Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres










Base de données
Gamme d'année
5.
Cell Metab ; 32(4): 561-574.e7, 2020 10 06.
Article de Anglais | MEDLINE | ID: mdl-33027675

RÉSUMÉ

Aberrant redox signaling underlies the pathophysiology of many chronic metabolic diseases, including type 2 diabetes (T2D). Methodologies aimed at rebalancing systemic redox homeostasis have had limited success. A noninvasive, sustained approach would enable the long-term control of redox signaling for the treatment of T2D. We report that static magnetic and electric fields (sBE) noninvasively modulate the systemic GSH-to-GSSG redox couple to promote a healthier systemic redox environment that is reducing. Strikingly, when applied to mouse models of T2D, sBE rapidly ameliorates insulin resistance and glucose intolerance in as few as 3 days with no observed adverse effects. Scavenging paramagnetic byproducts of oxygen metabolism with SOD2 in hepatic mitochondria fully abolishes these insulin sensitizing effects, demonstrating that mitochondrial superoxide mediates induction of these therapeutic changes. Our findings introduce a remarkable redox-modulating phenomenon that exploits endogenous electromagneto-receptive mechanisms for the noninvasive treatment of T2D, and potentially other redox-related diseases.


Sujet(s)
Diabète de type 2/thérapie , Champs électromagnétiques/effets indésirables , Animaux , Diabète de type 2/sang , Diabète de type 2/métabolisme , Femelle , Homéostasie , Humains , Mâle , Souris , Souris de lignée C57BL , Espèces réactives de l'oxygène/métabolisme , Cellules cancéreuses en culture
6.
Redox Biol ; 32: 101518, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32278283

RÉSUMÉ

Selenium is a metalloid trace element essential for maintaining the optimal redox environment in cells and tissues. It is structurally incorporated into over 25 selenoproteins and enzymes. The glutathione peroxidase (GPx) family of enzymes has a critical role in human health because of its antioxidant function. The recommended daily allowance (RDA) for selenium intake in humans was established to maximize the activity of GPx in plasma. Suboptimal availability of selenium can limit the expression and activities of GPxs leading to a compromised redox environment. This can cause detrimental oxidative distress that could be prevented by increasing the availability of selenium. In cell culture studies, the medium is typically deficient in selenium; supplementation with selenium can increase selenoenzyme activities. However, the optimal level of supplementation in cell culture media has not been well characterized. We performed dose-response experiments for the activities of GPx1 and GPx4 vs. the level of selenium supplementation in cell culture medium. For this, we advanced an assay to determine the activities of both GPx1 and GPx4 efficiently in a single run. During the optimization process, we found that the observed activities of GPx1 and GPx4 depend greatly on the pH of the assay buffer; the observed activities increase with increasing pH, with pH 8 being optimal. Using the combination assay, we also found that the expression and activities for both GPx1 and GPx4 can be maximized in exponentially growing cells by supplementing cell culture media with ≈ 200 nM seleno-l-methionine, without concerns for toxicity. Optimizing the availability of selenium in cell culture to maximize the expression and activities GPx1 and GPx4 may allow for better translation of information from cell culture work to in vivo settings.


Sujet(s)
Sélénium , Glutathione peroxidase/génétique , Humains , Phospholipid hydroperoxide glutathione peroxidase , ARN messager , Sélénoprotéines , Glutathione Peroxydase GPX1
7.
Cancer Res ; 80(7): 1401-1413, 2020 04 01.
Article de Anglais | MEDLINE | ID: mdl-32041838

RÉSUMÉ

Pharmacologic ascorbate treatment (P-AscH-, high-dose, intravenous vitamin C) results in a transient short-term increase in the flux of hydrogen peroxide that is preferentially cytotoxic to cancer cells versus normal cells. This study examines whether an increase in hydrogen peroxide is sustained posttreatment and potential mechanisms involved in this process. Cellular bioenergetic profiling following treatment with P-AscH- was examined in tumorigenic and nontumorigenic cells. P-AscH- resulted in sustained increases in the rate of cellular oxygen consumption (OCR) and reactive oxygen species (ROS) in tumor cells, with no changes in nontumorigenic cells. Sources for this increase in ROS and OCR were DUOX 1 and 2, which are silenced in pancreatic ductal adenocarcinoma, but upregulated with P-AscH- treatment. An inducible catalase system, to test causality for the role of hydrogen peroxide, reversed the P-AscH--induced increases in DUOX, whereas DUOX inhibition partially rescued P-AscH--induced toxicity. In addition, DUOX was significantly downregulated in pancreatic cancer specimens compared with normal pancreas tissues. Together, these results suggest that P-AscH--induced toxicity may be enhanced by late metabolic shifts in tumor cells, resulting in a feed-forward mechanism for generation of hydrogen peroxide and induction of metabolic stress through enhanced DUOX expression and rate of oxygen consumption. SIGNIFICANCE: A high dose of vitamin C, in addition to delivering an acute exposure of H2O2 to tumor cells, activates DUOX in pancreatic cancer cells, which provide sustained production of H2O2.


Sujet(s)
Acide ascorbique/pharmacologie , Carcinome du canal pancréatique/thérapie , Dual oxydases/métabolisme , Peroxyde d'hydrogène/métabolisme , Tumeurs du pancréas/thérapie , Administration par voie intraveineuse , Animaux , Acide ascorbique/usage thérapeutique , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Lignée cellulaire tumorale , Traitement médicamenteux adjuvant/méthodes , Relation dose-effet des médicaments , Régulation négative/génétique , Dual oxydases/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Oxygène/métabolisme , Consommation d'oxygène/effets des médicaments et des substances chimiques , Pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Duodénopancréatectomie , Espèces réactives de l'oxygène/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Free Radic Biol Med ; 150: 1-11, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32032663

RÉSUMÉ

Therapies for lung cancer patients initially elicit desirable responses, but the presence of hypoxia and drug resistant cells within tumors ultimately lead to treatment failure. Disulfiram (DSF) is an FDA approved, copper chelating agent that can target oxidative metabolic frailties in cancer vs. normal cells and be repurposed as an adjuvant to cancer therapy. Clonogenic survival assays showed that DSF (50-150 nM) combined with physiological levels of Cu (15 µM CuSO4) was selectively toxic to H292 NSCLC cells vs. normal human bronchial epithelial cells (HBEC). Furthermore, cancer cell toxicity was exacerbated at 1% O2, relative to 4 or 21% O2. This selective toxicity of DSF/Cu was associated with differential Cu ionophore capabilities. DSF/Cu treatment caused a >20-fold increase in cellular Cu in NSCLCs, with nearly two-fold higher Cu present in NSCLCs vs. HBECs and in cancer cells at 1% O2vs. 21% O2. DSF toxicity was shown to be dependent on the retention of Cu as well as oxidative stress mechanisms, including the production of superoxide, peroxide, lipid peroxidation, and mitochondrial damage. DSF was also shown to selectively (relative to HBECs) enhance radiation and chemotherapy-induced NSCLC killing and reduce radiation and chemotherapy resistance in hypoxia. Finally, DSF decreased xenograft tumor growth in vivo when combined with radiation and carboplatin. These results support the hypothesis that DSF could be a promising adjuvant to enhance cancer therapy based on its apparent ability to selectively target fundamental differences in cancer cell oxidative metabolism.


Sujet(s)
Disulfirame , Tumeurs du poumon , Lignée cellulaire tumorale , Cuivre , Disulfirame/pharmacologie , Humains , Hypoxie , Tumeurs du poumon/traitement médicamenteux , Oxydoréduction
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...