Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 11(12): e0166948, 2016.
Article in English | MEDLINE | ID: mdl-27935997

ABSTRACT

Many germ line diseases stem from a relatively minor disturbance in mutant protein endoplasmic reticulum (ER) 3D assembly. Chaperones are recruited which, on failure to correct folding, sort the mutant for retrotranslocation and cytosolic proteasomal degradation (ER-associated degradation-ERAD), to initiate/exacerbate deficiency-disease symptoms. Several bacterial (and plant) subunit toxins, retrograde transport to the ER after initial cell surface receptor binding/internalization. The A subunit has evolved to mimic a misfolded protein and hijack the ERAD membrane translocon (dislocon), to effect cytosolic access and cytopathology. We show such toxins compete for ERAD to rescue endogenous misfolded proteins. Cholera toxin or verotoxin (Shiga toxin) containing genetically inactivated (± an N-terminal polyleucine tail) A subunit can, within 2-4 hrs, temporarily increase F508delCFTR protein, the major cystic fibrosis (CF) mutant (5-10x), F508delCFTR Golgi maturation (<10x), cell surface expression (20x) and chloride transport (2x) in F508del CFTR transfected cells and patient-derived F508delCFTR bronchiolar epithelia, without apparent cytopathology. These toxoids also increase glucocerobrosidase (GCC) in N370SGCC Gaucher Disease fibroblasts (3x), another ERAD-exacerbated misfiling disease. We identify a new, potentially benign approach to the treatment of certain genetic protein misfolding diseases.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/drug effects , Endoplasmic Reticulum/drug effects , Protein Folding/drug effects , Toxins, Biological/pharmacology , Blotting, Western , Cholera Toxin/pharmacology , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/prevention & control , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Microscopy, Fluorescence , Models, Biological , Mutation , Protein Transport/drug effects , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/prevention & control , Shiga Toxins/pharmacology , Toxins, Biological/classification
2.
PLoS One ; 7(12): e51739, 2012.
Article in English | MEDLINE | ID: mdl-23251616

ABSTRACT

The colonic epithelium continuously regenerates with transitions through various cellular phases including proliferation, differentiation and cell death via apoptosis. Human colonic adenocarcinoma (Caco-2) cells in culture undergo spontaneous differentiation into mature enterocytes in association with progressive increases in expression of glutathione S-transferase alpha-1 (GSTA1). We hypothesize that GSTA1 plays a functional role in controlling proliferation, differentiation and apoptosis in Caco-2 cells. We demonstrate increased GSTA1 levels associated with decreased proliferation and increased expression of differentiation markers alkaline phosphatase, villin, dipeptidyl peptidase-4 and E-cadherin in postconfluent Caco-2 cells. Results of MTS assays, BrdU incorporation and flow cytometry indicate that forced expression of GSTA1 significantly reduces cellular proliferation and siRNA-mediated down-regulation of GSTA1 significantly increases cells in S-phase and associated cell proliferation. Sodium butyrate (NaB) at a concentration of 1 mM reduces Caco-2 cell proliferation, increases differentiation and increases GSTA1 activity 4-fold by 72 hours. In contrast, 10 mM NaB causes significant toxicity in preconfluent cells via apoptosis through caspase-3 activation with reduced GSTA1 activity. However, GSTA1 down-regulation by siRNA does not alter NaB-induced differentiation or apoptosis in Caco-2 cells. While 10 mM NaB causes GSTA1-JNK complex dissociation, phosphorylation of JNK is not altered. These findings suggest that GSTA1 levels may play a role in modulating enterocyte proliferation but do not influence differentiation or apoptosis.


Subject(s)
Glutathione Transferase/pharmacology , Alkaline Phosphatase/metabolism , Apoptosis/drug effects , Butyrates/pharmacology , Caco-2 Cells , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Enzyme Activation/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , S Phase/drug effects , Transfection
3.
Toxicol Lett ; 214(1): 53-62, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22906494

ABSTRACT

Glutathione S-transferases (GSTs) act as modulators of mitogen-activated protein kinase signal transduction pathways via a mechanism involving protein-protein interactions. We have demonstrated that GSTA1 forms complexes with JNK and modifies JNK activation during cellular stress, but the factors that influence complex association and dissociation are unknown. We hypothesized that menadione causes dissociation of GSTA1-JNK complexes, activates JNK, and the consequences of menadione exposure depend on GSTA1 expression. We demonstrate that menadione causes GSTA1-JNK dissociation and JNK activation in preconfluent Caco-2 cells, whereas postconfluent cells are resistant to this effect. Moreover, preconfluent cells are more sensitive than postconfluent cells to menadione-induced cytotoxicity. Activation of JNK is transient since removal of menadione causes GSTA1 to re-associate with JNK reducing cytotoxicity. Over-expression and knockdown of GSTA1 did not alter JNK activation by menadione or sensitivity to menadione-induced cytotoxicity. These results indicate that GSTA1-JNK complex integrity does not affect the ability of menadione to activate JNK. N-acetyl cysteine prevents GSH depletion and blocks menadione-induced complex dissociation, JNK activation and inhibits menadione-induced cytotoxicity. JNK activation and inhibits menadione-induced cytotoxicity. The data suggest that the mechanism of menadione-induced JNK activation involves the production of reactive oxygen species, likely superoxide anion, and intracellular GSH levels play an important role in preventing GSTA1-JNK complex dissociation, subsequent JNK activation and induction of cytotoxicity.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Glutathione Transferase/metabolism , MAP Kinase Kinase 4/metabolism , Vitamin K 3/toxicity , Acetylcysteine/pharmacology , Aldehydes/metabolism , Antifibrinolytic Agents/pharmacology , Caco-2 Cells , Dose-Response Relationship, Drug , Down-Regulation , Glutathione/metabolism , Glutathione Transferase/genetics , Humans , Lipid Peroxidation , MAP Kinase Kinase 4/genetics , Malondialdehyde/metabolism , Oxidative Stress , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...