Distinct mitochondrial defects trigger the integrated stress response depending on the metabolic state of the cell

  1. Eran Mick
  2. Denis V Titov
  3. Owen S Skinner
  4. Rohit Sharma
  5. Alexis A Jourdain
  6. Vamsi K Mootha  Is a corresponding author
  1. Howard Hughes Medical Institute and Department of Molecular Biology, Massachusetts General Hospital, United States
  2. Broad Institute, United States
  3. Department of Systems Biology, Harvard Medical School, United States
5 figures, 1 table and 3 additional files

Figures

Interrogating the genomic response to mitochondrial dysfunction in proliferating and differentiated muscle cells.

(A) Phase contrast images of proliferating C2C12 myoblasts (left), post-mitotic cells cultured in low-serum media for 24 hr following confluence (middle), and differentiated myotubes (right). Scale bar denotes 200 μm. (B) Schematic of the ETC/OXPHOS system and the site of action of small-molecule inhibitors. Pier, piericidin; Anti, antimycin; Oligo, oligomycin. (C) Oxygen consumption rate (OCR) of myoblasts and myotubes acutely treated with mitochondrial inhibitors, normalized in each well to OCR prior to treatment (Untr., untreated). Mean ± SD, N = 7–9 from two experiments. The Games-Howell test was used for all pairwise comparisons. ns, p>0.05; ***, p<0.001. (D) Principal component analysis (PCA) of gene expression levels derived from RNA-seq in myoblasts and myotubes treated with inhibitors for 10 hr. N = 3.

Figure 2 with 1 supplement
Mechanistically distinct mitochondrial inhibitors trigger the ISR and depress proliferative gene expression in myoblasts.

(A) PCA of gene expression levels derived from RNA-seq in myoblasts treated with inhibitors for 10 hr. N = 3 (same samples as in Figure 1D). Pier, piericidin; Anti, antimycin; Oligo, oligomycin. Detailed results are provided in Supplementary file 1. (B) Enriched features in the promoters of the 500 genes with the most positive PC1 weights and the 500 genes with the most negative weights, based on iRegulon analysis of transcription factor binding motifs and ChIP-seq peaks. TF, transcription factor; NES, normalized enrichment score; # Targets, number of gene targets. Detailed results are provided in Supplementary file 2. (C) Gene set enrichment analysis of REACTOME pathways using PC1 weights as the gene ranks. Detailed results are provided in Figure 2—source data 1. (D) Volcano plot relating each gene’s PC1 weight (x-axis) and jackstraw P-value for significance of association with PC1 (y-axis). (E) Fold-change from DMSO derived from RNA-seq for representative ATF4/DDIT3 target genes strongly upregulated along PC1 (Atf3, activating transcription factor 3; Gdf15, growth/differentiation factor 15; Ddit3, DNA damage-inducible transcript 3). Mean ± SD. (F) Fold-change from DMSO derived from RNA-seq for representative cell cycle pathway genes (Ccne1, G1/S-specific cyclin-E1; Tk1, thymidine kinase, cytosolic) and a cholesterol pathway gene (Idi1, isopentenyl-diphosphate delta isomerase 1) strongly downregulated along PC1. Mean ± SD.

Figure 2—figure supplement 1
Additional data on transcriptional and protein markers of the ISR following mitochondrial inhibitor treatments in myoblasts.

(A) Heatmap of mean expression fold-changes derived from RNA-seq for upregulated ATF4/DDIT3 target genes most associated with PC1 (jackstraw p-value<0.0001). Atf4 was also included. Data was min-max normalized such that in each row, the treatment that elicited the maximum upregulation from the DMSO baseline was assigned the value one and the remaining treatments were assigned the fraction of the maximum upregulation they elicited. Rows (genes) were then hierarchically clustered based on Pearson correlation using Ward agglomeration. The first three branching points in the dendrogram define Groups 1–3, as referenced in the main text. Genes in red are used throughout this manuscript as markers of the ISR. See also Supplementary file 1. (B) Western blot of ATF4 and (p-)eIF2α in myoblasts treated for 6 hr as indicated. Pier, piericidin; Anti, antimycin; Oligo, oligomycin.

Figure 3 with 1 supplement
Oxidizing cytosolic NADH/NAD+ is sufficient to ablate ISR activation by complex I inhibition in myoblasts.

(A) Schematic of localization and activity of LbNOX and mitoLbNOX. IMS, intermembrane space. (B) Schematic of localization and activity of NDI1. (C) NADH/NAD+ in extracts of cells expressing luciferase (control), LbNOX, mitoLbNOX or NDI1 and treated with inhibitors for 1 hr. Data is normalized to DMSO in control cells. Mean ± SD, N = 5–6 from two experiments. Welch’s t-test (two-tailed) was used to compare each pair of treatments within control cells, as well as each treatment in the other cells with its equivalent in control cells (significance notations with no connecting lines), followed by Holm’s correction for multiple testing. (D) Media [lactate]/[pyruvate] following 2 hr treatments in control, LbNOX, mitoLbNOX or NDI1 cells. Data is normalized to DMSO in control cells. Mean ± SD, N = 5–6 from three experiments. Statistical analysis as in C. (E) qPCR of Ddit3 following 10 hr treatments in control, LbNOX, mitoLbNOX or NDI1 cells. Data is presented as fold-change from DMSO in control cells. Mean ± SD, N = 5–6 from two experiments. Statistical analysis as in C on the ΔΔCt values. (F) PCA of gene expression levels derived from RNA-seq in control, LbNOX, mitoLbNOX or NDI1 cells following 10 hr treatments, as indicated. Detailed results are provided in Supplementary files 1 and 2. (G) Fold-change from DMSO derived from RNA-seq for Atf3 and Ddit3 in control and LbNOX cells treated for 48 hr with chloramphenicol (CAP) in the presence of uridine. Mean ± SD, N = 2. (H) qPCR of Atf4 and Trp53inp1 in control and LbNOX cells following 10 hr antimycin treatment, with or without uridine (uri). Data is presented as fold-change from DMSO in control cells without uridine. Mean ± SD, N = 2. ns, p>0.05; *, p<0.05; **, p<0.01; ***, p<0.001.

Figure 3—figure supplement 1
Additional data on bioenergetics and metabolism in myoblast cell lines.

(A) Oxygen consumption rate (OCR) of control, LbNOX, mitoLbNOX or NDI1 cells sequentially treated with piericidin and antimycin. Mean ± SD, N = 4–5. (B) Peak intensities of glycerol 3-phosphate and succinate in extracts of control cells treated with inhibitors for 1 hr. Data is normalized to DMSO. Mean ± SD, N = 3.

Figure 4 with 1 supplement
Complex I inhibition in myoblasts triggers the ISR through the kinase GCN2 due to an aspartate and asparagine deficiency.

(A) Secreted [lactate] following 2 hr piericidin treatment in control (Ctrl), LbNOX (Lb) or NDI1 cells. Data is normalized to DMSO (-) separately for each cell line. Mean ± SD, N = 6 from three experiments (same samples as in Figure 3D). Welch’s t-test (two-tailed) was used to compare control with LbNOX and NDI1 cells, followed by Holm’s correction for multiple testing. (B) Adenylate energy charge following 1 hr piericidin treatment in control, LbNOX or NDI1 cells. Mean ± SD, N = 5–6 from two experiments. The Games-Howell test was used to make all pairwise comparisons. Notations with no connecting lines relate to the equivalent treatment in control cells. (C) Fold-change (x-axis) and statistical significance (y-axis) of metabolite differential abundance following 1 hr piericidin treatment in extracts of control or LbNOX cells. N = 3. P-Ser, phosphoserine; Asp, aspartate; Asn, asparagine; Cit, citrate; Suc, succinate; αKG, α-ketoglutarate; IMP, inosine monophosphate; AICAR, 5-aminoimidazole-4-carboxamide ribonucleotide; R5P, ribose 5-phosphate; G3P/DHAP, glyceraldehyde 3-phosphate and/or dihydroxyacetone phosphate; P-Cr, phosphocreatine; Pro, proline. Detailed results are provided in Figure 4—source data 1. (D) Intracellular [aspartate] following 1 hr piericidin treatment in control or LbNOX cells. Data is normalized to DMSO in control cells. Mean ± SD, N = 6 from two experiments (includes samples shown in C). The Games-Howell test was used for all pairwise comparisons. (E) Peak intensity of intracellular asparagine following 1 hr piericidin treatment in control cells, with or without aspartate, and in LbNOX cells. Data is normalized to DMSO in control cells. Mean ± SD, N = 6 from two experiments. The Games-Howell test was used for all pairwise comparisons. (F) qPCR of Ddit3 following 10 hr piericidin treatment in control cells, with or without aspartate, and in LbNOX cells. Data is presented as fold-change from DMSO in control cells. Mean ± SD, N = 8 from three experiments. The Games-Howell test was used to make all pairwise comparisons of ΔΔCt values. (G) Media [lactate]/[pyruvate] following 2 hr piericidin treatment, with or without aspartate, in control cells. Data is normalized to DMSO without aspartate. Mean ± SD, N = 6 from three experiments. Welch’s t-test (two-tailed) was used to compare each treatment with and without aspartate, followed by Holm’s correction. (H) Proliferative rate (doublings in 24 hr) of control cells, with or without aspartate, and of LbNOX cells following piericidin treatment. Data is normalized to DMSO in control cells. Mean ± SD, N = 5–6 from three experiments. The Games-Howell test was used for all pairwise comparisons. (I) qPCR of Atf3 following 10 hr pyruvate withdrawal, with or without aspartate, in Ndufa9-KO C2C12 myoblasts. Data is presented as fold-change from the condition with pyruvate (+). Mean ± SD, N = 3. (J) qPCR of GDF15 following 10 hr piericidin treatment, with or without pyruvate or aspartate, in primary human skeletal myoblasts. Data is presented as fold-change from DMSO. Mean ± SD, N = 3. (K) qPCR of Atf3 following 10 hr piericidin or tunicamycin (Tuni) treatment, with or without GCN2iB, in control cells. Data is presented as fold-change from DMSO. Mean ± SD, N = 6-7. Welch’s t-test (two-tailed) was used to compare each treatment with and without GCN2iB, followed by Holm’s correction. (L) Western blot of (p-)GCN2, ATF4 and (p-)eIF2α following 6 hr piericidin treatment in the indicated conditions in LbNOX cells. LbNOX expression was induced only where indicated. (M) qPCR of Atf3 and Gdf15 in the same cells and conditions shown in L. Data is presented as fold-change from DMSO. Mean ± SD, N = 2–3. GiB, GCN2iB. (N) Model for ISR activation by complex I inhibition in myoblasts. ns, p>0.05; *, p<0.05; **, p<0.01; ***, p<0.001.

Figure 4—figure supplement 1
Additional data on metabolic consequences that trigger the ISR in myoblasts.

(A) Secreted [lactate] following 2 hr inhibitor treatments in control or LbNOX cells. Data is normalized to DMSO separately in each cell line. Mean ± SD, N = 6 from three experiments (expanded version of Figure 4A). (B) Adenylate energy charge following 1 hr treatments in control or LbNOX cells. Mean ± SD, N = 5–6 from two experiments (expanded version of Figure 4B). (C) Scatter plot of intracellular metabolite fold-changes between piericidin and DMSO in control cells (x-axis) or LbNOX cells (y-axis). N = 3 replicates (same data as in Figure 4C). (D) Intracellular [aspartate] following 1 hr treatments in control or LbNOX cells. Mean ± SD, N = 6 from two experiments (expanded version of Figure 4D). (E) Peak intensity of intracellular asparagine following 1 hr treatments in control or LbNOX cells. Mean ± SD, N = 6 from two experiments (expanded version of Figure 4E). (F)-(I) qPCR of ISR-related transcripts following 10 hr treatments, with or without aspartate, in control cells. Data is presented as fold-change from DMSO without aspartate. Mean ± SD, N = 3. Ppp1r15a, protein phosphatase 1 regulatory subunit 15A. (J) qPCR of Atf3 following 10 hr piericidin treatment, with or without pyruvate or aspartate, in primary mouse embryonic fibroblasts. Data is presented as fold-change from DMSO (-). Mean ± SD, N = 3. (K) qPCR of Atf3 following 10 hr piericidin or tunicamycin treatment, with or without aspartate, GCN2iB or the PERK inhibitor GSK2656157, in control cells. Data is presented as fold-change from DMSO. Mean ± SD, N = 3. (L) Ratio of p-eIF2α to total eIF2α, measured by western blot, following 6 hr piericidin treatment in LbNOX cells. LbNOX expression was induced only where indicated. Data is normalized separately in each blot to the DMSO-treated sample not expressing LbNOX. Mean ± SD, N = 4 from four experiments. A one-sample t-test was used to examine whether the mean of DMSO-treated samples expressing LbNOX differed from 1, and Welch’s t-test (two-tailed, paired by blot) was used to compare the piericidin-treated samples with and without LbNOX expression. Holm’s correction was then applied. ns, p>0.05; **, p<0.01.

Figure 5 with 1 supplement
ATP synthase inhibition triggers the ISR in myotubes in a manner related to inner-membrane hyperpolarization.

(A) PCA of gene expression levels derived from RNA-seq in myotubes treated with inhibitors for 10 hr. N = 3 (same samples as in Figure 1D). Detailed results are provided in Supplementary file 1. (B) qPCR of Ddit3 following 10 hr treatments in myotubes. Data is presented as fold-change from DMSO. Mean ± SD, N = 6 from two experiments. The Games-Howell test was used for all pairwise comparisons of ΔΔCt values. (C) NADH/NAD+ in myotube extracts following 1 hr treatments. Data is normalized to DMSO. Mean ± SD, N = 8 from two experiments. The Games-Howell test was used for all pairwise comparisons. (D) Media [lactate]/[pyruvate] following 2 hr treatments in myotubes expressing GFP, LbNOX or mitoLbNOX using adenoviral transduction. Data is normalized to DMSO in GFP. Mean ± SD, N = 6 from three experiments. Welch’s t-test (two-tailed) was used to compare each pair of treatments within GFP cells, as well as each treatment in the other cells with its equivalent in GFP cells (significance notations with no connecting lines), followed by Holm’s correction for multiple testing. (E) Secreted [lactate] following 2 hr treatments in myotubes. Data is normalized to DMSO. Mean ± SD, N = 6 from three experiments (same samples as in D). (F) Adenylate energy charge following 1 hr treatments in myotubes. Mean ± SD, N = 7–8 from two experiments. The Games-Howell test was used for all pairwise comparisons. (G) Intracellular [aspartate] following 1 hr treatments in myotubes. Data is normalized to DMSO. Mean ± SD, N = 7–8 from two experiments. The Games-Howell test was used for all pairwise comparisons. (H) Peak intensity of intracellular asparagine following 1 hr treatments in myotubes. Data is normalized to DMSO. Mean ± SD, N = 8 from two experiments. The Games-Howell test was used for all pairwise comparisons. (I) Fold-change from DMSO derived from RNA-seq for Atf3, Ddit3 and Gdf15 in myotubes treated for 10 hr with oligomycin alone, or in combination with piericidin, BAM15 or 5% O2. Mean ± SD, N = 3. See also Supplementary file 1. (J) Model for ISR activation by ATP synthase inhibition in myotubes. ns, p>0.05; *, p<0.05; **, p<0.01; ***, p<0.001.

Figure 5—figure supplement 1
Additional data on ISR activation in myotubes.

(A) qPCR of Ddit3 following 10 hr inhibitor treatments in C2C12 myoblasts, post-mitotic cells and myotubes. Data is normalized to DMSO separately in each panel. Mean ± SD, N = 3 (myoblast samples are a subset of those previously shown in Figure 3E and myotube samples are a subset of those previously shown in Figure 5B). (B) qPCR of Ddit3 in myotubes treated for 48 hr with chloramphenicol (CAP). Data is presented as fold-change from DMSO. Mean ± SD, N = 5–6. (C) qPCR of Ddit3 in myotubes treated for 10 hr with DMSO or oligomycin in the presence of the indicated concentration of BAM15. Data is normalized to DMSO (-) with no BAM15. Mean ± SD, N = 2. (D) qPCR of Ddit3 in myotubes expressing GFP or LbNOX (Lb) and treated with oligomycin for 10 hr. Data is normalized to DMSO (-) in GFP. Mean ± SD, N = 3. Welch’s t-test (two-tailed) was used to compare oligomycin-treated samples expressing GFP and LbNOX. ns, p>0.05. See also Supplementary file 1. (E) qPCR of GDF15 in primary human myotubes treated for 10 hr with oligomycin alone, in combination with piericidin or in combination with BAM15. Data is presented as fold-change from DMSO. Mean ± SD, N = 3–6.

Tables

Key resources table
Reagent type
(species) or resource
DesignationSource or
reference
IdentifiersAdditional
information
Cell line (Mus musculus)C2C12ATCCCRL-1772
Cell line (M. musculus)Ndufa9-knockout C2C12PMID:27622560
Cell line (M. musculus)Primary mouse embryonic fibroblasts (MEF)LonzaM-FB-481
Cell line (Homo sapiens)Primary human skeletal myoblasts (HSkM)Thermo Fisher ScientificA11440
Recombinant DNA reagentDoxycycline-inducible luciferase (pLVX-TRE3G-Luc)Takara Bio (formerly Clontech)Sold as part of 631187
Recombinant DNA reagentDoxycycline-inducible LbNOXPMID:27124460Available without inducible system on Addgene (#75285)
Recombinant DNA reagentDoxycycline-inducible mitoLbNOXPMID:27124460Available without inducible system on Addgene (#74448)
Recombinant DNA reagentDoxycycline-inducible Saccharomyces cerevisiae NDI1PMID:27124460
OtherHuman adenovirus type5 expressing eGFPVector Biolabs1060-HT
OtherHuman adenovirus type5 expressing eGFP and LbNOXVector Biolabs
(PMID:27124460)
OtherHuman adenovirus type5 expressing eGFP and mitoLbNOXVector Biolabs
(PMID:27124460)
OtherDulbecco’s Modified Eagle’s Medium (DMEM), 4 mM L-Glutamine, no glucose, no sodium pyruvateThermo Fisher Scientific11966–025
OtherDialyzed fetal bovine serum (dFBS)Thermo Fisher Scientific26400–044
OtherDialyzed horse serumValley BiomedicalAS3053-DI
OtherHorse serum, New Zealand originThermo Fisher Scientific16050–130
Chemical compound, drugD-(+)-Glucose solutionSigmaG8769Working concentration 10 mM
Chemical compound, drugDoxycycline hyclateSigmaD9891300 ng/ml
Chemical compound, drugPiericidin ASanta CruzSc-2022870.5 μM
Chemical compound, drugAntimycin ASigmaA86740.5 μM
Chemical compound, drugOligomycin ASigma753511 μM
Chemical compound, drugBAM15TimTecST0563880.5 μM (or as indicated)
Chemical compound, drugChloramphenicolSigmaC037820 μg/ml
Chemical compound, drugTunicamycinSigmaT77651 μg/ml
Chemical compound, drugL-Aspartic acidSigmaA925610 mM
Chemical compound, drugL-Asparagine monohydrateSigmaA83810.5 mM
Chemical compound, drugSodium pyruvate solutionThermo Fisher Scientific11360–0701 mM
Chemical compound, drugUridineSigmaU3003200 μM
Chemical compound, drugGCN2iBAcme Bioscience
(PMID:30061420)
0.5 μM
Chemical compound, drugGSK2656157Cayman Chemical173720.25 μM
Chemical compound, drugSodium DL-Lactate-3,3,3-d3CDN IsotopesD6556
Chemical compound, drugSodium pyruvate-13C3Sigma490717
Chemical compound, drugAdenosine-15N5 5′-MonophosphateSigma900382
Chemical compound, drugAdenosine-13C5 5’-DiphosphateToronto Research ChemicalsA281697
Chemical compound, drugAdenosine-13C10 5′-TriphosphateSigma710695
Chemical compound, drugL-Aspartic acid-1,4-13C2Cambridge Isotope LaboratoriesCLM-4455
Chemical compound, drugFormic acid, LC/MS gradeThermo Fisher ScientificA117-50
Chemical compound, drugAmmonium hydrogen carbonate, LC/MS gradeSigma5.33005
Commercial assay or kitTaqMan gene expression master mixThermo Fisher Scientific4369514
Commercial assay or kitUbr3 mouse TaqMan probeThermo Fisher ScientificMm01328979_m1
Commercial assay or kitAtf3 mouse TaqMan probeThermo Fisher ScientificMm00476033_m1
Commercial assay or kitDdit3 mouse TaqMan probeThermo Fisher ScientificMm01135937_g1
Commercial assay or kitAtf4 mouse TaqMan probeThermo Fisher ScientificMm00515325_g1
Commercial assay or kitGdf15 mouse TaqMan probeThermo Fisher ScientificMm00442228_m1
Commercial assay or kitPpp1r15a mouse TaqMan probeThermo Fisher ScientificMm01205601_g1
Commercial assay or kitTrp53inp1 mouse TaqMan probeThermo Fisher ScientificMm00458142_g1
Commercial assay or kitTBP human TaqMan probeThermo Fisher ScientificHs00427620_m1
Commercial assay or kitGDF15 human TaqMan probeThermo Fisher ScientificHs00171132_m1
AntibodyGCN2 (rabbit, polyclonal)Cell Signaling Technology33021:500 dilution
AntibodyPhospho-GCN2 (Thr899) (rabbit, monoclonal)Abcamab758361:500 dilution
AntibodyATF4 (rabbit, monoclonal)Cell Signaling Technology118151:500 dilution
AntibodyeIF2α (mouse, monoclonal)Cell Signaling Technology21031:500 dilution
AntibodyPhospho-eIF2α (Ser51) (rabbit, monoclonal)Cell Signaling Technology35971:500 dilution
AntibodyActin (mouse, monoclonal)SigmaA47001:3000 dilution

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Eran Mick
  2. Denis V Titov
  3. Owen S Skinner
  4. Rohit Sharma
  5. Alexis A Jourdain
  6. Vamsi K Mootha
(2020)
Distinct mitochondrial defects trigger the integrated stress response depending on the metabolic state of the cell
eLife 9:e49178.
https://doi.org/10.7554/eLife.49178