Abstract Pyruvate metabolism defects lead to severe neuropathies such as the Leigh syndrome (LS) but the molecular mechanisms underlying neuronal cell death remain poorly understood. Here, we unravel a connection between pyruvate metabolism and the regulation of the epitranscriptome that plays an essential role during brain development. Using genetically engineered mouse model and primary neuronal cells, we identify the transcription factor E4F1 as a key coordinator of AcetylCoenzyme A (AcCoA) production by the pyruvate dehydrogenase complex (PDC) and its utilization as an essential co-factor by the Elongator complex to acetylate tRNAs at the wobble position uridine 34 (U[34]). E4F1-mediated direct transcriptional regulation of Dlat and Elp3, two genes encoding key subunits of the PDC and of the Elongator complex, respectively, ensures proper translation fidelity and cell survival in the central nervous system (CNS) during mouse embryonic development. Furthermore, analysis of PDH-deficient cells highlight a crosstalk linking the PDC to ELP3 expression that is perturbed in LS patients. Subject terms: tRNAs, Neurodegeneration __________________________________________________________________ The mechanisms by which pyruvate metabolism defects lead to neurological symptoms in Leigh syndrome patients remain unclear. Here, the authors show a link between pyruvate metabolism and translation fidelity that is important for brain development. Introduction The Leigh syndrome (LS, OMIM#256000) is a severe inborn neurodegenerative encephalopathy affecting one in forty thousand newborns that stems from mitochondrial deficiencies in pyruvate metabolism or in the electron transport chain (ETC). Mutations in over ninety-five genes encoded by either the nuclear or the mitochondrial genomes have been linked to this inborn metabolic disorder, including genes encoding key components or regulators of the pyruvate dehydrogenase (PDH) complex (PDC). PDC is a mitochondrial multisubunit complex that converts pyruvate into AcetylCoenzymeA (AcCoA) to fuel the tri-carboxylic acid (TCA) cycle. Most LS patients display chronic lactate acidemia and bilateral necrotizing lesions of the basal ganglia and the brain stem. Affected children also develop a broad range of motor and neurological symptoms which lead to premature death during early childhood, including muscular atonia, growth and mental retardation, ataxia and vision loss^[112]1. No efficient cure has been identified so far for PDC-linked mitochondrial disorders although few patients have shown a partial improvement of their clinical symptoms under a ketogenic diet, a regimen that metabolically bypasses the PDC complex, or upon its pharmacological stimulation by dichloro-acetate (DCA)^[113]2. Paradoxically, despite most of the genetic alterations implicated in the LS have been identified, the molecular mechanisms underlying the clinical symptoms of these children, and in particular their neurological defects, remain poorly understood. Converging evidence suggests that deregulation of the multifunctional protein E4F1 is associated to PDH deficiency and to the LS. The E4F1 protein was originally identified as a transcriptional regulator of the viral E4 gene that is hijacked by the oncoprotein E1A during adenoviral infection^[114]3. Since, E4F1 was also characterized as an atypical E3 ligase of the tumor suppressor p53 that controls its transcriptional activities independently of degradation^[115]4. Beside its role in regulating the balance between cell proliferation and cell death^[116]4–[117]6, E4F1 was also identified as a major regulator of the PDC complex. Thus, E4F1 directly binds to the promoter of several genes encoding essential subunits or regulators of the PDC complex, including DLAT and DLD, the E2 and E3 core subunits of the PDC, the regulatory subunit of the PDH phosphatase complex PDPR, the pyruvate transporter of the inner mitochondrial membrane MPC1, and the SLC25A19 protein involved in mitochondrial uptake of thiamine pyrophosphate, an essential co-factor of the PDC^[118]7–[119]10. A potential connection between E4F1 and the LS was identified through the clinical management of two siblings of an italian family who developed clinical symptoms reminiscent of those observed in LS patients, including muscular hypotonia, microcephaly, hyperlactacidemia and organic aciduria associated to impaired PDH activity in their skeletal muscles. Brain MRI-imaging revealed symmetric necrotic lesions in the subthalamic nuclei and in the substantia nigra that are common in LS patients^[120]11. Strikingly, extensive genetic analyzes of these patients failed to identify any of the pathogenic variants previously linked to LS but highlighted a homozygous E4F1^K144Q missense mutation. Consistent with a pivotal role of E4F1 in pyruvate metabolism and LS pathogenesis, knock-out (KO) mice lacking E4f1 in their skeletal muscles exhibit decreased PDH activity and display phenotypes that are reminiscent of some of the clinical symptoms of LS patients, including chronic lactate acidemia and exercice intolerance^[121]8. However, no major neuropathological abnormalities were observed in this animal model, suggesting that the neuropathy of LS patients results from tissue specific defects in the central nervous system (CNS). Here, we generated mice lacking E4F1 in the CNS to investigate its role during neuronal development. Detailed molecular characterization of this animal model during embryonic development and of E4F1-deficient primary fibroblasts demonstrated that in addition to its role in pyruvate metabolism, E4F1 is also a key transcriptional regulator of Elp3, a gene encoding the catalytic subunit of the Elongator complex. The Elongator complex is an acetyl-transferase which targets a subset of tRNAs in the acceptor stem loop at the wobble position uridine 34 (U[34]) to promote a stepwise cascade of chemical modifications (methoxy-carbonyl-methylation and thiolation) to produce mature tRNAs and thereby ensure protein translation fidelity^[122]12–[123]14. Our data indicate that perturbation of E4F1 activities during embryonic development results in severe brain defects and microcephaly associated to impaired tRNAs U[34] modifications, translation defects and the induction of an integrated stress response (ISR) leading to neuronal cell death. Moreover, we identified a crosstalk between the PDC and ELP3 expression that is perturbed in cells isolated from LS patients. Altogether, our data highlight the importance of the links between pyruvate metabolism and the epitranscriptome to ensure proteome homeostasis in the CNS. Results E4F1 plays a critical role in neuronal development To evaluate the roles of E4f1 during brain development, we generated animals lacking E4F1 in their CNS by crossing E4f1 constitutive and conditional KO (cKO) mice with transgenic animals expressing the CRE recombinase under the control of the Nestin (Nes) promoter that is predominantly active in all neuronal and glial progenitors from day 11 of embryonic development (E11) onwards, and with a reporter strain allowing Cre-mediated expression of the green fluorescent protein (GFP) from the Rosa26 locus (Supplementary Figure [124]S1A)^[125]15–[126]18. In these compound mutant animals (hereafter referred to as E4f1^(Nes)KO) and their control littermates (CTL), immunohistochemistry (IHC) analysis of the GFP reporter on head or whole embryo sagittal sections isolated at different developmental stages confirmed that Cre-mediated recombination was mainly restricted to the CNS and the spinal cord (Fig. [127]1A and Supplementary Figure [128]S1B). The efficiency and the specificity of Cre-mediated inactivation of E4f1 in the CNS was confirmed at E14.5 by RT-qPCR and immunoblotting, respectively (Fig. [129]1B, C). E4f1^(Nes)KO mice were represented at the expected mendelian ratio at E14.5, E16.5 and E18.5 and showed no obvious growth developmental abnormalities, but were absent at P1, indicating that E4F1 deficiency in the CNS leads to perinatal death (Fig. [130]1D and Supplementary Figure [131]S1C,D). Moreover, E4f1^(Nes)KO E18.5 embryos exhibited microcephaly, a developmental defect also observed in some LS patients^[132]1 including those harboring the homozygous E4F1^A430C mutation which encodes an hypomorpic allele of E4F1, as shown by our complementation assays in E4f1^cKO Mouse Embryonic Fibroblasts (MEFs) (Supplementary Figure [133]S1E–G). Fig. 1. E4F1 plays a critical role in corticogenesis during mouse embryonic development. [134]Fig. 1 [135]Open in a new tab A Immunohistochemistry (IHC) analysis of GFP expression on brain sagittal sections prepared from E14.5, E16.5 and E18.5 E4f1^(Nes)KO and CTL littermates. Scale bars, 2.5 mm. Images are representative of experiments performed on n = 4 animals/group. B RT-qPCR analysis of E4f1 mRNA levels in the indicated tissues in E18.5 E4f1^(Nes)KO and CTL embryos (n = 6 for brain samples and n = 3 for other tissues). C Immunoblot analysis of E4F1 and ACTIN protein levels in the brain of E14.5 E4f1^(Nes)KO and CTL embryos. Histobars represent the quantification of n = 4 immunoblots performed on independent samples for each genotype. D Numbers of E4f1^(Nes)KO and CTL embryos identified at different developmental stages. Expected numbers were calculated based on a Mendelian distribution. E, F Analysis of brain structures in E14.5 and E16.5 E4f1^(Nes)KO embryos by High Resolution Episcopic Microscopy (HREM). E Histobars represent the relative volume of the neuroepithelium tissue and of brain ventricles of E4f1^(Nes)KO and CTL embryos at E14.5 (n = 3) and E16.5 (n = 3 for CTL and n = 5 for E4f1^(Nes)KO). F Representative HREM images showing the 3D volumic reconstruction of different areas of the brain (forebrain in green, midbrain in blue and hindbrain in red) and of ventricles (lateral ventricles in yellow, third ventricle in pink, mesencephalic vesicle in orange and fourth ventricle in red) of E16.5 E4f1^(Nes)KO and CTL littermates (n ≥ 4). Scale bars, 280 μm. Data are presented as mean + standard error of the mean (SEM) for Fig. 1B or mean + standard deviation (SD) for Fig. 1C, E from the indicated number of animals. Statistical significance was evaluated using unpaired two-sided Student’s t-test (ns, not significant). Next, we performed high-resolution episcopic microscopy (HREM) and generated three-dimension (3D) volumic brain reconstructions of E14.5 and E16.5 E4f1^(Nes)KO embryos and CTL littermates to evaluate the morphology of different regions of the CNS (Fig. [136]1E,F and Supplementary Figure [137]S2A,B). These analyzes revealed that the forebrain, midbrain and hindbrain neuroepithelium of E16.5 E4f1^(Nes)KO embryos presented large areas of degeneration. These defects were illustrated by patchy losses of neuroepithelium or as areas of cavitation leading to alterations in the structure of all regions of the brain. The space between the brain and the cranium was also enlarged in E4F1-deficient mutants when compared to control littermates. 3D reconstructions showed that the volume of the total and subdivided neural tissues (forebrain, midbrain, hindbrain) was smaller in E4f1^(Nes)KO E16.5 embryos, whereas the volume of the total and subdivided ventricles was bigger in E4F1-deficient animals than in control littermates. Minor brain lesions were detected in the forebrain, but not in the midbrain and hindbrain, at E14.5, suggesting that the brain defects resulting from E4F1 deficiency started at E14.5 and worsened at later stages of embryonic development (Fig. [138]1E,F and supplementary Figure [139]S2A,B). Histological analyzes of hematoxylin and eosin (H&E)-stained sagittal sections of E14.5 and E16.5 embryos confirmed the structural lesions observed by HREM and showed that these brain defects started at E14.5 and increased at later developmental stages. The thickness of the cortical plate (CP), which is enriched in neurons, was reduced in E14.5 E4f1^(Nes)KO embryos. E4F1 deficiency in the CNS resulted in decreased cellularity of all cortical layers (Supplementary Figure [140]S2C). No major infiltration by CD45 positive immune cells was observed at E16.5 nor at E18.5 (Supplementary Figure [141]S2D). Moreover, IHC and immunoblot analyzes showed enhanced cleavage of the apoptotic marker caspase 3 (C-CASP3) at E16.5 and E18.5, but very mild increase at E14.5 (Fig. [142]2A-C). Protein levels of cleaved caspase 8, a regulator of the extrinsic apoptic pathway and of other forms of cell death, was not induced in the brain of E16.5 E4f1^(Nes)KO embryos (Supplementary Figure [143]S2E). To identify the neural populations in which E4F1 plays an important role, we determined in coronal brain sections prepared from E14.5 embryos the number of apical and intermediate progenitors, as well as that of neurons, identified on the basis of SOX2, TBR2 and TBR1 expression, respectively. The number of TBR2 positive intermediate progenitors, and that of TBR1 neurons decreased in the brain of E4f1^(Nes)KO E14.5 embryos, whereas the number of SOX2 positive apical progenitors remained comparable to that of control animals. We also evaluated the expression of C-CASP3 and of the proliferation marker Ki67 (Fig. [144]2D). These in situ analyzes showed that E4F1 deficiency impaired the proportion of cycling progenitors, likely explaining the decreased cellularity observed in various regions of the neuroepithelium. Moreover, and consistently with our IHC analyzes, no C-CASP3 staining was observed at E14.5, suggesting that the proliferation defects of E4F1-deficient progenitors at E14.5 preceded the neuronal cell death detected at later stages of development. These data indicate that E4F1 plays a critical role in neuronal proliferation and cell survival during mid-corticogenesis and that perturbation of E4F1 functions in the CNS results in microcephaly and perinatal death. Fig. 2. E4f1 inactivation impairs neuronal development during mid-corticogenesis. [145]Fig. 2 [146]Open in a new tab A Left panels: immunoblot analysis of uncleaved and cleaved CASPASE 3 (CASP3 and C-CASP3, respectively) and ACTIN protein levels in the brain of E14.5, E16.5 and E18.5 E4f1^(Nes)KO and CTL embryos. Histobars represent the quantification of immunoblot analyzes of the ratio C-CASP3/CASP3 protein levels in the brain of E4f1^(Nes)KO and CTL embryos at the indicated developmental stage (n = 4 animals/group). B IHC analysis of C-CASP3 protein in brain sagittal sections prepared from E14.5, E16.5 and E18.5 E4f1^(Nes)KO and CTL embryos. Insets show images at higher magnification of the forebrain neuroepithelium. Images are representative of experiments performed on n = 5 animals/group for E14.5, n = 6 CTL animals and n = 7 E4f1^(Nes)KO animals for E16.5, and n = 4 CTL animals and n = 3 E4f1^(Nes)KO animals for E18.5. Scale bars, 100 μm and 20 μm for the insets. Black stars point at C-CASP3 positive cells. Fb= forebrain; LV= lateral ventricule. C Histobars represent the number of C-CASP3 positive cells in different brain regions of E4f1^(Nes)KO and CTL embryos at the indicated developmental stage (n = 5 animals/group for E14.5, n = 6 CTL animals and n = 7 E4f1^(Nes)KO animals for E16.5, and n = 4 CTL animals and n = 3 E4f1^(Nes)KO animals for E18.5). D Upper panel: schematic representation of embryonic neurogenesis and of the dorso-lateral region of the medial plane used to determine the relative numbers of the different neuronal progenitors and their respective proliferation and survival rates by immunofluorescence (BioRender Created in BioRender. David, A. (2025) [147]https://BioRender.com/s82s985; NIAID Visual & Medical Arts. 30/08/2024. Pyramidal Neuron. NIAID NIH BIOART Source. bioart.niaid.nih.gov/bioart/424; NIAID Visual & Medical Arts. 16/09/2024. Fibroblast. NIAID NIH BIOART Source. bioart.niaid.nih.gov/bioart/152; NIAID Visual & Medical Arts. 29/08/2024. Outer Radial Glial Cell. NIAID NIH BIOART Source. bioart.niaid.nih.gov/bioart/400). The different protein markers used to distinguish apical and intermediate progenitors and neurons are indicated. Lower panel: immunofluorescence (IF) analysis of SOX2, TBR2, TBR1, C-CASP3 and Ki67 protein levels in brain coronal sections prepared from E14.5 E4f1^(Nes)KO and CTL embryos. Scale bars, 50 μm. Histobars represent the number of positive cells for the indicated protein marker expressed as % of the control (n = 4 for SOX2, TBR1, TBR2 analysis, and n = 3 for C-CASP3, Ki67 analysis). Data are presented as mean + standard deviation (SD) for Fig. 2A or mean + standard error of mean (SEM) for Fig. 2C, D from the indicated number of animals. Statistical significance was evaluated using unpaired two-sided Student’s t-test (ns, not significant). Identification of an E4F1 core transcriptional program operating in the CNS To gain insights into the molecular mechanisms underlying impaired neurogenesis in E4F1-deficient animals, we compared the gene expression profile of total brains harvested from E4f1^(Nes)KO and CTL embryos at E14.5 using RNA sequencing (RNA-seq). Upon E4f1 inactivation in the CNS, 185 genes displayed a fold change superior to 2 with a false discovery rate (FDR) inferior to 0.05, among which 116 were significantly down-regulated and 69 were up-regulated. Among the top 35 differentially expressed genes with the lowest false discovery rate ((FDR)-adjusted p-values) in E4F1-deficient brains were Wdr7, Kif1bp, Neurl4, Fktn, Dlat, Znhit6, Senp8, Tube1, Elp3, Leo1, Tti2, Dph5, Taz, Prbm1, Ddi2, Mrfap1, Taz, Dnajc19, Mrpl15, Nsun5, Eef1g and Rad52 (Fig. [148]3A, B, Supplementary Figure [149]S3A and Supplementary Data [150]1). Interestingly, all these genes were previously identified as E4F1-direct target genes in mouse fibroblasts and embryonic stem (ES) cells as well as in human breast cancer cells by chromatin immunoprecipitation (ChIP) combined with next generation sequencing (ChIP-seq)^[151]7,[152]8,[153]10. Gene set enrichment analysis (GSEA) indicated that this E4F1-signature was significantly down-regulated in the brain of E4f1^(Nes)KO embryos, highlighting an E4F1 core transcriptional program operating in various cell types (Fig. [154]3C, Supplementary Figure [155]S3B and Supplementary Data [156]2). Impaired expression of a subset of genes composing this core E4F1 program was confirmed by RT-qPCR on mRNAs extracted from total brains or forebrains of E14.5 E4f1^(Nes)KO embryos (Fig. [157]3D and Supplementary Figure [158]S3C). Of note, GSEA and pathway enrichment analysis (PEA) also indicated that in addition to the deregulation of this E4F1 core program, an ISR was induced in E4F1-deficient brains (Fig. [159]3E,F and Supplementary Figure [160]S3D). Fig. 3. Gene expression profile of E4F1-deficient neurons. [161]Fig. 3 [162]Open in a new tab A,B RNA-seq analysis of brains prepared from E14.5, E4f1^(Nes)KO and CTL embryos (n = 3 for each genotype). A Heatmap representing the relative mRNA levels (fold change, log[2] ratio) of the differentially expressed genes (DEG) with the most statistically significant False Discovery Rate (likelihood ratio tests (LRT) - FDR-adjusted p-values). B Volcano plot depicting the significance (FDR) and magnitude of difference (fold change, log[2] ratio) of DEG identified in E14.5 E4f1^(Nes)KO embryos. C Gene set enrichment analysis (GSEA) of RNA-seq data showing decreased expression of E4F1 target genes. The normalized enrichment score (NES) and false discovery rate (FDR) are indicated. P-values were corrected using Benjamini–Hochberg’ FDR. D RT-qPCR analysis of E4f1 mRNA levels and those of a subset of its direct target genes (Ndufs5, Dnajc19, Taz, Wdr7, Neurl4) in total RNAs prepared from the forebrain of E14.5 E4f1^(Nes)KO and CTL embryos (n = 5). Data are presented as mean + standard error of the mean (SEM) for the indicated number of animals. Statistical significance was evaluated using unpaired two-sided Student’s t-test. E List of the top 10 pathways identified by pathway enrichment analysis (PEA) as differentially modulated in E14.5 E4f1^(Nes)KO embryos. The size of the histobars represents the number of genes in the indicated category with an FDR ≤ 0,001. F GSEA showing the induction of the Integrated Stress Response (ISR) signature in E14.5 E4f1^(Nes)KO embryos. P-values were corrected using Benjamini–Hochberg’ FDR. E4F1 regulates the PDC complex and pyruvate metabolism in the CNS Among the top three most deregulated genes identified by RNA-seq in the brain of E4f1^(Nes)KO embryos was Dlat, an E4F1-direct target gene encoding the E2 subunit of the PDC complex. Interestingly, RT-qPCR analysis of mRNAs prepared from E14.5 E4f1^(Nes)KO forebrains indicated that the role of E4F1 in the regulation of the PDC in neurons extended to other E4F1-direct target genes encoding several key subunits or regulators of the PDC, including Dld, Mpc1, Pdpr and Slc25a19 (Fig. [163]4A). Furthermore, immunoblot analyzes confirmed that E4f1 inactivation in the CNS impinged on DLAT and MPC1 protein levels, whereas those of PDHA1, the E1 subunit of the PDC complex, that were evaluated as a control, as well as DLD, remained unaffected (Fig. [164]4B). Consistent with this expression profile, PDH activity was significantly impaired in the brain of E4f1^(Nes)KO embryos and resulted in decreased amounts of AcCoA (Fig. [165]4C, D). Paradoxically, the abundance of histone H3 acetylation marks previously linked to nuclear PDC did not decrease in E4F1-deficient brains, suggesting that the pool of pyruvate-derived AcCoA poorly contributes to histone acetylation in the CNS (Supplementary Figure [166]S3E)^[167]19,[168]20. Furthermore, circulating lactate levels were significantly higher in E14.5 E4f1^(Nes)KO embryos than in CTL animals (Fig. [169]4E) and IHC analyzes indicated that the protein levels of the lactate transporter MCT4 was upregulated in the brain of E4F1-deficient embryos (Fig. [170]4F). These data show that E4f1 inactivation in the CNS impacts on pyruvate oxidation by the PDC and favors the redirection of the glycolytic flux towards lactate production, a feature reminiscent of the metabolic reprogramming occurring in LS patients. Fig. 4. E4F1 regulates pyruvate metabolism in the CNS. [171]Fig. 4 [172]Open in a new tab A RT-qPCR analysis of E4f1 mRNA levels and those of genes encoding key subunits or regulators of the pyruvate dehydrogenase complex (PDC) (Dlat, Dld, Mpc1, Pdpr, Slc25a19 and Pdha1) in the forebrain of E14.5 E4f1^(Nes)KO and CTL embryos (a minimum of n = 4 animals/group was used to perform these analysis). B Representative immunoblots showing E4F1, DLAT, MPC1, DLD, PDHA1, and ACTIN (loading control) protein levels in the brain of E14.5 E4f1^(Nes)KO and CTL embryos. Right panel: Histobars represent the quantification of immunoblots performed on n = 4 independent samples/group. C Pyruvate Dehydrogenase (PDH) activity in protein extracts prepared from E18.5 E4f1^(Nes)KO and CTL embryos (n = 5 animals/group). D Relative AcetylCoenzyme A (AcCoA) levels in brain from E18.5 E4f1^(Nes)KO and CTL embryos measured by HR-MS (n = 4 animals). E Blood lactate levels in E18.5 E4f1^(Nes)KO (n = 43) and CTL (n = 99) embryos. F IF analysis of MCT4 protein levels in brain sagittal sections prepared from E14.5 E4f1^(Nes)KO and CTL embryos (n = 5 animals/group). Fb= forebrain; Mb= middle brain; LV= lateral ventricule. Scale bars, 50 μm. Data are presented as mean + standard error of mean (SEM) for Fig. 4A, C or mean + standard deviation (SD) for Fig. 4B, D, E from the indicated number of animals. Statistical significance was evaluated using unpaired two-sided Student’s t-test (ns, not significant). E4f1 controls the Elongator complex and U[34] codon-biased translation in the CNS Strikingly, our RNA-seq datasets indicated that Elp3 mRNA levels were also strongly reduced in E4F1-deficient brains (Fig. [173]3A, B). Elp3 encodes the catalytic subunit of the Elongator complex, an acetyl-transferase that was previously implicated in neurogenesis^[174]21. This prompted us to further investigate the links between E4F1 and the Elongator complex. Analysis of previous ChIP-seq experiments performed in murine embryonic fibroblasts (MEFs) and in ES cells suggested that Elp3 is a direct target gene of E4F1 (Fig. [175]5A)^[176]7,[177]22. Quantitative ChIP (qChIP) experiments performed with a previously validated polyclonal anti-E4F1 antibody^[178]7 confirmed that E4F1 binds to the promoter region of Elp3 on its canonical binding site in neuronal N2A cells (Fig. [179]5B). We then determined by RT-qPCR the mRNA levels of different subunits of the Elongator complex in the brain of E4f1^(Nes)KO embryos. Consistent with the role of E4F1 in the direct regulation of Elp3 transcription, Elp3 mRNA levels were reduced in E14.5 E4F1-deficient brains, whereas those of Elp1, Elp2 and Elp4, remained unchanged (Fig. [180]5C). At later developmental stages (E16.5 and E18.5), the mRNA levels of all these core subunits of the Elongator complex were affected by E4F1 deficiency (Supplementary Figure [181]S4A). Immunoblots showed that E4f1 inactivation also impacted on ELP3 protein levels (Fig. [182]5D). Furthermore, and in line with previous reports showing that the stability of the catalytic subunit ELP3 and that of the scaffold ELP1 are interdependent^[183]21,[184]23, ELP1 protein levels also decreased in the brain of E14.5 E4f1^(Nes)KO embryos (Fig. [185]5D). Fig. 5. E4f1 controls the expression and activity of the Elongator complex. [186]Fig. 5 [187]Open in a new tab A E4F1 ChIP-seq read densities in transformed MEFs (tMEFs) and mouse ES cells (MESC) at the Elp3 locus. The sequence and the position of the E4F1 consensus motif in the Elp3 locus are indicated. Arrows indicate gene orientation. B Quantitative ChIP (qChIP) experiments on the Elp3 promoter region using an anti-E4F1 polyclonal antibody (E4F1) or an irrelevant antibody (irr) and chromatin prepared from neuronal N2A cells (n = 3 independent experiments). Results are represented as the relative ratio between the mean value of immunoprecipitated chromatin (calculated as a percentage of the input) with the anti-E4F1 antibody and the one obtained with the control antibody. C RT-qPCR analysis of Elp1, Elp2, Elp3, and Elp4 mRNA levels in total RNAs prepared from the brain of E14.5 E4f1^(Nes)KO and CTL embryos (n = 5 animals/group). D Immunoblot analysis showing ELP1, ELP3 and ACTIN (loading control) protein levels in the brain of E14.5 E4f1^(Nes)KO and CTL embryos. Right panel: histobars represent the quantification of immunoblots performed on n = 4 and n = 5 independent samples for ELP1 and ELP3 respectively. E Liquid-chromatography coupled to tandem mass spectrometry (LC-MS/MS) analysis of the 5-carbamoylmethyl uridine (ncm^5U[34]), 5-methoxy-carbonyl-methyl-2-thio uridine (mcm^5s^2U[34]), methyl-3-cytosine (m^3C) and methylguanosine (Gm) tRNAs modifications, normalized to the total amount of Uridine (U), Cytosine (C) or Guanosine (G) (n = 5 independent samples/group). Data are presented as mean + standard error of the mean (SEM) from the indicated number of samples. Statistical significance was evaluated using unpaired two-sided Student’s t-test (ns, not significant). Next, we explored the impact of E4F1-deficiency on the activity of the Elongator complex. In mammalian cells, the Elongator complex promotes the acetylation of a subset of tRNAs (tRNAs^Gln[UUG], tRNAs^Glu[UUC] and tRNA^Lys[UUU]) on the uridine located at position 34 of the anti-codon stem loop (U[34]). This acetylation reaction initiates a stepwise cascade that first leads to the attachment of a carboxy-methyl group (cm^5U[34]), followed by a methylation step involving the Alkylation repair homolog 8 complex (ALKBH8/TRM112) to generate 5-methoxycarbonylmethyl uridine (mcm^5U[34]), and then the addition of a thiol group by the Cytosolic thiouridylase proteins 1/2 (CTU1/2) enzymes (mcm^5s^2U[34]). From ELP3-acetylated tRNAs, another branch of the pathway generates 5-carbamoylmethyl uridine (ncm^5U[34]) through a yet unknown enzyme (Supplementary Figure [188]S4B)^[189]13,[190]14,[191]24. Liquid-chromatography coupled to tandem mass spectrometry (LC-MS/MS) analysis of tRNAs purified from E14.5 E4f1^(Nes)KO and CTL brains showed that the relative abundance of ncm^5U[34] and mcm^5s^2U[34] decreased in E4F1-deficient neurons, whereas that of other tRNAs modifications not linked to the Elongator complex, including 3-methylcytosine (m^3C) and methylguanosine (Gm), remained unchanged (Fig. [192]5E). RT-qPCR analyzes showed that the total amount of tRNAs targeted by the Elongator complex, tRNAs^Gln[UUG], tRNAs^Glu[UUC] and tRNA^Lys[UUU], were comparable in the brain of E14.5 E4f1^(Nes)KO and CTL embryos, demonstrating that the decreased abundance of mcm^5s^2U[34] and ncm^5U[34] in E4F1-deficient neurons was not the consequence of an impaired production of these tRNAs (Supplementary Figure [193]S4C). Since the Elongator complex has also been involved in tubulin and histone acetylation, we evaluated the ratio of acetylated/total tubulin (α-TUB-Ac/TUB) and that of acetylated histone H3 on lysine 14/total Histone H3 (H3K14Ac/H3)^[194]25,[195]26. We failed to detect any significant decrease in α-TUB-Ac nor in H3K14Ac in the brain of E14.5 E4f1^(Nes)KO embryos, indicating that E4F1 deficiency in the CNS, and the associated impaired activity of the Elongator complex, preferentially impacted on tRNAs acetylation (Supplementary Figure [196]S4D). Because changes in Elongator-dependent tRNAs U[34] modifications have been linked to codon-biased translation defects, we next performed polysome profiling experiments^[197]27. We isolated total cytosolic mRNAs, as well as mRNAs from pooled light polysomal (1 to 3 ribosomes) and heavy polysomal (more than 3 ribosomes) fractions prepared from the brain of E14.5 E4f1^(Nes)KO embryos and control littermates and then determined by next generation sequencing the abundance of these transcripts (Fig. [198]6A and Supplementary Data [199]3). We first focused on a group of genes exhibiting similar total transcript levels but differential mRNA abundance in either the light or the heavy fractions purified from E4F1-deficient brains. Bioinformatic analysis of codons content showed that these mRNAs are significantly enriched in two U[34]-codons (AAA, GAA) in comparison to transcripts of the whole transcriptome (Fig. [200]6B). Moreover, 12% (282 out of 2288) of the corresponding proteins, including KCNA3, contain at least one penta-hydrophilic motif previously linked to protein aggregation and degradation upon codon-dependent translation defects linked to the Elongator complex (Fig. [201]6C)^[202]28. We confirmed by immunoblotting and RTqPCR that E4F1-deficiency in the brain of E14.5 embryos impacted on KCNA3 protein, but not mRNA, levels (Fig. [203]6D and Supplementary Figure [204]S4E). Altogether, these data support the notion that E4f1 inactivation in the CNS leads to perturbed activity of the Elongator complex and translation defects that are linked, at least in part, to impaired tRNAs U[34] modifications. Fig. 6. E4F1 regulates U[34] codon-biased translation in the CNS. [205]Fig. 6 [206]Open in a new tab A Polysome profiling analysis of brains of E14.5 E4f1^(Nes)KO and CTL embryos (n = 5 animals/group). Dot plots represent transcript levels (Log[2] fold change) between E4f1^(Nes)KO and CTL embryos in the pooled light (less then 3 ribosomes, left panel) or heavy (more than 3 ribosomes, right panel) polysomal fractions, according to their abundance in the total cytosolic fraction. The lists indicate the 16 genes which transcript abundance is the most deregulated (log[2]FC) in either the light or the heavy polysomal fractions prepared from E4F1-deficient brains but showing similar total transcript levels. B Codon enrichment in transcripts regulated at the post-transcriptional level in the brain of E14.5 E4f1^(Nes)KO and CTL embryos. U[34]-linked codons are indicated in green. Codon enrichment was assessed by χ^2 tests. C Bioinformatic analysis showing that 12% of the proteins (282 out of 2288) corresponding to genes regulated at the post-transcriptional level in the brain of E14.5 E4f1^(Nes)KO embryos contain at least one penta-hydrophilic amino-acid sequence previously linked to protein aggregation and degradation upon U[34] codon-dependent translation defects. Presence of the penta-hydrophilic sequence was determined by AME (Analysis of Motif Enrichment). D Immunoblot analysis showing KCNA3 and HSP90 (loading control) protein levels in the brain of E14.5 E4f1^(Nes)KO and CTL embryos. Immunoblot is representative of 3 independent experiments. E4F1 deficiency triggers the ISR that leads to neuronal cell death Previous findings showed that genetic inactivation of Elp3 during corticogenesis leads to the induction of the ISR mediated in part by ATF4, a member of the Activating Transcription Factor (ATF) family of proteins^[207]21. Consistent with these findings, IHC analysis of brain sections prepared from E14.5 E4f1^(Nes)KO embryos confirmed the induction of ATF4 protein levels (Fig. [208]7A). Furthermore, the induction of this ATF4-driven transcriptional response correlated with the activation of its upstream regulator eIF2a, as evidenced by its increased phosphorylation. Co-staining experiments with antibodies recognizing glial (GFAP) or neuronal (NeuN) markers indicated that both cell types exhibited increased phospho-eIF2a levels (Fig. [209]7B). In agreement with these data, RNA-seq and RT-qPCR analyzes indicated that E4F1 deficiency in the CNS led to increased mRNA levels of ATF4-target genes implicated in the ISR, including Atf3, Atf5, Eif4ebp1, Asns, Sesn2, Slc6a9, Chop, Chac1, Trib3, Mthfd2, Slc3a2 and Slc7a5 (Figs. [210]3, [211]7C and Supplementary Data [212]1). The ISR involves the derepression of integral ER membrane proteins which activate independent signaling pathways^[213]29. Similar to Elp3 cKO mice, the mRNA levels of Xbp1 and its spliced transcripts Xbp1-u and Xbp1-s, as well as those of Atf6, remained unchanged in the brain of E14.5 E4f1^(Nes)KO embryos, supporting the notion that E4f1 deficiency triggered specifically an ATF4-driven response (Fig. [214]7C and Supplementary Figure [215]S4F). Furthermore, ten days upon genetic inactivation of E4f1 in vitro, ISR-related genes, including Asns, Eif4ebp, Sens2, Trib3 and Slc7a3, were also induced at the mRNA level in primary cortical cultures composed of neurons and glial cells but not in MEFs, indicating that the induction of the ISR triggered by E4F1 deficiency occurred in a cell autonomous and in a cell type specific manner (Fig. [216]7D and Supplementary Figure [217]S4G). To evaluate whether the neuronal cell death observed at E16.5 and E18.5 was linked to the induction of the ISR, we daily administered ISRIB, a selective ISR inhibitor, to pregnant females from E10.5 onwards. The induction of the ISR-related genes Atf5, Eif4ebp, Chac1 and Trib3 in the brain of E14.5 E4f1^(Nes)KO embryos was impeded by ISRIB (Fig. [218]7E). Furthermore, immunoblot analysis of protein extracts prepared from ISRIB or mock-treated E16.5 embryos indicated that cleaved-caspase 3 levels in the brain of E4F1-deficient embryos were reduced upon inhibition of eIF2a (Fig. [219]7F). However, administration of ISRIB was insufficient to rescue the neonatal lethality of E4f1^(Nes)KO mice (Fig. [220]7G). These data support a model in which perturbation of E4F1-mediated control of the Elongator complex impacts on proteome homeostasis leading to the induction of an ISR and ultimately to neuronal cell death. Fig. 7. E4F1 deficiency triggers the ISR that leads to neuronal cell death. [221]Fig. 7 [222]Open in a new tab A IHC analysis of ATF4 protein levels in brain sagittal sections prepared from E14.5 E4f1^(Nes)KO and CTL embryos. Data are representative of n = 4 independent experiments. Scale bars, 100 μm. B Immunofluorescence (IF) analysis of phospho-eIF2a (p-eIF2a), NeuN and GFAP protein levels in brain sagittal sections prepared from E14.5 E4f1^(Nes)KO and CTL embryos. Data are representative of n = 4 independent experiments. Scale bars, 50 μm. C Relative mRNA levels of genes related to the ISR response in the forebrain of E14.5 E4f1^(Nes)KO and CTL embryos determined by RT-qPCR (n = 5 independent samples/group). D Relative mRNA levels of genes related to the ISR response in populations of E4f1^cKO and CTL primary neurons and glial cells determined by RT-qPCR, 10 days after E4f1 inactivation in vitro (n = 6 independent populations of cells/group). E Relative mRNA levels of genes related to the ISR response in the brain of mock or ISRIB -treated E14.5 E4f1^(Nes)KO and CTL embryos determined by RT-qPCR (n = 5 independent samples/group). F Upper panel: immunoblot analysis showing Cleaved-Caspase-3 (C-CASP3) and TUBULIN (loading control) protein levels in the brain of mock or ISRIB -treated E16.5 E4f1^(Nes)KO and CTL embryos. Lower panel: histobars represent the quantification of immunoblots performed on n = 5 independent samples/group. G Numbers of ISRIB-treated E4f1^(Nes)KO and CTL embryos identified at birth (P1). Expected numbers were calculated based on a normal mendelian distribution. Data are presented as mean + standard error of the mean (SEM) for Fig. 7C or mean + standard deviation (SD) for Fig. 7D–F from the indicated number of samples. Statistical significance was evaluated using unpaired two-sided Student’s t-test (ns, not significant). E4F1 coordinates pyruvate-derived AcCoA production by the PDC and tRNAs U[34] acetylation The key role of E4F1 in pyruvate metabolism and in the regulation of the Elongator complex prompted us to evaluate whether E4F1 coordinates, at the transcriptional level, the production of pyruvate-derived AcCoA by the PDC and its utilization by the Elongator complex to promote tRNAs acetylation and ensure translation fidelity. To challenge this hypothesis, we first evaluated Dlat and Elp3 expression upon acute E4f1 inactivation in MEFs. E4F1-deficient MEFs exhibited impaired Dlat and Elp3 mRNA and protein levels (Fig. [223]8A, B). Moreover, and consistent with our findings in E4f1^(Nes)KO embryos, these cells also exhibited decreased amounts of mcm^5s^2U[34] and ncm^5U[34] (Fig. [224]8C). Next, we performed stable isotope tracing experiments to evaluate whether pyruvate-derived AcCoA produced by the PDC serves as a co-factor and a carbon source for the Elongator complex to acetylate tRNAs at U[34]. MEFs were incubated in presence of ^13C uniformally labeled pyruvate ([U-^13C]pyruvate) for 6 h and ^13C-enrichment was measured by LC-MS/MS in the ionized fragments of ncm^5U[34] and mcm^5s^2U[34] that were predicted to incorporate the two ^13C-labeled carbons of the acetyl-group provided by pyruvate-derived ^13C-AcCoA into the side chain of the uridine ring upon acetylation by the Elongator complex (Fig. [225]8D). The m + 2 isotopologues of ncm^5U[34] and mcm^5s^2U[34] were significantly detected in MEFs incubated with^13C-pyruvate for 6 h, a process affected upon pre-incubation with UK-5099, a pharmacological inhibitor of the pyruvate mitochondrial transporter MPC1 that indirectly blocks PDH activity (Fig. [226]8E and Supplementary Figure [227]S5A). In the same experimental set-up, ^13C-acetate, another alternative source of AcCoA, contributed to a much lesser extent to U[34] tRNAs modifications, suggesting that pyruvate and acetate are channeled differently in MEFs to fuel specific acetylation reactions (Supplementary Figure [228]S5B). These data demonstrate that pyruvate-derived AcCoA produced by the PDC is an important co-factor for Elongator-mediated acetylation reactions targeting U[34] tRNAs. Fig. 8. E4F1 coordinates AcCoA production by the PDC and its utilization by the Elongator complex to ensure U[34] tRNAs acetylation. [229]Fig. 8 [230]Open in a new tab A RT-qPCR analysis of E4f1, Dlat and Elp3 mRNA levels in E4f1^cKO and CTL MEFs (n = 4 independent populations of MEFs/group), 7 days after activation of the Cre recombinase. B Left panel: immunoblot analysis of E4F1, DLAT, ELP3 and TUBULIN protein levels in the same cells than in A. Right panel: histobars represent the quantification of immunoblots performed on independent samples (n = 4 populations/group). C LC-MS/MS analysis of ncm^5U[34], mcm^5s^2U[34] and Um in tRNAs purified from the same cells than in A and normalized to the total amount of Uridine (U) (n = 4 independent populations of MEFs/group). D Schematic representation of pyruvate contribution to U[34] tRNAs modifications when cells are incubated with uniformly labeled [U-^13C]-pyruvate (NIAID Visual & Medical Arts. 29/08/2024. Mitochondria. NIAID NIH BIOART Source. bioart.niaid.nih.gov/bioart/352). E Stable isotope tracing experiments in MEFs incubated with DMSO or the MPC1 inhibitor UK-5099 and cultured in the presence of [U-^13C]-pyruvate. ^13C-enrichment in ncm^5U[34] and mcm^5s^2U[34] was determined by LC-MS/MS in two independent experiments performed in triplicates. Data are represented as the relative ratio between the ^13C-labeled m + 2 isotopologue and the corresponding unlabeled U[34] modification. F Immunoblot analysis of PDHA1, ELP3 and TUBULIN protein levels in MEFs transduced with control or Pdha1 shRNAs lentiviruses (shPdha1 #1 and #2). Right panel: histobars represent the quantification of immunoblots performed on n = 4 independent samples. G Immunoblot analysis of ELP3 and ACTIN protein levels in human skin fibroblasts isolated from Leigh Sydrome (LS) patients harboring mutations in the indicated genes or from age-matched healthy donors. Immunoblot was performed in 2 independent experiments. H Immunoblot analysis of PDHA1, ELP3 and ACTIN protein levels in human skin fibroblasts isolated from LS patients harboring mutations in the PDHA1 gene transducted with an empty control or a WT-PDHA1 encoding retrovirus. Right panel: histobars represent the quantification of immunoblots performed on cells isolated from n = 3 LS patients. Data are presented as mean + standard error of the mean (SEM) for Fig. 8A, C or mean + standard deviation (SD) for Fig. 8B, E, F and H from the indicated number of samples. Statistical significance was evaluated using unpaired two-sided Student’s t-test (ns, not significant). PDH deficiency impacts on ELP3 expression To further understand the relationship between pyruvate metabolism and the Elongator complex, we depleted PDHA1 in MEFs using two independent short hairpin RNAs (shRNAs). Strikingly, Pdha1 knock-down (KD) cells displayed decreased ELP3 protein, but not mRNA, levels (Fig. [231]8F and Supplementary Figure [232]S6A). Comparable decrease of ELP3 protein levels were obtained upon shRNA-mediated depletion of Dlat or pharmacological inhibition of PDC activity by β-Fluoropyruvate, unraveling a crosstalk between the PDC and ELP3 expression (Supplementary Figure [233]S6B-C). Furthermore, skin fibroblasts isolated from LS patients harboring mutations in the PDHA1 or the E4F1 genes, but not in those with mutations in SURF1 or NDUFS6 displayed reduced ELP3 protein levels (Fig. [234]8G). Since the LS is a mendelian disease, we next evaluated the effect of ectopic wild-type (WT) PDHA1 on endogenous ELP3 protein levels in these patient-derived PDHA1-mutated cells. Ectopic expression of WT-PDHA1 increased ELP3 protein levels in these PDHA1-mutated LS fibroblasts, confirming the notion that ELP3 and PDC expression are tightly coordinated (Fig. [235]8H). Discussion The LS is a severe pediatric neurodegenerative disease linked to either PDC or ETC deficiencies. The prognosis for LS patients is very poor and current treatments provide limited improvement of their clinical symptoms. The paucity of animal models recapitulating the genetic alterations found in LS patients has limited studies aiming at understanding the molecular defects underlying their clinical symptoms and the development of efficient treatments. Moreover, because the relative contribution of cell autonomous versus systemic metabolic defects to the various clinical manifestations associated with LS remain elusive, the development of genetically engineered mouse models allowing tissue specific inactivation of genes related to the LS is of major interest. A brain specific Pdha1-cKO mouse model was previously generated in which PDH deficiency led to the expected metabolic reprogramming including impaired glucose-derived flux into the TCA cycle and increased lactate production. These mice also exhibited defective synthesis of neuro-transmitters linked to the TCA cycle including glutamate that was associated to neuronal excitability defects, cerebral hypotrophy and epilepsy which led to post-natal death around P25. In the same animal model, acetate and propionate were found to be efficient anaplerotic sources for the TCA which compensate, at least in part, for the lack of glucose oxidation by the PDC^[236]30,[237]31. Interestingly, combining a ketogenic diet with the systemic administration of propionate mitigated the growth defects and the neuronal phenotypes of these mice but only delayed the post-natal death of these animals by a few weeks. Hence, a better understanding of the molecular defects associated to PDH deficiency is still needed to develop more efficient therapies. The detailed molecular characterization of mice in which we inactivated E4f1, a gene encoding a key transcriptional regulator of the PDC, in both neuronal and glial progenitors during early embryonic development uncovered an unexpected link between the PDC and the control of translation fidelity. Despite these mice display a reduction but not a complete inhibition of PDH activity, all E4F1-deficient animals died within hours after birth, likely because of reduced maternal-pup interactions and inability to feed. A comparable neonatal death was described in other mouse models developing embryonic neuronal defects, such as those resulting from the genetic inactivation of the splicing factor Tra2b or Tsc1, a component of the tuberous sclerosis complex^[238]32,[239]33. Noteworthy, and consistent with our previous findings that E4F1 is a key regulator of the cell cycle and of the intra-S checkpoint^[240]5,[241]7,[242]10, E4F1-deficient animals died earlier than Pdha1 brain-specific cKOs, strongly suggesting that E4F1 functions in the CNS extend beyond the regulation of the PDC. Using E4f1-brain specific cKO mice, we unraveled a network involving E4F1, the PDC and ELP3 that links pyruvate metabolism to translation fidelity. We don’t exclude the possibility that the deregulation of other E4F1-target genes, including Nsun5 (a methyl-transferase targeting ribosomal RNAs) or Eef1g (a translation elongation factor), also contributes to the translation defects observed in E4F1-deficient animals. Further investigations will be required to evaluate whether these different components of the E4F1 core transcriptional program synergistically contribute to the translation of the same subset or of different pools of mRNAs. Nevertheless, our data indicate that these mistranslated transcripts display a codon content bias towards U[34] codons and that most of the corresponding proteins contain the hydrophilic motif previously associated with protein aggregation occurring upon perturbations of U[34]-enzymes. Altogether, these results support the notion that impaired activity of the Elongator complex plays a pivotal role in the translation defects observed in the CNS of E4F1-deficient embryos. Finally, our data also highlighted a crosstalk between the PDC and ELP3, independent of E4F1, that is perturbed in cells isolated from PDH-deficient patients, suggesting that impairement of protein translation fidelity is a common feature of cells exhibiting impaired PDH. How metabolic perturbations lead to changes in gene expression is a field of active research. Many studies highlighted the role of different metabolites, including AcCoA and S-Adenosyl-Methionine (SAM), in the regulation of the epigenome. By serving as essential co-factors of acetyltransferases and methyltransferases, these metabolites directly contribute to DNA or histone modifications including acetylation or methylation, respectively^[243]34–[244]36. Although it is known for decades that some of these acetyl- or methyl- transferases also target RNAs, including mRNAs, tRNAs, or ribosomal RNAs, the importance of their metabolic co-factors in regulating the epitranscriptome remains poorly understood^[245]37. Previous work showed that serine metabolism in mitochondria provides methyl donors to produce the taurinomethyluridine base at the wobble position of some mitochondrial tRNAs and control mitochondrial translation of OXPHOS-related transcripts encoded by the mitochondrial genome^[246]38. In our study, we validated an innovative stable isotope tracing approach to track the metabolic fate of pyruvate-derived carbons into U[34] tRNAs modifications and demonstrated that AcCoA produced by the PDC directly contributes to tRNAs modifications linked to the Elongator complex. Strikingly, in the same experimental set-up, ^13C-labeled acetate contributed much less to tRNAs U[34] acetylation, suggesting that these metabolic substrates are channeled differently to fuel different pools of AcCoA that are utilized for specific acetylation reactions. Our data support a model where E4F1 regulates a transcriptional program coordinating AcCoA production by the PDC and its utilization by the Elongator complex to promote tRNAs U[34] modifications. Perturbation of this program impaired protein translation fidelity and associated with the induction of an ISR that ultimately led to neuronal cell death. Interestingly, despite E4F1-mediated control of Elp3 occurred in different cell types including MEFs, the induction of the ISR was only observed in the CNS in vivo and in neuronal cells in vitro. The mechanisms driving this cell-type specific and cell autonomous effect remain to be identified. It is noteworthy that some antibiotics, including tetracyclines, have been shown to improve the fitness of nutrient-deprived cells isolated from patients suffering mitochondrial diseases with ETC deficiencies. Strikingly, tetracycline analogs, such as doxycycline, selectively attenuated mitochondrial translation in these cells, and when administrated to Ndufs4-mutant animals, improved their neuronal defects and prolonged significantly their lifespan. Although these antibiotics induced a clear ATF4-driven cellular response, their protective effects were rather attributed to the induction of a mitohormetic response which restored a proper redox balance and thereby improved cell survival^[247]39. These results suggest that modulating mitochondrial translation, a process amenable to pharmacological intervention, plays a pivotal role in protecting cells exhibiting ETC deficiency. In E4F1 brain-specific cKO embryos, in which we demonstrated impaired PDH activity, pharmacological inhibition of eIF2a mitigated the induction of the ISR and diminished neuronal cell death, suggesting that either the intensity or the duration of the ATF4-response may lead to different outputs in these neuronal cells. Interestingly, previous studies also suggested that inhibiting mTORC1 activity, activating a hypoxic response or modulating NAD metabolism, can partly rescue phenotypes associated to respiratory chain deficiencies^[248]40–[249]42. The efficacy of these approaches in PDH-deficient-related mitochondrial diseases has not been investigated yet but our data pave the way for further studies evaluating whether the interplay between the PDC and the Elongator complex also represents a new avenue for therapeutic intervention. Methods Study Approval All mouse procedures were approved by the ethic committee for animal warefare of the region Languedoc Roussillon (Comité d’Ethique en Expérimentation Animal Languedoc-Roussillon), an accredited institution of the French Minister for Education, Research and Innovation (agreement number #18030-201812111250227). For human fibroblasts, informed consent was obtained from all investigated subjects according to the Declaration of Helsinki principles and was approved by Research Ethics committees (agreement numbers DC 2009-939 from the “Comité d’Ethique de L’hopital Bicêtre Assistance Publique Hopitaux de Paris” and [250]GGP15041 from the “Comitato Etico Regione Lombardia”). Animal housing The following strains were used in this study: E4f1^tm1.1Llca, E4f1^tm1Pisc, Tg(Nes-cre)^1Kln, Gt(ROSA)26Sor^tm1(cre/ERT2)Tyj^[251]15–[252]18. Mice were interbred and maintained on a mix 129 Sv/J; C57Bl/6 J background and were housed in a pathogen free barrier facility (room temperature 22 °C; relative humidity 55%, and a 12-h-light–dark cycle). Animal housing and euthanasia were performed in accordance with the 3 R rules. Mice were maintained under chow diet (A03, Safe) containing 22 kcal% protein, 65 kcal% carbohydrate and 13 kcal% fat. Mice were genotyped by PCR on tail genomic DNA using Red-N extract kit (Sigma, XNAT), with primers indicated in Supplementary Table [253]1. Pregnant females were treated with ISRIB (2.5 mg/kg/day, Sigma, SML0843) by intraperitoneal injections from E10.5 onwards. All mouse strains are available upon request. Patient material Skin fibroblasts were isolated from LS patients or aged-match control individuals (between 1 month and 5 years of age) recruited at Paris Kremlin Bicêtre hospital or at the Foundation IRCSS Institute of Neurology Carlo Besta (Milan, Italy). The sex of the patients was not considered as a variable in this study although all experiments were conducted on fibroblasts isolated from both male and female Leigh syndrome patients and control individuals. Cell culture MEFs were isolated from E13.5 E4f1^-/flox; Rosa26-CRE^ER embryos and cultured for no more than 3 passages in DMEM complemented with 10% of heat-inactivated fetal bovine serum (FBS, Fisher Scientific). In vitro E4f1 inactivation in these primary cells was obtained upon incubation with 4-hydroxy-tamoxifen (1 μM, Sigma, H7904) overnight (E4f1^cKO). For stable isotope tracing experiments, MEFs were cultured for the last 24 h in serum-free media (custom DMEM/F12 containing 5.5 mM Glucose; 1 mM Glutamine; 0.2 mM Branched Chain Amino Acids and a mix of growth factors (ATCC, #PCS201040)), in presence of the MPC-1 inhibitor UK5099 (50μM, Sigma, PZ0160) for 12 h and with 1 mM of universally labeled ^13C-pyruvate (Sigma, 490717) or ^13C-Acetate (Sigma, 282014) for the last 6 hours. Skin fibroblasts isolated from LS patients or control individuals were cultured for no more than 8 passages in DMEM complemented with 10% of heat-inactivated fetal bovine serum (FBS, Fisher Scientific). The N2A murine neuroblastoma cell line (provided by Nguyen Lab) was cultured in DMEM/F12 complemented with 10% of heat-inactivated fetal bovine serum (FBS, Fisher Scientific). Primary cortical cultures (composed of neurons and glial cells) were prepared from the brain of P1 E4f1^flox/flox; Rosa26-CRE^ER newborns and cultured in BrainPhys medium (Stem Cell Technologies, 05790) supplemented with 2% B27 (Gibco, 17504044) and 0.25% Glutamax (Gibco, 35050037)^[254]43. Low passage populations of primary skin fibroblasts isolated from LS patients and age-matched control individuals (between P6 and P10), or MEFs of interest (P1–P3) can be provided upon request. RNA extraction, RNA-seq and quantitative PCR Total RNAs were isolated from various organs or from cells using TRIzol Reagent (Invitrogen, 15596018). For RNA-seq, mRNA libraries were generated following the manufacturer recommendations (mRNA hyper prep from ROCHE). Bar-coded libraries were pooled and sequenced on the Novaseq6000 200 cycles Illumina sequencer, with a sequencing depth corresponding to a minimum of 40 million of 2x100bp paired-end reads per sample after demultiplexing. Quantitative real time PCR (RT-qPCR) was performed on a LightCycler 480 (Roche). cDNAs were synthesized from 500 ng of total RNA using the SuperScript™ III Reverse Transcriptase (Invitrogen, 18080044). The relative mRNA copy number was calculated using Ct values and was normalized to 18S and tubulin β5 mRNA levels. Primers used for RT-qPCR analyzes are listed in the Supplementary Data [255]3. For RNA-sequencing, STAR v2.5.3a was used to align reads on the reference genome mm10. Gene expression was determined with RSEM 1.2.28 on RefSeq catalog, prior to normalization and differential analysis with DESeq2 bioconductor package using likelihood ratio tests (LRT). Multiple hypothesis adjusted p-values were calculated with the Benjamini–Hochberg procedure to control FDR. GSEA and PEA were performed with fgsea bioconductor package on Hallmark gene sets from MSigDB collections, completed with some custom gene sets. As for differential expression analysis, p-values were corrected using Benjamini–Hochberg’ FDR. Polysome profiling Entire brain from E14.5 embryos were lysed in polysome lysis buffer (7% sucrose, 50 mM Tris-HCl pH 7.5, 5 mM MgCl[2], 25 mM KCl, 1% NP40, 10 U/mL RNAseOUT (Invitrogen, 10777019) supplemented with protease inhibitors and 20 μg/mL Emetin (Sigma, E2375)) by hard shaking with 1.4 mm ceramic spheres (Lysing matrix D MPBio, 1169130-CF) in a FastPrep instrument (MPBio). 30 μg of total RNA were loaded on 15–50% sucrose gradient and ultracentrifuged at 2,10,000×g for 2.5 h at 4 °C in a SW41 rotor (Beckman Coulter). Approximately 10% of the cytosolic RNAs were kept as an input. The different fractions were separated through a live optical density (OD) 254 nm UV spectrometer and collected with an ISCO (Lincoln, NE) density gradient fractionation system. The absorbance at 254 nm was measured continuously as a function of gradient depth. Total cytosolic mRNAs, as well as mRNAs associated with 1 to 3 ribosomes (Light polysomal fraction) and mRNAs associated with more than 3 ribosomes (Heavy polysomal fraction) were extracted using TRIzol LS (Invitrogen, 10296028) according to the manufacturer instructions. For sequencing, mRNA libraries were generated following the manufacturer recommendations (mRNA hyper prep from ROCHE). Bar-coded libraries were pooled and sequenced on a Novaseq6000 200 cycles Illumina sequencer, with a sequencing depth corresponding to a minimum of 80 million of 2 × 100 bp paired-end reads per sample after demultiplexing. Polysome profiling datasets were pre-processed using the nf-core RNA-seq pipeline v3.11.0^[256]44. Initial quality control of raw reads was conducted with FastQC v0.11.9. Trim Galore v0.6.7 was used for reads trimming, with parameters set to a minimum read length of 25 bp, a quality threshold of Phred score 30, and a stringency of 10. Additionally, 1 bp was clipped from both 5’ ends of R1 and R2 reads. Reads were aligned to the mouse reference genome (GRCm38) using STAR v2.7.9a. Transcript-level quantification was performed using Salmon v1.10.1, and gene-level summarization was carried out using tximport v1.32.0. Post-alignment quality was assessed with RSeQC v3.0.1 to ensure data integrity and alignment accuracy. For statistical analysis, datasets from polysome fractions (light, heavy, and total) were analyzed independently using R v4.4.0. Differential expression (DE) analysis for each fraction was conducted with edgeR v4.2.1, comparing gene expression between E4f1^(Nes)KO and CTL animals. Normalized count data were fitted to a generalized linear model (GLM) according to E4f1 status as a cofactor (KO or WT). After estimating the signal dispersion, the abundance of transcripts in a given fraction was considered significantly different in likelihood ratio tests when exhibiting an absolute fold-change ≥ 1.2 and a FDR-adjusted p-value < 0.2. To integrate results from the different fractions, the corresponding genes were categorized as controlled at the post-transcriptional level (differential abundance of their transcripts in either the heavy and/or the light polysomal fractions, but not in the total fraction), at the transcriptional level (differential abundance of their transcripts in the total fraction but not in the heavy nor in the light polysomal fractions), and at both the transcriptional and post-trancriptional (Mix) levels (differential abundance of their transcripts in the total fraction and in the heavy and/or in the light polysomal fractions). Codon usage bias was assessed by comparing codon frequencies in transcripts controlled at the post-trancriptional level in E4F1-deficient cells to those in transcripts from the entire GRCm38 transcriptome using χ^2 tests. In the corresponding proteins, enrichment of the specific hydrophilic motif linked to U[34]-dependent translational defects was conducted using the AME (Analysis of Motif Enrichment)^[257]45 tool from the MEME suite v1.12.0, using the occurrence of this motif in the full GRCm38 proteome as a reference. Measurement of PDH activity and blood lactate levels PDH activity was measured using Pyruvate Dehydrogenase Activity Assay Kit (MAK183-1KT, Sigma) as recommended by the manufacturer. Lactate levels were measured in blood using a lactometer (EKF Diagnostics). AcetylCoEnzyme A (AcCoA) quantification Brains from E14.5 embryos were lysed in 10% trichloroacetic acid (TCA) diluted in HPLC-grade water (Fisher Scientific) precooled at 4 °C. Samples were centrifuged and supernatants were loaded onto SPE columns (Oasis HLB 3cc; Waters WAT094226). Columns were washed with HPLC-grade water, then eluted with HPLC-grade MeOH with 25 mM ammonium acetate. Samples were dried with unheated speed vac, resuspended in HPLC-grade water, transfered onto HPLC glass vials and placed in the autosampler at 4 °C. LC–high resolution mass spectrometry quantification was performed using a Vanquish chromatographic system coupled with a Q-Exactive Plus mass spectrometer (Thermo Fisher Scientific) fitted with an electrospray source. Chromatographic separation was performed on an InfinityLab Poroshell HPH-C18 column (2.7 μm, 3.0 × 100 mm; Agilent) maintained at 30 °C. Identification was determined by extracting the accurate mass of acyl-CoAs with a mass accuracy of 5 ppm and comparing the retention time to standards injected in the same sequence (mix of 5 μM acyl-CoA). Peak areas of acyl-CoAs were divided by peak area of the internal standard used, MES, and by the weight of the corresponding brain. Data treatment was done with Xcalibur Freestyle Software (Thermo Fisher Scientific). Protein extraction and western blotting Total protein extracts were prepared from total brains isolated from E14.5, E16.5 or E18.5 E4f1^(Nes)KO and CTL embryos lysed in Laemmli buffer (80 mM Tris pH = 6,8, 2% SDS, 12% sucrose, 2% β-mercaptoethanol, bromophenol blue) and immunoblotting was performed using the following antibodies: anti- E4F1 (1/1000)^[258]46, C-CASP3 (Cell Signaling, 9661S, lot 43, 1/1000), DLAT (Santa Cruz, sc-271534, lot C1813, 1/500), MPC1 (Sigma, HPA045119, lot E105936, 1/1000), DLD (Santa Cruz, sc-365977, lot F1611, 1/1000), PDHE1 (Life Technologies, 459400, lot J4161, 1/500), ELP3 (Abcam, ab190907, lot GR3180892-3, 1/500), ELP1 (Abcam, ab62498, lot GR3369590, 1/500), TUBULIN (Sigma, T6199, lot 115M4796V, 1/1000), ACTIN (Sigma, A3854, lot 100M4782, 1/7000), Acetylated-TUBULIN (Santa Cruz, sc23950, lot E2218, 1/500), Histone H3K9Ac (Cell Signaling, 9649S, lot 13, 1/1000), Histone H3K23Ac (Abcam, ab61234, lot GR82698-4, 1/1000), Histone H3K27Ac (Abcam, ab4729, lot 17, 1/1000), Histone H3K14Ac (Cell Signaling, 7627, 1/1000), total Histone H3 (Cell Signaling, 4499S, lot 9, 1/1000), HSP90 (Cell Signaling, 13998S, lot 2, 1/1000), KCNA3 (Diagomics, AF6702, lot 23, 1/500) and Cleaved-Caspase 8 (Proteintech, 66093-1, lot 10023946, 1/1000). LC-MS detection of tRNA modifications Total RNAs were isolated from brains of E14.5 E4f1^(Nes)KO and CTL embryos or from MEFs using TRIzol Reagent (Invitrogen, 15596018). tRNAs were purified on an agarose gel. 400 ng of tRNAs were digested with 0.001U of nuclease P1 (Sigma, N8630) in 25 mM of ammonium acetate pH 5.3 for 2 hours at 42 °C. Samples were then treated with 0.001U of alkaline phosphatase (Sigma, P4252) in 250 mM ammonium acetate pH 5.3 at 37 °C for 2 h. The nucleoside solution was diluted twice and filtered with 0.22 µm filters (Millex®-GV, Millipore, SLGVR04NL). 5 µL of each sample was injected in triplicate in a LC-MS/MS. For stable isotope labeling experiments performed in MEFs using ^13C-sodium pyruvate (Sigma, 490717) or ^13C-sodium acetate (Sigma, 282014), 4 ug of total RNA were digested with 0.002 U of nuclease P1 and alkaline phosphatase by following the same procedure. Nucleosides were then separated using Nexera LC-40 systems (Shimadzu) and Synergi™ Fusion-RP C18 column (4 µm particle size, 250 mm × 2 mm, 80 Å) (Phenomenex, 00G-4424-B0). Mobile phase was composed of 5 mM ammonium acetate pH5.3 (solvent A) and acetonitrile (solvent B). The 30 minutes elution gradient started with 100% phase A followed by a linear gradient until reaching 8% solvent B at 13 min. Solvent B was further increased to 40% over the next 10 min. After 2 min, solvent B was decreased back to 0% at 25.5 minutes. Initial conditions were recovered by rinsing the LC-system with 100% solvent A for additional 4.5 min. The flow rate was set at 0.4 mL/min and the column temperature at 35 °C. Analysis was performed using a triple quadrupole 8060NX (Shimadzu corporation, Kyoto, Japan) in positive ion mode. Peak areas were measured using Skyline 21.2 software (MacCoss Lab Software; [259]https://skyline.ms) and the ratio of modified nucleosides on non-modified nucleosides was calculated. Quantitative chromatin immunoprecipitation (qChIP) assays For qChIP assays in cells, N2A cells were incubated with 1% formaldehyde / 1% paraformaldehyde for 5 min. Nuclei were isolated and chromatin was extracted and sonicated (epishear, Active Motif). qChIPs on the Elp3 locus were carried out using a control irrelevant IgG or an affinity-purified rabbit anti-E4F1 polyclonal antibody^[260]7,[261]22,[262]46. Primers used for qChIP assays are listed in the supplementary table [263]1. Lentivirus production Lentiviral and retroviral particles encoding Flag-tagged human WT-PDHA1, the PDHA1^C91A mutant, human E4F1^WT or the E4F1^K144Q mutant, and shRNAs targeting mouse Pdha1 (TRCN0000041914, TRCN0000041914, Sigma) or mouse Dlat (TRCN0000041608 and TRCN0000041609, Sigma) were produced in HEK293T packaging cells. 72 h after transfection, viral supernatants were harvested and added on MEFs overnight in presence of polybrene (5 μg/mL, Sigma). Selection of transduced cells was performed 48 h after transduction with puromycin (2 μg/mL, Invitrogen, J67236.8EQ) or hygromycin (50 μg/mL, Invitrogen, 10687010). Immunohistochemistry, immunofluorescence and histological analyzes The brain of E14.5, E16.5, E18.5 E4f1^(Nes)KO and CTL embryos were dissected in 0.1 M phosphate-buffered saline pH7.4 (PBS) and were fixed at 4 °C in 4% paraformaldehyde (PFA) for one hour. Fixed samples were cryoprotected overnight in 20% sucrose in PBS at 4 °C, embedded in OCT Compound (VWR International, 361603E) and sectioned (14 µm) onto slides (SuperFrost Plus, VWR International, 631-0108) using a cryostat. Entire head samples were fixed in 4% neutral-buffered formalin (24 h), decalcificated in 10% EDTA for 3 days and paraffin embedded. FFPE tissues were sectioned (4μm sections) and processed for immunohistochemistry (IHC) or hematoxylin and eosin (H/E) staining. For GFP (Invitrogen, A10262,, lot 2260907, 1/100), Cleaved-Caspase 3 (Cell Signaling, 9661S, lot 43, 1/100) and CD45 (Biosciences, 14-051-82, lot 4316815, 1/500) stainings, IHC was performed on a VENTANA Discovery Ultra automated staining instrument (Ventana Medical Systems) according to the manufacturer’s instructions. The number of Cleaved-Caspase3 positive cells were quantified on whole-slides digitized sections (Panoramic 250 Flash II/3DHISTECH whole-slide digital microscope) scanned at a 0.22 µm/pixel resolution, using Definiens Developer XD software (Definiens, Munich, Germany). IF stainings were processed manually. Briefly, brain sections were pretreated with target retrieval solution (DAKO, 20 min, 95 °C) prior incubation with the following primary antibodies overnight at 4 °C: anti- MCT4 (Proteintech, 22787-1-AP, lot 00049309, 1/200), ATF4 (Cell Signaling, 11815, lot 6, 1/200), phospho-eIF2a (Cliniscience, [264]P04387, lot [265]P04387, 1/500), Ki67 (abcam, Ab15580, lot GR342706-1, 1/500), SOX2 (Santa Cruz, Sc17320, lot #H0516, 1/500), TBR2 (eBiosciences, 14-4875-82, lot 2504948, 1/250), TBR1 (Abcam, ab31940, lot GR3182037-1, 1/500), GFAP (Avès Labs, AB2313547, lot 4937976, 1/200) and NeuN (Biolegend, 834501, lot B352650, 1/500). After washing, sections were incubated for 1 h at room temperature with either anti-rabbit, anti-rat or anti-goat secondary antibodies coupled to Alexa-488, Alexa-647 or Alexa-555 (Life Technologies). Nuclei were counterstained with DAPI (Sigma, D9542) and slides were washed in PBST and coverslipped using Mowiol mounting solution. For images analyzes, sections were visualized on a Thunder epifluorescent microscope (Nikon). High-Resolution Episcopic Microscopy (HREM) E14.5 embryos and E16.5 heads were fixed in Bouin’s fixative for 24 h, then washed and stored in 70% EtOH. Fixed samples were gradually dehydrated in an increasing series of ethanol concentrations and were embedded in a methacrylate resin (JB-4 kit, Polysciences, Warrington, PA) containing eosin and acridin orange as contrasts agents^[266]47. The resin blocks were sectioned on the Histo3D system to generate data by repeated removal of 7 µm thick sections^[267]48. Resulting HREM data with a voxel size of 8 × 8 × 7 µm^3 (E14.5 whole embryos) or 6.5 × 6.5 × 7 µm^3 (E16.5 heads) were generated from approximately 1000 or 800 aligned images, respectively. All HREM images were converted into a volume dataset and segmented using Avizo 9.4.0 software (Thermofisher Scientific, France) to create 2- and 3-dimensional (3D) images, according to the Allen brain atlas. Statistical analysis In vivo studies were performed on a sufficient number of animals per genotype with a minimum of 5 animals per experimental group. The in vitro data were obtained from 3 to 5 independent experiments as indicated in the figure legends. Statistical significance was evaluated using unpaired bilateral Student’s t-test. P-values inferior or equal to 0.05 were considered statistically significant. Reporting summary Further information on research design is available in the [268]Nature Portfolio Reporting Summary linked to this article. Supplementary information [269]Supplementary Information^ (5.6MB, pdf) [270]41467_2024_55444_MOESM2_ESM.pdf^ (117.6KB, pdf) Description of Additional Supplementary Files [271]Supplementary Data 1^ (2.2MB, xlsx) [272]Supplementary Data 2^ (9.5KB, xlsx) [273]Supplementary Data 3^ (4.5MB, xlsx) [274]Reporting Summary^ (142.5KB, pdf) Source data [275]Source Data^ (32.5MB, xlsx) Acknowledgements