Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
J Cell Mol Med ; 17(12): 1598-607, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24238063

ABSTRACT

Podocytes are highly differentiated glomerular epithelial cells that contribute to the glomerular barrier function of kidney. A role for autophagy has been proposed in maintenance of their cellular integrity, but the mechanisms controlling autophagy in podocytes are not clear. The present study tested whether CD38-mediated regulation of lysosome function contributes to autophagic flux or autophagy maturation in podocytes. Podocytes were found to exhibit a high constitutive level of LC3-II, a robust marker of autophagosomes (APs), suggesting a high basal level of autophagic activity. Treatment with the mTOR inhibitor, rapamycin, increased LC3-II and the content of both APs detected by Cyto-ID Green staining and autophagolysosomes (APLs) measured by acridine orange staining and colocalization of LC3 and Lamp1. Lysosome function inhibitor bafilomycin A1 increased APs, but decreased APLs content under both basal and rapamycin-induced conditions. Inhibition of CD38 activity by nicotinamide or silencing of CD38 gene produced the similar effects to that bafilomycin A1 did in podocytes. To explore the possibility that CD38 may control podocyte autophagy through its regulation of lysosome function, the fusion of APs with lysosomes in living podocytes was observed by co-transfection of GFP-LC3B and RFP-Lamp1 expression vectors. A colocalization of GFP-LC3B and RFP-Lamp1 upon stimulation of rapamycin became obvious in transfected podocytes, which could be substantially blocked by nicotinamide, CD38 shRNA, and bafilomycin. Moreover, blockade of the CD38-mediated regulation by PPADS completely abolished rapamycin-induced fusion of APs with lysosomes. These results indicate that CD38 importantly control lysosomal function and influence autophagy at the maturation step in podocytes.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Autophagy , Kidney Glomerulus/cytology , Lysosomes/metabolism , Podocytes/cytology , Podocytes/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , Autophagy/drug effects , Calcium Signaling/drug effects , Gene Silencing/drug effects , Lysosomes/drug effects , Membrane Fusion/drug effects , Mice , NADP/analogs & derivatives , NADP/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Podocytes/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Sirolimus/pharmacology , Transcription Factor TFIIH , Transcription Factors/metabolism , Ubiquitinated Proteins/metabolism
2.
Cell Physiol Biochem ; 31(6): 925-37, 2013.
Article in English | MEDLINE | ID: mdl-23817226

ABSTRACT

BACKGROUND/AIMS: In addition to their action of lowering blood cholesterol levels, statins modulate biological characteristics and functions of arterial myocytes such as viability, proliferation, apoptosis, survival and contraction. The present study tested whether simvastatin, as a prototype statin, enhances autophagy in coronary arterial myocytes (CAMs) to thereby exert their beneficial effects in atherosclerosis. METHODS AND RESULTS: Using flow cytometry, we demonstrated that simvastatin significantly increased the autophagsome formation in CAMs. Western blot analysis confirmed that simvastatin significantly increased protein expression of typical autophagy markers LC3B and Beclin1 in these CAMs. Confocal microscopy further demonstrated that simvastatin increased fusion of autophagosomes with lysosomes, which was blocked by autophagy inhibitor 3-methyladenine or silencing of Atg7 genes. Simvastatin reduced mammalian target of rapamycin (mTOR) activity, which was reversed by Rac1-GTPase overexpression and the mTOR agonist phosphatidic acid. Moreover, both Rac1-GTPase overexpression and activation of mTOR by phosphatidic acid drastically blocked simvastatin-induced autophagosome formation in CAMs. Interestingly, simvastatin increased protein expression of a contractile phenotype marker calponin in CAMs, which was blocked by autophagy inhibitor 3-methyladenine. Simvastatin markedly reduced proliferation of CAMs under both control and proatherogenic stimulation. However, this inhibitory effect of simvastatin on CAM proliferation was blocked by by autophagy inhibitor 3-methyladenine or silencing of Atg7 genes. Lastly, animal experiments demonstrated that simvastatin increased protein expression of LC3B and calponin in mouse coronary arteries. CONCLUSION: Our results indicate that simvastatin inhibits the Rac1-mTOR pathway and thereby increases autophagy in CAMs which may stabilize CAMs in the contractile phenotype to prevent proliferation and growth of these cells.


Subject(s)
Autophagy/drug effects , Coronary Vessels/cytology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Muscle Cells/drug effects , Simvastatin/pharmacology , TOR Serine-Threonine Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy-Related Protein 7 , Calcium-Binding Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Muscle Cells/cytology , Muscle Cells/metabolism , Phenotype , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , rac1 GTP-Binding Protein/antagonists & inhibitors , Calponins
SELECTION OF CITATIONS
SEARCH DETAIL
...