RT Journal Article SR Electronic T1 Translational profiling of macrophages infected with Leishmania donovani identifies mTOR- and eIF4A-sensitive immune-related transcripts JF bioRxiv FD Cold Spring Harbor Laboratory SP 2019.12.20.884338 DO 10.1101/2019.12.20.884338 A1 Visnu Chaparro A1 Louis-Philippe Leroux A1 Laia Masvidal A1 Julie Lorent A1 Tyson E. Graber A1 Aude Zimmermann A1 Guillermo Arango Duque A1 Albert Descoteaux A1 Tommy Alain A1 Ola Larsson A1 Maritza Jaramillo YR 2019 UL http://biorxiv.org/content/early/2019/12/20/2019.12.20.884338.abstract AB The protozoan parasite Leishmania donovani (L. donovani) causes visceral leishmaniasis, a chronic infection which is fatal when untreated. While previous studies showed that L. donovani reprograms transcription to subvert host cell functions, it remains unclear whether the parasite also alters host mRNA translation to establish a successful infection. To assess this, we compared transcriptome-wide translation in primary mouse macrophages infected with L. donovani promastigotes or amastigotes using polysome-profiling. This identified ample selective changes in translation (3,127 transcripts) which were predicted to target central cellular functions by inducing synthesis of proteins related to chromatin remodeling and RNA metabolism while inhibiting those related to intracellular trafficking and antigen presentation. Parallel quantification of protein and mRNA levels for a set of transcripts whose translation was activated upon L. donovani infection (Papbpc1, Eif2ak2, and Tgfb) confirmed, as indicated by polysome-profiling, increased protein levels despite largely unaltered mRNA levels. Mechanistic in silico analyses suggested activated translation depending on the kinase mTOR (e.g. Pabpc1) and the RNA helicase eIF4A (e.g. Tgfb) during infection. Accordingly, treatment with mTOR inhibitors torin-1 or rapamycin reversed L. donovani-induced PABPC1 without affecting corresponding transcript levels. Similarly, the production of TGF-β decreased in presence of the eIF4A inhibitor silvestrol despite unaltered Tgfb mRNA levels. Consistent with parasite modulation of host eIF4A-sensitive translation to promote infection, silvestrol suppressed L. donovani replication within macrophages. In contrast, parasite survival was favored under mTOR inhibition. In summary, infection-associated changes in translation of mTOR- and eIF4A-sensitive mRNAs contribute to modulate mRNA metabolism and immune responses in L. donovani-infected macrophages. Although the net outcome of such translation programs favours parasite propagation, individual translation programs appear to have opposing roles during L. donovani infection, thereby suggesting their selective targeting as key for therapeutic effects.Author Summary Fine-tuning the efficiency of mRNA translation into proteins allows cells to tailor their responses to stress without the need for synthesizing new mRNA molecules. It is well established that the protozoan parasite Leishmania donovani alters transcription of specific genes to subvert host cell functions. However, discrepancies between transcriptomic and proteomic data suggest that post-transcriptional regulatory mechanisms also contribute to modulate host gene expression programs during L. donovani infection. Herein, we report that one third of protein-coding mRNAs expressed in macrophages are differentially translated upon infection with L. donovani. Our computational analyses reveal that subsets of mRNAs encoding functionally related proteins share the same directionality of translational regulation, which is likely to impact metabolic and microbicidal activity of infected cells. We also show that upregulated translation of transcripts that encode central regulators of mRNA metabolism and inflammation is sensitive to the activation of mTOR or eIF4A during infection. Finally, we observe that inhibition of eIF4A activity reduces parasite survival within macrophages while selective blockade of mTOR has the opposite effect. Thus, our study points to a dual role for translational control of host gene expression during L. donovani infection and suggests that novel regulatory nodes could be targeted for therapeutic intervention.