417 results on '"Garry R Buettner"'
Search Results
2. Peroxiporin Expression Is an Important Factor for Cancer Cell Susceptibility to Therapeutic H2O2: Implications for Pharmacological Ascorbate Therapy.
- Author
-
Dieanira Erudaitius, Andrew Huang, Sarah Kazmi, Garry R Buettner, and Victor G J Rodgers
- Subjects
Medicine ,Science - Abstract
Cancer cell toxicity to therapeutic H2O2 varies widely depending on cell type. Interestingly, it has been observed that different cancer cell types have varying peroxiporin expression. We hypothesize that variation in peroxiporin expression can alter cell susceptibility to therapeutic H2O2 concentrations. Here, we silence peroxiporin aquaporin-3 (AQP3) on the pancreatic cancer cell line MIA PaCa-2 and compare clonogenic survival response to the wild-type. The results showed a significantly higher surviving fraction in the clonogenic response for siAQP3 MIA PaCa-2 cells at therapeutic H2O2 doses (P < 0.05). These results suggest that peroxiporin expression is significant in modulating the susceptibility of cancer cells to ascorbate therapy.
- Published
- 2017
- Full Text
- View/download PDF
3. Moles of a Substance per Cell Is a Highly Informative Dosing Metric in Cell Culture.
- Author
-
Claire M Doskey, Thomas J van 't Erve, Brett A Wagner, and Garry R Buettner
- Subjects
Medicine ,Science - Abstract
The biological consequences upon exposure of cells in culture to a dose of xenobiotic are not only dependent on biological variables, but also the physical aspects of experiments e.g. cell number and media volume. Dependence on physical aspects is often overlooked due to the unrecognized ambiguity in the dominant metric used to express exposure, i.e. initial concentration of xenobiotic delivered to the culture medium over the cells. We hypothesize that for many xenobiotics, specifying dose as moles per cell will reduce this ambiguity. Dose as moles per cell can also provide additional information not easily obtainable with traditional dosing metrics.Here, 1,4-benzoquinone and oligomycin A are used as model compounds to investigate moles per cell as an informative dosing metric. Mechanistic insight into reactions with intracellular molecules, differences between sequential and bolus addition of xenobiotic and the influence of cell volume and protein content on toxicity are also investigated.When the dose of 1,4-benzoquinone or oligomycin A was specified as moles per cell, toxicity was independent of the physical conditions used (number of cells, volume of medium). When using moles per cell as a dose-metric, direct quantitative comparisons can be made between biochemical or biological endpoints and the dose of xenobiotic applied. For example, the toxicity of 1,4-benzoquinone correlated inversely with intracellular volume for all five cell lines exposed (C6, MDA-MB231, A549, MIA PaCa-2, and HepG2).Moles per cell is a useful and informative dosing metric in cell culture. This dosing metric is a scalable parameter that: can reduce ambiguity between experiments having different physical conditions; provides additional mechanistic information; allows direct comparison between different cells; affords a more uniform platform for experimental design; addresses the important issue of repeatability of experimental results, and could increase the translatability of information gained from in vitro experiments.
- Published
- 2015
- Full Text
- View/download PDF
4. A randomized trial of pharmacological ascorbate, gemcitabine, and nab-paclitaxel for metastatic pancreatic cancer
- Author
-
Kellie L. Bodeker, Brian J. Smith, Daniel J. Berg, Chandrikha Chandrasekharan, Saima Sharif, Naomi Fei, Sandy Vollstedt, Heather Brown, Meghan Chandler, Amanda Lorack, Stacy McMichael, Jared Wulfekuhle, Brett A. Wagner, Garry R. Buettner, Bryan G. Allen, Joseph M. Caster, Barbara Dion, Mandana Kamgar, John M. Buatti, and Joseph J. Cullen
- Subjects
Pancreatic neoplasms ,Ascorbic acid ,Controlled clinical trial ,Gemcitabine ,Nab-paclitaxel ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Background: Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have poor 5-year survival. Pharmacological ascorbate (P-AscH-, high dose, intravenous, vitamin C) has shown promise as an adjunct to chemotherapy. We hypothesized adding P-AscH- to gemcitabine and nab-paclitaxel would increase survival in patients with metastatic PDAC. Methods: Patients diagnosed with stage IV pancreatic cancer randomized 1:1 to gemcitabine and nab-paclitaxel only (SOC, control) or to SOC with concomitant P-AscH−, 75 g three times weekly (ASC, investigational). The primary outcome was overall survival with secondary objectives of determining progression-free survival and adverse event incidence. Quality of life and patient reported outcomes for common oncologic symptoms were captured as an exploratory objective. Thirty-six participants were randomized; of this 34 received their assigned study treatment. All analyses were based on data frozen on December 11, 2023. Results: Intravenous P-AscH- increased serum ascorbate levels from micromolar to millimolar levels. P-AscH- added to the gemcitabine + nab-paclitaxel (ASC) increased overall survival to 16 months compared to 8.3 months with gemcitabine + nab-paclitaxel (SOC) (HR = 0.46; 90 % CI 0.23, 0.92; p = 0.030). Median progression free survival was 6.2 (ASC) vs. 3.9 months (SOC) (HR = 0.43; 90 % CI 0.20, 0.92; p = 0.029). Adding P-AscH- did not negatively impact quality of life or increase the frequency or severity of adverse events. Conclusions: P-AscH− infusions of 75 g three times weekly in patients with metastatic pancreatic cancer prolongs overall and progression free survival without detriment to quality of life or added toxicity (ClinicalTrials.gov number NCT02905578).
- Published
- 2024
- Full Text
- View/download PDF
5. A reciprocal relationship between mitochondria and lipid peroxidation determines the chondrocyte intracellular redox environment
- Author
-
Madeline R. Hines, Piedad C. Gomez-Contreras, Suryamin Liman, Alexandria M. Wilson, Kevin J. Lu, Jaycie A. O'Neill, Jacob S. Fisher, Douglas C. Fredericks, Brett A. Wagner, Garry R. Buettner, Holly Van Remmen, and Mitchell C. Coleman
- Subjects
Lipid peroxidation ,Glutathione peroxidase 4 ,Mitochondria ,Cartilage ,Chondrocyte ,Immuno-spin trapping ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
In orthopedic research, many studies have applied vitamin E as a protective antioxidant or used tert-butyl hydroperoxide to induce oxidative injury to chondrocytes. These studies often support the hypothesis that joint pathology causes oxidative stress and increased lipid peroxidation that might be prevented with lipid antioxidants to improve cell survival or function and joint health; however, lipid antioxidant supplementation was ineffective against osteoarthritis in clinical trials and animal data have been equivocal. Moreover, increased circulating vitamin E is associated with increased rates of osteoarthritis. This disconnect between benchtop and clinical results led us to hypothesize that oxidative stress-driven paradigms of chondrocyte redox function do not capture the metabolic and physiologic effects of lipid antioxidants and prooxidants on articular chondrocytes. We used ex vivo and in vivo cartilage models to investigate the effect of lipid antioxidants on healthy, primary, articular chondrocytes and applied immuno-spin trapping techniques to provide a broad indicator of high levels of oxidative stress independent of specific reactive oxygen species. Key findings demonstrate lipid antioxidants were pro-mitochondrial while lipid prooxidants decreased mitochondrial measures. In the absence of injury, radical formation was increased by lipid antioxidants; however, in the presence of injury, radical formation was decreased. In unstressed conditions, this relationship between chondrocyte mitochondria and redox regulation was reproduced in vivo with overexpression of glutathione peroxidase 4. In mice aged 18 months or more, overexpression of glutathione peroxidase 4 significantly decreased the presence of pro-mitochondrial peroxisome proliferation activated receptor gamma and deranged the relationship between mitochondria and the redox environment. This complex interaction suggests strategies targeting articular cartilage may benefit from adopting more nuanced paradigms of articular chondrocyte redox metabolism.
- Published
- 2024
- Full Text
- View/download PDF
6. Ascorbate mediates the non-enzymatic reduction of nitrite to nitric oxide
- Author
-
Juan Du, Milos R. Filipović, Brett A. Wagner, and Garry R. Buettner
- Subjects
Ascorbate ,Nitric oxide ,and Nitrite ,Biochemistry ,QD415-436 - Abstract
Nitric oxide (NO•) generated by nitric oxide synthases is involved in many physiological and pathophysiological processes. However, non-enzymatic formation of NO• also occurs in vivo. Here we investigated the production of NO• from nitrite, as facilitated by ascorbate, over the pH range of 2.4–7.4. Using a nitric oxide electrode, we observed at low pH a rapid generation of NO• from nitrite and ascorbate that slows with increasing pH. The formation of NO• was confirmed by its reaction with oxyhemoglobin. In the ascorbate/nitrite system a steady-state level of NO• was achieved, suggesting that a futile redox cycle of nitrite-reduction by ascorbate and NO•-oxidation by dioxygen was established. However, at pH-values of around 7 and greater, the direct reduction of nitrite by ascorbate is very slow; thus, this route to the non-enzymatic production of NO• is not likely to be significant process in vivo in environments having a pH around 7.4. The production of nitric oxide by nitrite and ascorbate would be important only in areas of lower pH, e.g. stomach/digestive system, sites of inflammation, and areas of hypoxia such as tumor tissue. In patients receiving very large doses of ascorbate delivered by intravenous infusion, plasma levels of ascorbate on the order of 20 - 30 mM can be achieved. After infusion, levels of nitrate and nitrite in plasma were unchanged. Thus, in blood and tissue that maintain a pH of about 7.4, the reduction of nitrite to nitric oxide by ascorbate appears to be insignificant, even at very large, pharmacological levels of ascorbate.
- Published
- 2023
- Full Text
- View/download PDF
7. Stability of aqueous solutions of ascorbate for basic research and for intravenous administration
- Author
-
Brett A. Wagner and Garry R. Buettner
- Subjects
Ascorbate ,Metal-catalysis ,Vitamin C stability ,Pharmacological ascorbate stability ,Biochemistry ,QD415-436 - Abstract
Ascorbate (vitamin C) can rapidly oxidize in many near-neutral pH, aqueous solutions. We report on the stability of ascorbate solutions prepared for infusion into patients using standard pharmacy protocols, for example, 75 g of ascorbate/L in water for infusion. The concentration of ascorbate was monitored for changes over time using direct UV–Vis spectroscopy. The pH of the solution was about 5.7 with no significant change over 24 h. There was only an approximate loss of 1% per day over the first 3 days of storage. This information allows decisions on how far ahead of need such preparations can be made. We also provide laboratory approaches to minimize or control the rate of oxidation of ascorbate solutions for use in chemical and biochemical studies as well as preclinical animal studies. The goal is to have the amount of ascorbate intended to be used in experiments be the actual amount available.
- Published
- 2023
- Full Text
- View/download PDF
8. The role of mitochondria in pharmacological ascorbate-induced toxicity
- Author
-
Juan Du, Amanda N. Pope, Brianne R. O’Leary, Brett A. Wagner, Prabhat C. Goswami, Garry R. Buettner, and Joseph J. Cullen
- Subjects
Medicine ,Science - Abstract
Abstract At pharmacological levels, ascorbate (P-AscH-) acts as a pro-oxidant by generating H2O2, depleting ATP in sensitive cells leading to cell death. The aim of this study was to determine the role of ATP production by oxidative phosphorylation or glycolysis in mechanisms of resistance to P-AscH–induced cell death. Pancreatic cancer cells were used to generate ρ0 cells by mitochondrial overexpression of the Y147A mutant uracil-N-glycosylase or Herpes Simplex Virus protein. The ρ0 phenotype was confirmed by probing for mitochondrial DNA, mitochondrial DNA-encoded cytochrome c oxidase subunit 2, and monitoring the rate of oxygen consumption. In ρ0 cells, glycolysis accounted for 100% of ATP production as there was no mitochondrial oxygen consumption. Even though the activities of H2O2-removing antioxidant enzymes were similar in both the parental and ρ0 clones, P-AscH- -induced clonogenic cell death in ρ0 cells showed more resistance than the parental cell line. In addition, P-AscH- induced more DNA damage and more consumption of NAD+ and greater decreases in the production of ATP in the parental cell line compared to the ρ0 cells. Thus, cancer cells that largely use oxidative phosphorylation to generate ATP may be more sensitive to P-AscH- compared with cells that are glycolysis-dependent.
- Published
- 2022
- Full Text
- View/download PDF
9. Pharmacologic Ascorbate Radiosensitizes Pancreatic Cancer but Radioprotects Normal Tissue: The Role of Oxidative Stress-Induced Lipid Peroxidation
- Author
-
Gloria Y. Chen, Brianne R. O’Leary, Juan Du, Rory S. Carroll, Garett J. Steers, Garry R. Buettner, and Joseph J. Cullen
- Subjects
pancreatic cancer ,radioprotection ,ascorbate ,lipid peroxidation ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The toxicity of ionizing radiation limits its effectiveness in the treatment of pancreatic ductal adenocarcinoma. Pharmacologic ascorbate (P-AscH−) has been shown to radiosensitize pancreatic cancer cells while simultaneously radioprotecting normal cells. We hypothesize that P-AscH− protects the small intestine while radiosensitizing pancreatic cancer cells partially through an oxidative stress mechanism. Duodenal samples from pancreaticoduodenectomy specimens of patients who underwent radio-chemotherapy ± P-AscH− and mouse tumor and jejunal samples treated with radiation ± P-AscH− were evaluated. Pancreatic cancer and non-tumorigenic cells were treated with radiation ± P-AscH− to assess lipid peroxidation. To determine the mechanism, pancreatic cancer cells were treated with selenomethionine or RSL3, an inhibitor of glutathione peroxidase 4 (GPx4). Radiation-induced decreases in villi length and increases in 4-HNE immunofluorescence were reversed with P-AscH− in human duodenum. In vivo, radiation-induced decreases in villi length and increased collagen deposition were reversed in P-AscH−-treated jejunal samples. P-AscH− and radiation increased BODIPY oxidation in pancreatic cancer cells but not in non-tumorigenic cells. Selenomethionine increased GPx4 protein and activity in pancreatic cancer and reversed P-AscH−-induced toxicity and lipid peroxidation. RSL3 treatment inhibited GPx4 activity and increased lipid peroxidation. Differences in oxidative stress may play a role in radioprotecting normal cells while radiosensitizing pancreatic cancer cells when treated with P-AscH−.
- Published
- 2024
- Full Text
- View/download PDF
10. Depletion of Labile Iron Induces Replication Stress and Enhances Responses to Chemoradiation in Non-Small-Cell Lung Cancer
- Author
-
Khaliunaa Bayanbold, Mekhla Singhania, Melissa A. Fath, Charles C. Searby, Jeffrey M. Stolwijk, John B. Henrich, Casey F. Pulliam, Joshua D. Schoenfeld, Kranti A. Mapuskar, Sei Sho, Joseph M. Caster, Bryan G. Allen, Garry R. Buettner, Maria Spies, Prabhat C. Goswami, Michael S. Petronek, and Douglas R. Spitz
- Subjects
ferritin heavy chain ,iron chelator ,deferoxamine ,labile iron pool ,cell cycle ,replication stress ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The intracellular redox-active labile iron pool (LIP) is weakly chelated and available for integration into the iron metalloproteins that are involved in diverse cellular processes, including cancer cell-specific metabolic oxidative stress. Abnormal iron metabolism and elevated LIP levels are linked to the poor survival of lung cancer patients, yet the underlying mechanisms remain unclear. Depletion of the LIP in non-small-cell lung cancer cell lines using the doxycycline-inducible overexpression of the ferritin heavy chain (Ft-H) (H1299 and H292), or treatment with deferoxamine (DFO) (H1299 and A549), inhibited cell growth and decreased clonogenic survival. The Ft-H overexpression-induced inhibition of H1299 and H292 cell growth was also accompanied by a significant delay in transit through the S-phase. In addition, both Ft-H overexpression and DFO in H1299 resulted in increased single- and double-strand DNA breaks, supporting the involvement of replication stress in the response to LIP depletion. The Ft-H and DFO treatment also sensitized H1299 to VE-821, an inhibitor of ataxia telangiectasis and Rad2-related (ATR) kinase, highlighting the potential of LIP depletion, combined with DNA damage response modifiers, to alter lung cancer cell responses. In contrast, only DFO treatment effectively reduced the LIP, clonogenic survival, cell growth, and sensitivity to VE-821 in A549 non-small-cell lung cancer cells. Importantly, the Ft-H and DFO sensitized both H1299 and A549 to chemoradiation in vitro, and Ft-H overexpression increased the efficacy of chemoradiation in vivo in H1299. These results support the hypothesis that the depletion of the LIP can induce genomic instability, cell death, and potentiate therapeutic responses to chemoradiation in NSCLC.
- Published
- 2023
- Full Text
- View/download PDF
11. Pharmacologic Ascorbate and DNMT Inhibitors Increase DUOX Expression and Peroxide-Mediated Toxicity in Pancreatic Cancer
- Author
-
Garett J. Steers, Brianne R. O’Leary, Juan Du, Brett A. Wagner, Rory S. Carroll, Frederick E. Domann, Prabhat C. Goswami, Garry R. Buettner, and Joseph J. Cullen
- Subjects
ascorbic acid ,pharmacologic ascorbate ,pancreatic cancer ,epigenetics ,DNA methyltransferase (DNMT) ,ten-eleven translocation (TET) methylcytosine dioxygenase ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Recent studies have demonstrated an important role for vitamin C in the epigenetic regulation of cancer-related genes via DNA demethylation by the ten-eleven translocation (TET) methylcytosine dioxygenase enzymes. DNA methyltransferase (DNMT) reverses this, increasing DNA methylation and decreasing gene expression. Dual oxidase (DUOX) enzymes produce hydrogen peroxide (H2O2) in normal pancreatic tissue but are silenced in pancreatic cancer (PDAC). Treatment of PDAC with pharmacologic ascorbate (P-AscH−, intravenous, high dose vitamin C) increases DUOX expression. We hypothesized that inhibiting DNMT may act synergistically with P-AscH− to further increase DUOX expression and cytotoxicity of PDAC. PDAC cells demonstrated dose-dependent increases in DUOX mRNA and protein expression when treated with DNMT inhibitors. PDAC cells treated with P-AscH− + DNMT inhibitors demonstrated increased DUOX expression, increased intracellular oxidation, and increased cytotoxicity in vitro and in vivo compared to either treatment alone. These findings suggest a potential therapeutic, epigenetic mechanism to treat PDAC.
- Published
- 2023
- Full Text
- View/download PDF
12. Pharmacological ascorbate improves the response to platinum-based chemotherapy in advanced stage non-small cell lung cancer
- Author
-
Muhammad Furqan, Taher Abu-Hejleh, Laura M. Stephens, Stacey M. Hartwig, Sarah L. Mott, Casey F. Pulliam, Michael Petronek, John B. Henrich, Melissa A. Fath, Jon C. Houtman, Steven M. Varga, Kellie L. Bodeker, Aaron D. Bossler, Andrew M. Bellizzi, Jun Zhang, Varun Monga, Hariharasudan Mani, Marina Ivanovic, Brian J. Smith, Margaret M. Byrne, William Zeitler, Brett A. Wagner, Garry R. Buettner, Joseph J. Cullen, John M. Buatti, Douglas R. Spitz, and Bryan G. Allen
- Subjects
Non-small cell ,Ascorbate ,Vitamin C ,Platinum ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Purpose: Platinum-based chemotherapy with or without immunotherapy is the mainstay of treatment for advanced stage non-small cell lung cancer (NSCLC) lacking a molecular driver alteration. Pre-clinical studies have reported that pharmacological ascorbate (P-AscH-) enhances NSCLC response to platinum-based therapy. We conducted a phase II clinical trial combining P-AscH- with carboplatin-paclitaxel chemotherapy. Experimental design: Chemotherapy naïve advanced stage NSCLC patients received 75 g ascorbate twice per week intravenously with carboplatin and paclitaxel every three weeks for four cycles. The primary endpoint was to improve tumor response per Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 compared to the historical control of 20%. The trial was conducted as an optimal Simon's two-stage design. Blood samples were collected for exploratory analyses. Results: The study enrolled 38 patients and met its primary endpoint with an objective response rate of 34.2% (p = 0.03). All were confirmed partial responses (cPR). The disease control rate was 84.2% (stable disease + cPR). Median progression-free and overall survival were 5.7 months and 12.8 months, respectively. Treatment-related adverse events (TRAE) included one grade 5 (neutropenic fever) and five grade 4 events (cytopenias). Cytokine and chemokine data suggest that the combination elicits an immune response. Immunophenotyping of peripheral blood mononuclear cells demonstrated an increase in effector CD8 T-cells in patients with a progression-free survival (PFS) ≥ 6 months. Conclusions: The addition of P-AscH- to platinum-based chemotherapy improved tumor response in advanced stage NSCLC. P-AscH- appears to alter the host immune response and needs further investigation as a potential adjuvant to immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
13. Pharmacological ascorbate induces sustained mitochondrial dysfunction
- Author
-
Rory S. Carroll, Juan Du, Brianne R. O'Leary, Garett Steers, Prabhat C. Goswami, Garry R. Buettner, and Joseph J. Cullen
- Subjects
Physiology (medical) ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
14. Magnetic resonance imaging (MRI) of pharmacological ascorbate-induced iron redox state as a biomarker in subjects undergoing radio-chemotherapy
- Author
-
Cameron M. Cushing, Michael S. Petronek, Kellie L. Bodeker, Sandy Vollstedt, Heather A. Brown, Emyleigh Opat, Nancy J. Hollenbeck, Thomas Shanks, Daniel J. Berg, Brian J. Smith, Mark C. Smith, Varun Monga, Muhammad Furqan, Matthew A. Howard, Jeremy D. Greenlee, Kranti A. Mapuskar, Joel St-Aubin, Ryan T. Flynn, Joseph J. Cullen, Garry R. Buettner, Douglas R. Spitz, John M. Buatti, Bryan G. Allen, and Vincent A. Magnotta
- Subjects
Quantitative imaging ,GBM ,T2* ,QSM ,Pharmacological ascorbate ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Pharmacological ascorbate (P-AscH-) combined with standard of care (SOC) radiation and temozolomide is being evaluated in a phase 2 clinical trial (NCT02344355) in the treatment of glioblastoma (GBM). Previously published data demonstrated that paramagnetic iron (Fe3+) catalyzes ascorbate's oxidation to form diamagnetic iron (Fe2+). Because paramagnetic Fe3+ may influence relaxation times observed in MR imaging, quantitative MR imaging of P-AscH--induced changes in redox-active Fe was assessed as a biomarker for therapy response.Gel phantoms containing either Fe3+ or Fe2+ were imaged with T2* and quantitative susceptibility mapping (QSM). Fifteen subjects receiving P-AscH- plus SOC underwent T2* and QSM imaging four weeks into treatment. Subjects were scanned: pre-P-AscH- infusion, post-P-AscH- infusion, and post-radiation (3–4 h between scans). Changes in T2* and QSM relaxation times in tumor and normal tissue were calculated and compared to changes in Fe3+ and Fe2+ gel phantoms. A GBM mouse model was used to study the relationship between the imaging findings and the labile iron pool.Phantoms containing Fe3+ demonstrated detectable changes in T2* and QSM relaxation times relative to Fe2+ phantoms. Compared to pre-P-AscH-, GBM T2* and QSM imaging were significantly changed post-P-AscH- infusion consistent with conversion of Fe3+ to Fe2+. No significant changes in T2* or QSM were observed in normal brain tissue. There was moderate concordance between T2* and QSM changes in both progression free survival and overall survival. The GBM mouse model showed similar results with P-AscH- inducing greater changes in tumor labile iron pools compared to the normal tissue. Conclusions: T2* and QSM MR-imaging responses are consistent with P-AscH- reducing Fe3+ to Fe2+, selectively in GBM tumor volumes and represent a potential biomarker of response. This study is the first application using MR imaging in humans to measure P-AscH--induced changes in redox-active iron.
- Published
- 2021
- Full Text
- View/download PDF
15. Simultaneous detection of the enzyme activities of GPx1 and GPx4 guide optimization of selenium in cell biological experiments
- Author
-
Jeffrey M. Stolwijk, Kelly C. Falls-Hubert, Charles C. Searby, Brett A. Wagner, and Garry R. Buettner
- Subjects
Selenium ,GPx4 ,GPx1 ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
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.
- Published
- 2020
- Full Text
- View/download PDF
16. Augmentation of intracellular iron using iron sucrose enhances the toxicity of pharmacological ascorbate in colon cancer cells
- Author
-
Kristin E. Brandt, Kelly C. Falls, Joshua D. Schoenfeld, Samuel N. Rodman, Zhimin Gu, Fenghuang Zhan, Joseph J. Cullen, Brett A. Wagner, Garry R. Buettner, Bryan G. Allen, Daniel J. Berg, Douglas R. Spitz, and Melissa A. Fath
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Pharmacological doses (> 1 mM) of ascorbate (a.k.a., vitamin C) have been shown to selectively kill cancer cells through a mechanism that is dependent on the generation of H2O2 at doses that are safely achievable in humans using intravenous administration. The process by which ascorbate oxidizes to form H2O2 is thought to be mediated catalytically by redox active metal ions such as iron (Fe). Because intravenous iron sucrose is often administered to colon cancer patients to help mitigate anemia, the current study assessed the ability of pharmacological ascorbate to kill colon cancer cells in the presence and absence of iron sucrose.In vitro survival assays showed that 10 mM ascorbate exposure (2 h) clonogenically inactivated 40â80% of exponentially growing colon cancer cell lines (HCT116 and HT29). When the H2O2 scavenging enzyme, catalase, was added to the media, or conditionally over-expressed using a doxycycline inducible vector, the toxicity of pharmacological ascorbate was significantly blunted. When colon cancer cells were treated in the presence or absence of 250 µM iron sucrose, then rinsed, and treated with 10 mM ascorbate, the cells demonstrated increased levels of labile iron that resulted in significantly increased clonogenic cell killing, compared to pharmacological ascorbate alone. Interestingly, when colon cancer cells were treated with iron sucrose for 1 h and then 10 mM ascorbate was added to the media in the continued presence of iron sucrose, there was no enhancement of toxicity despite similar increases in intracellular labile iron. The combination of iron chelators, deferoxamine and diethylenetriaminepentaacetic acid, significantly inhibited the toxicity of either ascorbate alone or ascorbate following iron sucrose. These observations support the hypothesis that increasing intracellular labile iron pools, using iron sucrose, can be used to increase the toxicity of pharmacological ascorbate in human colon cancer cells by a mechanism involving increased generation of H2O2. Keywords: Oxidative stress, Redox active iron, Iron sucrose, Catalase, Deferoxamine, Chelators
- Published
- 2018
- Full Text
- View/download PDF
17. In vitro Cytotoxicity and Pharmacokinetic Evaluation of Pharmacological Ascorbate in Dogs
- Author
-
Margaret L. Musser, Alyssa L. Mahaffey, Melissa A. Fath, Garry R. Buettner, Brett A. Wagner, Benjamin K. Schneider, Yeon-Jung Seo, Jonathan P. Mochel, and Chad M. Johannes
- Subjects
canine ,vitamin C ,high-dose ,osteosarcoma ,intravenous ,Veterinary medicine ,SF600-1100 - Abstract
Background: High-dose, pharmacological ascorbate (P-AscH−) is preferentially cytotoxic to human cancer cells in vitro. Investigations on the efficacy of P-AscH− as an adjuvant treatment for multiple human cancers are on-going, but has yet to be formally investigated in dogs. The primary objectives of this study were to determine the pharmacokinetic (PK) profile of P-AscH− in healthy Beagle dogs and the effects of P-AscH− on canine osteosarcoma cells in vitro.Methods: Eight purpose-bred, healthy, spayed female Beagle dogs, between 20 and 21 months old, and 8–10 kg were administered two doses of P-AscH− (550 or 2,200 mg/kg) via intravenous infusion over 6 h, on separate days. Plasma ascorbate concentrations were measured at 12 time points during and after infusion for PK analysis using nonlinear mixed-effects (NLME) and non-compartmental analysis (NCA). Clonogenic assays were performed on 2 canine osteosarcoma cell lines (D-17 and OSCA-8) and canine primary dermal fibroblasts after exposure to high concentrations of ascorbate (75 pmoles/cell).Results: Plasma ascorbate levels in the dogs peaked at approximately 10 mM following 2,200 mg/kg and returned to baseline 6–8 h after dosing. Minor adverse effects were seen in two dogs. Ascorbate (75 pmoles/cell) significantly decreased survival in both the osteosarcoma cell lines (D-17 63% SD 0.010, P = 0.005; OSCA-8 50% SD 0.086, P = 0.026), compared to normal fibroblasts (27% SD 0.056).Conclusions: Pharmacological ascorbate is preferentially cytotoxic to canine-derived cancer cells. High levels of ascorbate can be safely administered to dogs. Further studies are needed to determine the effects of P-AscH− on canine patients.
- Published
- 2019
- Full Text
- View/download PDF
18. Pharmacological Ascorbate Enhances Chemotherapies in Pancreatic Ductal Adenocarcinoma
- Author
-
Brianne R. O'Leary, Elena K. Ruppenkamp, Garett J. Steers, Juan Du, Rory S. Carroll, Brett A. Wagner, Garry R. Buettner, and Joseph J. Cullen
- Subjects
Paclitaxel ,Hepatology ,Endocrinology, Diabetes and Metabolism ,Leucovorin ,Antineoplastic Agents ,Ascorbic Acid ,Irinotecan ,Oxaliplatin ,Pancreatic Neoplasms ,Mice ,Endocrinology ,Antineoplastic Combined Chemotherapy Protocols ,Internal Medicine ,Animals ,Humans ,Fluorouracil ,Carcinoma, Pancreatic Ductal - Abstract
Pharmacological ascorbate (P-AscH - , high-dose, intravenous vitamin C) has shown promise as an adjuvant therapy for pancreatic ductal adenocarcinoma (PDAC) treatment. The objective of this study was to determine the effects of P-AscH - when combined with PDAC chemotherapies.Clonogenic survival, combination indices, and DNA damage were determined in human PDAC cell lines treated with P-AscH - in combination with 5-fluorouracil, paclitaxel, or FOLFIRINOX (combination of leucovorin, 5-fluorouracil, irinotecan, oxaliplatin). Tumor volume changes, overall survival, blood analysis, and plasma ascorbate concentration were determined in vivo in mice treated with P-AscH - with or without FOLFIRINOX.P-AscH - combined with 5-fluorouracil, paclitaxel, or FOLFIRINOX significantly reduced clonogenic survival in vitro. The DNA damage, measured by γH2AX protein expression, was increased after treatment with P-AscH - , FOLFIRINOX, and their combination. In vivo, tumor growth rate was significantly reduced by P-AscH - , FOLFIRINOX, and their combination. Overall survival was significantly increased by the combination of P-AscH - and FOLFIRINOX. Treatment with P-AscH - increased red blood cell and hemoglobin values but had no effect on white blood cell counts. Plasma ascorbate concentrations were significantly elevated in mice treated with P-AscH - with or without FOLFIRINOX.The addition of P-AscH - to standard of care chemotherapy has the potential to be an effective adjuvant for PDAC treatment.
- Published
- 2022
- Full Text
- View/download PDF
19. Catalase Modulates the Radio-Sensitization of Pancreatic Cancer Cells by Pharmacological Ascorbate
- Author
-
Juan Du, Rory S. Carroll, Garett J. Steers, Brett A. Wagner, Brianne R. O’Leary, Chris S. Jensen, Garry R. Buettner, and Joseph J. Cullen
- Subjects
pharmacological ascorbate ,DNA damage ,DNA repair ,pancreatic cancer ,vitamin C ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Pancreatic cancer cells (PDACs) are more susceptible to an oxidative insult than normal cells, resulting in greater cytotoxicity upon exposure to agents that increase pro-oxidant levels. Pharmacological ascorbate (P-AscH−), i.e., large amounts given intravenously (IV), generates significant fluxes of hydrogen peroxide (H2O2), resulting in the killing of PDACs but not normal cells. Recent studies have demonstrated that P-AscH− radio-sensitizes PDAC but is a radioprotector to normal cells and tissues. Several mechanisms have been hypothesized to explain the dual roles of P-AscH− in radiation-induced toxicity including the activation of nuclear factor-erythroid 2-related factor 2 (Nrf2), RelB, as well as changes in bioenergetic profiles. We have found that P-AscH− in conjunction with radiation increases Nrf2 in both cancer cells and normal cells. Although P-AscH− with radiation decreases RelB in cancer cells vs. normal cells, the knockout of RelB does not radio-sensitize PDACs. Cellular bioenergetic profiles demonstrate that P-AscH− with radiation increases the ATP demand/production rate (glycolytic and oxidative phosphorylation) in both PDACs and normal cells. Knocking out catalase results in P-AscH− radio-sensitization in PDACs. In a phase I trial where P-AscH− was included as an adjuvant to the standard of care, short-term survivors had higher catalase levels in tumor tissue, compared to long-term survivors. These data suggest that P-AscH− radio-sensitizes PDACs through increased peroxide flux. Catalase levels could be a possible indicator for how tumors will respond to P-AscH−.
- Published
- 2021
- Full Text
- View/download PDF
20. Tumor cells have decreased ability to metabolize H2O2: Implications for pharmacological ascorbate in cancer therapy
- Author
-
Claire M. Doskey, Visarut Buranasudja, Brett A. Wagner, Justin G. Wilkes, Juan Du, Joseph J. Cullen, and Garry R. Buettner
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Ascorbate (AscH−) functions as a versatile reducing agent. At pharmacological doses (P-AscH−; [plasma AscH−] ≥≈20 mM), achievable through intravenous delivery, oxidation of P-AscH− can produce a high flux of H2O2 in tumors. Catalase is the major enzyme for detoxifying high concentrations of H2O2. We hypothesize that sensitivity of tumor cells to P-AscH− compared to normal cells is due to their lower capacity to metabolize H2O2. Rate constants for removal of H2O2 (kcell) and catalase activities were determined for 15 tumor and 10 normal cell lines of various tissue types. A differential in the capacity of cells to remove H2O2 was revealed, with the average kcell for normal cells being twice that of tumor cells. The ED50 (50% clonogenic survival) of P-AscH− correlated directly with kcell and catalase activity. Catalase activity could present a promising indicator of which tumors may respond to P-AscH−.
- Published
- 2016
- Full Text
- View/download PDF
21. Oxidation of ferumoxytol by ionizing radiation releases iron. An electron paramagnetic resonance study
- Author
-
Michael S Petronek, Douglas R Spitz, Garry R Buettner, and Bryan G Allen
- Subjects
Radiation ,Iron ,Neoplasms ,Radiation, Ionizing ,Health, Toxicology and Mutagenesis ,Electron Spin Resonance Spectroscopy ,Humans ,Radiology, Nuclear Medicine and imaging ,Hydrogen Peroxide ,Oxidation-Reduction ,Ferrosoferric Oxide - Abstract
Ferumoxytol (FMX) is an iron oxide nanoparticle that is FDA approved for the treatment of iron deficiency anemia. FMX contains an Fe3O4 core. Currently, the redox chemistry of Fe3O4 nanoparticles remains relatively unexplored. FMX has recently gained interest as an anti-cancer agent. Ionizing radiation (IR) is a treatment modality employed to treat several types of cancer. Utilizing electron paramagnetic resonance (EPR) spectroscopy, we found that the products produced from the radiolysis of water can oxidize the Fe3O4 core of FMX. Because of the limited diffusion of the HO2• and HO• produced, these highly oxidizing species have little direct effect on FMX oxidation. We have determined that H2O2 is the primary oxidant of FMX. In the presence of labile Fe2+, we found that reducing species generated from the radiolysis of H2O are able to reduce the Fe3+ sites of the Fe3O4 core. Importantly, we also have shown that IR stimulates the release of ferric iron from FMX. Because of its release of iron, FMX may serve as an adjuvant to enhance radiotherapy.
- Published
- 2022
- Full Text
- View/download PDF
22. Changes in metabolic landscapes shape divergent but distinct mutational signatures and cytotoxic consequences of redox stress
- Author
-
Natalya P Degtyareva, Victoria C Placentra, Scott A Gabel, Leszek J Klimczak, Dmitry A Gordenin, Brett A Wagner, Garry R Buettner, Geoffrey A Mueller, Tatyana I Smirnova, and Paul W Doetsch
- Subjects
Genetics ,Molecular Biology - Abstract
Mutational signatures discerned in cancer genomes, in aging tissues and in cells exposed to toxic agents, reflect complex processes underlying transformation of cells from normal to dysfunctional. Due to its ubiquitous and chronic nature, redox stress contributions to cellular makeover remain equivocal. The deciphering of a new mutational signature of an environmentally-relevant oxidizing agent, potassium bromate, in yeast single strand DNA uncovered a surprising heterogeneity in the mutational signatures of oxidizing agents. NMR-based analysis of molecular outcomes of redox stress revealed profound dissimilarities in metabolic landscapes following exposure to hydrogen peroxide versus potassium bromate. The predominance of G to T substitutions in the mutational spectra distinguished potassium bromate from hydrogen peroxide and paraquat and mirrored the observed metabolic changes. We attributed these changes to the generation of uncommon oxidizing species in a reaction with thiol-containing antioxidants; a nearly total depletion of intracellular glutathione and a paradoxical augmentation of potassium bromate mutagenicity and toxicity by antioxidants. Our study provides the framework for understanding multidimensional processes triggered by agents collectively known as oxidants. Detection of increased mutational loads associated with potassium bromate-related mutational motifs in human tumors may be clinically relevant as a biomarker of this distinct type of redox stress.
- Published
- 2023
- Full Text
- View/download PDF
23. Data from Pharmacologic Ascorbate Primes Pancreatic Cancer Cells for Death by Rewiring Cellular Energetics and Inducing DNA Damage
- Author
-
Garry R. Buettner, Joseph J. Cullen, Stacia L. Koppenhafer, David J. Gordon, Juan Du, Brett A. Wagner, Adrienne R. Gibson, Claire M. Doskey, and Visarut Buranasudja
- Abstract
The clinical potential of pharmacologic ascorbate (P-AscH−; intravenous delivery achieving mmol/L concentrations in blood) as an adjuvant in cancer therapy is being reevaluated. At mmol/L concentrations, P-AscH− is thought to exhibit anticancer activity via generation of a flux of H2O2 in tumors, which leads to oxidative distress. Here, we use cell culture models of pancreatic cancer to examine the effects of P-AscH− on DNA damage, and downstream consequences, including changes in bioenergetics. We have found that the high flux of H2O2 produced by P-AscH− induces DNA damage. In response to this DNA damage, we observed that PARP1 is hyperactivated. Using our unique absolute quantitation, we found that P-AscH− mediated the overactivation of PARP1, which results in consumption of NAD+, and subsequently depletion of ATP leading to mitotic cell death. We have also found that Chk1 plays a major role in the maintenance of genomic integrity following treatment with P-AscH−. Hyperactivation of PARP1 and DNA repair are ATP-consuming processes. Using a Seahorse XF96 analyzer, we demonstrated that the severe decrease in ATP after challenging with P-AscH− is because of increased demand, not changes in the rate of production. Genetic deletion and pharmacologic inhibition of PARP1 preserved both NAD+ and ATP; however, the toxicity of P-AscH− remained. These data indicate that disruption of bioenergetics is a secondary factor in the toxicity of P-AscH−; damage to DNA appears to be the primary factor.Implications:Efforts to leverage P-AscH− in cancer therapy should first focus on DNA damage.
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Data from Pharmacologic Ascorbate Primes Pancreatic Cancer Cells for Death by Rewiring Cellular Energetics and Inducing DNA Damage
- Author
-
Garry R. Buettner, Joseph J. Cullen, Stacia L. Koppenhafer, David J. Gordon, Juan Du, Brett A. Wagner, Adrienne R. Gibson, Claire M. Doskey, and Visarut Buranasudja
- Abstract
S1. There is differential susceptibility to P-AscH- across cell lines. S2. Bolus addition of hydrogen peroxide depletes intracellular ATP and NAD+ in MIA PaCa-2 and PANC-1 cells. S3. Pharmacological ascorbate depletes intracellular NADH in MIA PaCa-2 cells, but not in PANC-1, 339, and H6c7 cells. S4. Pharmacological inhibition of PARP1 by olaparib enhances the cytotoxicity of P-AscH- in MIA PaCa-2 cells. S5. A combination of P-AscH- and prexasertib synergistically inhibits cell proliferation of MIA PaCa-2 cells. Supplementary Table S1. Normal pancreatic cells are more resistant to P-AscH- than pancreatic cancer cells. Supplementary Table S2. The ED50's for depletion of ATP and NAD+ correlate with kcell for the removal of H2O2. Supplementary Table S3. The gene target and primer pairs used for QPCR.
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Tables from First-in-Human Phase I Clinical Trial of Pharmacologic Ascorbate Combined with Radiation and Temozolomide for Newly Diagnosed Glioblastoma
- Author
-
John M. Buatti, Douglas R. Spitz, Brian J. Smith, Mindi J. TenNapel, Garry R. Buettner, Brett A. Wagner, Daniel Berg, Joseph J. Cullen, Karra A. Jones, Joseph M. Caster, Steven N. Seyedin, Kranti A. Mapuskar, Matthew A. Howard, Jeremy D. Greenlee, Sandy Vollstedt, Nancy Hollenbeck, Heather Brown, Thomas Carlisle, Raymond Hohl, Sonia Sandhu, Varun Monga, Mark C. Smith, Kellie L. Bodeker, and Bryan G. Allen
- Abstract
Supplementary Table 1. Description of radiation treatment volumes (GTV, CTV, and PTV) Supplementary Table 2. Description of dose to organs at risk and treatment interruptions Supplemental Table 3. Subject count for key steps within the RT-phase of the clinical trial Supplemental Table 4. Subject count for key steps within the ADJ-phase of the clinical trial
- Published
- 2023
- Full Text
- View/download PDF
26. Figure S2 from Dual Oxidase-Induced Sustained Generation of Hydrogen Peroxide Contributes to Pharmacologic Ascorbate-Induced Cytotoxicity
- Author
-
Joseph J. Cullen, Prabhat C. Goswami, Garry R. Buettner, Douglas R. Spitz, Rory S. Carroll, Matthew S. Alexander, Kelly C. Falls-Hubert, Jeffrey M. Stolwijk, Brett A. Wagner, Amanda L. Kalen, Ehab H. Sarsour, Juan Du, Brianne R. O'Leary, and Adrienne R. Gibson
- Abstract
Increased DUOX1 and DUOX2 expression is reversed by catalase.
- Published
- 2023
- Full Text
- View/download PDF
27. Table S1 from Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer
- Author
-
Joseph J. Cullen, Bryan G. Allen, Heather A. Brown, Sandy Vollstedt, Kellie L. Bodeker, Daniel J. Berg, John M. Buatti, Douglas R. Spitz, Brianne R. O’Leary, Katherine Gibson-Corley, Juan Du, Brett A. Wagner, Garry R. Buettner, Samuel R. Schroeder, Justin G. Wilkes, and Matthew S. Alexander
- Abstract
Radiation induced intestinal damage histology and collagen deposition information.
- Published
- 2023
- Full Text
- View/download PDF
28. Data from Loss of SOD3 (EcSOD) Expression Promotes an Aggressive Phenotype in Human Pancreatic Ductal Adenocarcinoma
- Author
-
James J. Mezhir, Douglas R. Spitz, Frederick E. Domann, Michael D. Henry, Jeffery Keene, Robert A. Beardsley, Wendy Cozen, Brenda Y. Hernandez, Charles F. Lynch, Garry R. Buettner, Brett A. Wagner, Sean Altekruse, Adam J. Case, Bryan G. Allen, Anna M. Button, Jennifer E. Hrabe, Andrew M. Bellizzi, Melissa A. Fath, and Brianne R. O'Leary
- Abstract
Purpose: Pancreatic ductal adenocarcinoma (PDA) cells are known to produce excessive amounts of reactive oxygen species (ROS), particularly superoxide, which may contribute to the aggressive and refractory nature of this disease. Extracellular superoxide dismutase (EcSOD) is an antioxidant enzyme that catalyzes the dismutation of superoxide in the extracellular environment. This study tests the hypothesis that EcSOD modulates PDA growth and invasion by modifying the redox balance in PDA.Experimental Design: We evaluated the prognostic significance of EcSOD in a human tissue microarray (TMA) of patients with PDA. EcSOD overexpression was performed in PDA cell lines and animal models of disease. The impact of EcSOD on PDA cell lines was evaluated with Matrigel invasion in combination with a superoxide-specific SOD mimic and a nitric oxide synthase (NOS) inhibitor to determine the mechanism of action of EcSOD in PDA.Results: Loss of EcSOD expression is a common event in PDA, which correlated with worse disease biology. Overexpression of EcSOD in PDA cell lines resulted in decreased invasiveness that appeared to be related to reactions of superoxide with nitric oxide. Pancreatic cancer xenografts overexpressing EcSOD also demonstrated slower growth and peritoneal metastasis. Overexpression of EcSOD or treatment with a superoxide-specific SOD mimic caused significant decreases in PDA cell invasive capacity.Conclusions: These results support the hypothesis that loss of EcSOD leads to increased reactions of superoxide with nitric oxide, which contributes to the invasive phenotype. These results allow for the speculation that superoxide dismutase mimetics might inhibit PDA progression in human clinical disease. Clin Cancer Res; 21(7); 1741–51. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
29. Supplementary Legend from First-in-Human Phase I Clinical Trial of Pharmacologic Ascorbate Combined with Radiation and Temozolomide for Newly Diagnosed Glioblastoma
- Author
-
John M. Buatti, Douglas R. Spitz, Brian J. Smith, Mindi J. TenNapel, Garry R. Buettner, Brett A. Wagner, Daniel Berg, Joseph J. Cullen, Karra A. Jones, Joseph M. Caster, Steven N. Seyedin, Kranti A. Mapuskar, Matthew A. Howard, Jeremy D. Greenlee, Sandy Vollstedt, Nancy Hollenbeck, Heather Brown, Thomas Carlisle, Raymond Hohl, Sonia Sandhu, Varun Monga, Mark C. Smith, Kellie L. Bodeker, and Bryan G. Allen
- Abstract
Supplementary Legend
- Published
- 2023
- Full Text
- View/download PDF
30. Figure S1 from Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer
- Author
-
Joseph J. Cullen, Bryan G. Allen, Heather A. Brown, Sandy Vollstedt, Kellie L. Bodeker, Daniel J. Berg, John M. Buatti, Douglas R. Spitz, Brianne R. O’Leary, Katherine Gibson-Corley, Juan Du, Brett A. Wagner, Garry R. Buettner, Samuel R. Schroeder, Justin G. Wilkes, and Matthew S. Alexander
- Abstract
The effect of ascorbate and catalase on clonogenic cell survival in PDAC.
- Published
- 2023
- Full Text
- View/download PDF
31. Data from Mechanisms of Ascorbate-Induced Cytotoxicity in Pancreatic Cancer
- Author
-
Joseph J. Cullen, Charles M. Knudson, Changbin Du, Agshin F. Taghiyev, Sih-han Wang, Garry R. Buettner, Brett A. Wagner, Mark Levine, Sean M. Martin, and Juan Du
- Abstract
Purpose: Pharmacologic concentrations of ascorbate may be effective in cancer therapeutics. We hypothesized that ascorbate concentrations achievable with i.v. dosing would be cytotoxic in pancreatic cancer for which the 5-year survival is Experimental Design: Pancreatic cancer cell lines were treated with ascorbate (0, 5, or 10 mmol/L) for 1 hour, then viability and clonogenic survival were determined. Pancreatic tumor cells were delivered s.c. into the flank region of nude mice and allowed to grow at which time they were randomized to receive either ascorbate (4 g/kg) or osmotically equivalent saline (1 mol/L) i.p. for 2 weeks.Results: There was a time- and dose-dependent increase in measured H2O2 production with increased concentrations of ascorbate. Ascorbate decreased viability in all pancreatic cancer cell lines but had no effect on an immortalized pancreatic ductal epithelial cell line. Ascorbate decreased clonogenic survival of the pancreatic cancer cell lines, which was reversed by treatment of cells with scavengers of H2O2. Treatment with ascorbate induced a caspase-independent cell death that was associated with autophagy. In vivo, treatment with ascorbate inhibited tumor growth and prolonged survival.Conclusions: These results show that pharmacologic doses of ascorbate, easily achievable in humans, may have potential for therapy in pancreatic cancer. Clin Cancer Res; 16(2); 509–20
- Published
- 2023
- Full Text
- View/download PDF
32. Supplementary Figures 1-3 from Loss of SOD3 (EcSOD) Expression Promotes an Aggressive Phenotype in Human Pancreatic Ductal Adenocarcinoma
- Author
-
James J. Mezhir, Douglas R. Spitz, Frederick E. Domann, Michael D. Henry, Jeffery Keene, Robert A. Beardsley, Wendy Cozen, Brenda Y. Hernandez, Charles F. Lynch, Garry R. Buettner, Brett A. Wagner, Sean Altekruse, Adam J. Case, Bryan G. Allen, Anna M. Button, Jennifer E. Hrabe, Andrew M. Bellizzi, Melissa A. Fath, and Brianne R. O'Leary
- Abstract
Supplementary Figures 1-3. S1: Creation and characterization of EcSOD stably expressing PDA cell lines.; S2: Activity of EcSOD activity from stably expression PDA cell lines; S3: L-NNA decreases NOS activity in PDA cells.
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Figure 1 from First-in-Human Phase I Clinical Trial of Pharmacologic Ascorbate Combined with Radiation and Temozolomide for Newly Diagnosed Glioblastoma
- Author
-
John M. Buatti, Douglas R. Spitz, Brian J. Smith, Mindi J. TenNapel, Garry R. Buettner, Brett A. Wagner, Daniel Berg, Joseph J. Cullen, Karra A. Jones, Joseph M. Caster, Steven N. Seyedin, Kranti A. Mapuskar, Matthew A. Howard, Jeremy D. Greenlee, Sandy Vollstedt, Nancy Hollenbeck, Heather Brown, Thomas Carlisle, Raymond Hohl, Sonia Sandhu, Varun Monga, Mark C. Smith, Kellie L. Bodeker, and Bryan G. Allen
- Abstract
Supplemental Figure 1: P-AscH- treatment in combination with RT and TMZ decreases the systemic the systemic oxidaitive stress marker 4-hydroxy-2-nonenal modified proteins. S1A-SAB.
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Figure Legends from Loss of SOD3 (EcSOD) Expression Promotes an Aggressive Phenotype in Human Pancreatic Ductal Adenocarcinoma
- Author
-
James J. Mezhir, Douglas R. Spitz, Frederick E. Domann, Michael D. Henry, Jeffery Keene, Robert A. Beardsley, Wendy Cozen, Brenda Y. Hernandez, Charles F. Lynch, Garry R. Buettner, Brett A. Wagner, Sean Altekruse, Adam J. Case, Bryan G. Allen, Anna M. Button, Jennifer E. Hrabe, Andrew M. Bellizzi, Melissa A. Fath, and Brianne R. O'Leary
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
35. Data from Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer
- Author
-
Joseph J. Cullen, Bryan G. Allen, Heather A. Brown, Sandy Vollstedt, Kellie L. Bodeker, Daniel J. Berg, John M. Buatti, Douglas R. Spitz, Brianne R. O’Leary, Katherine Gibson-Corley, Juan Du, Brett A. Wagner, Garry R. Buettner, Samuel R. Schroeder, Justin G. Wilkes, and Matthew S. Alexander
- Abstract
Chemoradiation therapy is the mainstay for treatment of locally advanced, borderline resectable pancreatic cancer. Pharmacologic ascorbate (P-AscH−, i.e., intravenous infusions of ascorbic acid, vitamin C), but not oral ascorbate, produces high plasma concentrations capable of selective cytotoxicity to tumor cells. In doses achievable in humans, P-AscH− decreases the viability and proliferative capacity of pancreatic cancer via a hydrogen peroxide (H2O2)-mediated mechanism. In this study, we demonstrate that P-AscH− radiosensitizes pancreatic cancer cells but inhibits radiation-induced damage to normal cells. Specifically, radiation-induced decreases in clonogenic survival and double-stranded DNA breaks in tumor cells, but not in normal cells, were enhanced by P-AscH−, while radiation-induced intestinal damage, collagen deposition, and oxidative stress were also reduced with P-AscH− in normal tissue. We also report on our first-in-human phase I trial that infused P-AscH− during the radiotherapy “beam on.” Specifically, treatment with P-AscH− increased median overall survival compared with our institutional average (21.7 vs. 12.7 months, P = 0.08) and the E4201 trial (21.7 vs. 11.1 months). Progression-free survival in P-AscH−–treated subjects was also greater than our institutional average (13.7 vs. 4.6 months, P < 0.05) and the E4201 trial (6.0 months). Results indicated that P-AscH− in combination with gemcitabine and radiotherapy for locally advanced pancreatic adenocarcinoma is safe and well tolerated with suggestions of efficacy. Because of the potential effect size and minimal toxicity, our findings suggest that investigation of P-AscH− efficacy is warranted in a phase II clinical trial.Significance:These findings demonstrate that pharmacologic ascorbate enhances pancreatic tumor cell radiation cytotoxicity in addition to offering potential protection from radiation damage in normal surrounding tissue, making it an optimal agent for improving treatment of locally advanced pancreatic adenocarcinoma.
- Published
- 2023
- Full Text
- View/download PDF
36. Data from First-in-Human Phase I Clinical Trial of Pharmacologic Ascorbate Combined with Radiation and Temozolomide for Newly Diagnosed Glioblastoma
- Author
-
John M. Buatti, Douglas R. Spitz, Brian J. Smith, Mindi J. TenNapel, Garry R. Buettner, Brett A. Wagner, Daniel Berg, Joseph J. Cullen, Karra A. Jones, Joseph M. Caster, Steven N. Seyedin, Kranti A. Mapuskar, Matthew A. Howard, Jeremy D. Greenlee, Sandy Vollstedt, Nancy Hollenbeck, Heather Brown, Thomas Carlisle, Raymond Hohl, Sonia Sandhu, Varun Monga, Mark C. Smith, Kellie L. Bodeker, and Bryan G. Allen
- Abstract
Purpose:Standard treatment for glioblastoma (GBM) includes surgery, radiation therapy (RT), and temozolomide (TMZ), yielding a median overall survival (OS) of approximately 14 months. Preclinical models suggest that pharmacologic ascorbate (P-AscH−) enhances RT/TMZ antitumor effect in GBM. We evaluated the safety of adding P-AscH− to standard RT/TMZ therapy.Patients and Methods:This first-in-human trial was divided into an RT phase (concurrent RT/TMZ/P-AscH−) and an adjuvant (ADJ) phase (post RT/TMZ/P-AscH− phase). Eight P-AscH− dose cohorts were evaluated in the RT phase until targeted plasma ascorbate levels were achieved (≥20 mmol/L). In the ADJ phase, P-AscH− doses were escalated in each subject at each cycle until plasma concentrations were ≥20 mmol/L. P-AscH− was infused 3 times weekly during the RT phase and 2 times weekly during the ADJ phase continuing for six cycles or until disease progression. Adverse events were quantified by CTCAE (v4.03).Results:Eleven subjects were evaluable. No dose-limiting toxicities occurred. Observed toxicities were consistent with historical controls. Adverse events related to study drug were dry mouth and chills. Targeted ascorbate plasma levels of 20 mmol/L were achieved in the 87.5 g cohort; diminishing returns were realized in higher dose cohorts. Median progression-free survival (PFS) was 9.4 months and median OS was 18 months. In subjects with undetectable MGMT promoter methylation (n = 8), median PFS was 10 months and median OS was 23 months.Conclusions:P-AscH−/RT/TMZ is safe with promising clinical outcomes warranting further investigation.
- Published
- 2023
- Full Text
- View/download PDF
37. Supplementary Methods, Table 1, Figures 1-7 from Manganese Superoxide Dismutase Regulates a Metabolic Switch during the Mammalian Cell Cycle
- Author
-
Prabhat C. Goswami, Garry R. Buettner, Philip Reigan, Sujatha Venkataraman, Leena Chaudhuri, Timothy D. Veenstra, Zhen Xiao, Amanda L. Kalen, and Ehab H. Sarsour
- Abstract
PDF file - 964K, Results presented in this section show: (1) A minimal difference in cell cycle phase distributions in MnSOD (+/+), and (-/-) MEFs following re-entry into the proliferative cycle (Table I) (2) An increase in MnSOD protein levels and activity in MnSOD overexpressing MB231 human mammary epithelial cancer cells (Figure 1) (3) An inhibition of MnSOD activity in quiescent normal human fibroblasts was associated with an increase in glucose consumption and percent S-phase (Figure 2) (4) Cell cycle phase-associated increase in cellular ROS levels was absent in MnSOD (-/-) MEFs (Figure 3) (5) Cell cycle phase specific increase in DHE-fluorescence is primarily due to an increase in cellular steady-state levels of superoxide (Figure 4) (6) Mass spectrometry results identifying lysine and arginine methylation pattern of MnSOD during quiescence and proliferation; species conservation of lysine and arginine methylation sites in MnSOD (Figure 5) (7) Site directed mutagenesis approach to mutate lysine 89 and 202 of MnSOD, and engineer expression vectors of wild-type and K-to-A mutant carrying human MnSOD cDNAs (Figure 6). (8) Computer modeling of MnSOD-methylation pattern (Figure 7)
- Published
- 2023
- Full Text
- View/download PDF
38. Supplementary Figure Legend, Tables 1 - 2 from Manganoporphyrins Increase Ascorbate-Induced Cytotoxicity by Enhancing H2O2 Generation
- Author
-
Joseph J. Cullen, Garry R. Buettner, Zita A. Sibenaller, Juan Du, Anna M. Button, Jessemae L. Welsh, Cameron M. Cushing, Brett A. Wagner, Samuel R. Schroeder, and Malvika Rawal
- Abstract
PDF file - 102K, Table 1. AscH- and MnT4MPyP are synergistic as seen by Combination Index (CI) and Dose Reduction Index (DRI). Table 2. Summary statistics for tumor volume measurements in the generalized estimating equations analysis.
- Published
- 2023
- Full Text
- View/download PDF
39. Supplemental Figure S5 from Pharmacological Ascorbate Radiosensitizes Pancreatic Cancer
- Author
-
Joseph J. Cullen, Garry R. Buettner, Douglas R. Spitz, Prabhat C. Goswami, James Mezhir, Susan Tsai, Brian Smith, Sarah L. Mott, Anna M. Button, Robert K. Strother, Claire M. Doskey, Amanda L. Kalen, Brett A. Wagner, Bryan G. Allen, Zita A. Sibenaller, Jessemae L. Welsh, John A. Cieslak, and Juan Du
- Abstract
Supplemental Figure S5. Bioluminescence imaging microscopy.
- Published
- 2023
- Full Text
- View/download PDF
40. Supplemental Table S2 from Pharmacological Ascorbate Radiosensitizes Pancreatic Cancer
- Author
-
Joseph J. Cullen, Garry R. Buettner, Douglas R. Spitz, Prabhat C. Goswami, James Mezhir, Susan Tsai, Brian Smith, Sarah L. Mott, Anna M. Button, Robert K. Strother, Claire M. Doskey, Amanda L. Kalen, Brett A. Wagner, Bryan G. Allen, Zita A. Sibenaller, Jessemae L. Welsh, John A. Cieslak, and Juan Du
- Abstract
Supplemental Table S2. Summary statistics for the survival analysis.
- Published
- 2023
- Full Text
- View/download PDF
41. Data from Pharmacological Ascorbate Radiosensitizes Pancreatic Cancer
- Author
-
Joseph J. Cullen, Garry R. Buettner, Douglas R. Spitz, Prabhat C. Goswami, James Mezhir, Susan Tsai, Brian Smith, Sarah L. Mott, Anna M. Button, Robert K. Strother, Claire M. Doskey, Amanda L. Kalen, Brett A. Wagner, Bryan G. Allen, Zita A. Sibenaller, Jessemae L. Welsh, John A. Cieslak, and Juan Du
- Abstract
The toxicity of pharmacologic ascorbate is mediated by the generation of H2O2 via the oxidation of ascorbate. Because pancreatic cancer cells are sensitive to H2O2 generated by ascorbate, they would also be expected to become sensitized to agents that increase oxidative damage such as ionizing radiation. The current study demonstrates that pharmacologic ascorbate enhances the cytotoxic effects of ionizing radiation as seen by decreased cell viability and clonogenic survival in all pancreatic cancer cell lines examined, but not in nontumorigenic pancreatic ductal epithelial cells. Ascorbate radiosensitization was associated with an increase in oxidative stress–induced DNA damage, which was reversed by catalase. In mice with established heterotopic and orthotopic pancreatic tumor xenografts, pharmacologic ascorbate combined with ionizing radiation decreased tumor growth and increased survival, without damaging the gastrointestinal tract or increasing systemic changes in parameters indicative of oxidative stress. Our results demonstrate the potential clinical utility of pharmacologic ascorbate as a radiosensitizer in the treatment of pancreatic cancer. Cancer Res; 75(16); 3314–26. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
42. Supplementary Figure 1 from Manganoporphyrins Increase Ascorbate-Induced Cytotoxicity by Enhancing H2O2 Generation
- Author
-
Joseph J. Cullen, Garry R. Buettner, Zita A. Sibenaller, Juan Du, Anna M. Button, Jessemae L. Welsh, Cameron M. Cushing, Brett A. Wagner, Samuel R. Schroeder, and Malvika Rawal
- Abstract
PDF file - 123K, Supplementary Figure 1. AsPC-1 and Panc-1 cells treated with MnT2EPyP or MnTBAP in combination with AscH-.
- Published
- 2023
- Full Text
- View/download PDF
43. The relationship between vitamin C status, the gut-liver axis, and metabolic syndrome
- Author
-
Maret G. Traber, Garry R. Buettner, and Richard S. Bruno
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Metabolic syndrome (MetS) is a constellation of cardiometabolic risk factors, which together predict increased risk of more serious chronic diseases. We propose that one consequence of dietary overnutrition is increased abundance of Gram-negative bacteria in the gut that cause increased inflammation, impaired gut function, and endotoxemia that further dysregulate the already compromised antioxidant vitamin status in MetS. This discussion is timely because “healthy” individuals are no longer the societal norm and specialized dietary requirements are needed for the growing prevalence of MetS. Further, these lines of evidence provide the foundational basis for investigation that poor vitamin C status promotes endotoxemia, leading to metabolic dysfunction that impairs vitamin E trafficking through a mechanism involving the gut-liver axis. This report will establish a critical need for translational research aimed at validating therapeutic approaches to manage endotoxemia—an early, but inflammation-inducing phenomenon, which not only occurs in MetS, but is also prognostic of more advanced metabolic disorders including type 2 diabetes mellitus, as well as the increasing severity of nonalcoholic fatty liver diseases.
- Published
- 2019
- Full Text
- View/download PDF
44. Understanding the Redox Biology of Selenium in the Search of Targeted Cancer Therapies
- Author
-
Jeffrey M. Stolwijk, Rohan Garje, Jessica C. Sieren, Garry R. Buettner, and Yousef Zakharia
- Subjects
selenium ,selenomethionine ,selenoproteins ,cancer ,glutathione peroxidases ,thioredoxin reductases ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Selenium (Se) is an essential trace nutrient required for optimal human health. It has long been suggested that selenium has anti-cancer properties. However, clinical trials have shown inconclusive results on the potential of Se to prevent cancer. The suggested role of Se in the prevention of cancer is centered around its role as an antioxidant. Recently, the potential of selenium as a drug rather than a supplement has been uncovered. Selenium compounds can generate reactive oxygen species that could enhance the treatment of cancer. Transformed cells have high oxidative distress. As normal cells have a greater capacity to meet oxidative challenges than tumor cells, increasing the flux of oxidants with high dose selenium treatment could result in cancer-specific cell killing. If the availability of Se is limited, supplementation of Se can increase the expression and activities of Se-dependent proteins and enzymes. In cell culture, selenium deficiency is often overlooked. We review the importance of achieving normal selenium biology and how Se deficiency can lead to adverse effects. We examine the vital role of selenium in the prevention and treatment of cancer. Finally, we examine the properties of Se-compounds to better understand how each can be used to address different research questions.
- Published
- 2020
- Full Text
- View/download PDF
45. TREM-1 is required for enhanced OpZ-induced superoxide generation following priming
- Author
-
Shubha Murthy, Sankar Baruah, Jayden L Bowen, Kathy Keck, Brett A Wagner, Garry R Buettner, David B Sykes, and Julia Klesney-Tait
- Subjects
Mice ,Neutrophils ,Superoxides ,Immunology ,Zymosan ,Animals ,Immunology and Allergy ,Cell Biology ,Triggering Receptor Expressed on Myeloid Cells-1 ,Respiratory Burst - Abstract
Inflammatory agents, microbial products, or stromal factors pre-activate or prime neutrophils to respond to activating stimuli in a rapid and aggressive manner. Primed neutrophils exhibit enhanced chemotaxis, phagocytosis, and respiratory burst when stimulated by secondary activating stimuli. We previously reported that Triggering Receptor Expressed on Myeloid cells-1 (TREM-1) mediates neutrophil effector functions such as increased superoxide generation, transepithelial migration, and chemotaxis. However, it is unclear whether TREM-1 is required for the process of priming itself or for primed responses to subsequent stimulation. To investigate this, we utilized in vitro and in vivo differentiated neutrophils that were primed with TNF-α and then stimulated with the particulate agonist, opsonized zymosan (OpZ). Bone marrow progenitors isolated from WT and Trem-1–/– mice were transduced with estrogen regulated Homeobox8 (ER-Hoxb8) fusion transcription factor and differentiated in vitro into neutrophils following estrogen depletion. The resulting neutrophils expressed high levels of TREM-1 and resembled mature in vivo differentiated neutrophils. The effects of priming on phagocytosis and oxidative burst were determined. Phagocytosis did not require TREM-1 and was not altered by priming. In contrast, priming significantly enhanced OpZ-induced oxygen consumption and superoxide production in WT but not Trem-1–/– neutrophils indicating that TREM-1 is required for primed oxidative burst. TREM-1-dependent effects were not mediated during the process of priming itself as priming enhanced degranulation, ICAM-1 shedding, and IL-1ß release to the same extent in WT and Trem-1–/- neutrophils. Thus, TREM-1 plays a critical role in primed phagocytic respiratory burst and mediates its effects following priming.
- Published
- 2022
- Full Text
- View/download PDF
46. Hydrogen Peroxide Mediates Artemisinin-Derived C-16 Carba-Dimer-Induced Toxicity of Human Cancer Cells
- Author
-
Amanda L. Kalen, Brett A. Wagner, Ehab H. Sarsour, Maneesh G. Kumar, Jessica L. Reedy, Garry R. Buettner, Nabin C. Barua, and Prabhat C. Goswami
- Subjects
artemisinin ,carba-dimer ,aliphatic nitro chemistry ,oxidative stress ,cyclin d1 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
This study used a nitroaliphatic chemistry approach to synthesize a novel artemisinin-derived carba-dimer (AG-1) and determined its anti-proliferative effects in human normal and cancer cells. AG-1 treatments selectively inhibit proliferation of cancer cells compared to normal human fibroblasts. Compared to artemisinin, AG-1 is more toxic to human breast, prostate, head−neck, pancreas and skin cancer cells; 50% inhibition (IC50) 123 µM in AG-1 vs. 290 µM in artemisinin-treated breast cancer cells. AG-1 treatment decreased (~5 folds) cyclin D1 protein expression that correlated with an increase in the percentage of cells in the G1-phase, suggesting a G1 delay. AG-1-induced toxicity was independent of the DNA damage at 72 h post-treatment, as measured by micronuclei frequency and γH2AX protein levels. Results from electron paramagnetic resonance spectroscopy showed Fe-catalyzed formation of AG-1 carbon-centered radicals in a cell-free system. Flow cytometry analysis of H2DCF-DA oxidation showed a significant increase in the steady-state levels of reactive oxygen species (ROS) in AG-1-treated cells. Pre-treatment with N-acetyl-l-cysteine and antioxidant enzymes (superoxide dismutase and catalase) significantly suppressed AG-1-induced toxicity, suggesting that superoxide and hydrogen peroxide contribute to AG-1-induced toxicity in human cancer cells. AG-1 represents a novel class of anti-cancer drug that is more potent than its parent compound, artemisinin.
- Published
- 2020
- Full Text
- View/download PDF
47. Prolonged Reactive Oxygen Species Production following Septic Insult
- Author
-
Vladimir P. Badovinac, Garry R. Buettner, Isaac J. Jensen, Roger R. Berton, Patrick W. McGonagill, Thomas S. Griffith, Elvia E. Silva, and Brett A. Wagner
- Subjects
Adult ,Male ,Antioxidant ,Adolescent ,medicine.medical_treatment ,Lymphocyte ,Immunology ,Ascorbic Acid ,Severity of Illness Index ,Antioxidants ,Article ,Immature Monocyte ,Sepsis ,Mice ,Young Adult ,Immune system ,Disease severity ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Aged ,Aged, 80 and over ,chemistry.chemical_classification ,Reactive oxygen species ,Vitamin C ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,Healthy Volunteers ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Case-Control Studies ,Leukocytes, Mononuclear ,Female ,Reactive Oxygen Species ,business - Abstract
The dysregulated host response and organ damage following systemic infection that characterizes a septic event predisposes individuals to a chronic immunoparalysis state associated with severe transient lymphopenia and diminished lymphocyte function, thereby reducing long-term patient survival and quality of life. Recently, we observed lasting production of reactive oxygen species (ROS) in mice that survive sepsis. ROS production is a potent mechanism for targeting infection, but excessive ROS production can prove maladaptive by causing organ damage, impairing lymphocyte function, and promoting inflammaging, concepts paralleling sepsis-induced immunoparalysis. Notably, we observed an increased frequency of ROS-producing immature monocytes in septic hosts that was sustained for greater than 100 days postsurgery. Recent clinical trials have explored the use of vitamin C, a potent antioxidant, for treating septic patients. We observed that therapeutic vitamin C administration for sepsis limited ROS production by monocytes and reduced disease severity. Importantly, we also observed increased ROS production by immature monocytes in septic patients both at admission and ∼28 days later, suggesting a durable and conserved feature that may influence the host immune response. Thus, lasting ROS production by immature monocytes is present in septic patients, and early intervention strategies to reduce it may improve host outcomes, potentially reducing sepsis-induced immunoparalysis.
- Published
- 2021
- Full Text
- View/download PDF
48. An assay for the rate of removal of extracellular hydrogen peroxide by cells
- Author
-
Brett A. Wagner, Jordan R. Witmer, Thomas J. van't Erve, and Garry R. Buettner
- Subjects
Hydrogen peroxide ,Kinetics ,Erythrocyte ,Quantitative redox biology ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Cells have a wide range of capacities to remove extracellular hydrogen peroxide. At higher concentrations of extracellular H2O2 (micromolar) the rate of removal can be approximated by a rate equation that is first-order in the concentration of H2O2 and cell density. Here we present a method to determine the observed rate constant for the removal of extracellular H2O2 on a per cell basis. In the cells examined, when exposed to 20 μM H2O2, these rate constants (kcell) range from 0.46×10−12 s−1 cell−1 L for Mia-PaCa-2 cells (human pancreatic carcinoma) to 10.4×10−12 s−1 cell−1 L for U937 cells (human histiocytic lymphoma). For the relatively small red blood cell kcell=2.9×10−12 s−1 cell−1 L. These rate constants, kcell, can be used to compare the capacity of cells to remove higher levels of extracellular H2O2, as often presented in cell culture experiments. They also provide a means to estimate the rate of removal of extracellular H2O2, rate=−kcell [H2O2] (cells L−1), and the half-life of a bolus of H2O2. This information is essential to optimize experimental design and interpret data from experiments that expose cells to extracellular H2O2.
- Published
- 2013
- Full Text
- View/download PDF
49. The latency of peroxisomal catalase in terms of effectiveness factor for pancreatic and glioblastoma cancer cell lines in the presence of high concentrations of H2O2: Implications for the use of pharmacological ascorbate in cancer therapy
- Author
-
Garry R. Buettner, Dieanira Erudaitius, and Victor G. J. Rodgers
- Subjects
0301 basic medicine ,Biochemistry & Molecular Biology ,Cell ,Antineoplastic Agents ,Medical Biochemistry and Metabolomics ,Biochemistry ,Cell Line ,Medicinal and Biomolecular Chemistry ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Physiology (medical) ,Pancreatic cancer ,medicine ,Extracellular ,Humans ,Latency (engineering) ,Cancer ,Tumor ,biology ,Chemistry ,Hydrogen Peroxide ,Peroxisome ,Catalase ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Cancer cell ,Cancer research ,biology.protein ,Biochemistry and Cell Biology ,Glioblastoma ,Digestive Diseases ,030217 neurology & neurosurgery - Abstract
Previous research has identified variation in cancer cell line response to high levels of extracellular H2O2 (eH2O2) exposure. This directly contributes to our understanding cellular efficacy of pharmacological ascorbate (P-AscH-) therapy. Here we investigate the factors contributing to latency of peroxisomal catalase of a cell and the importance of latency in evaluating cell exposure to eH2O2. First, we develop a mathematical framework for the latency of catalase in terms of an effectiveness factor, ηeff, to describe the catalase activity in the presence of high levels of eH2O2. A simplified relationship emerges, [Formula: see text] when mprp/Dij≪1, where mp,rp, and [Formula: see text] are the experimentally determined peroxisome permeability, average peroxisome radius, and the pseudo first-order reaction rate constant, respectively. [Formula: see text] is the catalase concentration in the peroxisome and k2=1.7x107M-1s-1. Next, previously published parameters are used to determine the latency effect of the cell lines: normal pancreatic cells (H6c7), pancreatic cancer cells (MIA PaCa-2), and glioblastoma cells (LN-229, T98G, and U-87), all which vary in their susceptibility to exposure to high eH2O2. The results show that effectiveness is not significantly different except for the most susceptible, MIA PaCa-2cell line, which is higher when compared to all other cell lines. This result is counterintuitive and further implies that latency, as a single parameter, is ineffective in forecasting cell line susceptibility to P-AscH- therapy equivalent eH2O. Thus, further research remains necessary to identify why cancer cells vary in susceptibility to P-AscH- therapy.
- Published
- 2020
- Full Text
- View/download PDF
50. Disulfiram causes selective hypoxic cancer cell toxicity and radio-chemo-sensitization via redox cycling of copper
- Author
-
Jeffrey M. Stolwijk, Ann Tomanek-Chalkley, Mengshi Li, Samuel N. Rodman, Michael L. McCormick, Garry R. Buettner, Kelly C. Falls-Hubert, Michael K. Schultz, Michael G. Anderson, Val C. Sheffield, Bryan G. Allen, Kai Gui, Shane R. Solst, Brian Wels, Hartmut Schmidt, Vanessa Sandfort, Aimee L. Butler, Douglas R. Spitz, and Charles Searby
- Subjects
0301 basic medicine ,Lung Neoplasms ,medicine.disease_cause ,Biochemistry ,Article ,Superoxide dismutase ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Lipid oxidation ,Cell Line, Tumor ,Physiology (medical) ,Disulfiram ,medicine ,Humans ,Hypoxia ,biology ,Superoxide ,Carboplatin ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Toxicity ,Cancer cell ,Cancer research ,biology.protein ,Oxidation-Reduction ,Copper ,Oxidative stress ,medicine.drug - Abstract
Therapy for lung cancer patients includes surgery, radiation, and chemotherapy. While treatments initially illicit desirable responses, 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 versus 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) is selectively toxic to lung cancer cells versus normal bronchial epithelial cells. Furthermore, cancer cell toxicity is exacerbated at 1% O2, relative to 4 or 21% O2. This selective toxicity of DSF/Cu can be attributed to its differential Cu ionophore capabilities. DSF/Cu treatment significantly increased total cellular Cu levels, with higher Cu present in tumor versus normal cells and in cancer cells at 1% O2 versus 21% O2. DSF toxicity was shown to be dependent on Cu by inhibiting toxicity with a Cu chelator, bathocuproinedisulfonic acid (BCS), which inhibits redox cycling. Toxicity was also shown to be dependent on oxidative stress mechanisms that are initiated by Cu, including the production of superoxide and peroxide, which were inhibited by a superoxide dismutase (SOD) mimetic, GC4419, and overexpression of catalase. DSF/Cu treatment also induced lipid oxidation, increased mitochondrial superoxide production, and decreased mitochondrial membrane potential, which were detected by C11-BODIPY, MitoSox, and JC-1. In addition to its selective toxicity to hypoxic cancer cells, DSF/Cu also enhanced radio-chemotherapy-induced cancer cell killing and reduced therapy resistance at hypoxia. DSF also decreased xenograft tumor growth in vivo when combined with radiation and carboplatin. These results support the hypothesis that DSF is a promising adjuvant for cancer therapy based on its apparent ability to selectively target fundamental differences in cancer cell oxidative metabolism.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.