandial hyperglycemia is implicated as a risk factor predisposing to vascular complications. This study was designed to assess recurrent short-term increases in glucose on markers of renal fibrogenesis. Human renal cortical fibroblasts were exposed to fluctuating short-term (2 h) increases to 15 mM d-glucose, three times a day over 72 h, on a background of 5 mM d-glucose. To determine whether observed changes were due to fluctuating osmolality, identical experiments were undertaken with cells exposed to l-glucose. Parallel experiments were performed in cells exposed to 5 mM d-glucose and constant exposure to either 15 or 7.5 mM d-glucose. Fluctuating d-glucose increased extracellular matrix, as measured by proline incorporation (P < 0.05), collagen IV (P < 0.005), and fibronectin production (P < 0.001), in association with increased tissue inhibitor of matrix metalloproteinase (MMP) (P < 0.05). Sustained exposure to 15 mM d-glucose increased fibronectin (P < 0.001), in association with increased MMP-2 (P = 0.01) and MMP-9 activity (P < 0.05), suggestive of a protective effect on collagen matrix accumulation. Transforming growth factor-beta(1) (TGF-beta(1)) mRNA was increased after short-term (90 min) exposure to 15 mM glucose (P < 0.05) and after 24-h exposure to 7.5 mM ? (P < 0.05). Normalization of TGF-beta(1) secretion occurred within 48 h of constant exposure to an elevated glucose. Fluctuating l-glucose also induced TGF-beta(1) mRNA and a profibrotic profile, however, to a lesser extent than observed with exposure to fluctuating d-glucose. The results suggest that exposure to fluctuating glucose concentrations increases renal interstitial fibrosis compared with stable elevations in d-glucose. The effects are, in part, due to the inherent osmotic changes.


Subject(s)
Blood Glucose/metabolism , Kidney/pathology , Cells, Cultured , Collagen Type IV/biosynthesis , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/metabolism , Fibrosis , Glucose/administration & dosage , Glucose/metabolism , Humans , Kidney Cortex/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Proline/metabolism , RNA, Messenger/metabolism , Time Factors , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
9.
Am J Physiol Endocrinol Metab ; 285(4): E708-17, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12783777

ABSTRACT

The mitogen-activated protein (MAP) kinases contribute to altered cell growth and function in a variety of disease states. However, their role in the endothelial complications of diabetes mellitus remains unclear. Human endothelial cells were exposed for 72 h to 5 mM (control) or 25 mM (high) glucose or 5 mM glucose plus 20 mM mannitol (osmotic control). The roles of p38 and p42/44 MAP kinases in the high glucose-induced growth effects were determined by assessment of phosphorylated MAP kinases and their downstream activators by Western blot and by pharmacological inhibition of these MAP kinases. Results were expressed as a percentage (means +/- SE) of control. High glucose increased the activity of total and phosphorylated p38 MAP kinase (P < 0.001) and p42/44 MAP kinase (P < 0.001). Coexposure of p38 MAP kinase blocker with high glucose reversed the antiproliferative but not the hypertrophic effects associated with high-glucose conditions. Transforming growth factor (TGF)-beta1 increased the levels of phosphorylated p38 MAP kinase, and p38 MAP kinase blockade reversed the antiproliferative effects of this cytokine. The high glucose-induced increase in phosphorylated p38 MAP kinase was reversed in the presence of TGF-beta1 neutralizing antibody. Although hyperosmolarity also induced antiproliferation (P < 0.0001) and cell hypertrophy (P < 0.05), there was no change in p38 activity, and therefore inhibition of p38 MAP kinase had no influence on these growth responses. Blockade of p42/44 MAP kinase had no effect on the changes in endothelial cell growth induced by either high glucose or hyperosmolarity. High glucose increased p42/44 and p38 MAP kinase activity in human endothelial cells, but only p38 MAP kinase mediated the antiproliferative growth response through the effects of autocrine TGF-beta1. High glucose-induced endothelial cell hypertrophy was independent of activation of the MAP kinases studied. In addition, these effects were independent of any increase in osmolarity associated with high-glucose exposure.


Subject(s)
Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Glucose/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Glucose/metabolism , Humans , Plant Proteins , Umbilical Cord/blood supply , Umbilical Cord/metabolism , p38 Mitogen-Activated Protein Kinases
10.
Am J Physiol Renal Physiol ; 285(4): F748-57, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12799307

ABSTRACT

The progression of renal disease correlates strongly with hypertension and the degree of proteinuria, suggesting a link between excessive Na+ reabsorption and exposure of the proximal tubule to protein. The present study investigated the effects of albumin on cell growth and Na+ uptake in primary cultures of human proximal tubule cells (PTC). Albumin (1.0 mg/ml) increased cell proliferation to 134.1 +/- 11.8% (P < 0.001) of control levels with no change in levels of apoptosis. Exposure to 0.1 and 1.0 mg/ml albumin increased total 22Na+ uptake to 119.1 +/- 6.3% (P = 0.005) and 115.6 +/- 5.3% (P < 0.006) of control levels, respectively, because of an increase in Na+/H+ exchanger isoform 3 (NHE3) activity. This was associated with an increase in NHE3 mRNA to 161.1 +/- 15.1% (P < 0.005) of control levels in response to 0.1 mg/ml albumin. Using confocal microscopy with a novel antibody raised against the predicted extracellular NH2 terminus of human NHE3, we observed in nonpermeabilized cells that exposure of PTC to albumin (0.1 and 1.0 mg/ml) increased NHE3 at the cell surface to 115.4 +/- 2.7% (P < 0.0005) and 122.4 +/- 3.7% (P < 0.0001) of control levels, respectively. This effect was paralleled by significant increases in NHE3 in the subplasmalemmal region as measured in permeabilized cells. These albumin-induced increases in expression and activity of NHE3 in PTC suggest a possible mechanism for Na+ retention in response to proteinuria.


Subject(s)
Albumins/pharmacology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Sodium-Hydrogen Exchangers/metabolism , Apoptosis/drug effects , Cell Division/drug effects , Cells, Cultured , Fluorescent Antibody Technique , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/physiology , Microscopy, Confocal , Osmolar Concentration , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium/pharmacokinetics , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics
11.
Am J Physiol Cell Physiol ; 284(6): C1374-86, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12540377

ABSTRACT

Human endothelial cells were exposed to 5 mM glucose (control), 25 mM (high) glucose, or osmotic control for 72 h. TGF-beta1 production, cell growth, death, and cell cycle progression, and the effects of TGF-beta1 and TGF-beta neutralization on these parameters were studied. High glucose and hyperosmolarity increased endothelial TGF-beta1 secretion (P < 0.0001) and bioactivity (P < 0.0001). However, high glucose had a greater effect on reducing endothelial cell number (P < 0.001) and increasing cellular protein content (P < 0.001) than the osmotic control. TGF-beta antibody only reversed the antiproliferative and hypertrophic effects of high glucose. High glucose altered cell cycle progression and cyclin-dependent kinase inhibitor expression independently of hyperosmolarity. High glucose increased endothelial cell apoptosis (P < 0.01), whereas hyperosmolarity induced endothelial cell necrosis (P < 0.001). TGF-beta antibody did not reverse the apoptotic effects observed with high glucose. Exogenous TGF-beta1 mimicked the increased S phase delay but not endoreduplication observed with high glucose. High glucose altered endothelial cell growth, apoptosis, and cell cycle progression. These growth effects occurred principally via a TGF-beta1 autocrine pathway. In contrast, apoptosis and endoreduplication occurred independently of this cytokine and hyperosmolarity.


Subject(s)
Autocrine Communication , Endothelium, Vascular/metabolism , Glucose/metabolism , Proto-Oncogene Proteins c-bcl-2 , Transforming Growth Factor beta/metabolism , Antibodies/metabolism , Apoptosis/physiology , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cell Division/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Diabetes Mellitus/metabolism , Endothelium, Vascular/cytology , Enzyme Inhibitors/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Necrosis , Osmolar Concentration , Proto-Oncogene Proteins/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein
12.
Pflugers Arch ; 444(5): 644-53, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12194018

ABSTRACT

We used replication-deficient adenoviruses overexpressing antisense against G(q) class alpha-subunits to determine the roles of G(q) and G(11) in mediating M(3)-receptor-coupled Ca(2+) mobilization in intact HT29 human colonic carcinoma epithelial cells. Western blot analysis and confocal microscopy showed that the viruses expressing antisense directed against the alpha-subunits of G(q) or G(11) produced isoform-specific reductions in the levels of these alpha-subunits. Fura-2 was used to measure changes in the Ca(2+) response following activation of the M(3) receptors by carbachol. The G(alpha)(q) antisense virus suppressed the peak Ca(2+) response by 70%, whereas the G(alpha)(11) antisense virus reduced it by 34%. We then used co-infection with both viruses to determine the effect of concomitant suppression of both G(alpha)(q) and G(alpha)(11). Overexpression of antisense to both alpha-subunits reduced by approximately 50% the levels of both G(alpha)(q) and G(alpha)(11). It also almost completely inhibited the Ca(2+) response to carbachol. These data show that both G(q) and G(11) are involved in mediating the action of the M(3) receptor on cytosolic Ca(2+) in HT29 cells. Furthermore, they suggest that the coupling of the M(3) receptor to these G proteins is specific, in that G(alpha)(q) cannot substitute for G(alpha)(11), and vice versa.


Subject(s)
Calcium Signaling/physiology , Epithelial Cells/physiology , Heterotrimeric GTP-Binding Proteins/genetics , Receptors, Muscarinic/metabolism , Adenoviridae/genetics , DNA, Antisense , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Expression Regulation, Viral , HT29 Cells , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Intestinal Mucosa/cytology , Receptor, Muscarinic M3
13.
Am J Physiol Lung Cell Mol Physiol ; 281(6): L1369-78, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11704532

ABSTRACT

The protease-activated receptor (PAR)-2 is present on the smooth muscle and epithelium of human airways and can be activated by mast cell tryptase, trypsin, or the PAR-2 activating peptide (AP). Trypsin and the PAR-2 AP induced contractions in human isolated airways, and these contractions were potentiated in the presence of the cyclooxygenase (COX) inhibitor indomethacin. Trypsin also increased the contractions to histamine in airways from sensitized (allergic) patients but not from nonsensitized (nonallergic) patients. Tryptase purified from human lung, skin and lung recombinant beta-tryptases, trypsin, and the PAR-2 AP all increased DNA synthesis in human airway smooth muscle (HASM) cells. Activation of PAR-2 by tryptase, trypsin, and the PAR-2 AP did not induce PGE(2) release from HASM cells. Trypsin and the PAR-2 AP increased the levels of intracellular calcium in HASM cells, with desensitization evident after treatment with either agonist. In conclusion, activation of PAR-2 can induce contractions of human airways, potentiate contractions to histamine, and induce proliferation and therefore may contribute to airway diseases such as asthma.


Subject(s)
Bronchi/metabolism , Inflammation Mediators/pharmacology , Muscle, Smooth/metabolism , Receptors, Thrombin/metabolism , Serine Endopeptidases/pharmacology , Adult , Bronchi/cytology , Bronchi/drug effects , Bronchoconstriction/drug effects , Bronchoconstriction/physiology , Calcium/metabolism , Cell Division/drug effects , Cells, Cultured , Dinoprostone/metabolism , Humans , Middle Aged , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Oligopeptides/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Receptor, PAR-2 , Tryptases
14.
Am J Respir Cell Mol Biol ; 23(4): 555-9, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11017922

ABSTRACT

We hypothesized that an atypical isoform of protein kinase (PK) C, PKC-zeta, is essential for proliferation of human airway smooth muscle (HASM) cells in primary culture. Recombinant replication-deficient E1-deleted adenoviruses (100 plaque-forming units [pfu]/cell) expressing the antisense of PKC-zeta and the wild-type PKC-zeta (Ad-CMV-PKC-zeta) were added to actively growing cells that were subsequently incubated for 48 h in platelet-derived growth factor (PDGF) 40 ng/mL or 10% fetal bovine serum (FBS). Expression of the antisense at a virus concentration of 100 pfu/cell produced a significant (n = 3, P<0.05) decrease in the mean manual cell count in the presence of PDGF to 37+/-5% relative to that in cells with no virus (100%), whereas in cells infected with virus containing no construct, this figure was 102+/-13%. The increase in cell number in response to FBS, however, was not affected by the presence of the antisense. Corresponding values for cells in 10% FBS were 100+/-22%, 85+/-22%, and 122+/-18%. Western blotting revealed decreased levels of PKC-zeta protein, but not PKC-alpha or PKC-epsilon protein, in cells infected with the antisense when compared with levels in control cells. Thus, in HASM cells, PKC-zeta is involved in proliferation in response to PDGF, but not in response to FBS, for which alternate signal transduction pathways independent of PKC-zeta must exist.


Subject(s)
Cell Division/drug effects , Isoenzymes/genetics , Muscle, Smooth/drug effects , Oligonucleotides, Antisense/pharmacology , Protein Kinase C/genetics , Trachea/drug effects , Adenoviridae/genetics , Animals , Cattle , Cell Line , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Muscle, Smooth/cytology , Trachea/cytology , Up-Regulation
15.
Neuropharmacology ; 39(11): 2054-66, 2000 Aug 23.
Article in English | MEDLINE | ID: mdl-10963749

ABSTRACT

The P2X(1) purinergic receptor subtype occurs on smooth muscle cells of the vas deferens and urinary bladder where it is localized in two different size receptor clusters, with the larger beneath autonomic nerve terminal varicosities. We have sought to determine whether these synaptic-size clusters only form in the presence of varicosities and whether they are labile when exposed to agonists. P2X(1) and a chimera of P2X(1) and green fluorescent protein (GFP) were delivered into cells using microinjection, transient transfection or infection with a replication-deficient adenovirus. The P2X(1)-GFP chimera was used to study the time course of P2X(1) receptor clustering in plasma membranes and the internalization of the receptor following prolonged exposure to ATP. Both P2X(1) and P2X(1)-GFP clustered in the plasma membranes of Xenopus oocytes, forming patches 4-6 microm in diameter. Human embryonic kidney 293 (HEK293) cells, infected with the adenovirus, possessed P2X(1) antibody-labeled regions in the membrane colocalized with GFP fluorescence. The ED(50) for the binding of alpha,beta-methylene adenosine triphosphate (alpha,beta-meATP) to the P2X(1)-GFP chimera was similar to native P2X(1) receptors. ATP-generated whole-cell currents in oocytes or HEK293 cells expressing either P2X(1) or P2X(1)-GFP were similar. Exposure of HEK293 cells to alpha, beta-meATP for 10-20 min in the presence of 5 microM monensin led to the disappearance of P2X(1)-GFP fluorescence from the surface of the cells. These observations using the P2X(1)-GFP chimera demonstrate that P2X(1) receptors spontaneously form synaptic-size clusters in the plasma membrane that are internalized on exposure to agonists.


Subject(s)
Indicators and Reagents/metabolism , Luminescent Proteins/metabolism , Membrane Potentials/physiology , Receptors, Purinergic P2/metabolism , Recombinant Fusion Proteins/metabolism , Adenosine Triphosphate/pharmacology , Adenoviridae/genetics , Animals , Cell Line , Green Fluorescent Proteins , Humans , Membrane Potentials/drug effects , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2X , Recombinant Fusion Proteins/genetics , Transcription, Genetic/genetics , Transfection , Xenopus
16.
Immunol Cell Biol ; 78(4): 375-86, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10947862

ABSTRACT

Plasma membrane-spanning G-protein-linked receptors transduce approximately 60% of all extracellular stimuli in higher animals. Many G-protein-linked receptor pathways are yet to be elucidated, with the receptor, G-protein or effector system as yet unidentified. In addition, many fundamental issues pertaining to G-protein signalling remain unresolved, such as the factors governing the specificity of G-protein receptor coupling and the control of signal amplitude in response to G-protein activation. In order to address some of these issues, the use of replication-deficient adenoviruses as gene transfer vectors for investigations of G-protein signalling has been developed, facilitating dissection of G-protein-linked signal transduction pathways in an extensive range of cultured cells, as well as in vivo. The present review focuses on the versatility and utility of adenoviruses for the investigation of signalling by heterotrimeric G-proteins and explores some of the recent advances in adenoviral technology as they relate to the study of signal transduction.


Subject(s)
Adenoviridae/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Signal Transduction , Adenoviridae/growth & development , Angiotensins/metabolism , Animals , Cells, Cultured , Endocytosis , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Gene Transfer Techniques , Genetic Vectors , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Receptors, Adrenergic/metabolism , Receptors, LHRH/metabolism , Receptors, Muscarinic/metabolism , Receptors, Odorant/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y2 , Receptors, Serotonin/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Receptors, Vasopressin/metabolism , Recombinant Proteins/metabolism , Virus Replication/genetics
17.
Cell Calcium ; 27(5): 247-55, 2000 May.
Article in English | MEDLINE | ID: mdl-10859591

ABSTRACT

Using Fura-2 to measure changes in intracellular calcium ([Ca(2+)](i)), we show that P(2U)receptors in HT29 cells trigger an increase in [Ca(2+)](i)by pertussis toxin-insensitive G proteins. We then use replication-deficient adenoviruses expressing wild-type and dominant negative mutants of G(alpha q)and G(alpha i2), antisense directed against G(alpha q)or the C-terminal fragment of beta-adrenergic receptor kinase (beta ARK-CT) to identify these G proteins. We find the [Ca(2+)](i)response to UTP is not affected by increased expression of the wild-type G(alpha q), wild-type G(alpha i2)or beta ARK-CT, while it is blocked by over-expression of dominant negative G(alpha q). The timecourse of the UTP response is, however, altered by wild-type G(alpha q)and is only weakly inhibited by antisense G(alpha q). This suggests that the P(2U)response is mediated, at least partially, by a G protein distinct from G(alpha q). In contrast, the M(3)muscarinic response is inhibited by over-expression of antisense against G(alpha q), or over-expression of beta ARK-CT, a finding in agreement with our previous observation that the muscarinic response in HT29 cells is mediated by the beta gamma-subunits of G(q). We also find that P(2U)and M(3)receptors do not control identical Ca(2+)stores, suggesting that differential activation of G proteins can lead to Ca(2+)release from distinct stores.


Subject(s)
Calcium/metabolism , GTP-Binding Proteins/metabolism , Receptors, Muscarinic/metabolism , Receptors, Purinergic P2/metabolism , Adenoviridae , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA, Antisense/pharmacology , GTP-Binding Proteins/drug effects , HT29 Cells/drug effects , Humans , Receptor, Muscarinic M3 , Receptors, Muscarinic/drug effects , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2Y2 , Uridine Triphosphate/pharmacology , beta-Adrenergic Receptor Kinases
18.
J Biol Chem ; 275(37): 29107-12, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10869366

ABSTRACT

The distribution of P2X receptors on neurons in rat superior cervical ganglia and lability of P2X receptors on exposure to agonists were determined. Antibody labeling of each P2X subtype P2X(1)-P2X(7) showed neurons isolated into culture possessed primarily P2X(2) subunits with others occurring in order P2X(7) > P2X(6) > P2X(3) > P2X(1) > P2X(5) > P2X(4). Application of ATP and alpha,beta-meATP to neurons showed they possessed a predominantly nondesensitizing P2X receptor type insensitive to alpha,beta-meATP, consistent with immunohistochemical observations. P2X(1)-green fluorescent protein (GFP) was used to study the time course of P2X(1) receptor clustering in plasma membranes of neurons and internalization of receptors following prolonged exposure to ATP. At 12-24 h after adenoviral infection, P2X(1)-GFP formed clusters about 1 microm diameter in the neuron membrane. Application of ATP and alpha,beta-meATP showed these neurons possessed a predominantly desensitizing P2X receptor type sensitive to alpha,beta-meATP. Infection converted the major functional P2X receptor type in the membrane to P2X(1). Exposure of infected neurons to alpha,beta-meATP for less than 60 s led to the disappearance of P2X(1)-GFP fluorescence from the cell surface that was blocked by monensin, indicating the chimera is normally endocytosed into these organelles on exposure to agonist.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Receptors, Purinergic P2/analysis , Superior Cervical Ganglion/chemistry , Adenosine Triphosphate/pharmacology , Animals , Cells, Cultured , Purinergic P2 Receptor Agonists , Rabbits , Rats , Receptors, Purinergic P2/physiology , Superior Cervical Ganglion/virology
19.
FEBS Lett ; 459(3): 443-7, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10526181

ABSTRACT

Regulation of amiloride-sensitive epithelial Na(+) channels (ENaC) is a prerequisite for coordination of electrolyte transport in epithelia. Downregulation of Na(+) conductance occurs when the intracellular Na(+) concentration is increased during reabsorption of electrolytes, known as feedback inhibition. Recent studies have demonstrated the involvement of alphaG(0) and alphaG(i2) proteins in the feedback control of ENaC in mouse salivary duct cells. In this report, we demonstrate that Na(+) feedback inhibition is also present in Xenopus oocytes after expression of rat alpha,beta, gamma-ENaC. Interfering with intracellular alphaG(0) or alphaG(i2) signaling by coexpression of either constitutively active alphaG(0)/alphaG(i2) or dominant negative alphaG(0)/alphaG(i2) and by coinjecting sense or antisense oligonucleotides for alphaG(0) had no impact on Na(+) feedback. Moreover, no evidence for involvement of the intracellular G protein cascade was found in experiments in which a regulator of G protein signaling (RGS3) or beta-adrenergic receptor kinase (betaARK) was coexpressed together with alpha,beta, gamma-ENaC. Although some experiments suggest the presence of an intracellular Na(+) receptor, we may conclude that Na(+) feedback in Xenopus oocytes is different from that described for salivary duct cells in that it does not require G protein signaling.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Oocytes/metabolism , Proto-Oncogene Proteins/metabolism , Sodium Channels/metabolism , Animals , Epithelium/metabolism , Feedback , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go , Mice , Rats , Sodium/metabolism , Xenopus laevis
20.
Electrophoresis ; 20(10): 2065-70, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10451116

ABSTRACT

Human salivary gland epithelial cells, a continuous cell line derived from an irradiated human salivary gland and human embryonic kidney cell line human embryonic kidney (HEK)293 were examined for the purpose of establishing whether they expressed endogenous P2X ionotropic receptors at any stage in their cycles. HSG cells were found to express P2X1-6 subtypes using both Western blotting and immunofluorescence labeling. HEK293 cells had no detectable levels of P2X1-3 and P2X6 under normal circumstances along with very low levels of P2X4 and P2X5 but when the cells were grown past confluence then all subtypes were expressed on the surface membrane with the exception of P2X2. The results are discussed in terms of the likely influence of ATP acting as an intercellular signaling molecule.


Subject(s)
Blotting, Western , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Kidney/chemistry , Kidney/embryology , Receptors, Purinergic P2/analysis , Salivary Glands/chemistry , Antibody Specificity , Cell Line , Epithelial Cells/chemistry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...