Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 24.
Published in final edited form as: Nat Cell Biol. 2024 May 13;26(6):892–902. doi: 10.1038/s41556-024-01414-x

Polyglutamine-mediated ribotoxicity disrupts proteostasis and stress responses in Huntington’s disease

Ranen Aviner 1,2,4, Ting-Ting Lee 1, Vincent B Masto 1, Kathy H Li 3, Raul Andino 2, Judith Frydman 1
PMCID: PMC12288859  NIHMSID: NIHMS2087603  PMID: 38741019

Abstract

Huntington’s disease (HD) is a neurodegenerative disorder caused by expansion of a CAG trinucleotide repeat in the Huntingtin (HTT) gene, encoding a homopolymeric polyglutamine (polyQ) tract. Although mutant HTT (mHTT) protein is known to aggregate, the links between aggregation and neurotoxicity remain unclear. Here we show that both translation and aggregation of wild-type HTT and mHTT are regulated by a stress-responsive upstream open reading frame and that polyQ expansions cause abortive translation termination and release of truncated, aggregation-prone mHTT fragments. Notably, we find that mHTT depletes translation elongation factor eIF5A in brains of symptomatic HD mice and cultured HD cells, leading to pervasive ribosome pausing and collisions. Loss of eIF5A disrupts homeostatic controls and impairs recovery from acute stress. Importantly, drugs that inhibit translation initiation reduce premature termination and mitigate this escalating cascade of ribotoxic stress and dysfunction in HD.


Huntington’s disease (HD) is a progressive, age-dependent neurodegenerative disorder characterized by motor, cognitive and psychiatric symptoms1. It is caused by expansion of a CAG repeat segment in exon 1 of the Huntingtin (HTT) gene (herein HTT-ex1), encoding a variable-length polyglutamine (polyQ) tract. Repeats longer than 35 glutamines are pathogenic, and length is inversely correlated with age of onset2. CAG expansions in other genes cause similar neurodegenerative disorders (the so-called triplet disorders), including spinocerebral ataxias3, suggesting common mechanisms of toxicity. Still, it remains unclear how CAG expansions in a single gene cause such widespread dysfunction that affects multiple cellular pathways, including DNA replication and repair, transcription, splicing, mitochondrial function, autophagy and stress responses1,2,4.

One common hypothesis argues that toxicity is mediated by the mutant protein. Indeed, longer polyQ tracts tend to form oligomers, aggregates and inclusion bodies associated with HD-like symptoms1,2,4. Both soluble oligomers and insoluble aggregates of mutant HTT (mHTT) can sequester other proteins, for example, molecular chaperones57, depleting them from the cellular pool and disrupting proteostasis. However, aggregate or inclusion formation can also be neuroprotective8, and neurons from affected individuals often lack aggregates2, suggesting a more complex relationship between repeat expansion, aggregation and neurodegeneration.

An alternative hypothesis posits that CAG repeats mediate RNA-based toxicity by affecting RNA structure and translation fidelity or sequestering RNA-binding proteins9. Indeed, patients with pathogenic CAG expansions interspersed with the synonymous glutamine codon, CAA, show delayed disease onset10. As these variants produce an identical polyQ tract, their protective effect cannot be solely due to changes in aggregation propensity of the encoded protein.

In this Article, to reconcile these observations, we explored links between repeat expansion, translation, protein aggregation and neurotoxicity in HD. Using a combination of cellular and animal models, ribosome profiling (ribo-seq) and proteomic analyses, we find that translation and aggregation of mHTT are dynamically regulated by an upstream open reading frame (uORF) and show that CAG expansions trigger abortive termination of mHTT translation, thus releasing truncated fragments. Furthermore, mHTT interacts with and sequesters translation factor eIF5A, promoting widespread ribosome pausing across the transcriptome. This causes ribo- and proteotoxicity, which can be mitigated by small-molecule inhibitors of translation initiation.

Results

uORF dynamically regulates HTT translation

Human HTT messenger RNA harbours a uORF (Fig. 1a) that suppresses translation of a downstream reporter when ectopically expressed11. This uORF is evolutionarily conserved from mouse to human in size and distance from the main open reading frame (ORF) (Fig. 1b), and features an optimal translation initiation context12. uORFs are generally known to inhibit the translation of downstream ORFs13. To determine whether this uORF inhibits HTT translation in vivo, we analysed previously published ribo-seq datasets. In cultured human cells treated with either lactimidomycin (LTM) or harringtonine (Harr.)14, which arrest ribosomes at translation start sites, we find initiating ribosomes on both the uORF and main ORF of HTT mRNA (Fig. 1c and Extended Data Fig. 1a). In cells treated with cycloheximide (CHX), which blocks translation elongation, ribosomes occupy the entire length of the uORF but not flanking regions (Fig. 1c and Extended Data Fig. 1a), suggesting that ribosomes translate the uORF and terminate ahead of the main HTT ORF. Similar ribosome traces were also found in mouse hippocampus and cultured primary neurons15 (Fig. 1d), as well as other tissues and cell types (Extended Data Fig. 1b).

Fig. 1 |. uORF modulates translation of HTT under normal and stress conditions.

Fig. 1 |

a,b, A schematic illustration of mammalian HTT mRNA (a) and its 5′ UTR (b). c, HTT uORF is translated in culture. Ribosome profiling (ribo-seq) traces of initiating (A-site positions, top) and elongating (footprints, bottom) ribosomes on the 5′ region of HTT mRNA in HCT116 cells (n = 1 each). Footprints on the uORF and main ORF are in purple and grey, respectively. d, HTT uORF is translated in mouse brains. Ribo-seq traces of elongating ribosomes on Htt mRNA in mouse hippocampus (top, representative of n = 15 mice) and primary neurons isolated from mouse hippocampus (bottom, n = 1). e, The TE of Htt compared with known uORF-suppressed transcripts in mouse hippocampus, calculated as ribosome density on the main ORF divided by transcript abundance (n = 15 mice). Centre lines show medians, box limits indicate upper and lower quartiles, and whiskers extend to 1.5 times the interquartile range. fh, Htt translation is induced during acute ER stress: WT mouse striatal cells were treated with either 1 μM Tg or dimethyl sulfoxide (DMSO) for 1 h, and lysates were fractionated on 10–50% sucrose gradients (n = 3), and shown are representative traces of absorbance at 254 nm (f); qPCR analysis of specific mRNAs in fractions from f (g); immunoblots of Htt protein accumulation (D7F7 antibody) during acute ER stress (h). Mean ± s.d. of three independent repeats for qPCR and representative blots and densitometry from three independent repeats. P, P value of a two-sided Student’s t-test comparing heavy polysome fractions from stressed and unstressed cells. Source numerical data and unprocessed blots are available in Source data.

If the uORF suppresses translation of the main ORF in vivo, this should be reflected by low translation efficiency (TE), calculated as ribosome occupancy on the main ORF divided by RNA abundance. Indeed, the TE of Htt in mouse hippocampus is lower than that of highly expressed proteins and similar to that of Atf4 and Ppp1r15a (GADD34) (Fig. 1e), which are translationally repressed by their uORFs under basal conditions16. Taken together, these observations suggest that the endogenous uORF inhibits translation of HTT in vivo by reducing initiation on the main ORF.

uORF-regulated transcripts, including Atf4 and Ppp1r15a, are known to increase their translation during conditions that trigger the integrated stress response (ISR), including endoplasmic reticulum (ER) stress and neuronal differentiation. This occurs through a mechanism that involves uORF bypassing and/or re-initiation1618. To determine whether Htt translation is upregulated during stress, we analysed ribo-seq datasets from mouse embryonic fibroblasts undergoing acute ER stress19 or SH-SY5Y neuroblastoma cells undergoing differentiation17. In response to stress, more ribosomes bypass the uORF on both ATF4 and HTT (Extended Data Fig. 1c). To confirm this, we monitored the effect of thapsigargin (Tg)-induced ER stress on Htt translation in neuron-like cells isolated from mouse striatum (STHdh striatal cells)20—the region of the brain most affected in patients with HD1. We measured the association of specific transcripts with translating polysomes by sucrose gradient fractionation and quantitative polymerase chain reaction (qPCR). Tg treatment reduced global translation, seen as ribosome run-off in polysome profiles (Fig. 1f), and displaced the housekeeping β-actin (Actb) mRNA from polysomes (Fig. 1g), consistent with a general attenuation of translation initiation during stress. In contrast, Htt and Atf4 mRNAs responded to Tg treatment by shifting to heavier polysome fractions (Fig. 1g), as expected for uORF-repressed transcripts that are translationally induced during stress. Furthermore, immunoblot analysis confirmed that Htt protein levels increase in Tg-treated striatal cells (Fig. 1h).

uORF modulates aggregation of mHTT

Because HTT aggregation is concentration dependent2, we next tested whether the presence of a functional uORF in HTT mRNA affects the solubility of the encoded protein. Expression of HTT-ex1–GFP variants with and without the endogenous 5′ untranslated region (UTR) of human HTT revealed that the uORF delays formation of inclusions in transfected 293T cells (Fig. 2ac and Extended Data Fig. 2a). This effect was lost by mutating the uORF AUG to AAG, preventing translation initiation (Fig. 2ac and Extended Data Fig. 2a). Immunoblot analysis of the soluble and insoluble fractions of transfected cells confirmed that the uORF reduces the fraction of insoluble mHTT-ex1 in both 293T and mouse striatal cells (Fig. 2d and Extended Data Fig. 2b,c).

Fig. 2 |. uORF reduces truncation and aggregation of mHTT protein.

Fig. 2 |

ac, HTT uORF reduces inclusion body formation in transfected 293T cells: immunofluorescence analysis of 293T cells expressing HTT-ex1–GFP plasmid variants with or without the endogenous 5′ UTR of human HTT, imaged at 40 h post-transfection (a); quantification of cells with inclusions at 40 h post-transfection (b) and at 0 to 60 h post-transfection (c). Mean ± s.d. of cells with GFP-positive inclusions, counted manually from 15 random fields of each condition, using 20× magnification. About 500 cells were counted for each condition. Specific cell counts can be found in Source data. P, P value of a two-sided Student’s t-test. ND, not detected. hpt, hours post-tranfection. d, uORF reduces mHTT truncation and aggregation. Immunoblots of soluble and insoluble fractions from 293T cells transfected with HTT-ex1–GFP for 24 h (n = 4), probed with either GFP- or HTT-specific (MAB5492) antibodies. WB, western blot. Pink arrowheads indicate truncated mHTT species. e, mHTT incurs abortive termination that is mitigated by mild inhibition of translation initiation using 4EGI-1. Flow cytometry (FACS) analysis of 293T cells transfected with tandem fluorescent reporters of GFP and mCherry, separated by test sequences. K0, no polylysines; K20, 20 consecutive lysines; Q25 and Q97, HTT-ex1 with polyQ(25) or polyQ(97). 12.5 μM 4EGI-1 was added 40 h post-transfection for 6 h. Mean ± s.d., n = 3. P, P value of a two-sided Student’s t-test. Source numerical data and unprocessed blots are available in Source data.

Interestingly, the uORF also reduced formation of truncated mHTT-ex1 fragments lacking the C-terminal GFP, which were detected only in the insoluble fraction of transfected cells (Fig. 2d, pink arrowheads, and Extended Data Fig. 2b). Such neurotoxic mHTT-ex1 truncated fragments are also detected in brains of patients with HD and mouse models1. To test whether mHTT-ex1 truncations originate from premature translation termination on mHTT-ex1, we used a previously established translation stalling reporter, GFP–KAAA(20)–mCherry21. Upon transfection, the ratio of mCherry to GFP, translated from a single ORF and measured by flow cytometry, provides a proxy for abortive termination. We replaced the polylysine stalling sequence with HTT-ex1 and found that translation of polyQ(97) incurs a higher rate of abortive termination compared with polyQ(25) (Fig. 2e and Extended Data Fig. 2d). Inhibitors of translation initiation reduce premature termination on the polylysine reporter by lowering ribosome density on the coding region22,23. Abortive termination of polyQ(97) was also reduced by mild suppression of translation initiation using 4EGI-1 (Fig. 2e and Extended Data Fig. 2e,f), an eIF4A1 inhibitor that ameliorates motor symptoms in a mouse model of HD24. Mild suppression of initiation also reduced the ratio of truncated to full-length mHTT-ex1 found in the insoluble fraction of transfected 293T cells (Extended Data Fig. 2e). We conclude that attenuating mHTT-ex1 translation initiation, either pharmacologically or via its uORF, reduces premature translation termination and release of aggregation-prone fragments.

Ribosome collisions on mHTT cause premature translation termination

Since persistent ribosome collisions can lead to abortive termination25, we asked if CAG expansions increase the risk of ribosome collisions on mHTT-ex1. Indeed, disomes can form on glutamine repeats in yeast26 and polyproline tracts, such as the one just downstream of the polyQ in HTT, are known to cause collisions27,28. We were unable to test this on endogenous HTT mRNA due to its low abundance and low TE, as well as the inability to confidently map CAG repeats by ribo-seq. To address this gap, we developed a viral delivery platform to express, label and detect HTT nascent chains (NCs) in cells. We introduced the coding sequence for wild-type (WT) polyQ(8) or mutant polyQ(73) human HTT-ex1 into the single ORF of poliovirus (Fig. 3a, top). Upon infection, poliovirus rapidly shuts off cellular mRNA translation and reprogrammes ribosomes to translate viral RNA from an internal ribosome entry site29. In the engineered virus, HTT-ex1 is translated as part of the viral polyprotein and then released by proteolytic cleavage once the downstream viral protease is fully synthesized.

Fig. 3 |. Ribosome collisions cause abortive translation termination of mHTT.

Fig. 3 |

a, The experimental design of the viral delivery system to express, label and monitor HTT-ex1 NCs. WT polyQ(8) or mutant polyQ(73) HTT-ex1 was cloned into the single ORF of poliovirus genome. Infection with the engineered viruses triggers rapid shut-off of host mRNA translation and homogenous, synchronized translation of HTT-ex1 as part of the viral polypeptide. Infected cells are treated with puromycin to label nascent polypeptide chains (NCs) and analysed by immunoblotting. b, Immunoblots of Huh7 cells infected with the engineered viruses for 3.5 h and treated with 1 μM puromycin for 15 min before collection (n = 5). The pink box shows nascent virus–HTT polyproteins before cleavage by the viral protease. WB, western blot. c, Lysates from cells infected as above were digested with RNase I, fractionated on 10–50% sucrose gradients to separate RNase-resistant collided ribosomes, and treated with puromycin to label NCs (n = 3). Top: absorbance profiles at 254 nm. Bottom: immunoblot of matching fractions. d, Lysates from cells infected as above were fractionated on 10–50% sucrose gradients and treated with puromycin to label NCs (n = 3). e, Ribosomes were extracted from the fractions in d (pink box) and subjected to proteomic analysis (n = 3). Shown are individual (circles) and averages (lines) of MS-detected viral peptides mapping either N- or C-terminally to the HTT-ex1 insert. f, Box (left) and volcano (right) analyses of polysome interactors identified by MS analysis from e (n = 3). P, Mann–Whitney P value. Centre lines show medians, box limits indicate upper and lower quartiles, and whiskers extend to 1.5 times the interquartile range. g, Lysates from cells infected as above were fractionated on 10–50% sucrose gradients and analysed by immunoblotting (n = 3). Top: absorbance profiles at 254 nm. Bottom: immunoblots of eIF5A. Source numerical data and unprocessed blots are available in Source data.

Consistent with a biosynthetic defect caused by polyQ expansion, viruses harbouring polyQ(73) replicated more slowly than their polyQ(8) counterparts (Extended Data Fig. 3a). To monitor ribosome distribution on HTT-ex1, we labelled NCs with puromycin, which becomes covalently incorporated by elongating ribosomes and can be detected using a puromycin-specific antibody30. If ribosomes are evenly distributed on the translated RNA, the size of puromycylated NCs should also be evenly distributed; in contrast, local accumulation of ribosomes on a specific region of the RNA will generate puromycylated NCs of a discrete size range. As predicted, expression of mHTT-ex1 in either Huh7 (Fig. 3b and Extended Data Fig. 3b) or SH-SY5Y neuroblastoma (Extended Data Fig. 3c) cells produced a strong smear of puromycylated NCs (Fig. 3b, top and middle), indicative of local ribosome build-up. The size of these puromycylated products is consistent with ribosome stalling on the expanded polyQ tract, before cleavage of N- and C-terminal viral sequences by the downstream viral protease.

Collided ribosomes form a tight interface that is resistant to RNase digestion31. If ribosome build-up on mHTT generates such an interface, the above polyQ(73)-puromycylated NCs should be associated with RNase-resistant disomes or higher-order polysomes. To test this, we infected cells as above, digested lysates with RNase I and performed sucrose gradient fractionation. The presence and size of NCs in each fraction were assessed by puromycin labelling. As predicted, more RNase-resistant trisomes were formed on polyQ(73) than polyQ(8) (Fig. 3c, top). Importantly, these trisomes were enriched for the puromycylated NCs specific to polyQ(73) (Fig. 3c and Extended Data Fig. 3d).

To isolate ribosomes that collide on mHTT-ex1, we infected cells as above, fractionated lysates on sucrose gradients and labelled NCs using puromycin. Based on puromycin immunoblots (Fig. 3d), we extracted ribosomes from fractions with polyQ(73)-specific collision events, and analysed their protein content by liquid chromatography–tandem mass spectrometry (LC–MS/MS). Consistent with premature termination on CAG expansions, less viral NCs were detected downstream of polyQ(73) as compared with polyQ(8) (Fig. 3e). This resulted in lower levels of the viral protein encoded downstream of mHTT-ex1 (Extended Data Fig. 3e). Taken together, we conclude that polyQ expansions in HTT increase the likelihood of ribosome collisions, abortive termination and release of aggregation-prone mHTT fragments.

mHTT depletes soluble eIF5A

Collisions recruit ribosome quality control (RQC) and protein quality control (PQC) factors to promote either resolution and continued elongation or premature termination and clearance25,32,33. To determine whether polyQ(73) translation attracts specific RQC and PQC factors, we compared the proteins that interact with polysomes translating either polyQ(73) or polyQ(8) HTT-ex1 (Supplementary Table 1). We find that ribosomes translating polyQ(73) mHTT recruit more RQC and PQC factors, including higher levels of proteasomes (Fig. 3f, left); collision-associated factors GCN1, ABCE1 and UFL1; and PQC factors involved in the clearance of mHTT, including UBR5 (ref. 34), VCP/p97 (ref. 35), HUWE1 (ref. 36), ADRM1 (ref. 37) and HSPB1/Hsp27 (ref. 38) (Fig. 3f, right).

Interestingly, despite similar interaction of translation elongation factors eEF1A1 and eEF2, we find that elongation factor eIF5A is markedly depleted from ribosomes translating polyQ(73) (Fig. 3f, pink, and Fig. 3g). Given the critical role of eIF5A in translation, proteostasis and neurodevelopment3941, as well as its involvement in TDP-43 aggregation42, we hypothesized that its depletion from ribosomes—potentially mediated by interactions with mHTT—may play a role in the aetiology of HD. To determine whether mHTT interacts with eIF5A, we transfected 293T cells with plasmids encoding Htt-ex1–GFP variants followed by GFP affinity purification. More eIF5A co-purified with polyQ(97) mHTT as compared with WT control (Fig. 4a). Similarly, more eIF5A interacted with mHTT-ex1 in mouse N2a cells43 (Fig. 4b) and in yeast44. Finally, eIF5A was also highly enriched in the insoluble fraction of N2a cells expressing mHTT-ex1 (ref. 45) (Fig. 4c), suggesting that its interactions with soluble mHTT lead to aggregation.

Fig. 4 |. polyQ expansions deplete eIF5A.

Fig. 4 |

a,b, eIF5A preferentially interacts with polyQ-expanded mHTT: immunoblots of GFP co-IP from 293T cells transfected with the indicated HTT-ex1–GFP variants (n = 2). WB, western blot (a); re-analysis of previously published proteomic data of GFP co-IP from mouse N2a neuroblastoma cells transfected with HTT-ex1–GFP variants (n = 3) (b). Mean ± s.d. P, P value of a two-sided Student’s t-test. c, eIF5A is enriched in aggregates from N2a cells transfected with mHTT-ex1. Re-analysis of previously published proteomic data in which mouse N2a neuroblastoma cells were transfected as above and the insoluble fraction was subjected to proteomic analysis (n = 3). d, The timeline of mHtt aggregation and symptom onset in the transgenic R6/2 mouse HD model. e, eIF5A is enriched in aggregates from symptomatic but not asymptomatic R6/2 mice. Re-analysis of previously published proteomic data in which soluble and insoluble brain proteomes from WT and R6/2 HD mice were analysed at different ages. The protein rank plot shows the t-test differences in the insoluble brain proteome R6/2 versus WT mice at 12 weeks. Inset: same analysis for early, intermediate and late ages (5, 8 and 12 weeks, respectively). n = 16, 12 and 12 for WT or n = 16, 12 and 16 for R6/2 at weeks 5, 8 and 12, respectively. For additional information, see ‘Statistics and reproducibility’ section. Centre lines show medians, box limits indicate upper and lower quartiles, and whiskers extend to 1.5 times the interquartile range. f, eIF5A levels are inversely correlated with the levels of mutant but not WT Htt, and only in symptomatic mice. Pairwise comparisons of eIF5A and Htt levels in the soluble brain proteome of WT and R6/2 mice (n = 16 and 12 for WT or n = 16 and 16 for R6/2 at weeks 5 and 12, respectively). r, Pearson’s correlation coefficient. g, Ribosomes from brains of aged HD mice show hyperubiquitination on sites associated with ribosome collisions. Re-analysis of previously published proteomic data in which total soluble brain proteomes and ubiquitin (Ub) remnant pulldowns from WT and zQ175 HD mice were analysed. Left: pairwise comparisons of eIF5A and Htt in 40-week-old WT and Q175 mice. Right: relative ubiquitinationof specific sites on ribosomal proteins, as determined by MS analysis (n = 4 mice of each genotype). Mean ± s.d. P, P value of a two-sided Student’s t-test; r, Pearson’s correlation coefficient. Source numerical data and unprocessed blots are available in Source data.

To relate these observations to HD pathology, we next examined a longitudinal dataset of soluble and insoluble brain proteomes from pre-symptomatic and symptomatic R6/2 HD mice, expressing mHtt with polyQ(>140), compared with age-matched WT controls6 (Fig. 4d). eIF5A was highly abundant in the insoluble fraction of aged, symptomatic R6/2 HD brains, but not that of young animals (Fig. 4e). Analysis of soluble translation elongation factors revealed that eIF5A—but not other elongation factors—becomes anti-correlated with soluble mHtt as R6/2 mice get older and develop symptoms (Fig. 4f and Extended Data Fig. 4a). Interestingly, at the pre-symptomatic age of 8 weeks, we observed an increase in the soluble levels of eIF5A and RQC factors Gcn1, Ltn1 and Drg2 (Extended Data Fig. 4b,c), potentially reflecting an early adaptive response to ribotoxic stress.

eIF5A facilitates translation elongation across ribosomal stall sites, particularly proline-rich sequences27,46. Knockdown of eIF5A increases ribosome pausing and collisions in yeast and human cells46,47. Therefore, we hypothesized that the reduction in soluble eIF5A in older HD mice should result in increased ribosome collisions throughout the transcriptome. Since ribosome collisions lead to ubiquitination of specific ribosomal subunits48,49, we searched for these hallmark ubiquitination events in brain proteomics and ubiquitomics data of another HD mouse model (zQ175 (ref. 50)). This model showed a similar inverse correlation between soluble mHTT and eIF5A levels (Fig. 4g, left), as well as increased collision-related ubiquitination of ribosomal proteins48,49 in aged HD animals (Fig. 4g).

Loss of eIF5A affects translation elongation dynamics

To better define how loss of eIF5A affects translation and cellular responses in HD, we used striatal cells from mice expressing two copies of full-length Htt with either polyQ(7) or polyQ(111) tracts20. First, we confirmed that expression of mHtt is associated with lower levels of soluble eIF5A (Fig. 5a) and higher levels of 40S ribosomal protein ubiquitination (Fig. 5b), similar to HD mouse brains. Next, we treated lysates with RNase I, isolated RNase-resistant polysomes and analysed their content by LC–MS/MS (Supplementary Table 1). As expected, more RNase-resistant polysomes were recovered from cells expressing mHtt (Fig. 5c, black). Furthermore, polyQ(111) polysomes were depleted for eIF5A and enriched for the collided ribosome recognition complex Gcn1–Gcn20–Rbg33 (Fig. 5c, pink). We next performed ribosome profiling and pause score analysis. As reported51, we find mHtt increases the rate of ribosome stalling (Fig. 5d,e). These stalling events were indeed enriched for PPX amino acid triplet motifs within the ribosomal active site (Fig. 5e and Supplementary Table 1). PPX motifs are known to depend on eIF5A for efficient elongation39, confirming that eIF5A deficiency is linked to ribosome stalling in HD. We next examined the mRNAs affected by increased stalling in polyQ(111) cells. These disproportionately encode proteins involved in proteostasis, including ribosomal and proteasomal proteins, as well as the chaperonin TRiC/CCT and factors involved in myelination and glycolysis (Fig. 5f,g and Supplementary Table 1). Consistent with ribosome pausing leading to stalled NC aggregation52, the protein products of mRNAs encountering increased stalling in polyQ(111) cells were enriched in the insoluble brain proteome of aged R6/2 mice (Extended Data Fig. 5a). Stalling and aggregation were also accompanied by disruption of major proteostasis machines. Thus, the soluble brain proteome of 8- and 12-week-old R6/2 mice exhibits an early and persistent loss of 1:1 stoichiometry among core ribosomal proteins and proteasomal subunits (Extended Data Fig. 5bd). The stoichiometry loss cannot be explained by increased aggregation of specific proteins, because neither ribosomal nor proteasomal proteins were enriched in the insoluble brain proteome of 8-week-old HD mice (Extended Data Fig. 5e). These analyses indicate that the increase in aberrant translation pausing due to eIF5A depletion disproportionally affects mRNAs encoding key proteostasis complexes, thereby impacting their biogenesis. We propose that these events seed a vicious cycle of proteostasis dysfunction that contributes to HD symptoms. Supporting this notion, mutations affecting ribosomal and proteasomal stoichiometry were linked to neurodegeneration53. Taken together, we conclude that mHTT-ex1 protein interacts with and sequesters eIF5A, altering translation elongation dynamics on hundreds of mRNAs and disrupting proteostasis.

Fig. 5 |. Loss of eIF5A disrupts global translation elongation dynamics.

Fig. 5 |

a, Soluble eIF5A levels are lower in mouse striatal cells expressing mHtt. Immunoblots of cytoplasmic lysates of striatal cells expressing either polyQ(7) or polyQ(111) full-length Htt (n = 4). Hyp, hypusinated eIF5A. P, P value of a two-sided Student’s t-test. b, Rps3 is hyperubiquitinated in striatal cells expressing mHtt. Striatal cell lysates were fractionated on 10–50% sucrose gradients, and fractions were collected for immunoblots (n = 4). Top: absorbance profiles at 254 nm. Bottom: anti-Rps3 immunoblot. Ub, ubiquitin. c, mHtt increases ribosome collisions. Striatal cells were lysed, digested with RNase I and fractionated on 10–50% sucrose gradients. Proteomic analysis was performed on fractions of the gradient harbouring collided, RNase-resistant ribosomes (n = 3). Source numerical data are available in Supplementary Table 2. d, Ribo-seq analysis was performed on 80S monosomes from RNase-digested striatal cell lysates (n = 2). Shown is the distribution of pause scores for each genotype. P, two-sided Mann–Whitney P value. Centre lines show medians, box limits indicate upper and lower quartiles, and whiskers extend to 1.5 times the interquartile range. e, A pairwise comparison of individual pause sites. Inset: amino acid motif enriched in paused ribosomes. Source numerical data are available in Supplementary Table 3. In d and e, mHtt increases ribosome pausing on eIF5A-dependent sites. f, Fisher enrichment analysis of Gene Ontology (GO) functions enriched among pause-containing transcripts. Source numerical data are available in Supplementary Table 4. g, Ribo-seq traces of elongating ribosomes on select transcripts. The pink arrows indicate pause sites. Source numerical data and unprocessed blots are available in Source data.

Loss of eIF5A impedes recovery from acute stress

ER stress and activation of the unfolded protein response are common features of HD, and their modulation can affect neurotoxicity54. Indeed, we find that polyQ(111) striatal cells are hypersensitive to Tg-induced ER stress (Fig. 6a), as reported55. One important aspect of the stress response is phosphorylation of translation initiation factor eIF2a (EIF2S1), leading to ISR induction. eIF2a phosphorylation induces translation of uORF-repressed factors such as ATF4 and GADD34/PPP1R15A, which promote recovery from stress16. Because the yeast eIF5A plays a central role in translation of uORF-repressed stress effectors18,40, we hypothesized that depletion of eIF5A by mHTT may impair the translational induction of these protective factors, such as ATF4. Indeed, translation of Atf4 at steady state is lower in polyQ(111) cells, despite similar levels of Atf4 mRNA and higher basal phosphorylation of eIF2a (Extended Data Fig. 6a). This is associated with higher ribosome occupancy on the Atf4 uORF (Extended Data Fig. 6b), suggesting that translation of the main ORF is not induced despite eIF2a phosphorylation. Importantly, Atf4 accumulation in response to Tg was slower and weaker in polyQ(111) cells (Fig. 6b), potentially accounting for their increased susceptibility to stress (Fig. 6a).

Fig. 6 |. Loss of eIF5A disrupts recovery from acute stress.

Fig. 6 |

a,b, Striatal cells expressing mHtt fail to launch a protective stress recovery programme: striatal cells expressing either polyQ(7) or polyQ(111) Htt were treated with increasing concentrations of Tg, and viability was assessed at 24 h (n = 4), with data presented as mean ± s.d. (a); striatal cells were treated with 1 μM Tg for the indicated times and analysed by immunoblot (n = 4) (b). c, WT polyQ(7) striatal cells were treated with increasing concentrations of GC7, an eIF5A inhibitor, for 48 h. Before immunoblot analysis, 1 μM Tg was added for an additional 4 h (n = 3). d, Mutant polyQ(111) cells were transfected with plasmids encoding human eIF5A and its essential modifying enzyme, DHPS, followed by treatment with 1 μM Tg for the indicated times (n = 2). Source numerical data and unprocessed blots are available in Source data.

To further assess if eIF5A is required for ISR-mediated induction of protective Atf4 in striatal neurons, we next treated WT polyQ(7) cells with eIF5A inhibitor GC7, followed by stress induction by Tg. On its own, GC7 caused a slight elevation in Atf4 levels (Fig. 6c, left). Importantly, when combined with Tg treatment, GC7 dampened Atf4 production in a dose-dependent manner (Fig. 6c, right). Next, we tested whether supplementing eIF5A boosts the weak Atf4 response to Tg stress in polyQ(111) cells. We overexpressed eIF5A and its essential modifying enzyme deoxyhypusine synthase (Dhps) in polyQ(111) cells followed by treatment with Tg. The eIF5A supplementation increased Atf4 protein levels during ER stress in HD cells (Fig. 6d), albeit to a modest extent. We conclude that eIF5A activity is required for the upregulation of ISR-regulated protective factors in response to ER stress in striatal cells and that mHtt expression dampens this response. Supporting this conclusion, symptom onset in HD mice is associated with eIF5A depletion from the soluble proteome and with increased collision-related ubiquitination of ribosomal proteins (Fig. 4eg). Taken together, our data indicate that interventions to reduce the loss of eIF5A or otherwise increase its activity could be attractive therapeutic strategies in HD.

Reducing translation initiation mitigates ribotoxicity in HD

If mHTT sensitizes cells to ribosome collisions, HD cells should be more sensitive to drugs that increase collisions by slowing down elongation and less sensitive to drugs that decrease collisions by slowing down initiation. Indeed, polyQ(111) cells were hypersensitive to low doses of drugs that trigger collisions, for example, emetine (EME), anisomycin (ANS) and CHX56 (Fig. 7a). By contrast, polyQ(111) cells were less sensitive to low doses of translation initiation inhibitors Rocaglamide A (RocA) and 4EGI-1 (Fig. 7b). This is consistent with our earlier observation that 4EGI-1 reduced abortive translation termination in 293T cells transfected with mHTT-ex1 (Fig. 2e and Extended Data Fig. 2e), as well as the ameliorative effects of 4EGI-1 in R6/1 mice24. Finally, we speculated that the lower rate of translation initiation in polyQ(111) cells51 (Extended Data Fig. 6a) could reduce ribosome collisions and abortive termination. We transfected polyQ(7) and polyQ(111) cells with GFP–mCherry stalling reporters harbouring either K0 or K20 sequences and measured the relative fluorescence. As predicted, premature termination on the K20 motif—a hard staller—was lower in polyQ(111) cells (Fig. 7c). We propose that the protective effects of reducing translation initiation in HD are mediated by lower risk of collisions, which alleviate ribotoxic stress and premature termination.

Fig. 7 |. mHtt expression alters striatal cell sensitivity to ribotoxicy.

Fig. 7 |

a,b, mHtt increases sensitivity to translation elongation inhibitors that induce ribosome collisions (a) and decreases sensitivity to translation initiation inhibitors that suppress collisions (b). Mean ± s.d. (n = 3). c, mHtt expression in striatal cells is associated with increased readthrough of ribosome stalling K20 motif. Flow cytometry (FACS) analysis of polyQ(7) and polyQ(111) cells transfected with tandem fluorescent reporters of GFP and mCherry separated by the indicated test sequences. K0, no polylysines; K20, 20 consecutive lysines. Top: distribution of red/green ratios from a representative sample. Bottom: bar graphs of median red/green ratios. Mean ± s.d. (n = 3). d, Summary model. Source numerical data are available in Source data.

Discussion

Experimental therapies for HD have attempted to lower mHTT protein levels by antisense oligonucleotides or clear aggregates by antibodies, with limited success. Our study uncovers targets for lowering mHTT levels and countering its cellular toxicity. We find that CAG expansions cause abortive termination due to ribosome collisions, releasing neurotoxic mHTT-ex1 fragments. In young healthy cells, a combination of uORF-mediated attenuation of translation initiation and robust RQC and PQC probably limit or resolve collisions and clear mHTT fragments, preventing ribotoxic and proteotoxic stress. With age, cells become more susceptible to collisions and aggregation52, exacerbating the ribotoxic effects of mHTT. As mHTT protein fragments accumulate, they sequester eIF5A and other RQC factors. This, in turn, causes a decline in neuronal proteostasis, leading to deleterious effects on cellular fitness, including aberrant stress responses and additional proteostasis disruption. Eventually, ribosome collisions throughout the transcriptome disrupt biogenesis of key proteostasis machines and dampen induction of protective stress responses (Fig. 7d).

Our model may explain several observations from patients with HD, including the link between CAG repeat length, purity and age of onset10. Longer expansions can accommodate a larger number of rapidly elongating ribosomes, increasing the risk of collisions and accelerating ribotoxicity. Given the important role of localized protein synthesis in neurons57, it is tempting to speculate that such ribotoxicity may have an outsized effect in neurons as compared with other cell types. Our findings provide a mechanism for production of neurotoxic, aggregation-prone mHTT-ex1 fragments in patients with HD2 in addition to missplicing of mHTT intron 1 (ref. 58) and cleavage of mHTT protein by caspases59. Thus, ribosome collisions on the expanded polyQ can induce premature termination and release of truncated mHTT-ex1 fragments. Finally, the presence of polyalanine and polyserine protein products in patients with HD, thought to occur through repeat associated non-AUG (RAN) translation60, may also occur via frameshifting following stalling on mHTT-ex1. Since ribosome collisions induce frameshifting in bacteria61 and yeast62, it is possible that frameshifting on mHTT in mammals generates mixed polyQ, polyalanine and polyserine tracts, depending on the frame.

Our analyses indicate that, as disease progresses, ribotoxicity in HD extends beyond translation of mHTT itself. mHTT causes a progressive, age-dependent depletion of eIF5A in HD cells and mouse brains. Importantly, eIF5A depletion is recapitulated within hours of ectopic mHTT expression by our engineered viruses. In the absence of sufficient eIF5A, elongating ribosomes stall more frequently, leading to collisions that promote NC degradation or aggregation. This generates a constant load of ribotoxic stress that prevents efficient recovery from acute insults. We propose that mHTT-mediated ribotoxicity initiates a cascade of dysfunction that is augmented by eIF5A loss, leading to altered elongation kinetics throughout the translatome and an insufficient response to stress. Importantly, our findings suggest that pharmacological interventions that reduce ribosome collisions and/or enhance eIF5A function could be beneficial and delay symptom onset in HD. Finally, the nuanced and complex interplay between stress, translation initiation and translation elongation in the regulation of mHTT biogenesis uncovered here highlights the importance of improved mechanistic understanding when designing HD therapies.

Online content

Any methods, additional references, Nature Portfolio reporting summaries, source data, extended data, supplementary information, acknowledgements, peer review information; details of author contributions and competing interests; and statements of data and code availability are available at https://doi.org/10.1038/s41556-024-01414-x.

Methods

Cell cultures

Huh7 human hepatocellular carcinoma cells were a kind gift from Raul Andino. HEK293T human embryonic kidney cells (CRL-3216) and SH-SY5Y human neuroblastoma cells (CRL-2266) were from ATCC. STHdh Q7/Q7 (CH00097) and Q111/Q111 (CH00095) mouse striatal neurons were from Coriell Institute for Medical Research. Huh7 and SH-SY5Y cells were grown in Dulbecco’s modified Eagle medium (DMEM)/F-12 1:1 medium (Thermo Fisher) supplemented with 10% foetal bovine serum (FBS), 100 units ml−1 penicillin and 100 mg ml−1 streptomycin. HEK293T human embryonic kidney cells and STHdh Q7/Q7 and Q111/Q111 striatal neuron cells were grown in DMEM high-glucose medium (Thermo Fisher) supplemented as above. Cells were grown at 37 °C (Huh7, 293T and SH-SY5Y) or 32 °C (STHdh) in a 5% CO2 incubator.

Plasmids and oligonucleotides

Plasmids and oligonucleotide sequences are provided in Supplementary Table 1.

Analysis of published ribosome profiling data

gWIPS-viz63 and RPFdb64 webservers were used to search for relevant ribosome profiling datasets with coverage on Htt uORF and main coding sequence (CDS). Relevant mapped reads were downloaded from gWIPS-viz in BigWig format and visualized using Integrated Genome Viewer version 2.8.2.

Polysome profiles

A total of 1–5 × 107 striatal neurons or Huh7 cells were collected by scraping in ice-cold phosphate-buffered saline (PBS), centrifuged 1,000g for 5 min at 4 °C, resuspended in PBS and centrifuged again. Cell pellets were flash frozen in liquid nitrogen. Pellets were thawed on ice and resuspended in 200 μl polysome buffer (25 mM Tris–HCl pH 7.5, 25 mM KCl, 10 mM MgCl2, 2 mM dithiothreitol (DTT) and cOmplete EDTA-free protease inhibitor cocktail (Millipore Sigma)). Triton X-100 and sodium deoxycholate were added to a final concentration of 1% each, and the samples were incubated on ice for 20 min, passed ten times through a 26 G needle and centrifuged at 20,000g for 10 min at 4 °C to remove cell debris. For RNase-resistance assays, 100 U RNase I (Thermo Fisher) was added per 100 μg total RNA in clarified lysates. Digestion was performed for 45 min at room temperature and terminated by addition of 200 U Superase-In (Thermo Fisher). Lysates were loaded on 10–50% sucrose gradients in polysome buffer (or polysome buffer with 500 mM KCl, for RNase-treated samples) and subjected to ultracentrifugation at 36,000 rpm (average 160,030g) in an SW41.Ti swinging bucket rotor (Beckman Coulter) for 150 min at 4 °C. Equal volume fractions were collected using Gradient Station (BioComp) with continuous monitoring of ribosomal RNA (rRNA) at UV254.

qRT–PCR

To extract RNA from sucrose gradient fractions, 1 μl pellet paint (Millipore Sigma) and 500 μl phenol:chloroform:isoamyl alcohol (25:24:1) were added to each 500 μl fraction and incubated for 5 min at room temperature. After centrifugation at 12,000g for 15 min, 4 °C, the top phase was removed and subjected to another round of extraction as above. Four-hundred microlitres of the top phase was combined with 600 μl isopropanol and centrifuged at 12,000g for 30 min, 4 °C. The pellet was washed with 1 ml 75% ice-cold ethanol, air dried and resuspended in 20 μl RNase-free water. Complementary DNA was synthesized using the High Capacity cDNA Reverse Transcription Kit (Thermo Fisher), according to the manufacturer’s instructions, using 5 μl of RNA from each fraction. Quantitative real-time PCR (qRT–PCR) analysis was performed using SensiFast SYBR (BioLine) and gene-specific primers (Supplementary Table 1), according to the manufacturer’s instructions. To estimate relative abundance of specific mRNAs in each gradient fraction, each Ct value was divided by the sum of Ct values across all gradient fractions.

SDS–PAGE and immunoblotting

Cells were washed twice with ice-cold PBS and either lysed on plate with RIPA buffer (Thermo Fisher) or scraped in PBS, pelleted by centrifugation for 5 min at 1,000g, 4 °C, and resuspended in polysome buffer. Each lysis buffer was supplemented with fresh 2 mM DTT and cOmplete EDTA-free protease inhibitor cocktail. RIPA buffer was also supplemented with 50 units ml−1 benzonase (Millipore Sigma) to remove DNA. For ubiquitination analysis of ribosomal proteins, 100 μM PR-619 and 5 mM N-ethylmaleimide (NEM, Selleck Chemicals) were included in the lysis buffer. For detection of phosphorylated proteins, PhosSTOP phosphatase inhibitor cocktail (Roche) was added at a ratio of one tablet per 10 ml buffer. Lysis was performed on ice for 20 min, and lysates were clarified by centrifugation for 10 min at 12,000g, 4 °C. Then, 4× Laemmli sample buffer (Bio-Rad) supplemented with fresh 10% 2-mercaptoethanol was added to a final concentration of 1×. Afterwards, 15–30 μg of each sample was resolved on 4–20% (Bio-Rad) or 12% (homemade) SDS–PAGE, and transferred to 0.2 or 0.45 μm polyvinylidene fluoride membranes pre-soaked in methanol for 30 s. Membranes were blocked with 4% bovine serum albumin (BSA; Millipore Sigma) in Tris-buffered saline supplemented with 0.1% Tween-20 (Millipore Sigma) (TBST) for 1 h at room temperature, and then probed with specific primary antibodies for 2 h at room temperature. Primary antibodies were diluted in 4% BSA/TBST supplemented with 0.02% sodium azide as follows: rabbit anti-HTT D7F7 (Cell Signaling 5656S, 1:1,000, used to detect full-length HTT), mouse anti-HTT 2B4 (Millipore Sigma MAB5492, 1:1,000, used to detect HTT-ex1), mouse anti-PolyQ MW1 (Developmental Studies Hybridoma Bank, 1:1,000), mouse anti-puromycin (Millipore Sigma MABE343, 1:2,500), mouse anti-eIF2a (Abcam ab5369, 1:1,000), rabbit anti-p-eIF2a S51 (Cell Signaling 9721, 1:1,000), rabbit anti-RPS2 (Bethyl Laboratories A303–794, 1:1,000), rabbit anti-RPS3 (Cell Signaling 9538, 1:1,000), rabbit anti-Atf4 (Cell Signaling 11815, 1:1,000), goat anti-Poliovirus VP1 (Thermo Fisher PA1–73124, 1:2,500), mouse anti-Poliovirus 3CD (homemade hybdridoma, 1:1,000), mouse anti-eIF5A (BD Sciences 611977, 1:1,000), rabbit anti-hypusine (Millipore Sigma ABS1064, 1:1,000) and rabbit anti-RPL13a (Cell Signaling 2765, 1:2,500). Secondary antibodies were diluted 1:10,000 in TBST. Western blot detection was done using ECL Plus Western Blotting Substrate (Thermo Fisher), and images were taken either by film radiography or Bio-Rad GelDoc imager. Alternatively, fluorescent western blot detection was performed using Li-Cor Odyssey Infrared imager. Densitometry analysis was performed using ImageJ version 1.52a.

Immunofluorescence analysis of HTT aggregation

Htt-ex1 Q25 or Q97 (ref. 65) was amplified by PCR and ligated into pcDNA3–GFP using NheI and XhoI. The endogenous 5′ UTR of human Htt (NM_001388492.1), either with a functional uORF (WT, ATG) or non-functional uORF (single nucleotide mutation, AAG), was synthesized by GenScript with flanking BmtI and HindIII sites and ligated into pcDNA3-mClover. For live cell imaging, 3 × 105 HEK293T cells were seeded in six-well plates. At 24 h, 2 μg DNA was transfected using Lipofectamine 2000 (Thermo Fisher), according to the manufacturer’s instructions. Transfected cells were monitored once every hour starting from 12 h post-transfection, using a Zeiss Axio Vert.A1 inverted microscope. For inclusion body counting, fixed cells were used. A total of 0.8 × 105 HEK293T cells were seeded on poly-L-ornithine (Millipore Sigma) pre-coated 12-mm coverslips. At 24 h, 400 ng DNA was transfected using Lipofecatmine 2000 (Thermo Fisher), according to the manufacturer’s instructions. Cells were fixed with 4% paraformaldehyde for 15 min at room temperature and washed twice with PBS for 5 min each. Cells were permeabilized with 0.1% Triton X-100 for 10 min at room temperature and washed twice with PBS. To label plasma membrane, coverslips were incubated with HCS CellMask Deep Red stain (Thermo Fisher) for 5 min at room temperature and washed twice with PBS. Coverslips were mounted with ProLong Glass Antifade Mountant with NucBlue Stain (Thermo Fisher), dried overnight and stored at −20 °C until image acquisition using a Zeiss LSM 700 laser scanning confocal microscope.

Generation of RIPA-soluble and RIPA-insoluble fractions

A total of 5 × 105 HEK293T cells were plated on poly-D-lysine-coated six-well plates. The following day, cells were transfected with 0.5–2 μg of the above plasmids using Lipofectamine 2000 (Thermo Fisher), according to the manufacturer’s instructions. At 8, 24 or 48 h post-transfection, cells were washed twice with ice-cold PBS and scraped into the same buffer. Cells were pelleted by centrifugation at 300g for 5 min at 4 °C and washed again with the same buffer. Pellets were resuspended in 50 μl RIPA lysis buffer (Thermo Fisher) and triturated through a 25 G syringe for ten times. Lysates were clarified by centrifugation at 1,500g for 10 min at 4 °C. RIPA-insoluble fractions were washed with RIPA buffer twice, followed by centrifugation at 1,500g for 10 min at 4 °C. Pellets were incubated in 20 μl formic acid at 56 °C for 1 h, with mixing at 1,000 rpm, and then dried overnight using a SpeedVac. Dried pellets were resuspended in 8 M urea, 150 mM NaCl and 20 mM Tris pH 8.0 and subjected to immunoblot analysis.

NC puromycylation

A total of 2.5 × 105 STHdh, Huh7 or SH-SY5Y cells in six-well plates were incubated with 1 μM puromycin (Thermo Fisher) for 15 min at 32 °C (STHdh) or 37 °C (Huh7 and SH-SY5Y), washed twice with PBS and lysed on-plate as described above. For sucrose gradients, each fraction was incubated with 1 μM puromycin for 15 min at 37 °C, followed by incubation with 7 μl StrataClean beads (Agilent) for 16 h at 4 °C with constant tumbling. Beads were pelleted by centrifugation at 1,000g for 5 min at 4 °C and resuspended in 30 μl 2× Laemmli buffer. Puromycylated samples were subjected to immunoblotting using an anti-puromycin antibody (Millipore Sigma).

Abortive termination reporters

The dual-fluorescence abortive termination reporters were a generous gift from Eric Bennett. pmGFP-P2A-K0-P2A-RFP was linearized by EcoNI and KpnI, and PCR-amplified human HTT-ex1 Q25 or Q97 was cloned in between GFP and mCherry, separated by self-cleaving P2A sites, using In-Fusion kit (Takara Bio). A total of 1 × 106 STHdh cells were plated on six-well plates. The following day, cells were transfected in triplicates with 2 μg dual-fluorescence plasmids using Lipofectamine 2000 (Thermo Fisher), according to the manufacturer’s instructions. At 24 h post-transfection, cells were washed twice with ice-cold PBS, trypsinized and washed with 1% BSA in ice-cold PBS. Cells were stained with LIVE/DEAD Fixable Violet Dead Cell Stain Kit (Thermo), diluted 1:1,000 in PBS, for 30 min on ice in the dark. Cells were washed twice with 1% BSA in ice-cold PBS and filtered using a 35-μm strainer mesh before flow cytometry analysis. A total of 50,000 cells per sample were analysed using a BD LSR II Flow Cytometer. The medium of GFP and mCherry intensity and mCherry-to-GFP ratio were calculated from the live singlet population. Flow cytometry data analysis was performed using FlowJo Ver. 9.

GFP co-immunoprecipitation

A total of 3 × 106 WT STHdh cells were plated in 10-cm plates. The following day, cells were transfected with 12 μg pcDNA3-Htt-ex1-GFP variants using Lipofectamine 2000 (Thermo Fisher), according to the manufacturer’s instructions. Sixteen hours post-transfection, cells were washed twice with ice-cold PBS, scraped and pelleted at 300g for 5 min at 4 °C. Pellets were lysed with immunoprecipitation (IP) lysis buffer (25 mM Tris–HCl pH 7.5, 50 mM NaCl, 10 mM MgCl2, 5% glycerol, 1% NP40, 1 mM DTT and complete protease inhibitor) for 10 min on ice, and then passed 10× through 25 G syringes. Lysates were clarified by centrifugation at 1,500g for 10 min at 4 °C. Three-hundred micrograms of total protein and 10 μl ChromoTek GFP-Trap magnetic agarose beads (Proteintech) were used per sample. Beads were washed three times with ice-cold IP lysis buffer. Lysates were added and incubated at 4 °C overnight, with shaking. Beads were washed 3× with ice-cold IP wash buffer (50 mM Tris–HCl pH 7.5, 150 mM NaCl, 10 mM MgCl2, 5% glycerol and 0.05% NP40). After the final wash, beads were resuspended in 1× Laemmli sample buffer and subjected to immunoblotting.

Ribosome profiling analysis

Ribosome footprints were prepared essentially as described66. Briefly, adherent cells were washed and collected in ice-cold PBS by scraping. Cells were pelleted by centrifugation at 1,000g for 5 min at 4 °C and pellets were resuspended in lysis buffer (50 mM HEPES–KOH pH 7.5, 140 mM KCl, 5 mM MgCl2 and 0.1% NP40) supplemented with 0.5 mM DTT and cOmplete EDTA-free protease inhibitor cocktail. Lysates were passed 10× through a 25 G needle and clarified by centrifugation at 20,000g for 10 min at 4 °C. Then, 25 U RNase I were added per 100 μg total RNA, and digestion was performed for 45 min at room temperature, and then terminated with 200 U Superase-In (Thermo Fisher). RNase-treated samples were fractionated on 10–50% sucrose gradients, and 80S monosome fractions were collected. RNA was extracted from each fraction using phenol:chloroform:isoamyl alcohol, as described above, and size selected for 16–34 nt on an 8% urea–PAGE. rRNA depletion was performed using Ribo-Zero rRNA Removal Kit (Illumina), according to the manufacturer’s instructions. All downstream steps were performed as described66. Libraries from two independent repeats were sequenced on a HiSeq 4000 (Illumina). After demultiplexing, sequencing reads were trimmed of adapter sequences and quality filtered using cutadapt (-a CTGTAGGCACCATCAAT -m1 -q20). rRNA was removed using Bowtie2 (--un). Remaining reads were aligned to Mus musculus genome assembly GRCm38 (mm10) using Hisat2 (--trim5 1). Read count matrices were generated using featureCounts and BigWig files were generated using bamCoverage and visualized on Integrated Genome Viewer. For ribosome stall site analysis, we used the standalone version of PausePred67 with the following parameters: window_size = 1,000, foldchange for pause = 20, read_lengths = 28,29,30, window coverage% = 10, upstream_seq and downstream_seq = 25, and offset = 12,12,12.

Generation and use of HTT-expressing virus

HTT-ex-1 Q8 or Q73 were amplified by PCR from gWizQ8 or gWizQ73 (kind gift from Donald Lo68) using primers with flanking XhoI and EcoRI sites, and cloned into prib(+)XpA-GFP69, encoding type 1 poliovirus Mahoney, by replacing the GFP.

Ten micrograms of plasmids were linearized with 50 U MluI for 2 h at 37 °C, followed by phenol:chloroform:isoamyl alcohol (25:24:1 v/v) extraction and in vitro transcription using MEGAscript T7 (Thermo Fisher), according to the manufacturer’s instructions. Ten micrograms of viral RNA was electroporated into 4 × 106 Huh7 cells in 0.4 ml PBS, 4-mm electroporation cuvette, at 270 V/960 μF (GenePulser, Bio-Rad). Cells were grown for 24 h at 37 °C, followed by 3× freeze–thaw cycles, and the medium was centrifuged at 2,500g for 5 min, 4 °C, to remove cell debris. Clarified virus-containing media (passage 0, P0) were aliquoted and frozen at −80 °C. P0 viruses were amplified once in Huh7 cells to generate P1 virus stock, which was processed and frozen as above. P1 stock was used for all experiments.

To determine virus titres, 1 × 105 Vero cells were seeded in six-well plates. The following day, tenfold serial dilutions of virus stocks/samples were prepared in media without FBS. Cells were washed with PBS, and 0.5 ml inoculum was added for 45 min at 37 °C. Two millilitres of overlay solution (1% agarose combined 1:1 v/v with 2× DMEM supplemented with 2% FBS) was added to each well and allowed to gel at room temperature. Two days later, 2 ml 2% formaldehyde was added for 1 h at room temperature. Cells were stained with 1 ml 0.1% crystal violet in 20% ethanol for 1 h at room temperature. For infections, Huh7 or SH-SY5Y cells were washed once with PBS and incubated with FBS-free DMEM supplemented with virus at a multiplicity of infection of 10. After 45 min at 37 °C, inoculum was replaced with fresh DMEM supplemented with 10% FBS.

Sample preparation for proteomic analysis

Proteins were extracted from isolated polysomes using methanol–chloroform precipitation. Four-hundred microlitres of methanol, 100 μl chloroform and 350 μl water were added sequentially to each 100 μl sample, followed by centrifugation at 14,000g for 5 min at room temperature. The top phase was removed and the protein interphase was precipitated by addition of 400 μl methanol, followed by centrifugation at 14,000g for 5 min at room temperature. Pellets were air dried and resuspended in 8 M urea and 25 mM ammonium bicarbonate (pH 7.5). Two to four micrograms of protein were subjected to reduction and alkylation with 10 mM DTT for 1 h at room temperature followed by 5 mM iodoacetamide for 45 min at room temperature, in the dark. The samples were then incubated with 1:50 enzyme-to-protein ratio of sequencing-grade trypsin (Promega) overnight at 37 °C. Peptides were desalted with μC18 Ziptips (Millipore Sigma), dried and resuspended in 10 μl 0.1% formic acid in water.

LC–MS/MS acquisition and data processing

Proteomic data were collected using Xcalibur 4.2 SP1 on LTQ-Elite (virus polysomes) and QE (RNase-digested polysomes) mass spectrometers (Thermo Fisher) coupled online to a nanoAcquity UPLC system (Waters Corporation) through an EASY-Spray nanoESI ion source (Thermo Fisher). Peptides were loaded onto an EASY-Spray column (75 μm × 15 cm column packed with 3 μm, 100 Å PepMap C18 resin) at 2% B (0.1% formic acid in acetonitrile) for 20 min at a flow rate of 600 nl min−1. Peptides were separated at 400 nl min−1 using a gradient from 2% to 25% B over 48 min (QE) or 220 min (LTQ) followed by a second gradient from 25% to 37% B over 8 min and then a column wash at 75% B and reequilibration at 2% B. Precursor scans were acquired in the Orbitrap analyser (QE: 350–1,500 m/z, resolution 70,000@200 m/z, automatic gain control (AGC) target 3 × 106; LTQ: 300–1,800 m/z, resolution 60,000@400 m/z, AGC target 2 × 106. The top ten or six (QE or LTQ) most intense, doubly charged or higher ions were isolated (4 m/z isolation window) and subjected to high-energy collisional dissociation (QE: 25 normalized collision energy (NCE); LTQ: 27.5 NCE), and the product ions were measured in the Orbitrap analyser (QE resolution: 17,500@200 m/z, AGC target 5 × 104; LTQ resolution: 15,000@400 m/z, AGC target 9 × 104).

Raw mass spectrometry (MS) data were processed using MaxQuant version 1.6.7.0 (ref. 70). MS/MS spectrum searches were performed using the Andromeda search engine71 against the forward and reverse human and mouse Uniprot databases (downloaded 28 August 2017 and 25 November 2020, respectively). Cysteine carbamidomethylation was chosen as fixed modification and methionine oxidation and N-terminal acetylation as variable modifications. Parent peptides and fragment ions were searched with maximal mass deviation of 6 ppm and 20 ppm, respectively. Mass recalibration was performed with a window of 20 ppm. Maximum allowed false discovery rate (FDR) was <0.01 at both the peptide and protein levels, based on a standard target–decoy database approach. The ‘calculate peak properties’ and ‘match between runs’ options were enabled.

All statistical tests were performed with Perseus version 1.6.7.0 using either ProteinGroups or Peptides output tables from MaxQuant. Potential contaminants, proteins identified in the reverse dataset and proteins identified only by site were filtered out. Intensity-based absolute quantification (iBAQ) was used to estimate absolute protein abundance. Two-sided Student’s t-test with a permutation-based FDR of 0.01 and S0 of 0.1 with 250 randomizations was used to determine statistically significant differences between grouped replicates. Categorical annotation was based on Gene Ontology Biological Process (GOBP), Molecular Function (GOMF) and Cellular Component (GOCC), as well as protein complex assembly by CORUM.

Cell viability

A total of 1 × 104 STHdh cells were seeded per well in a black-wall, clear-bottom 96-well plate. At 24 h, Tg (AG Scientific) was added for another 24 h. Cell viability was assayed using CellTiter-Glo (Promega), according to the manufacturer’s instructions. Luminescence was recorded using a BMG Labtech CLARIOstar Microplate Reader.

Statistics and reproducibility

No statistical methods were used to pre-determine sample sizes. Sample sizes (n) reflect independent replicates, except when otherwise indicated in the figure legends. For all proteomic analyses of R6/2 mouse brains (Hosp et al., related to Fig. 4e,f and Extended Data Figs. 4ac and 5a,ce), data were acquired from four, three and three WT mice and four, three and four R6/2 mice at weeks 5, 8 and 12, respectively. Data were acquired separately from four different brain regions of each animal at each age. Separate areas of the brain are plotted as independent datapoints. No data were excluded from the analyses, and the experiments were not randomized. The investigators were not blinded to allocation during experiments and outcome assessment.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

Extended Data

Extended Data Fig. 1 |. HTT translation initiation is dynamically regulated by its uORF.

Extended Data Fig. 1 |

(a) Ribo-seq traces of initiating ribosomes (A-sites, top) and elongating ribosomes (footprints, bottom) on human HTT uORF and start of main ORF. (b) Top, ribo-seq traces of elongating ribosomes on Htt uORF and start of main ORF in different organisms and tissues. Bottom, Htt protein level (low, moderate, high) in the indicated tissues, from ProteinAtlas. (c) Ribo-seq traces of elongating ribosomes on Htt and Atf4 mRNA in mouse embryonic fibroblasts before and after thapsigargin (Tg) treatment (left, n = 2) and SH-SY5Y neuroblasts before and after differentiation with retinoic acid (right, n = 2).

Extended Data Fig. 2 |. HTT uORF reduces truncation and aggregation of mHTT.

Extended Data Fig. 2 |

(a) 293 T cells were transfected with constructs expressing wild-type polyQ(25) or mutant polyQ(97) HTT-ex1 with a C-terminal fluorescent GFP. Shown are live cell images at the indicated times post-transfection. Images were obtained using 20x magnification. Representative images of 2 independent replicates. (b-c) Immunoblot analyses of 293 T (b) and mouse striatal cells (c) transfected as above and separated into soluble and insoluble fractions (n = 2). WB, western blot. (d) Flow cytometry analysis of transfected 293 T cells. (e) Effect of 4EGI-1 on global translation. 293 T cells were treated with increasing concentrations of 4EGI-1 for 24 h, followed by puromycin for 15 min (n = 2). Immunoblot analysis was performed on whole cell lysates using a puromycin-specific antibody. (f) 293 T cells were transfected with mHTT-ex1-GFP and increasing concentrations of 4EGI-1 were added 6 h post-transfection. Immunoblot analysis of the insoluble fraction was performed at 24 h post-transfection (n = 3). Means +/− s.d. P, p-value of a two-tailed Student t-test comparing 50 and 0 μM 4EGI-1. Unprocessed blots are available in source data.

Extended Data Fig. 3 |. Translation of mHTT from poliovirus genome is associated with ribosome collisions.

Extended Data Fig. 3 |

(a) Poliovirus replicates more slowly when engineered to express mutant versus wild-type HTT-ex1. Wild-type polyQ(8) and mutant polyQ(73) human HTT-ex1 were cloned into poliovirus genome. Infectious viruses were generated by transfection of in vitro transcribed RNA. Plaque assays were performed to determine virus titers (n = 3). Means +/− s.d. (left) and representative images of viral plaques (right). (b-c) Huh7 cells (b) and SH-SY5Y neuroblasts (c) were infected with engineered viruses. At the indicated times, puromycin was added to tissue culture media to label nascent polypeptide chains and lysates were analyzed by immunoblotting. Shown is a representative of 2 independent repeats. Purple boxes and arrowheads indicate virus-Htt polyproteins prior to cleavage by the viral protease. WB, western blot. (d) Quantification of blots from Fig. 3c (n = 3). Means +/− s.d. (e) Huh7 cells were infected with HTT-ex1-expressing viruses for 3.5 h, and ribosomes were isolated by ultracentrifugation on a sucrose cushion, followed by immunoblot analysis using antibodies specific to viral epitopes upstream and downstream of HTT-ex1 insert (n = 2). Unprocessed blots are available in source data.

Extended Data Fig. 4 |. HD mouse brains show age-dependent changes in levels of soluble translation and RQC factors.

Extended Data Fig. 4 |

(a) eIF5A, but not other elongation factors, is inversely correlated with mHTT levels in mouse brains at 12 weeks of age. Pearson’s correlation coefficients for wild-type or mutant Htt and the indicated translation elongation factors. Reanalysis of published MS analysis of soluble brain proteomes from 12-week-old R6/2 and control mice (n = 16/12). (b-c) Levels of RQC factors increase, while those of elongation factors decrease, in the soluble brain proteome of presymptomatic R6/2 HD mice. Shown are levels of the indicated proteins in wild-type and R6/2 HD cells at 5, 8 and 12 weeks of age (b) and their Student’s t-test differences between wild-type and R6/2 at 8 weeks. n = 16, 12 and 12 for WT or n = 16, 12 and 16 for R6/2 at weeks 5, 8 and 12, respectively. Means +/−s.d. (c). Histogram shows the distribution of t-test differences of the entire soluble proteome at 8 weeks.

Extended Data Fig. 5 |. Disruption of proteostasis machines in R6/2 mice.

Extended Data Fig. 5 |

(a) Proteins encoded by pause-containing transcripts detected in striatal cells expressing polyQ(111) mHtt are enriched in the insoluble brain proteome of aged 12-week-old R6/2 mice (n = 12/16 for WT and R6/2, respectively). (b) Kurtosis is a measure of tailedness or outliers relative to a normal distribution, and can be used to estimate stoichiometry of large molecular complexes. (c-d) Stoichiometry of ribosomal proteins (c) and proteasomal subunits (d) is disrupted in R6/2 brains starting at 8 weeks of age. Shown are kurtosis scores for all core ribosomal proteins or proteasomal subunits as measured by MS. n = 16, 12 and 12 for WT or n = 16, 12 and 16 for R6/2 at weeks 5, 8 and 12, respectively. (e) Ribosomal proteins are enriched in the insoluble brain proteome of symptomatic 12-week-old but not asymptomatic 8-week-old R6/2 mice. n = 16, 12 and 12 for WT or n = 16, 12 and 16 for R6/2 at weeks 5, 8 and 12, respectively. Proteasomal subunits were not detected in the insoluble brain proteome of either wild-type or R6/2 mice at any age. For a, c-e, center lines show medians; box limits—upper and lower quartiles; and whiskers extend 1.5 times the interquartile range.

Extended Data Fig. 6 |. Global and Atf4-specific translation levels in mouse striatal cells.

Extended Data Fig. 6 |

(a) Global translation is lower in striatal cells expressing polyQ(111) mHtt. Translation rates were monitored by puromycin labeling and quantified by densitometry (left, n = 3, Means +/−s.d.) and polysome profiles on 10–50% sucrose gradients (right, n = 2, representative traces). WB, western blot. (b) Translation efficiency of Atf4 is lower in polyQ(111) cells at steady-state. Means of values from RNA- and ribo-seq analyses (n = 2 each). (c) Ribo-seq traces of elongating ribosomes on Atf4 mRNA (n = 2). Unprocessed blots are available in source data.

Supplementary Material

Aviner Supplementary

The online version contains supplementary material available at https://doi.org/10.1038/s41556-024-01414-x.

Acknowledgements

We thank I. Munoz-Sanjuan and D. Marchionini, as well as members of the Frydman lab, for their helpful comments. This work was supported by NIH grants GM05643321, NS092525 to J.F. and AI36178, AI40085 and AI091575 to R. Andino and Cure Huntington’s Disease Initiative (CHDI) grant A-13887 to J.F.

Footnotes

Competing interests

The authors declare no competing interests.

Additional information

Extended data is available for this paper at https://doi.org/10.1038/s41556-024-01414-x.

Data availability

Previously published ribosome profiling analyses were from GSE87328 (Fig. 1c), GSE72064 (Fig. 1d,e), SRA160745, GSE74279 and GSE94460 (Extended Data Fig. 1a), GSE66715, GSE81283 and GSE36892 (Extended Data Fig. 1b) and GSE53743 and GSE119615 (Extended Data Fig. 1c). For analysis of previously published proteomic data, processed MS intensity tables were downloaded from the supplementary information section of the following published articles: Hosp et al.6, Supplementary Tables 1 and 3 (label-free quantification, LFQ); Sap et al.50, Supplementary Tables 1 and 3 (LFQ); Newcombe et al.43, Supplementary Table 2 (LFQ); and Sui et al.45, Supplementary Table 1 (raw intensities). No additional normalization of the data was performed. Statistical tests were done using two-sided Student’s t-test. Sequencing data were deposited in the Sequence Read Archive (SRA) database under BioProject number PRJNA730032. The MS proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE72 partner repository with the dataset identifier PXD026012. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  • 1.Bates GP et al. Huntington disease. Nat. Rev. Dis. Prim 1, 15005 (2015). [DOI] [PubMed] [Google Scholar]
  • 2.Finkbeiner S. Huntington’s disease. Cold Spring Harb. Perspect. Biol 3, a007476 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Paulson HL, Shakkottai VG, Clark HB & Orr HT Polyglutamine spinocerebellar ataxias—from genes to potential treatments. Nat. Rev. Neurosci 18, 613–626 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Jimenez-Sanchez M, Licitra F, Underwood BR & Rubinsztein DC Huntington’s disease: mechanisms of pathogenesis and therapeutic strategies. Cold Spring Harb. Perspect. Med 7, a024240 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Olzscha H. et al. Amyloid-like aggregates sequester numerous metastable proteins with essential cellular functions. Cell 144, 67–78 (2011). [DOI] [PubMed] [Google Scholar]
  • 6.Hosp F. et al. Spatiotemporal proteomic profiling of Huntington’s disease inclusions reveals widespread loss of protein function. Cell Rep. 21, 2291–2303 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Kim YE et al. Soluble oligomers of polyQ-expanded Huntingtin target a multiplicity of key cellular factors. Mol. Cell 63, 951–964 (2016). [DOI] [PubMed] [Google Scholar]
  • 8.Arrasate M, Mitra S, Schweitzer ES, Segal MR & Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 431, 805–810 (2004). [DOI] [PubMed] [Google Scholar]
  • 9.Nalavade R, Griesche N, Ryan DP, Hildebrand S. & Krauß S. Mechanisms of RNA-induced toxicity in CAG repeat disorders. Cell Death Dis. 4, e752 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lee J-M et al. CAG repeat not polyglutamine length determines timing of Huntington’s disease onset. Cell 178, 887–900.e14 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lee J. et al. An upstream open reading frame impedes translation of the huntingtin gene. Nucleic Acids Res. 30, 5110–5119 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kozak M. The scanning model for translation: an update. J. Cell Biol 108, 229–241 (1989). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zhang H, Wang Y. & Lu J. Function and evolution of upstream ORFs in eukaryotes. Trends Biochem. Sci 44, 782–794 (2019). [DOI] [PubMed] [Google Scholar]
  • 14.Fijałkowska D. et al. eIF1 modulates the recognition of suboptimal translation initiation sites and steers gene expression via uORFs. Nucleic Acids Res. 45, 7997–8013 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cho J. et al. Multiple repressive mechanisms in the hippocampus during memory formation. Science 350, 82–87 (2015). [DOI] [PubMed] [Google Scholar]
  • 16.Costa-Mattioli M. & Walter P. The integrated stress response: from mechanism to disease. Science 368, eaat5314 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rodriguez CM, Chun SY, Mills RE & Todd PK Translation of upstream open reading frames in a model of neuronal differentiation. BMC Genomics 20, 391 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Young SK & Wek RC Upstream open reading frames differentially regulate gene-specific translation in the integrated stress response. J. Biol. Chem 291, 16927–16935 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Reid DW et al. Complementary roles of GADD34- and CReP-containing eukaryotic initiation factor 2α phosphatases during the unfolded protein response. Mol. Cell. Biol 36, 1868–1880 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Trettel F. Dominant phenotypes produced by the HD mutation in STHdhQ111 striatal cells. Hum. Mol. Genet 9, 2799–2809 (2000). [DOI] [PubMed] [Google Scholar]
  • 21.Juszkiewicz S. & Hegde RS Initiation of quality control during poly(A) translation requires site-specific ribosome ubiquitination. Mol. Cell 65, 743–750.e4 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Juszkiewicz S. et al. Ribosome collisions trigger cis-acting feedback inhibition of translation initiation. eLife 9, e60038 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ferrin MA & Subramaniam AR Kinetic modeling predicts a stimulatory role for ribosome collisions at elongation stall sites in bacteria. eLife 6, e23629 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Creus-Muncunill J. et al. Increased translation as a novel pathogenic mechanism in Huntington’s disease. Brain 142, 3158–3175 (2019). [DOI] [PubMed] [Google Scholar]
  • 25.Yip MCJ & Shao S. Detecting and rescuing stalled ribosomes. Trends Biochem. Sci 46, 731–743 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zhao T. et al. Disome-seq reveals widespread ribosome collisions that promote cotranslational protein folding. Genome Biol. 22, 16 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Doerfel LK et al. EF-P is essential for rapid synthesis of proteins containing consecutive proline residues. Science 339, 85–88 (2013). [DOI] [PubMed] [Google Scholar]
  • 28.Huter P. et al. Structural basis for polyproline-mediated ribosome stalling and rescue by the translation elongation factor EF-P. Mol. Cell 68, 515–527.e6 (2017). [DOI] [PubMed] [Google Scholar]
  • 29.Gingras AC, Svitkin Y, Belsham GJ, Pause A. & Sonenberg N. Activation of the translational suppressor 4E-BP1 following infection with encephalomyocarditis virus and poliovirus. Proc. Natl Acad. Sci. USA 93, 5578–5583 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schmidt EK, Clavarino G, Ceppi M. & Pierre P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat. Methods 6, 275–277 (2009). [DOI] [PubMed] [Google Scholar]
  • 31.Arpat AB et al. Transcriptome-wide sites of collided ribosomes reveal principles of translational pausing. Genome Res. 30, 985–999 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sinha NK et al. EDF1 coordinates cellular responses to ribosome collisions. eLife 9, e58828 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Pochopien AA et al. Structure of Gcn1 bound to stalled and colliding 80S ribosomes. Proc. Natl Acad. Sci. USA 118, e2022756118 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Koyuncu S. et al. The ubiquitin ligase UBR5 suppresses proteostasis collapse in pluripotent stem cells from Huntington’s disease patients. Nat. Commun 9, 2886 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ghosh DK, Roy A. & Ranjan A. The ATPase VCP/p97 functions as a disaggregase against toxic Huntingtin-exon1 aggregates. FEBS Lett. 592, 2680–2692 (2018). [DOI] [PubMed] [Google Scholar]
  • 36.Zhou M. et al. HUWE1 amplifies ubiquitin modifications to broadly stimulate clearance of proteins and aggregates. Preprint at bioRxiv 10.1101/2023.05.30.542866 (2023). [DOI] [Google Scholar]
  • 37.Huang Z-N & Her L-S The ubiquitin receptor ADRM1 modulates HAP40-induced proteasome activity. Mol. Neurobiol 54, 7382–7400 (2017). [DOI] [PubMed] [Google Scholar]
  • 38.Wyttenbach A. Heat shock protein 27 prevents cellular polyglutamine toxicity and suppresses the increase of reactive oxygen species caused by huntingtin. Hum. Mol. Genet 11, 1137–1151 (2002). [DOI] [PubMed] [Google Scholar]
  • 39.Pelechano V. & Alepuz P. eIF5A facilitates translation termination globally and promotes the elongation of many non-polyproline-specific tripeptide sequences. Nucleic Acids Res.45, 7326–7338 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Manjunath H. et al. Suppression of ribosomal pausing by eIF5A is necessary to maintain the fidelity of start codon selection. Cell Rep. 29, 3134–3146.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Faundes V. et al. Impaired eIF5A function causes a Mendelian disorder that is partially rescued in model systems by spermidine. Nat. Commun 12, 833 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Smeltzer S. et al. Hypusination of Eif5a regulates cytoplasmic TDP-43 aggregation and accumulation in a stress-induced cellular model. Biochim. Biophys. Acta Mol. Basis Dis 1867, 165939 (2021). [DOI] [PubMed] [Google Scholar]
  • 43.Newcombe EA et al. Tadpole-like conformations of Huntingtin exon 1 are characterized by conformational heterogeneity that persists regardless of polyglutamine length. J. Mol. Biol 430, 1442–1458 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gruber A. et al. Molecular and structural architecture of polyQ aggregates in yeast. Proc. Natl Acad. Sci. USA 115, E3446–E3453 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sui X. et al. Widespread remodeling of proteome solubility in response to different protein homeostasis stresses. Proc. Natl Acad. Sci. USA 117, 2422–2431 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Schuller AP, Wu CC-C, Dever TE, Buskirk AR & Green R. eIF5A functions globally in translation elongation and termination. Mol. Cell 66, 194–205.e5 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Han P. et al. Genome-wide survey of ribosome collision. Cell Rep. 31, 107610 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Higgins R. et al. The unfolded protein response triggers site-specific regulatory ubiquitylation of 40S ribosomal proteins. Mol. Cell 59, 35–49 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Garshott DM, Sundaramoorthy E, Leonard M. & Bennett EJ Distinct regulatory ribosomal ubiquitylation events are reversible and hierarchically organized. eLife 9, e54023 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Sap KA et al. Global proteome and ubiquitinome changes in the soluble and insoluble fractions of Q175 Huntington mice brains. Mol. Cell. Proteom 18, 1705–1720 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Eshraghi M. et al. Mutant Huntingtin stalls ribosomes and represses protein synthesis in a cellular model of Huntington disease. Nat. Commun 12, 1461 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Stein KC, Morales-Polanco F, van der Lienden J, Rainbolt TK & Frydman J. Ageing exacerbates ribosome pausing to disrupt cotranslational proteostasis. Nature 601, 637–642 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zavodszky E, Peak-Chew S-Y, Juszkiewicz S, Narvaez AJ & Hegde RS Identification of a quality-control factor that monitors failures during proteasome assembly. Science 373, 998–1004 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shacham T, Sharma N. & Lederkremer GZ Protein misfolding and ER stress in Huntington’s disease. Front. Mol. Biosci 6, 20 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Duennwald ML & Lindquist S. Impaired ERAD and ER stress are early and specific events in polyglutamine toxicity. Genes Dev. 22, 3308–3319 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wu CC-C, Peterson A, Zinshteyn B, Regot S. & Green R. Ribosome collisions trigger general stress responses to regulate cell fate. Cell 182, 404–416.e14 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Glock C, Heumüller M. & Schuman EM mRNA transport & local translation in neurons. Curr. Opin. Neurobiol 45, 169–177 (2017). [DOI] [PubMed] [Google Scholar]
  • 58.Sathasivam K. et al. Aberrant splicing of HTT generates the pathogenic exon 1 protein in Huntington disease. Proc. Natl Acad. Sci. USA 110, 2366–2370 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Goldberg YP et al. Cleavage of huntingtin by apopain, a proapoptotic cysteine protease, is modulated by the polyglutamine tract. Nat. Genet 13, 442–449 (1996). [DOI] [PubMed] [Google Scholar]
  • 60.Bañez-Coronel M. et al. RAN translation in Huntington disease. Neuron 88, 667–677 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Smith AM, Costello MS, Kettring AH, Wingo RJ & Moore SD Ribosome collisions alter frameshifting at translational reprogramming motifs in bacterial mRNAs. Proc. Natl Acad. Sci. USA 116, 21769–21779 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Simms CL, Yan LL, Qiu JK & Zaher HS Ribosome collisions result in +1 frameshifting in the absence of no-go decay. Cell Rep. 28, 1679–1689.e4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Michel AM et al. GWIPS-viz: development of a ribo-seq genome browser. Nucleic Acids Res. 42, D859–D864 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wang H. et al. RPFdb v2.0: an updated database for genome-wide information of translated mRNA generated from ribosome profiling. Nucleic Acids Res. 47, D230–D234 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Duim WC, Jiang Y, Shen K, Frydman J. & Moerner WE Super-resolution fluorescence of Huntingtin reveals growth of globular species into short fibers and coexistence of distinct aggregates. ACS Chem. Biol 9, 2767–2778 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ingolia NT, Brar GA, Rouskin S, McGeachy AM & Weissman JS The ribosome profiling strategy for monitoring translation in vivo by deep sequencing of ribosome-protected mRNA fragments. Nat. Protoc 7, 1534–1550 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kumari R, Michel AM & Baranov PV PausePred and Rfeet: webtools for inferring ribosome pauses and visualizing footprint density from ribosome profiling data. RNA 24, 1297–1304 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Reinhart PH et al. Identification of anti-inflammatory targets for Huntington’s disease using a brain slice-based screening assay. Neurobiol. Dis 43, 248–256 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Xiao Y. et al. Poliovirus intrahost evolution is required to overcome tissue-specific innate immune responses. Nat. Commun 8, 375 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cox J. & Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol 26, 1367–1372 (2008). [DOI] [PubMed] [Google Scholar]
  • 71.Cox J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res 10, 1794–1805 (2011). [DOI] [PubMed] [Google Scholar]
  • 72.Perez-Riverol Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Aviner Supplementary

Data Availability Statement

Previously published ribosome profiling analyses were from GSE87328 (Fig. 1c), GSE72064 (Fig. 1d,e), SRA160745, GSE74279 and GSE94460 (Extended Data Fig. 1a), GSE66715, GSE81283 and GSE36892 (Extended Data Fig. 1b) and GSE53743 and GSE119615 (Extended Data Fig. 1c). For analysis of previously published proteomic data, processed MS intensity tables were downloaded from the supplementary information section of the following published articles: Hosp et al.6, Supplementary Tables 1 and 3 (label-free quantification, LFQ); Sap et al.50, Supplementary Tables 1 and 3 (LFQ); Newcombe et al.43, Supplementary Table 2 (LFQ); and Sui et al.45, Supplementary Table 1 (raw intensities). No additional normalization of the data was performed. Statistical tests were done using two-sided Student’s t-test. Sequencing data were deposited in the Sequence Read Archive (SRA) database under BioProject number PRJNA730032. The MS proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE72 partner repository with the dataset identifier PXD026012. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

RESOURCES