Redox Stress

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 56178 Experts worldwide ranked by ideXlab platform

Winter Zhang - One of the best experts on this subject based on the ideXlab platform.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Xiao Qu, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Winter Zhang
    Abstract:

    In KRAS -mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here, we investigated the biological consequences of these cooccurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular reactive oxygen species and decreased levels of ATP, NADPH/NADP + ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo . Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma. Significance: In KRAS -mutant non–small cell lung cancer, LKB1 loss results in enhanced energetic/Redox Stress, which is tolerated, in part, through cooccurring KEAP1/NRF2–dependent metabolic adaptations, thus enhancing glutamine dependence and vulnerability to glutaminase inhibition. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/13/3251/F1.large.jpg.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Winter Zhang, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Francesco Parlati
    Abstract:

    In KRAS-mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here we investigated the biological consequences of these co-occurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular ROS and decreased levels of ATP, NADPH/NADP+ ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo. Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma.

Ana Galancobo - One of the best experts on this subject based on the ideXlab platform.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Xiao Qu, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Winter Zhang
    Abstract:

    In KRAS -mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here, we investigated the biological consequences of these cooccurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular reactive oxygen species and decreased levels of ATP, NADPH/NADP + ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo . Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma. Significance: In KRAS -mutant non–small cell lung cancer, LKB1 loss results in enhanced energetic/Redox Stress, which is tolerated, in part, through cooccurring KEAP1/NRF2–dependent metabolic adaptations, thus enhancing glutamine dependence and vulnerability to glutaminase inhibition. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/13/3251/F1.large.jpg.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Winter Zhang, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Francesco Parlati
    Abstract:

    In KRAS-mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here we investigated the biological consequences of these co-occurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular ROS and decreased levels of ATP, NADPH/NADP+ ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo. Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma.

Francesco Parlati - One of the best experts on this subject based on the ideXlab platform.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Winter Zhang, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Francesco Parlati
    Abstract:

    In KRAS-mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here we investigated the biological consequences of these co-occurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular ROS and decreased levels of ATP, NADPH/NADP+ ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo. Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma.

Alissa Poteete - One of the best experts on this subject based on the ideXlab platform.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Xiao Qu, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Winter Zhang
    Abstract:

    In KRAS -mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here, we investigated the biological consequences of these cooccurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular reactive oxygen species and decreased levels of ATP, NADPH/NADP + ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo . Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma. Significance: In KRAS -mutant non–small cell lung cancer, LKB1 loss results in enhanced energetic/Redox Stress, which is tolerated, in part, through cooccurring KEAP1/NRF2–dependent metabolic adaptations, thus enhancing glutamine dependence and vulnerability to glutaminase inhibition. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/13/3251/F1.large.jpg.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Winter Zhang, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Francesco Parlati
    Abstract:

    In KRAS-mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here we investigated the biological consequences of these co-occurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular ROS and decreased levels of ATP, NADPH/NADP+ ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo. Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma.

Lindsey K Boroughs - One of the best experts on this subject based on the ideXlab platform.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Xiao Qu, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Winter Zhang
    Abstract:

    In KRAS -mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here, we investigated the biological consequences of these cooccurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular reactive oxygen species and decreased levels of ATP, NADPH/NADP + ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo . Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma. Significance: In KRAS -mutant non–small cell lung cancer, LKB1 loss results in enhanced energetic/Redox Stress, which is tolerated, in part, through cooccurring KEAP1/NRF2–dependent metabolic adaptations, thus enhancing glutamine dependence and vulnerability to glutaminase inhibition. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/13/3251/F1.large.jpg.

  • lkb1 and keap1 nrf2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in kras mutant lung adenocarcinoma
    Cancer Research, 2019
    Co-Authors: Ana Galancobo, Piyada Sitthideatphaiboon, Marlese Pisegna, Pei Hsuan Chen, Mirna Rodriguez, Winter Zhang, Lindsey K Boroughs, Alissa Poteete, Pan Tong, Francesco Parlati
    Abstract:

    In KRAS-mutant lung adenocarcinoma, tumors with LKB1 loss (KL) are highly enriched for concurrent KEAP1 mutations, which activate the KEAP1/NRF2 pathway (KLK). Here we investigated the biological consequences of these co-occurring alterations and explored whether they conferred specific therapeutic vulnerabilities. Compared with KL tumors, KLK tumors exhibited increased expression of genes involved in glutamine metabolism, the tricarboxylic acid cycle, and the Redox homeostasis signature. Using isogenic pairs with knockdown or overexpression of LKB1, KEAP1, and NRF2, we found that LKB1 loss results in increased energetic and Redox Stress marked by increased levels of intracellular ROS and decreased levels of ATP, NADPH/NADP+ ratio, and glutathione. Activation of the KEAP1/NRF2 axis in LKB1-deficient cells enhanced cell survival and played a critical role in the maintenance of energetic and Redox homeostasis in a glutamine-dependent manner. LKB1 and the KEAP1/NRF2 pathways cooperatively drove metabolic reprogramming and enhanced sensitivity to the glutaminase inhibitor CB-839 in vitro and in vivo. Overall, these findings elucidate the adaptive advantage provided by KEAP1/NRF2 pathway activation in KL tumors and support clinical testing of glutaminase inhibitor in subsets of KRAS-mutant lung adenocarcinoma.