Summary of Study ST002999

This data is available at the NIH Common Fund's National Metabolomics Data Repository (NMDR) website, the Metabolomics Workbench, https://www.metabolomicsworkbench.org, where it has been assigned Project ID PR001869. The data can be accessed directly via it's Project DOI: 10.21228/M84F0M This work is supported by NIH grant, U2C- DK119886.

See: https://www.metabolomicsworkbench.org/about/howtocite.php

This study contains a large results data set and is not available in the mwTab file. It is only available for download via FTP as data file(s) here.

Perform statistical analysis  |  Show all samples  |  Show named metabolites  |  Download named metabolite data  
Download mwTab file (text)   |  Download mwTab file(JSON)   |  Download data files (Contains raw data)
Study IDST002999
Study TitleMetabolomics and glucose and glutamine labeled isotope tracing analysis in murine lung adenocarcinoma cells in the context of normal or active NRF2 pathway as well as HDAC and glutaminase inhibitor treatment.
Study SummaryInterplay between metabolism and chromatin signaling are implicated in cancer progression. However, whether and how metabolic reprogramming in tumors generates chromatin vulnerabilities remain unclear. Lung adenocarcinoma (LUAD) tumors frequently harbor aberrant activation of the NRF2 antioxidant pathway which drives aggressive and chemo-resistant disease. Using a chromatin-focused CRISPR screen we report that NRF2 activation sensitizes LUAD cells to genetic and chemical inhibition of class I histone deacetylases (HDAC). This association is observed across cultured cells, mouse models and patient-derived xenografts. Integrative epigenomic, transcriptomic and metabolomic analysis demonstrates that HDAC inhibition causes widespread redistribution of H4ac and its reader protein, which transcriptionally downregulates metabolic enzymes. This results in reduced flux into amino acid metabolism and de novo nucleotide synthesis pathways that are preferentially required for the survival of NRF2-active cancer cells. Together, our findings suggest NRF2 activation as a potential biomarker for effective repurposing of HDAC inhibitors to treat solid tumors. In this metabolomics experiment we characterize the changes in metabolic pathway flux in KP LUAD cells in response to HDAC and glutaminase inhibition. This dataset includes metabolomics of U-C13 glucose tracing (1h and 24h) and U-C13 glutamine (8h) of mouse LUAD cell lines with Kras overexpression and p53 knock-out (KP), carrying empty vector (EV) or overexpression of NRF2dNeh2 (NRF2) and treated with DMSO, Romidepsin or CB-839. 3 technical replicates were done per condition in 2 separate experiments, 1: glucose tracing and 2: glutamine tracing.
Institute
Columbia University - Medical Center
DepartmentGenetics and Development
LaboratoryChao Lu
Last NameDimitris
First NameKaragiannis
Address622 W 168th St, New York, NY 10032
Emailkaragiannis_dimitrios@yahoo.gr
Phone+30 6982804931
Submit Date2023-12-05
Raw Data AvailableYes
Raw Data File Type(s)mzML
Analysis Type DetailLC-MS
Release Date2023-12-08
Release Version1
Karagiannis Dimitris Karagiannis Dimitris
https://dx.doi.org/10.21228/M84F0M
ftp://www.metabolomicsworkbench.org/Studies/ application/zip

Select appropriate tab below to view additional metadata details:


Project:

Project ID:PR001869
Project DOI:doi: 10.21228/M84F0M
Project Title:Metabolic Reprogramming by Histone Deacetylase Inhibition Preferentially Targets NRF2-activated tumors
Project Summary:Interplay between metabolism and chromatin signaling are implicated in cancer progression. However, whether and how metabolic reprogramming in tumors generates chromatin vulnerabilities remain unclear. Lung adenocarcinoma (LUAD) tumors frequently harbor aberrant activation of the NRF2 antioxidant pathway which drives aggressive and chemo-resistant disease. Using a chromatin-focused CRISPR screen we report that NRF2 activation sensitizes LUAD cells to genetic and chemical inhibition of class I histone deacetylases (HDAC). This association is observed across cultured cells, mouse models and patient-derived xenografts. Integrative epigenomic, transcriptomic and metabolomic analysis demonstrates that HDAC inhibition causes widespread redistribution of H4ac and its reader protein, which transcriptionally downregulates metabolic enzymes. This results in reduced flux into amino acid metabolism and de novo nucleotide synthesis pathways that are preferentially required for the survival of NRF2-active cancer cells. Together, our findings suggest NRF2 activation as a potential biomarker for effective repurposing of HDAC inhibitors to treat solid tumors.
Institute:Columbia University Medical Center
Department:Genetics and Development
Laboratory:Chao Lu
Last Name:Karagiannis
First Name:Dimitris
Address:630 West 168th Street, NEW YORK, NY, 10032, USA
Email:karagiannis_dimitrios@yahoo.gr
Phone:+30 6982804931

Subject:

Subject ID:SU003112
Subject Type:Cultured cells
Subject Species:Mus musculus
Taxonomy ID:10090

Factors:

Subject type: Cultured cells; Subject species: Mus musculus (Factor headings shown in green)

mb_sample_id local_sample_id Genotype Treatment Timepoint Tracer
SA326444EV_CB839_Gln_2Empty vector CB-839 8 hours U-C13 Glutamine
SA326445EV_CB839_Gln_3Empty vector CB-839 8 hours U-C13 Glutamine
SA326446EV_CB839_Gln_1Empty vector CB-839 8 hours U-C13 Glutamine
SA326447EV_DMSO_1h_3Empty vector DMSO 1 hour U-C13 Glucose
SA326448EV_DMSO_1h_1Empty vector DMSO 1 hour U-C13 Glucose
SA326449EV_DMSO_1h_2Empty vector DMSO 1 hour U-C13 Glucose
SA326450EV_DMSO_24h_2Empty vector DMSO 24 hours U-C13 Glucose
SA326451EV_DMSO_24h_1Empty vector DMSO 24 hours U-C13 Glucose
SA326452EV_DMSO_24h_3Empty vector DMSO 24 hours U-C13 Glucose
SA326453EV_DMSO_Gln_3Empty vector DMSO 8 hours U-C13 Glutamine
SA326454EV_DMSO_Gln_2Empty vector DMSO 8 hours U-C13 Glutamine
SA326455EV_DMSO_Gln_1Empty vector DMSO 8 hours U-C13 Glutamine
SA326456EV_Rom_1h_2Empty vector Romidepsin 1 hour U-C13 Glucose
SA326457EV_Rom_1h_1Empty vector Romidepsin 1 hour U-C13 Glucose
SA326458EV_Rom_1h_3Empty vector Romidepsin 1 hour U-C13 Glucose
SA326459EV_Rom_24h_2Empty vector Romidepsin 24 hours U-C13 Glucose
SA326460EV_Rom_24h_3Empty vector Romidepsin 24 hours U-C13 Glucose
SA326461EV_Rom_24h_1Empty vector Romidepsin 24 hours U-C13 Glucose
SA326462EV_ROM_Gln_2Empty vector Romidepsin 8 hours U-C13 Glutamine
SA326463EV_ROM_Gln_1Empty vector Romidepsin 8 hours U-C13 Glutamine
SA326464EV_ROM_Gln_3Empty vector Romidepsin 8 hours U-C13 Glutamine
SA326465QC2NA NA NA NA
SA326466QC3NA NA NA NA
SA326467QC4NA NA NA NA
SA326468QC1NA NA NA NA
SA326469QC_4NA NA NA NA
SA326470QC_2NA NA NA NA
SA326471QC5NA NA NA NA
SA326472QC_3NA NA NA NA
SA326473blank1NA NA NA NA
SA326474blank4NA NA NA NA
SA326475Blank_1NA NA NA NA
SA326476QC_1NA NA NA NA
SA326477blank2NA NA NA NA
SA326478blank3NA NA NA NA
SA326479NRF2_CB839_Gln_3NRF2 overexpression CB-839 8 hours U-C13 Glutamine
SA326480NRF2_CB839_Gln_2NRF2 overexpression CB-839 8 hours U-C13 Glutamine
SA326481NRF2_CB839_Gln_1NRF2 overexpression CB-839 8 hours U-C13 Glutamine
SA326482NRF2_DMSO_1h_1NRF2 overexpression DMSO 1 hour U-C13 Glucose
SA326483NRF2_DMSO_1h_3NRF2 overexpression DMSO 1 hour U-C13 Glucose
SA326484NRF2_DMSO_1h_2NRF2 overexpression DMSO 1 hour U-C13 Glucose
SA326485NRF2_DMSO_24h_3NRF2 overexpression DMSO 24 hours U-C13 Glucose
SA326486NRF2_DMSO_24h_2NRF2 overexpression DMSO 24 hours U-C13 Glucose
SA326487NRF2_DMSO_24h_1NRF2 overexpression DMSO 24 hours U-C13 Glucose
SA326488NRF2_DMSO_Gln_2NRF2 overexpression DMSO 8 hours U-C13 Glutamine
SA326489NRF2_DMSO_Gln_1NRF2 overexpression DMSO 8 hours U-C13 Glutamine
SA326490NRF2_DMSO_Gln_3NRF2 overexpression DMSO 8 hours U-C13 Glutamine
SA326491NRF2_Rom_1h_1NRF2 overexpression Romidepsin 1 hour U-C13 Glucose
SA326492NRF2_Rom_1h_2NRF2 overexpression Romidepsin 1 hour U-C13 Glucose
SA326493NRF2_Rom_1h_3NRF2 overexpression Romidepsin 1 hour U-C13 Glucose
SA326494NRF2_Rom_24h_2NRF2 overexpression Romidepsin 24 hours U-C13 Glucose
SA326495NRF2_Rom_24h_1NRF2 overexpression Romidepsin 24 hours U-C13 Glucose
SA326496NRF2_Rom_24h_3NRF2 overexpression Romidepsin 24 hours U-C13 Glucose
SA326497NRF2_ROM_Gln_3NRF2 overexpression Romidepsin 8 hours U-C13 Glutamine
SA326498NRF2_ROM_Gln_2NRF2 overexpression Romidepsin 8 hours U-C13 Glutamine
SA326499NRF2_ROM_Gln_1NRF2 overexpression Romidepsin 8 hours U-C13 Glutamine
Showing results 1 to 56 of 56

Collection:

Collection ID:CO003105
Collection Summary:Mouse lung adenocarcinoma (LUAD) cell lines were established as in a previous study by the Papagiannakopoulos lab (Romero et al. Nature Medicine 2017). Briefly, KrasLSL-G12D/+; p53flox/flox genetically engineered mice were intratracheally infected with pSECC lentiviral vectors expressing sgRNAs against Keap1 or tdTomato as a control. The mice developed LUAD tumors and cell lines were derived from them. In this experiment we used a cell line derived from control tumors (KP: Kras-mutant, p53-null). In this cell line we overexpressed an active form of NRF2 (NRF2) or introduced the empty vector (EV).
Sample Type:Cultured cells

Treatment:

Treatment ID:TR003121
Treatment Summary:For glucose tracing analysis, 2x105 KP cells (n=3) were plated in 6-well plates overnight in RPMI medium (Sigma). After 24 hours, the media was replaced with fresh RPMI medium containing DMSO or Romidepsin. At 48 hours the media was replaced with fresh glucose-free RPMI medium (Sigma) containing 10% dialyzed fetal bovine serum (Gibco), 2.0 g/L 13C6-glucose (Sigma) and DMSO or 5nM Romidepsin. Cells were harvested at 49 and 72 hours and processed as described below. For glutamine tracing, 2x105 KP cells (n=3) were plated in 6-well plates overnight in RPMI medium (Sigma). After 24 hours, the media was replaced with fresh RPMI medium containing DMSO, 1nM Romidepsin or 150nM CB-839. At 48 hours the media was replaced with fresh glutamine-free RPMI medium (Sigma) containing 10% dialyzed fetal bovine serum (Gibco), 2.0 g/L 13C6-glutamine (Cambridge Isotope Laboratories) and DMSO, 1nM Romidepsin or 150nM CB-839

Sample Preparation:

Sampleprep ID:SP003118
Sampleprep Summary:Cells were washed with cold PBS, lysed in 80% Ultra LC-MS acetonitrile (Thermo Scientific) supplemented with 20 µM deuterated 2-hydroxyglutarate (D-2-hydroxyglutaric-2,3,3,4,4-d5 acid (d5-2HG), Cambridge Isotope Laboratories) as an internal standard on ice for 15 minutes, and centrifuged for 10 minutes at 20,000 x g at 4 °C. 200 µL of supernatants were subjected to mass spectrometry analysis.

Combined analysis:

Analysis ID AN004926
Analysis type MS
Chromatography type HILIC
Chromatography system Agilent 1290 Infinity
Column Merck SeQuant ZIC-HILIC (150 x 2.1mm,5um)
MS Type ESI
MS instrument type QTOF
MS instrument name Agilent 6545 QTOF
Ion Mode NEGATIVE
Units Abundance (Extracted ion chromatogram core area)

Chromatography:

Chromatography ID:CH003718
Instrument Name:Agilent 1290 Infinity
Column Name:Merck SeQuant ZIC-HILIC (150 x 2.1mm,5um)
Column Temperature:35
Flow Gradient:0-1.5 min, 80% B; 1.5-7 min, 80% B to 50% B, 7-8.5 min, 50% B; 8.5-8.7 min, 50% B to 80% B, 8.7-13 min, 80% B
Flow Rate:0.3 mL/min
Solvent A:25 mM ammonium carbonate in water
Solvent B:acetonitrile
Chromatography Type:HILIC

MS:

MS ID:MS004669
Analysis ID:AN004926
Instrument Name:Agilent 6545 QTOF
Instrument Type:QTOF
MS Type:ESI
MS Comments:The overall runtime was 13 minutes, and the injection volume was 5 µL. The Agilent Q-TOF was operated in negative mode and the relevant parameters were as listed: ion spray voltage, 3500 V; nozzle voltage, 1000 V; fragmentor voltage, 125 V; drying gas flow, 11 L/min; capillary temperature, 325 °C; drying gas temperature, 350 °C; and nebulizer pressure, 40 psi. A full scan range was set at 50 to 1600 (m/z). The reference masses were 119.0363 and 980.0164. The acquisition rate was 2 spectra/s. Targeted analysis, isotopologues extraction (for the metabolic tracing study), and natural isotope abundance correction were performed by the Agilent Profinder B.10.00 Software (Agilent Technologies).
Ion Mode:NEGATIVE
  logo