39 results on '"Cynthia S. Lancaster"'
Search Results
2. Supplementary Figure 1 from Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Alex Sparreboom, Sharyn D. Baker, Jeffrey E. Rubnitz, Zhaoyuan Chen, Shuiying Hu, Zhili Zuo, Cynthia S. Lancaster, and Chaoxin Hu
- Abstract
PDF file - 46K
- Published
- 2023
3. Supplementary Figure 3 from Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Alex Sparreboom, Sharyn D. Baker, Jeffrey E. Rubnitz, Zhaoyuan Chen, Shuiying Hu, Zhili Zuo, Cynthia S. Lancaster, and Chaoxin Hu
- Abstract
PDF file - 255K
- Published
- 2023
4. Supplementary Figure Legends 1-5 from Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Alex Sparreboom, Sharyn D. Baker, Jeffrey E. Rubnitz, Zhaoyuan Chen, Shuiying Hu, Zhili Zuo, Cynthia S. Lancaster, and Chaoxin Hu
- Abstract
PDF file - 53K
- Published
- 2023
5. Data from Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Alex Sparreboom, Sharyn D. Baker, Jeffrey E. Rubnitz, Zhaoyuan Chen, Shuiying Hu, Zhili Zuo, Cynthia S. Lancaster, and Chaoxin Hu
- Abstract
OCTN2 is a bifunctional transporter that reabsorbs filtered carnitine in a sodium-dependent manner and secretes organic cations into urine as a proton antiport mechanism. We hypothesized that inhibition of OCTN2 by anticancer drugs can influence carnitine resorption. OCTN2-mediated transport inhibition by anticancer drugs was assessed using cells transfected with human OCTN2 (hOCTN2) or mouse Octn2 (mOctn2). Excretion of carnitine and acetylcarnitine was measured in urine collected from mice and pediatric patients with cancer before and after administration of etoposide. Five of 27 tested drugs (50–100 μmol/L) inhibited hOCTN2-mediated carnitine uptake by 42% to 85% (P < 0.001). Of these inhibitors, etoposide was itself a transported substrate of hOCTN2 and mOctn2. Etoposide uptake by hOCTN2 was reversed in the presence of excess carnitine. This competitive inhibitory mechanism was confirmed in an in silico molecular docking analysis. In addition, etoposide inhibited the transcellular apical-to-basolateral flux of carnitine in kidney cells. Etoposide was also associated with a significant urinary loss of carnitine in mice (∼1.5-fold) and in patients with cancer (∼2.4-fold). Collectively, these findings indicate that etoposide can inhibit hOCTN2 function, potentially disturb carnitine homeostasis, and that this phenomenon can contribute to treatment-related toxicities. Mol Cancer Ther; 11(4); 921–9. ©2012 AACR.
- Published
- 2023
6. Supplementary Figure 2 from Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Alex Sparreboom, Sharyn D. Baker, Jeffrey E. Rubnitz, Zhaoyuan Chen, Shuiying Hu, Zhili Zuo, Cynthia S. Lancaster, and Chaoxin Hu
- Abstract
PDF file - 107K
- Published
- 2023
7. Supplementary Figure 4 from Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Alex Sparreboom, Sharyn D. Baker, Jeffrey E. Rubnitz, Zhaoyuan Chen, Shuiying Hu, Zhili Zuo, Cynthia S. Lancaster, and Chaoxin Hu
- Abstract
PDF file - 172K
- Published
- 2023
8. Supplementary Figure Legends from Cisplatin-Induced Renal Injury Is Independently Mediated by OCT2 and p53
- Author
-
Alex Sparreboom, Giuliano Ciarimboli, Laura J. Janke, Eberhard Schlatter, Dorothea Zeeh, Lie Li, Alice A. Gibson, Ashley M. Kosloske, Edwin Hermann, Navjotsingh Pabla, Cynthia S. Lancaster, and Jason A. Sprowl
- Abstract
PDF file - 181K
- Published
- 2023
9. Supplementary Figure 2 from Cisplatin-Induced Renal Injury Is Independently Mediated by OCT2 and p53
- Author
-
Alex Sparreboom, Giuliano Ciarimboli, Laura J. Janke, Eberhard Schlatter, Dorothea Zeeh, Lie Li, Alice A. Gibson, Ashley M. Kosloske, Edwin Hermann, Navjotsingh Pabla, Cynthia S. Lancaster, and Jason A. Sprowl
- Abstract
PDF file - 339K, Comparative cisplatin-induced nephrotoxicity and urinary elimination of cisplatin in FVB and C57BL/6 mice.
- Published
- 2023
10. Data from Influence of Oct1/Oct2-Deficiency on Cisplatin-Induced Changes in Urinary N-Acetyl-β-D-Glucosaminidase
- Author
-
Alex Sparreboom, Ron H. Mathijssen, Oliver Zolk, Chaoxin Hu, Kelly K. Filipski, Cynthia S. Lancaster, Ashley M. Kosloske, and Ryan M. Franke
- Abstract
Purpose: This study aimed to test the influence of functional renal organic cation transporters (OCT2 in humans, Oct1 and Oct2 in mice) on biomarkers of cisplatin nephrotoxicity, such as urinary activity of N-acetyl-β-D-glucosaminidase (NAG).Experimental Design: Temporal cisplatin-induced nephrotoxicity was assessed by histopathology and biomarkers. Cisplatin-mediated NAG changes and survival were determined in wild-type and Oct1/2(-/-) mice. Identification of OCT2 inhibitors was done in transfected 293Flp-In cells, and the NCI60 cell line panel was used to assess contribution of OCT2 to cisplatin uptake in cancer cells.Results: Classical biomarkers such as blood urea nitrogen and serum creatinine were not elevated until 72 hours after cisplatin administration and substantial kidney damage had occurred. Oct1/2(-/-) mice had 2.9-fold lower NAG by 4 hours (P < 0.0001) and 2.3-fold increased survival (P = 0.0097). Among 16 agents, cimetidine strongly inhibited uptake of tetraethylammonium bromide (P = 0.0006) and cisplatin (P < 0.0001), but did not have an influence on cisplatin uptake in SK-OV-3 cells, the cancer line with the highest OCT2 mRNA levels. In wild-type mice, cimetidine inhibited cisplatin-induced NAG changes (P = 0.016 versus cisplatin alone) to a degree similar to that seen in Oct1/2(-/-) mice receiving cisplatin (P = 0.91). Cumulative NAG activity of >0.4 absorbance units (AU) was associated with 21-fold increased odds for severe nephrotoxicity (P = 0.0017), which was linked with overall survival (hazard ratio, 8.1; 95% confidence interval, 2.1-31; P = 0.0078).Conclusions: Cimetidine is able to inhibit OCT2-mediated uptake of cisplatin in the kidney, and subsequently ameliorate nephrotoxicity likely with minimal effect on uptake in tumor cells. Clin Cancer Res; 16(16); 4198–206. ©2010 AACR.
- Published
- 2023
11. Supplementary Figure 3 from Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Alex Sparreboom, Edwin Herrmann, Mary V. Relling, Ching-Hon Pui, Wenjian Yang, Ron H. J. Mathijssen, Denise Guckel, Vivian Massmann, Hermann Pavenstädt, Jason A. Sprowl, Ryan M. Franke, Eberhard Schlatter, Cynthia S. Lancaster, and Giuliano Ciarimboli
- Abstract
PDF file - 40K
- Published
- 2023
12. Data from Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Alex Sparreboom, Edwin Herrmann, Mary V. Relling, Ching-Hon Pui, Wenjian Yang, Ron H. J. Mathijssen, Denise Guckel, Vivian Massmann, Hermann Pavenstädt, Jason A. Sprowl, Ryan M. Franke, Eberhard Schlatter, Cynthia S. Lancaster, and Giuliano Ciarimboli
- Abstract
Purpose: Knowledge of transporters responsible for the renal secretion of creatinine is key to a proper interpretation of serum creatinine and/or creatinine clearance as markers of renal function in cancer patients receiving chemotherapeutic agents.Experimental Design: Creatinine transport was studied in transfected HEK293 cells in vitro and in wild-type mice and age-matched organic cation transporter 1 and 2–deficient [Oct1/2(−/−)] mice ex vivo and in vivo. Clinical pharmacogenetic and transport inhibition studies were done in two separate cohorts of cancer patients.Results: Compared with wild-type mice, creatinine clearance was significantly impaired in Oct1/2(−/−) mice. Furthermore, creatinine inhibited organic cation transport in freshly isolated proximal tubules from wild-type mice and humans, but not in those from Oct1/2(−/−) mice. In a genetic association analysis (n = 590), several polymorphisms around the OCT2/SLC22A2 gene locus, including rs2504954 (P = 0.000873), were significantly associated with age-adjusted creatinine levels. Furthermore, in cancer patients (n = 68), the OCT2 substrate cisplatin caused an acute elevation of serum creatinine (P = 0.0083), consistent with inhibition of an elimination pathway.Conclusions: Collectively, this study shows that OCT2 plays a decisive role in the renal secretion of creatinine. This process can be inhibited by OCT2 substrates, which impair the usefulness of creatinine as a marker of renal function. Clin Cancer Res; 18(4); 1101–8. ©2012 AACR.
- Published
- 2023
13. Supplementary Figure 1 from Cisplatin-Induced Renal Injury Is Independently Mediated by OCT2 and p53
- Author
-
Alex Sparreboom, Giuliano Ciarimboli, Laura J. Janke, Eberhard Schlatter, Dorothea Zeeh, Lie Li, Alice A. Gibson, Ashley M. Kosloske, Edwin Hermann, Navjotsingh Pabla, Cynthia S. Lancaster, and Jason A. Sprowl
- Abstract
PDF file - 117K, Pathways implicated in cisplatin-induced renal toxicity.
- Published
- 2023
14. Supplementary Figure 2 from Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Alex Sparreboom, Edwin Herrmann, Mary V. Relling, Ching-Hon Pui, Wenjian Yang, Ron H. J. Mathijssen, Denise Guckel, Vivian Massmann, Hermann Pavenstädt, Jason A. Sprowl, Ryan M. Franke, Eberhard Schlatter, Cynthia S. Lancaster, and Giuliano Ciarimboli
- Abstract
PDF file - 29K
- Published
- 2023
15. Supplementary Figure 1 from Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Alex Sparreboom, Edwin Herrmann, Mary V. Relling, Ching-Hon Pui, Wenjian Yang, Ron H. J. Mathijssen, Denise Guckel, Vivian Massmann, Hermann Pavenstädt, Jason A. Sprowl, Ryan M. Franke, Eberhard Schlatter, Cynthia S. Lancaster, and Giuliano Ciarimboli
- Abstract
PDF file - 52K
- Published
- 2023
16. Data from Influence of Polymorphic OATP1B-Type Carriers on the Disposition of Docetaxel
- Author
-
Alex Sparreboom, Ron H. J. Mathijssen, Sharyn D. Baker, Ron H. N. van Schaik, Walter J. Loos, Guoqing Du, Aisha L. Walker, Torben S. Mikkelsen, Thomas J. Corydon, Gitte H. Bruun, Alice A. Gibson, Shuiying Hu, Peter de Bruijn, Lena E. Friberg, Laure Elens, Bruno Hagenbuch, Amanda Obaidat, Cynthia S. Lancaster, and Anne-Joy M. de Graan
- Abstract
Purpose: Docetaxel is extensively metabolized by CYP3A4 in the liver but mechanisms by which the drug is taken up into hepatocytes remain poorly understood. We hypothesized that (i) liver uptake of docetaxel is mediated by the polymorphic solute carriers OATP1B1 and OATP1B3 and (ii) inherited genetic defects in this process may impair systemic drug elimination.Experimental Design: Transport of docetaxel was studied in vitro using various cell lines stably transfected with OATP1B1*1A (wild-type), OATP1B1*5 [c.521T>C (V174A); rs4149056], OATP1B3, or the mouse transporter Oatp1b2. Docetaxel clearance was evaluated in wild-type and Oatp1b2-knockout mice as well as in two cohorts of patients with multiple variant transporter genotypes (n = 213).Results: Docetaxel was found to be a substrate for OATP1B1, OATP1B3, and Oatp1b2 but was not transported by OATP1B1*5. Deficiency of Oatp1b2 in mice was associated with an 18-fold decrease in docetaxel clearance (P = 0.0099), which was unrelated to changes in intrinsic metabolic capacity in mouse liver microsomes. In patients, however, none of the studied common reduced function variants in OATP1B1 or OATP1B3 were associated with docetaxel clearance (P > 0.05).Conclusions: The existence of at least two potentially redundant uptake transporters in the human liver with similar affinity for docetaxel supports the possibility that functional defects in both of these proteins may be required to confer substantially altered disposition phenotypes. In view of the established exposure–toxicity relationships for docetaxel, we suggest that caution is warranted if docetaxel has to be administered together with agents that potently inhibit both OATP1B1 and OATP1B3. Clin Cancer Res; 18(16); 4433–40. ©2012 AACR.
- Published
- 2023
17. Supplementary Figure Legends 1-4 from Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Alex Sparreboom, Edwin Herrmann, Mary V. Relling, Ching-Hon Pui, Wenjian Yang, Ron H. J. Mathijssen, Denise Guckel, Vivian Massmann, Hermann Pavenstädt, Jason A. Sprowl, Ryan M. Franke, Eberhard Schlatter, Cynthia S. Lancaster, and Giuliano Ciarimboli
- Abstract
PDF file - 64K
- Published
- 2023
18. Data from Cisplatin-Induced Renal Injury Is Independently Mediated by OCT2 and p53
- Author
-
Alex Sparreboom, Giuliano Ciarimboli, Laura J. Janke, Eberhard Schlatter, Dorothea Zeeh, Lie Li, Alice A. Gibson, Ashley M. Kosloske, Edwin Hermann, Navjotsingh Pabla, Cynthia S. Lancaster, and Jason A. Sprowl
- Abstract
Purpose: Tubular secretion of cisplatin is abolished in mice deficient for the organic cation transporters Oct1 and Oct2 (Oct1/2−/−mice), and these animals are protected from severe cisplatin-induced kidney damage. Since tubular necrosis is not completely absent in Oct1/2−/−mice, we hypothesized that alternate pathways are involved in the observed injury.Experimental Design: Studies were done in wild-type, Oct1/2−/−, or p53-deficient animals, all on an FVB background, receiving cisplatin intraperitoneally at 15 mg/kg. Cisplatin metabolites were analyzed using mass spectrometry, and gene expression was assessed using Affymetrix microarrays and RT-PCR arrays.Results: KEGG pathway analyses on kidneys from mice exposed to cisplatin revealed that the most significantly altered genes were associated with the p53 signaling network, including Cdnk1a and Mdm2, in both wild-type (P = 2.40 × 10−11) and Oct1/2−/−mice (P = 1.92 × 10−8). This was confirmed by demonstrating that homozygosity for a p53-null allele partially reduced renal tubular damage, whereas loss of p53 in Oct1/2−/−mice (p53−/−/Oct1/2−/−) completely abolished nephrotoxicity. We found that pifithrin-α, an inhibitor of p53-dependent transcriptional activation, inhibits Oct2 and can mimic the lack of nephrotoxicity observed in p53−/−/Oct1/2−/−mice.Conclusions: These findings indicate that (i) the p53 pathway plays a crucial role in the kidney in response to cisplatin treatment and (ii) clinical exploration of OCT2 inhibitors may not lead to complete nephroprotection unless the p53 pathway is simultaneously antagonized. Clin Cancer Res; 20(15); 4026–35. ©2014 AACR.
- Published
- 2023
19. Supplementary Data from Influence of Oct1/Oct2-Deficiency on Cisplatin-Induced Changes in Urinary N-Acetyl-β-D-Glucosaminidase
- Author
-
Alex Sparreboom, Ron H. Mathijssen, Oliver Zolk, Chaoxin Hu, Kelly K. Filipski, Cynthia S. Lancaster, Ashley M. Kosloske, and Ryan M. Franke
- Abstract
Supplementary Figures S1-S2.
- Published
- 2023
20. Supplementary Figure 4 from Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Alex Sparreboom, Edwin Herrmann, Mary V. Relling, Ching-Hon Pui, Wenjian Yang, Ron H. J. Mathijssen, Denise Guckel, Vivian Massmann, Hermann Pavenstädt, Jason A. Sprowl, Ryan M. Franke, Eberhard Schlatter, Cynthia S. Lancaster, and Giuliano Ciarimboli
- Abstract
PDF file - 42K
- Published
- 2023
21. Supplementary Table 1 from Cisplatin-Induced Renal Injury Is Independently Mediated by OCT2 and p53
- Author
-
Alex Sparreboom, Giuliano Ciarimboli, Laura J. Janke, Eberhard Schlatter, Dorothea Zeeh, Lie Li, Alice A. Gibson, Ashley M. Kosloske, Edwin Hermann, Navjotsingh Pabla, Cynthia S. Lancaster, and Jason A. Sprowl
- Abstract
PDF file - 70K, Changes in gene expression of 25 genes associated with the p53 signaling pathway in kidneys isolated from wildtype and Oct1/2(-/-) FVB mice exposed to cisplatin (15 mg/kg cisplatin; i.p.).
- Published
- 2023
22. Supplementary Methods, Figures 1 - 4, Tables 1 - 3 from Influence of Polymorphic OATP1B-Type Carriers on the Disposition of Docetaxel
- Author
-
Alex Sparreboom, Ron H. J. Mathijssen, Sharyn D. Baker, Ron H. N. van Schaik, Walter J. Loos, Guoqing Du, Aisha L. Walker, Torben S. Mikkelsen, Thomas J. Corydon, Gitte H. Bruun, Alice A. Gibson, Shuiying Hu, Peter de Bruijn, Lena E. Friberg, Laure Elens, Bruno Hagenbuch, Amanda Obaidat, Cynthia S. Lancaster, and Anne-Joy M. de Graan
- Abstract
PDF file, 437KB, Description of determination of docetaxel concentrations; influence of phenol red on OATP1B1-mediated transport of docetaxel; characterization of paclitaxel concentration-dependent transport by OATP1B1 and OATP1B3; docetaxel clearance as a function of observed OATP1B1 (SLCO1B1) or OATP1B3 (SLCO1B3) diplotypes and genotypes; description of genotyped variants in OATP1B1 (SLCO1B1) and OATP1B3 (SLCO1B3); summary of patient demographics; and examples of OATP1B1 and OATP1B3 inhibitors.
- Published
- 2023
23. OATP1B1 polymorphism as a determinant of erythromycin disposition
- Author
-
Ron H.J. Mathijssen, Sharyn D. Baker, Cody J. Peer, Shuiying Hu, Gitte H. Bruun, Cynthia S. Lancaster, Thomas J. Corydon, William D. Figg, Torben Stamm Mikkelsen, Alice A. Gibson, Shelley Orwick, Alex Sparreboom, and Medical Oncology
- Subjects
Adult ,Male ,Organic anion transporter 1 ,Organic Anion Transporters ,Erythromycin ,Organic Anion Transporters, Sodium-Independent ,Pharmacology ,Article ,Cell Line ,Mice ,Cytochrome P-450 CYP3A ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Aged ,Aged, 80 and over ,Mice, Knockout ,Polymorphism, Genetic ,biology ,CYP3A4 ,Liver-Specific Organic Anion Transporter 1 ,Wild type ,Biological Transport ,Transporter ,Transfection ,Middle Aged ,Molecular biology ,Anti-Bacterial Agents ,Cell culture ,biology.protein ,Female ,medicine.drug - Abstract
Previous studies have demonstrated that the pharmacokinetic profile of erythromycin, a probe for CYP3A4 activity, is affected by inhibitors or inducers of hepatic solute carriers. We hypothesized that these interactions are mediated by OATP1B1 (gene symbol, SLCO1B1), a polypeptide expressed on the basolateral surface of hepatocytes. Using stably transfected Flp-In T-Rex293 cells, erythromycin was found to be a substrate for OATP1B1*1A (wild type) with a Michaelis-Menten constant of ~13 µmol/l, and that its transport was reduced by ~50% in cells expressing OATP1B1*5 (V174A). Deficiency of the ortholog transporter Oatp1b2 in mice was associated with a 52% decrease in the metabolic rate of erythromycin (P = 0.000043). In line with these observations, in humans the c.521T>C variant in SLCO1B1 (rs4149056), encoding OATP1B1*5, was associated with a decline in erythromycin metabolism (P = 0.0072). These results suggest that impairment of OATP1B1 function can alter erythromycin metabolism, independent of changes in CYP3A4 activity.
- Published
- 2012
24. Inhibition of OCTN2-Mediated Transport of Carnitine by Etoposide
- Author
-
Shuiying Hu, Jeffrey E. Rubnitz, Cynthia S. Lancaster, Zhaoyuan Chen, Sharyn D. Baker, Zhili Zuo, Alex Sparreboom, and Chaoxin Hu
- Subjects
Male ,Cancer Research ,Adolescent ,Organic Cation Transport Proteins ,Swine ,Cell Culture Techniques ,Pharmacology ,Transfection ,Article ,Cell Line ,Excretion ,Mice ,Carnitine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Transcellular ,Child ,Solute Carrier Family 22 Member 5 ,Acetylcarnitine ,Etoposide ,Kidney ,Organic cation transport proteins ,biology ,Biological Transport ,Antineoplastic Agents, Phytogenic ,Leukemia, Myeloid, Acute ,HEK293 Cells ,medicine.anatomical_structure ,Oncology ,Mediated transport ,biology.protein ,medicine.drug - Abstract
OCTN2 is a bifunctional transporter that reabsorbs filtered carnitine in a sodium-dependent manner and secretes organic cations into urine as a proton antiport mechanism. We hypothesized that inhibition of OCTN2 by anticancer drugs can influence carnitine resorption. OCTN2-mediated transport inhibition by anticancer drugs was assessed using cells transfected with human OCTN2 (hOCTN2) or mouse Octn2 (mOctn2). Excretion of carnitine and acetylcarnitine was measured in urine collected from mice and pediatric patients with cancer before and after administration of etoposide. Five of 27 tested drugs (50–100 μmol/L) inhibited hOCTN2-mediated carnitine uptake by 42% to 85% (P < 0.001). Of these inhibitors, etoposide was itself a transported substrate of hOCTN2 and mOctn2. Etoposide uptake by hOCTN2 was reversed in the presence of excess carnitine. This competitive inhibitory mechanism was confirmed in an in silico molecular docking analysis. In addition, etoposide inhibited the transcellular apical-to-basolateral flux of carnitine in kidney cells. Etoposide was also associated with a significant urinary loss of carnitine in mice (∼1.5-fold) and in patients with cancer (∼2.4-fold). Collectively, these findings indicate that etoposide can inhibit hOCTN2 function, potentially disturb carnitine homeostasis, and that this phenomenon can contribute to treatment-related toxicities. Mol Cancer Ther; 11(4); 921–9. ©2012 AACR.
- Published
- 2012
25. Pharmaceutical Relevance of Drug Transporter Malfunctions
- Author
-
Alex Sparreboom and Cynthia S. Lancaster
- Subjects
Pharmacology ,business.industry ,Medicine ,Pharmacology (medical) ,Relevance (information retrieval) ,Computational biology ,Drug transporter ,business - Published
- 2011
26. Effect of ABCC2 (MRP2) Transport Function on Erythromycin Metabolism
- Author
-
C.J. Peer, Cynthia S. Lancaster, Shelley Orwick, Ryan M. Franke, Ashley M. Kosloske, Ron H.J. Mathijssen, Sharyn D. Baker, Alice A. Gibson, Alex Sparreboom, William D. Figg, and Medical Oncology
- Subjects
Adult ,Male ,medicine.medical_specialty ,CYP3A ,Midazolam ,Erythromycin ,Pharmacology ,Article ,Cell Line ,Cohort Studies ,Mice ,Young Adult ,Dogs ,Internal medicine ,Cytochrome P-450 CYP3A ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Aged ,Antibacterial agent ,Mice, Knockout ,CYP3A4 ,biology ,Multidrug resistance-associated protein 2 ,Homozygote ,Genetic Variation ,Cytochrome P450 ,Middle Aged ,Multidrug Resistance-Associated Protein 2 ,Protein Transport ,Endocrinology ,biology.protein ,Female ,Multidrug Resistance-Associated Proteins ,Drug metabolism ,medicine.drug - Abstract
The macrolide antiobiotic erythromycin undergoes extensive hepatic metabolism and is commonly used as a probe for cytochrome P450 (CYP) 3A4 activity. By means of a transporter screen, erythromycin was identified as a substrate for the transporter ABCC2 (MRP2) and its murine ortholog, Abcc2. Because these proteins are highly expressed on the biliary surface of hepatocytes, we hypothesized that impaired Abcc2 function may influence the rate of hepatobiliary excretion and thereby enhance erythromycin metabolism. Using Abcc2 knockout mice, we found that Abcc2 deficiency was associated with a significant increase in erythromycin metabolism, whereas murine Cyp3a protein expression and microsomal Cyp3a activity were not affected. Next, in a cohort of 108 human subjects, we observed that homozygosity for a common reduced-function variant in ABCC2 (rs717620) was also linked to an increase in erythromycin metabolism but was not correlated with the clearance of midazolam. These results suggest that impaired ABCC2 function can alter erythromycin metabolism, independent of changes in CYP3A4 activity.
- Published
- 2011
27. Cisplatin-Induced Downregulation of OCTN2 Affects Carnitine Wasting
- Author
-
Zhili Zuo, Kelly K. Filipski, Cynthia S. Lancaster, Alex Sparreboom, Zhaoyuan Chen, Ryan M. Franke, Walter J. Loos, Shelley Orwick, Chaoxin Hu, and Medical Oncology
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Organic Cation Transport Proteins ,Down-Regulation ,Peroxisome proliferator-activated receptor ,Mice, Inbred Strains ,SLC22A5 ,Article ,Cell Line ,Mice ,SDG 3 - Good Health and Well-being ,Downregulation and upregulation ,Carnitine ,Internal medicine ,medicine ,Animals ,Humans ,PPAR alpha ,Solute Carrier Family 22 Member 5 ,Acetylcarnitine ,Mice, Knockout ,Cisplatin ,chemistry.chemical_classification ,Kidney ,Organic cation transport proteins ,Symporters ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Membrane Proteins ,Endocrinology ,medicine.anatomical_structure ,Oncology ,chemistry ,biology.protein ,Carrier Proteins ,medicine.drug - Abstract
Purpose: Carnitine is an essential cofactor for mitochondrial fatty acid oxidation that is actively reabsorbed by the luminal transporter Octn2 (Slc22a5). Because the nephrotoxic agent cisplatin causes urinary loss of carnitine in humans, we hypothesized that cisplatin may affect Octn2 function. Experimental Design: Excretion of carnitine and acetylcarnitine was measured in urine collected from mice with or without cisplatin administration. The transport of carnitine was assessed in cells that were transfected with OCT1 or OCT2. The effect of cisplatin treatment on gene expression was analyzed using a mouse GeneChip array and validated using quantitative reverse transcriptase-PCR. Results: In wild-type mice, urinary carnitine excretion at baseline was ∼3-fold higher than in mice lacking the basolateral cisplatin transporters Oct1 and Oct2 [Oct1/2(−/−) mice], indicating that carnitine itself undergoes basolateral uptake into the kidney. Transport of carnitine by OCT2, but not OCT1, was confirmed in transfected cells. We also found that cisplatin caused an increase in the urinary excretion of carnitine and acetylcarnitine in wild-type mice but not in Oct1/2(−/−) mice, suggesting that tubular transport of cisplatin is a prerequisite for this phenomenon. Cisplatin did not directly inhibit the transport of carnitine by Octn2 but downregulated multiple target genes of the transcription factor peroxisome proliferator activated receptor α, including Slc22a5, in the kidney of wild-type mice that were absent in Oct1/2(−/−) mice. Conclusion: Our study shows a pivotal role of Oct1 and Oct2 in cisplatin-related disturbances in carnitine homeostasis. We postulate that this phenomenon is triggered by deactivation of peroxisome proliferator activated receptor α and leads to deregulation of carnitine-shuttle genes. Clin Cancer Res; 16(19); 4789–99. ©2010 AACR.
- Published
- 2010
28. Influence of Oct1/Oct2-deficiency on cisplatin-induced changes in urinary N-acetyl-beta-D-glucosaminidase
- Author
-
Ryan M. Franke, Chaoxin Hu, Alex Sparreboom, Ron H.J. Mathijssen, Oliver Zolk, Kelly K. Filipski, Ashley M. Kosloske, Cynthia S. Lancaster, and Medical Oncology
- Subjects
Male ,Cancer Research ,Organic Cation Transport Proteins ,Urinary system ,Antineoplastic Agents ,Pharmacology ,Kidney ,Polymerase Chain Reaction ,Article ,Nephrotoxicity ,chemistry.chemical_compound ,Mice ,SDG 3 - Good Health and Well-being ,Cell Line, Tumor ,Acetylglucosaminidase ,medicine ,Animals ,Humans ,Cimetidine ,Enzyme Inhibitors ,Blood urea nitrogen ,Cisplatin ,Mice, Knockout ,Creatinine ,business.industry ,Kidney metabolism ,Organic Cation Transporter 2 ,medicine.anatomical_structure ,Oncology ,chemistry ,Biochemistry ,business ,Biomarkers ,medicine.drug ,Octamer Transcription Factor-1 - Abstract
Purpose: This study aimed to test the influence of functional renal organic cation transporters (OCT2 in humans, Oct1 and Oct2 in mice) on biomarkers of cisplatin nephrotoxicity, such as urinary activity of N-acetyl-β-D-glucosaminidase (NAG). Experimental Design: Temporal cisplatin-induced nephrotoxicity was assessed by histopathology and biomarkers. Cisplatin-mediated NAG changes and survival were determined in wild-type and Oct1/2(-/-) mice. Identification of OCT2 inhibitors was done in transfected 293Flp-In cells, and the NCI60 cell line panel was used to assess contribution of OCT2 to cisplatin uptake in cancer cells. Results: Classical biomarkers such as blood urea nitrogen and serum creatinine were not elevated until 72 hours after cisplatin administration and substantial kidney damage had occurred. Oct1/2(-/-) mice had 2.9-fold lower NAG by 4 hours (P < 0.0001) and 2.3-fold increased survival (P = 0.0097). Among 16 agents, cimetidine strongly inhibited uptake of tetraethylammonium bromide (P = 0.0006) and cisplatin (P < 0.0001), but did not have an influence on cisplatin uptake in SK-OV-3 cells, the cancer line with the highest OCT2 mRNA levels. In wild-type mice, cimetidine inhibited cisplatin-induced NAG changes (P = 0.016 versus cisplatin alone) to a degree similar to that seen in Oct1/2(-/-) mice receiving cisplatin (P = 0.91). Cumulative NAG activity of >0.4 absorbance units (AU) was associated with 21-fold increased odds for severe nephrotoxicity (P = 0.0017), which was linked with overall survival (hazard ratio, 8.1; 95% confidence interval, 2.1-31; P = 0.0078). Conclusions: Cimetidine is able to inhibit OCT2-mediated uptake of cisplatin in the kidney, and subsequently ameliorate nephrotoxicity likely with minimal effect on uptake in tumor cells. Clin Cancer Res; 16(16); 4198–206. ©2010 AACR.
- Published
- 2010
29. TOR Signaling Is a Determinant of Cell Survival in Response to DNA Damage
- Author
-
Mary-Ann Bjornsti, Changxian Shen, Bin Shi, Hong Guo, Padma Thimmaiah, and Cynthia S. Lancaster
- Subjects
Adult ,Saccharomyces cerevisiae Proteins ,Ribonucleoside Diphosphate Reductase ,Cell Survival ,DNA damage ,TORC1 signaling ,Cell Cycle Proteins ,Saccharomyces cerevisiae ,Genotoxic Stress ,TORC1 complex ,Protein Serine-Threonine Kinases ,Biology ,S Phase ,chemistry.chemical_compound ,Humans ,Cycloheximide ,Molecular Biology ,Checkpoint Kinase 2 ,Sirolimus ,Articles ,Cell Biology ,Methyl Methanesulfonate ,Molecular biology ,Methyl methanesulfonate ,TOR signaling ,Cell biology ,Protein Subunits ,chemistry ,Multiprotein Complexes ,Signal transduction ,DNA Damage ,Mutagens ,Signal Transduction ,Transcription Factors - Abstract
The conserved TOR (target of rapamycin) kinase is part of a TORC1 complex that regulates cellular responses to environmental stress, such as amino acid starvation and hypoxia. Dysregulation of Akt-TOR signaling has also been linked to the genesis of cancer, and thus, this pathway presents potential targets for cancer chemotherapeutics. Here we report that rapamycin-sensitive TORC1 signaling is required for the S-phase progression and viability of yeast cells in response to genotoxic stress. In the presence of the DNA-damaging agent methyl methanesulfonate (MMS), TOR-dependent cell survival required a functional S-phase checkpoint. Rapamycin inhibition of TORC1 signaling suppressed the Rad53 checkpoint-mediated induction of ribonucleotide reductase subunits Rnr1 and Rnr3, thereby abrogating MMS-induced mutagenesis and enhancing cell lethality. Moreover, cells deleted for RNR3 were hypersensitive to rapamycin plus MMS, providing the first demonstration that Rnr3 contributes to the survival of cells exposed to DNA damage. Our findings support a model whereby TORC1 acts as a survival pathway in response to genotoxic stress by maintaining the deoxynucleoside triphosphate pools necessary for error-prone translesion DNA polymerases. Thus, TOR-dependent cell survival in response to DNA-damaging agents coincides with increased mutation rates, which may contribute to the acquisition of chemotherapeutic drug resistance.
- Published
- 2007
30. Cisplatin-Induced Renal Injury is Independently Mediated by OCT2 and p53
- Author
-
Eberhard Schlatter, Lie Li, Navjotsingh Pabla, Ashley M. Kosloske, Dorothea Zeeh, Giuliano Ciarimboli, Laura J. Janke, Edwin Hermann, Cynthia S. Lancaster, Alice A. Gibson, Alex Sparreboom, and Jason A. Sprowl
- Subjects
Male ,Cancer Research ,Organic Cation Transport Proteins ,Antineoplastic Agents ,Real-Time Polymerase Chain Reaction ,Article ,Nephrotoxicity ,Mice ,Gene expression ,medicine ,Animals ,Benzothiazoles ,RNA, Messenger ,Oligonucleotide Array Sequence Analysis ,Cisplatin ,Homeodomain Proteins ,Mice, Knockout ,Kidney ,Organic cation transport proteins ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Organic Cation Transporter 2 ,Molecular biology ,Gene expression profiling ,medicine.anatomical_structure ,Real-time polymerase chain reaction ,Oncology ,biology.protein ,Mdm2 ,Female ,Kidney Diseases ,Tumor Suppressor Protein p53 ,Biomarkers ,medicine.drug ,Octamer Transcription Factor-1 ,Toluene - Abstract
Purpose: Tubular secretion of cisplatin is abolished in mice deficient for the organic cation transporters Oct1 and Oct2 (Oct1/2−/−mice), and these animals are protected from severe cisplatin-induced kidney damage. Since tubular necrosis is not completely absent in Oct1/2−/−mice, we hypothesized that alternate pathways are involved in the observed injury. Experimental Design: Studies were done in wild-type, Oct1/2−/−, or p53-deficient animals, all on an FVB background, receiving cisplatin intraperitoneally at 15 mg/kg. Cisplatin metabolites were analyzed using mass spectrometry, and gene expression was assessed using Affymetrix microarrays and RT-PCR arrays. Results: KEGG pathway analyses on kidneys from mice exposed to cisplatin revealed that the most significantly altered genes were associated with the p53 signaling network, including Cdnk1a and Mdm2, in both wild-type (P = 2.40 × 10−11) and Oct1/2−/−mice (P = 1.92 × 10−8). This was confirmed by demonstrating that homozygosity for a p53-null allele partially reduced renal tubular damage, whereas loss of p53 in Oct1/2−/−mice (p53−/−/Oct1/2−/−) completely abolished nephrotoxicity. We found that pifithrin-α, an inhibitor of p53-dependent transcriptional activation, inhibits Oct2 and can mimic the lack of nephrotoxicity observed in p53−/−/Oct1/2−/−mice. Conclusions: These findings indicate that (i) the p53 pathway plays a crucial role in the kidney in response to cisplatin treatment and (ii) clinical exploration of OCT2 inhibitors may not lead to complete nephroprotection unless the p53 pathway is simultaneously antagonized. Clin Cancer Res; 20(15); 4026–35. ©2014 AACR.
- Published
- 2014
31. Oxaliplatin-induced neurotoxicity is dependent on the organic cation transporter OCT2
- Author
-
Hugh Giovinazzo, Laura J. Janke, Cynthia S. Lancaster, Guoqing Du, Giuliano Ciarimboli, Alice A. Gibson, Alex Sparreboom, Anthony F. Shields, Guido Cavaletti, Jason A. Sprowl, Sprowl, J, Ciarimboli, G, Lancaster, C, Giovinazzo, H, Gibson, A, Du, G, Janke, L, Cavaletti, G, Shields, A, and Sparreboom, A
- Subjects
Nervous system ,Male ,Neurotoxicity Syndrome ,Organic Cation Transport Proteins ,Organoplatinum Compounds ,solute carrier ,Antineoplastic Agents ,Pharmacology ,chemotherapy ,Antineoplastic Agent ,Mice ,Ganglia, Spinal ,medicine ,Animals ,Humans ,Cytotoxicity ,Organic Cation Transport Protein ,Mice, Knockout ,Multidisciplinary ,Organic cation transport proteins ,biology ,Chemistry ,Animal ,Organoplatinum Compound ,Neurotoxicity ,Organic Cation Transporter 2 ,Biological Sciences ,medicine.disease ,digestive system diseases ,Oxaliplatin ,medicine.anatomical_structure ,Allodynia ,Toxicity ,biology.protein ,neuropathy ,Female ,Neurotoxicity Syndromes ,medicine.symptom ,medicine.drug ,Human ,Octamer Transcription Factor-1 - Abstract
Oxaliplatin is an integral component of colorectal cancer therapy, but its clinical use is associated with a dose-limiting peripheral neurotoxicity. We found that the organic cation transporter 2 (OCT2) is expressed on dorsal root ganglia cells within the nervous system where oxaliplatin is known to accumulate. Cellular uptake of oxaliplatin was increased by 16- to 35-fold in cells overexpressing mouse Oct2 or human OCT2, and this process was associated with increased DNA platination and oxaliplatin-induced cytotoxicity. Furthermore, genetic or pharmacologic knockout of Oct2 protected mice from hypersensitivity to cold or mechanical-induced allodynia, which are established tests to assess acute oxaliplatin-induced neurotoxicity. These findings provide a rationale for the development of targeted approaches to mitigate this debilitating toxicity.
- Published
- 2013
32. Modulation of OATP1B-type transporter function alters cellular uptake and disposition of platinum chemotherapeutics
- Author
-
Cynthia S. Lancaster, Alice A. Gibson, Alex Sparreboom, Jason A. Sprowl, Shuiying Hu, and Aisha L. Walker
- Subjects
Male ,Cancer Research ,Organic anion transporter 1 ,Gene Expression ,Organic Anion Transporters ,Antineoplastic Agents ,Pharmacology ,Organic Anion Transporters, Sodium-Independent ,Article ,chemistry.chemical_compound ,Mice ,Solute Carrier Organic Anion Transporter Family Member 1B3 ,Cell Line, Tumor ,Neoplasms ,Gene expression ,medicine ,Animals ,Humans ,Platinum ,Cisplatin ,biology ,Liver-Specific Organic Anion Transporter 1 ,Wild type ,Transporter ,Biological Transport ,Carboplatin ,Oxaliplatin ,Oncology ,chemistry ,Cell culture ,Drug Resistance, Neoplasm ,biology.protein ,medicine.drug - Abstract
Expression of the human organic anion transporting polypeptides OATP1B1 and OATP1B3 has been previously believed to be restricted to hepatocytes. Here we show that the gene encoding OATP1B3, but not OATP1B1, is abundantly expressed in multiple human solid tumors that include hepatocellular, lung, and ovarian carcinomas. Surprisingly, OATP1B3 gene expression in a panel of 60 human tumor cell lines was linked with sensitivity to multiple cytotoxic agents, including the platinum anticancer drugs cisplatin, carboplatin, and oxaliplatin. In addition, overexpression of OATP1B3 in mammalian cells increased cellular accumulation of platinum agents and decreased cell survival. In mice with a targeted disruption of the ortholog transporter Oatp1b2, the liver-to-plasma ratio of cisplatin was significantly reduced compared with wild-type mice, without concurrent changes in expression profiles of other transporter genes. Our findings indicate an unexpected role for tumoral and host OATP1B-type carriers in the toxicity and disposition of platinum anticancer drugs, and may provide a foundation for understanding the extensive interindividual pharmacodynamic variability seen with these drugs in patients. Mol Cancer Ther; 12(8); 1537–44. ©2013 AACR.
- Published
- 2013
33. Influence of polymorphic OATP1B-type carriers on the disposition of docetaxel
- Author
-
Guoqing Du, Sharyn D. Baker, Amanda Obaidat, Lena E. Friberg, Gitte H. Bruun, Torben Stamm Mikkelsen, Walter J. Loos, Alice A. Gibson, Aisha L. Walker, Alex Sparreboom, Thomas J. Corydon, Anne-Joy M. de Graan, Ron H. J. Mathijssen, Laure Elens, Shuiying Hu, Peter de Bruijn, Cynthia S. Lancaster, Bruno Hagenbuch, Ron H.N. van Schaik, Medical Oncology, Clinical Chemistry, and Erasmus MC other
- Subjects
Adult ,Male ,Cancer Research ,Organic anion transporter 1 ,Genotype ,Organic Anion Transporters ,Antineoplastic Agents ,Docetaxel ,Pharmacology ,Organic Anion Transporters, Sodium-Independent ,urologic and male genital diseases ,Article ,Cell Line ,Mice ,Solute Carrier Organic Anion Transporter Family Member 1B3 ,Cricetinae ,Neoplasms ,medicine ,Animals ,Humans ,neoplasms ,Aged ,Mice, Knockout ,Polymorphism, Genetic ,biology ,Human liver ,Liver-Specific Organic Anion Transporter 1 ,Disposition ,Middle Aged ,Phenotype ,Uptake transporters ,Oncology ,Liver ,Cell culture ,Inactivation, Metabolic ,biology.protein ,Hepatocytes ,Female ,Taxoids ,therapeutics ,medicine.drug - Abstract
Purpose: Docetaxel is extensively metabolized by CYP3A4 in the liver but mechanisms by which the drug is taken up into hepatocytes remain poorly understood. We hypothesized that (i) liver uptake of docetaxel is mediated by the polymorphic solute carriers OATP1B1 and OATP1B3 and (ii) inherited genetic defects in this process may impair systemic drug elimination. Experimental Design: Transport of docetaxel was studied in vitro using various cell lines stably transfected with OATP1B1*1A (wild-type), OATP1B1*5 [c.521T>C (V174A); rs4149056], OATP1B3, or the mouse transporter Oatp1b2. Docetaxel clearance was evaluated in wild-type and Oatp1b2-knockout mice as well as in two cohorts of patients with multiple variant transporter genotypes (n = 213). Results: Docetaxel was found to be a substrate for OATP1B1, OATP1B3, and Oatp1b2 but was not transported by OATP1B1*5. Deficiency of Oatp1b2 in mice was associated with an 18-fold decrease in docetaxel clearance (P = 0.0099), which was unrelated to changes in intrinsic metabolic capacity in mouse liver microsomes. In patients, however, none of the studied common reduced function variants in OATP1B1 or OATP1B3 were associated with docetaxel clearance (P > 0.05). Conclusions: The existence of at least two potentially redundant uptake transporters in the human liver with similar affinity for docetaxel supports the possibility that functional defects in both of these proteins may be required to confer substantially altered disposition phenotypes. In view of the established exposure–toxicity relationships for docetaxel, we suggest that caution is warranted if docetaxel has to be administered together with agents that potently inhibit both OATP1B1 and OATP1B3. Clin Cancer Res; 18(16); 4433–40. ©2012 AACR.
- Published
- 2012
34. Proximal Tubular Secretion of Creatinine by Organic Cation Transporter OCT2 in Cancer Patients
- Author
-
Cynthia S. Lancaster, Eberhard Schlatter, Mary V. Relling, Ron H.J. Mathijssen, Hermann Pavenstädt, Ching-Hon Pui, Wenjian Yang, Vivian Massmann, Denise Guckel, Jason A. Sprowl, Alex Sparreboom, Giuliano Ciarimboli, Edwin Herrmann, Ryan M. Franke, and Medical Oncology
- Subjects
Male ,Cancer Research ,Organic cation transport ,medicine.medical_specialty ,Organic Cation Transport Proteins ,Renal function ,Antineoplastic Agents ,Polymorphism, Single Nucleotide ,Article ,Cell Line ,Kidney Tubules, Proximal ,chemistry.chemical_compound ,Mice ,SDG 3 - Good Health and Well-being ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,Mice, Knockout ,Creatinine ,Organic cation transport proteins ,biology ,Cancer ,Organic Cation Transporter 2 ,Biological Transport ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Endocrinology ,Oncology ,chemistry ,Renal physiology ,biology.protein ,Female ,Cisplatin ,Ex vivo - Abstract
Purpose: Knowledge of transporters responsible for the renal secretion of creatinine is key to a proper interpretation of serum creatinine and/or creatinine clearance as markers of renal function in cancer patients receiving chemotherapeutic agents. Experimental Design: Creatinine transport was studied in transfected HEK293 cells in vitro and in wild-type mice and age-matched organic cation transporter 1 and 2–deficient [Oct1/2(−/−)] mice ex vivo and in vivo. Clinical pharmacogenetic and transport inhibition studies were done in two separate cohorts of cancer patients. Results: Compared with wild-type mice, creatinine clearance was significantly impaired in Oct1/2(−/−) mice. Furthermore, creatinine inhibited organic cation transport in freshly isolated proximal tubules from wild-type mice and humans, but not in those from Oct1/2(−/−) mice. In a genetic association analysis (n = 590), several polymorphisms around the OCT2/SLC22A2 gene locus, including rs2504954 (P = 0.000873), were significantly associated with age-adjusted creatinine levels. Furthermore, in cancer patients (n = 68), the OCT2 substrate cisplatin caused an acute elevation of serum creatinine (P = 0.0083), consistent with inhibition of an elimination pathway. Conclusions: Collectively, this study shows that OCT2 plays a decisive role in the renal secretion of creatinine. This process can be inhibited by OCT2 substrates, which impair the usefulness of creatinine as a marker of renal function. Clin Cancer Res; 18(4); 1101–8. ©2012 AACR.
- Published
- 2012
35. Distinct functional domains of Ubc9 dictate cell survival and resistance to genotoxic stress
- Author
-
Mary-Ann Bjornsti, Brenda A. Schulman, Robert C.A.M. van Waardenburg, Cynthia S. Lancaster, and David M. Duda
- Subjects
Saccharomyces cerevisiae Proteins ,Recombinant Fusion Proteins ,Ubiquitin-Protein Ligases ,Saccharomyces cerevisiae ,Molecular Sequence Data ,SUMO protein ,Genotoxic Stress ,Ubiquitin-conjugating enzyme ,Substrate Specificity ,Ubiquitin ,X-Ray Diffraction ,Humans ,Amino Acid Sequence ,Molecular Biology ,chemistry.chemical_classification ,DNA ligase ,biology ,Cell Biology ,Articles ,biology.organism_classification ,Cell biology ,Ubiquitin ligase ,Protein Structure, Tertiary ,chemistry ,Biochemistry ,Ubiquitin-Conjugating Enzymes ,biology.protein ,DNA Damage - Abstract
Covalent modification with SUMO alters protein function, intracellular localization, or protein-protein interactions. Target recognition is determined, in part, by the SUMO E2 enzyme, Ubc9, while Siz/Pias E3 ligases may facilitate select interactions by acting as substrate adaptors. A yeast conditional Ubc9P(123)L mutant was viable at 36 degrees C yet exhibited enhanced sensitivity to DNA damage. To define functional domains in Ubc9 that dictate cellular responses to genotoxic stress versus those necessary for cell viability, a 1.75-A structure of yeast Ubc9 that demonstrated considerable conservation of backbone architecture with human Ubc9 was solved. Nevertheless, differences in side chain geometry/charge guided the design of human/yeast chimeras, where swapping domains implicated in (i) binding residues within substrates that flank canonical SUMOylation sites, (ii) interactions with the RanBP2 E3 ligase, and (iii) binding of the heterodimeric E1 and SUMO had distinct effects on cell growth and resistance to DNA-damaging agents. Our findings establish a functional interaction between N-terminal and substrate-binding domains of Ubc9 and distinguish the activities of E3 ligases Siz1 and Siz2 in regulating cellular responses to genotoxic stress.
- Published
- 2006
36. Organic Anion Transporting Polypeptide 1B (OATP1B) Transporters Modulate Hydroxyurea Pharmacokinetics
- Author
-
Alex Sparreboom, Russell E. Ware, Aisha L. Walker, and Cynthia S. Lancaster
- Subjects
Kidney ,Immunology ,Transporter ,Cell Biology ,Hematology ,Biology ,Pharmacology ,Biochemistry ,In vitro ,Solute carrier family ,Organic anion-transporting polypeptide ,medicine.anatomical_structure ,Pharmacokinetics ,In vivo ,Fetal hemoglobin ,medicine ,biology.protein - Abstract
Abstract 1008 Background: Sickle cell is the most common inherited blood disorder affecting millions worldwide. Hydroxyurea is currently the only FDA approved drug for the treatment of sickle cell anemia (SCA) that alters disease pathophysiology. Known for inducing fetal hemoglobin production, hydroxyurea can decrease hospitalizations, vaso-occlusive pain episodes, and mortality associated with SCA. Although efficacious for many patients, a large degree of inter-patient variability in pharmacokinetic (PK) and pharmacodynamic (PD) properties of hydroxyurea has been noted, but sources of this variation remain largely undefined. Plasma transmembrane transporters have been shown to play an important role in the determination drug PK and PD of various xenobiotics. Our recent in vitro studies have demonstrated that hydroxyurea is a substrate for specific solute carrier (SLC) transporters including members of the organic anion transporting polypeptide 1B (OATP1B) subfamily which consists of OATP1B1, and OATP1B3 in humans, and one mouse orthologue Oatp1b2. These transporters are highly expressed on the sinusoidal membrane of hepatocytes, and function as uptake transporters bringing substrates from the blood into the liver. Limited reports have suggested that Oatp1b2 transcript is also found in kidney and intestine as well. In the present study we evaluated the uptake of the hydroxyurea by different haplotypes of hOATP1B3 transporter in vitro, as well as the influence of the Oatp1b2 transporter on hydroxyurea PK in vivo using a transporter knock-out mouse model. Methods: Transporter-mediated cellular uptake of hydroxyurea was determined in vitro by measuring [14C]-hydroxyurea accumulation in Xenopus laevis oocytes that over express two common haplotypes of OATP1B3. hOATP1B3*1 and the functional variant hOATP1B3*2 that contains the 334T>G and 699G>A SNPs were assessed. In vivo PK Studies were performed using a knockout model mouse model in which mOATP1b2 was eliminated from the DBA/LcaJ1 background. Mice were dose with 50mg/kg of [14C]-hydroxyurea via oral gavage or intravenous (i.v.) injection. Serial blood samples were collected at defined timepoints and concluded with terminal blood and tissue collections at 2 hours post injection. Hydroxyurea levels were determined based on radioactivity in plasma and liver, and kidney homogenate. Accumulation in the urine was also measured over a 3 day period. Results: In vitro kinetic studies did not show any differences between OATP1B3*1 and OATP1B3*2 haplotypes in time-, or concentration-dependent accumulation of hydroxyurea. The inhibitor substrates naringen and rifampin decreased hydroxyurea accumulation by at least 50% in OATP1B3*1 but did not decrease accumulation in OATP1B3*2. These in vitro studies suggest that the genetic mutations associated with these two haplotypes do not have a functional impact on the transport of hydroxyurea, but may have significant implications for potential drug-drug interaction. In vivo, Hydroxyurea PK was altered in the absence of the Oatp1b2 transporter. Plasma levels of hydroxyurea were similar after i.v. injections, but total exposure (AUC) in Oatp1b2-KO mice was decreased by 20% with an AUC of 2257 mg-min/ml compared to 2872 mg-min/ml observed in the wildtype mice (p=0.04). Hydroxyurea levels in the kidneys of Oatp1b2-KO mice were approximately 50% of wildtype with a mean accumulation of 360 pmol/g tissue (p= 0.0025). In addition, 30% less hydroxyurea was excreted in urine of Oatp1b2-KO mice by 72 hours (p=0.023). Accumulation of hydroxyurea in liver of Oatp1b2-KO mice and wildtype mice were 107 and 132 pmol/g tissue respectively, but this difference was not statistically significant. These results suggest that hydroxyurea PK is altered in the absence of Oatp1b2 transporter after oral administration. Conclusions: These studies provide the first in vivo evidence that specific transporters may be key determinants of hydroxyurea PK. Future studies are warranted to investigate the influence of human OATP1B and other transporters that modulate hydroxyurea PK. Identification of such transporters may help to elucidate factors such as genetic variations in the transporters and/or drug-drug interactions that may contribute to PK and PD variability observed in sickle cell patients. Disclosures: No relevant conflicts of interest to declare.
- Published
- 2012
37. Abstract 1522: Identification of pifithrin-α as a potent inhibitor of OCT2-mediated transport of cisplatin
- Author
-
Cynthia S. Lancaster, Ryan M. Franke, Kelly K. Filipski, Alex Sparreboom, and Ashley M. Kosloske
- Subjects
Cisplatin ,Cancer Research ,Kidney ,Organic cation transport proteins ,biology ,Pharmacology ,Pifithrin ,Solute carrier family ,Nephrotoxicity ,chemistry.chemical_compound ,medicine.anatomical_structure ,Oncology ,chemistry ,Apoptosis ,Renal physiology ,biology.protein ,medicine ,medicine.drug - Abstract
Members of the organic cation transporter (OCT) solute carrier family mediate the cellular uptake of a large number of structurally diverse molecules, including a variety of endogenous compounds and xenobiotics such as metformin, cimetidine, and cisplatin. OCT2 is highly expressed on the basolateral membrane of renal tubular cells, where it facilitates the renal secretion of substrate drugs. We previously found that the urinary excretion of cisplatin was drastically reduced in mice lacking the ortholog transporters Oct1 and Oct2 [Oct1/2(-/-) mice], and that, compared to wildtype mice, these animals were resistant to severe (grade 4) cisplatin-induced renal tubular necrosis. Since kidney damage was not completely abolished in Oct1/2(-/-) mice, we performed microarray analyses (Affymetrix Mouse Genome 430 v2.0) on kidney biopsies following cisplatin (10 mg/kg, i.p.) administration in order to further understand the mechanism of cisplatin-induced nephrotoxicity in these mice. Using a false-discovery rate of 5% and an average fold change of ≥2.0, we identified complex gene expression changes and a drug-response signature comprising 1063 up-regulated and 1072 down-regulated genes in wildtype mice that was strikingly different quantitatively in Oct1/2(-/-) mice. Next, we performed a KEGG pathway analysis to identify processes that are specifically altered in response to cisplatin. Ten out of 193 analyzed pathways showed significant (P0.13). During the course of our screening work on identifying inhibitors of OCT2-mediated transport of cisplatin, we observed that pifithrin-α, an investigational inhibitor of p53-dependent transcriptional activation and apoptosis, is a highly potent non-competitive inhibitor of OCT2-mediated transport of tetraethylammonium, a prototypical OCT2 substrate, with a Ki of 3.0 μM. Furthermore, pifithrin-α completely blocked OCT2-mediated transport of cisplatin in vitro even at a substrate-to-inhibitor concentration ratio of 5:1. Collectively, this study suggests that (i) the ability to mount an effective p53 response in response to cisplatin is differentially influencing treatment sensitivity in wildtype mice and Oct1/2(-/-) mice, and (ii) pifithrin-α should be explored as a unique dual OCT2/p53-inhibitor for preventing cisplatin nephrotoxicity. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1522.
- Published
- 2010
38. Abstract 3529: Cisplatin-induced downregulation of OCTN2 affects carnitine (vitamin BT) wasting
- Author
-
Ryan M. Franke, Chaoxin Hu, Cynthia S. Lancaster, Kelly K. Filipski, and Alex Sparreboom
- Subjects
Cisplatin ,Cancer Research ,medicine.medical_specialty ,Organic cation transport proteins ,biology ,Wild type ,Nephrotoxicity ,Endocrinology ,Oncology ,Downregulation and upregulation ,Internal medicine ,medicine ,biology.protein ,Carnitine ,Acetylcarnitine ,Homeostasis ,medicine.drug - Abstract
Carnitine is an essential cofactor for mitochondrial fatty acid oxidation. It is normally maintained at a steady level in the blood, and is retained in the body by a rescue mechanism that involves renal tubular reabsorption of filtered carnitine by OCTN2, a luminal organic cation transporter. The physiological significance of OCTN2 has been confirmed by the identification of mutations that cause a potentially lethal autosomal recessive disease known as primary systemic carnitine deficiency. Heterozygosity for OCTN2 mutations is associated with an intermediate carnitine-deficiency phenotype, suggesting that even partial loss of transporter function may be detrimental. The nephrotoxic agent cisplatin was previously reported to cause urinary loss of carnitine in cancer patients, although the mechanism by which this occurs remains unknown. We hypothesized that cisplatin may influence the function of OCTN2 and/or catalytic enzymes involved in carnitine homeostasis. In wildtype mice, we found that the urinary excretion of carnitine and acetylcarnitine at baseline over a 24-hour period was about ∼3 fold higher than in mice lacking the organic cation transporters Oct1 and Oct2 [Oct1/2(−/−) mice] (P Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3529.
- Published
- 2010
39. 502 SUMO conjugation and proteolysis regulate cell sensitivity to DNA topoisomerase I poisons
- Author
-
Hervé R. Jacquiau, Cynthia S. Lancaster, R. van Waardenburg, Brenda A. Schulman, and Mary-Ann Bjornsti
- Subjects
Cell sensitivity ,Cancer Research ,Sumo conjugation ,Oncology ,Biochemistry ,medicine.diagnostic_test ,Chemistry ,Proteolysis ,medicine ,DNA Topoisomerase I - Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.