Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cells ; 9(10)2020 10 15.
Article in English | MEDLINE | ID: mdl-33076330

ABSTRACT

The administration of adipose tissue-derived mesenchymal stem cells (ADMSCs) represents a promising therapeutic option after myocardial ischemia or myocardial infarction. However, their potential is reduced due to the high post-transplant cell mortality probably caused by oxidative stress and mitogen-deficient microenvironments. To identify protection strategies for ADMSCs, this study investigated the influence of the non-psychoactive phytocannabinoid cannabidiol (CBD) and the endocannabinoid analogue R(+)-methanandamide (MA) on the induction of heme oxygenase-1 (HO-1) and autophagy under serum-free conditions. At a concentration of 3 µM, CBD induced an upregulation of HO-1 mRNA and protein within 6 h, whereas for MA only a late and comparatively lower increase in the HO-1 protein could be detected after 48 h. In addition, both cannabinoids induced time- and concentration-dependent increases in LC3A/B-II protein, a marker of autophagy, and in metabolic activity. A participation of several cannabinoid-binding receptors in the effect on metabolic activity and HO-1 was excluded. Similarly, knockdown of HO-1 by siRNA or inhibition of HO-1 activity by tin protoporphyrin IX (SnPPIX) had no effect on CBD-induced autophagy and metabolic activity. On the other hand, the inhibition of autophagy by bafilomycin A1 led to a significant decrease in cannabinoid-induced metabolic activity and to an increase in apoptosis. Under these circumstances, a significant induction of HO-1 expression after 24 h could also be demonstrated for MA. Remarkably, inhibition of HO-1 by SnPPIX under conditions of autophagy deficit led to a significant reversal of apoptosis in cannabinoid-treated cells. In conclusion, the investigated cannabinoids increase metabolic viability of ADMSCs under serum-free conditions by inducing HO-1-independent autophagy but contribute to apoptosis under conditions of additional autophagy deficit via an HO-1-dependent pathway.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cannabinoids/pharmacology , Heme Oxygenase-1/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Arachidonic Acids/pharmacology , Cannabidiol/pharmacology , Caspase 3/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Macrolides/pharmacology , Metalloporphyrins/pharmacology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Oxidative Stress , Protoporphyrins/pharmacology , RNA, Small Interfering/genetics , Receptors, Cannabinoid/physiology , Reverse Transcriptase Polymerase Chain Reaction
2.
Cells ; 9(7)2020 07 16.
Article in English | MEDLINE | ID: mdl-32708634

ABSTRACT

Cannabidiol (CBD), a non-psychoactive cannabinoid, has been reported to mediate antioxidant, anti-inflammatory, and anti-angiogenic effects in endothelial cells. This study investigated the influence of CBD on the expression of heme oxygenase-1 (HO-1) and its functional role in regulating metabolic, autophagic, and apoptotic processes of human umbilical vein endothelial cells (HUVEC). Concentrations up to 10 µM CBD showed a concentration-dependent increase of HO-1 mRNA and protein and an increase of the HO-1-regulating transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). CBD-induced HO-1 expression was not decreased by antagonists of cannabinoid-activated receptors (CB1, CB2, transient receptor potential vanilloid 1), but by the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). The incubation of HUVEC with 6 µM CBD resulted in increased metabolic activity, while 10 µM CBD caused decreased metabolic activity and an induction of apoptosis, as demonstrated by enhanced caspase-3 cleavage. In addition, CBD triggered a concentration-dependent increase of the autophagy marker LC3A/B-II. Both CBD-induced LC3A/B-II levels and caspase-3 cleavage were reduced by NAC. The inhibition of autophagy by bafilomycin A1 led to apoptosis induction by 6 µM CBD and a further increase of the proapoptotic effect of 10 µM CBD. On the other hand, the inhibition of HO-1 activity with tin protoporphyrin IX (SnPPIX) or knockdown of HO-1 expression by Nrf2 siRNA was associated with a decrease in CBD-mediated autophagy and apoptosis. In summary, our data show for the first time ROS-mediated HO-1 expression in endothelial cells as a mechanism by which CBD mediates protective autophagy, which at higher CBD concentrations, however, can no longer prevent cell death inducing apoptosis.


Subject(s)
Autophagy , Cannabidiol/pharmacology , Heme Oxygenase-1/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/enzymology , Apoptosis/drug effects , Autophagy/drug effects , Cell Survival/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Models, Biological , NF-E2-Related Factor 2/metabolism , Protoporphyrins/pharmacology , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Time Factors
3.
Oncotarget ; 9(77): 34595-34616, 2018 Oct 02.
Article in English | MEDLINE | ID: mdl-30349652

ABSTRACT

Aberrant proliferation and migration of vascular smooth muscle cells (VSMC) have been closely linked to the development and progression of cardiovascular and cancer diseases. The cytoprotective enzyme heme oxygenase-1 (HO-1) has been shown to mediate anti-proliferative and anti-migratory effects in VSMC. This study investigates the effect of cannabidiol (CBD), a non-psychoactive cannabinoid, on HO-1 expression and disease-associated functions of human umbilical artery smooth muscle cells (HUASMC). HO-1 protein and mRNA were significantly increased by CBD in a time- and concentration-dependent manner. Although the expression of several cannabinoid-activated receptors (CB1, CB2, G protein-coupled receptor 55, transient receptor potential vanilloid 1) was verified in HUASMC, CBD was shown to induce HO-1 via none of these targets. Instead, the CBD-mediated increase in HO-1 protein was reversed by the glutathione precursor N-acetylcysteine, indicating the participation of reactive oxygen species (ROS) signaling; this was confirmed by flow cytometry-based ROS detection. CBD-induced HO-1 expression was accompanied by inhibition of growth factor-mediated proliferation and migration of HUASMC. However, neither inhibition of HO-1 activity nor knockdown of HO-1 protein attenuated CBD-mediated anti-proliferative and anti-migratory effects. Indeed, inhibition or depletion of HO-1 resulted in induction of apoptosis and intensified CBD-mediated effects on proliferation and migration. Collectively, this work provides the first indication of CBD-mediated enhancement of HO-1 in VSMC and potential protective effects against aberrant VSMC proliferation and migration. On the other hand, our data argue against a role of HO-1 in CBD-mediated inhibition of proliferation and migration while substantiating its anti-apoptotic role in oxidative stress-mediated cell fate.

4.
Oncotarget ; 9(33): 23149-23163, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29796178

ABSTRACT

Activation of the p38 mitogen-activated protein kinase (MAPK) pathway has been implicated in various detrimental events finally leading to endothelial dysfunction. The present study therefore investigates the impact of the p38 MAPK inhibitor SB202190 on the expression of the cytoprotective enzyme heme oxygenase-1 (HO-1) as well as metabolic activity, apoptosis and autophagy of endothelial cells. Using human umbilical vein endothelial cells (HUVEC) SB202190 was found to cause a time- and concentration-dependent induction of HO-1 protein. Induction of HO-1 protein expression was mimicked by SB203580, another p38 MAPK inhibitor, but not by SB202474, an inactive structural analogue of p38 MAPK inhibitors. HO-1 induction by both SB202190 and SB203580 was also demonstrated by analysis of mRNA expression. On the functional level, SB202190 was shown to increase metabolic activity and autophagy of HUVEC along with diminishing basal apoptosis. Treatment of cells with tin protoporphyrin IX (SnPPIX), a well-characterised HO-1 enzymatic inhibitor, or HO-1 siRNA left SB202190-modulated metabolic activity and autophagy virtually unaltered but caused a significant reversal of the anti-apoptotic action of SB202190. Conversely, however, HO-1 expression by SB202190 became completely suppressed by the autophagy inhibitor bafilomycin A1. Bafilomycin A1 likewise fully reversed effects of SB202190 on metabolic activity and apoptosis, albeit significantly inducing apoptosis per se. Collectively, this work demonstrates SB202190 to confer upstream induction of autophagy followed by HO-1 induction resulting in potential protective effects against apoptosis. On the other hand, our data oppose HO-1 to contribute to SB202190-mediated increases in metabolic activity and autophagy, respectively.

5.
Pancreas ; 28(1): 31-7, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14707727

ABSTRACT

INTRODUCTION: Cholecystokinin (CCK) is a peptide hormone and plays a major role both in the regulation of pancreatic enzyme secretion and growth of the gastrointestinal tract. The pancreatic CCK receptors are highly glycosylated membrane proteins that are able to bind plant lectins such as wheat germ agglutinin (WGA) and Ulex europaeus agglutinin (UEA-I). AIM AND METHODOLOGY: In preceding papers, we demonstrated an inhibition of CCK-8s induced Ca2+ signaling and secretion of rat pancreatic acini and AR42J cells by the lectins WGA and UEA-I (Pancreas 2001;23:368-374). Here we studied the influence of WGA, UEA-I, and 22 other lectins on 125I-CCK-8s binding on AR42J cells. A binding assay was used with 125I-CCK-8s and dexamethasone-stimulated AR42J cells, bearing CCK-A as well as CCK-B receptors. RESULTS: WGA inhibits 125I-CCK-8s binding in a dose-dependent manner. The binding is affected at concentrations of WGA >1 microg/mL. The EC50 for inhibition is 8 microg/mL. At a concentration of 25 microg/mL, WGA inhibits the hormone binding 70%. This inhibition can be abolished by the specific sugars for WGA N,N',N"-triacetylchitotriose and N-acetylglucosamine, but not by N-acetylneuraminic acid. UEA-I diminished hormone binding but without significance, although UEA-I binds to the fucose residues of receptor glycosylations. All other 22 lectins tested here were ineffective. CONCLUSION: The blockage of CCK receptors by WGA explains the inhibition of CCK-8s induced Ca2+ signaling and the secretion of pancreatic acinar cells and AR42J cells. Although the inhibitory effect of WGA is in agreement with the findings of Santer et al, the results with UEA-I are in contrast to those of Santer et al (1990), who described a strong increase in 125I-CCK-8s binding to isolated crude rat pancreatic cell membranes in the presence of UEA-I.


Subject(s)
Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/metabolism , Sincalide/metabolism , Wheat Germ Agglutinins/pharmacology , Acetylglucosamine/pharmacology , Animals , Benzodiazepinones/pharmacology , Binding, Competitive/drug effects , Cell Line, Tumor , Devazepide/pharmacology , Iodine Radioisotopes , Phenylurea Compounds/pharmacology , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin B/antagonists & inhibitors , Trisaccharides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...