De novo synthesized polyunsaturated fatty acids operate as both host immunomodulators and nutrients for Mycobacterium tuberculosis

  1. Thomas Laval
  2. Laura Pedró-Cos
  3. Wladimir Malaga
  4. Laure Guenin-Macé
  5. Alexandre Pawlik
  6. Véronique Mayau
  7. Hanane Yahia-Cherbal
  8. Océane Delos
  9. Wafa Frigui
  10. Justine Bertrand-Michel
  11. Christophe Guilhot
  12. Caroline Demangel  Is a corresponding author
  1. Immunobiology of Infection Unit, Institut Pasteur, INSERM U1224, Université de Paris, France
  2. Université de Paris, Sorbonne Paris Cité, France
  3. Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS-UPS UMR 5089, France
  4. Integrated Mycobacterial Pathogenomics Unit, Institut Pasteur, CNRS UMR 3525, Université de Paris, France
  5. Immunoregulation Unit, Institut Pasteur, INSERM U122, Université de Paris, France
  6. MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, France
  7. I2MC, Université de Toulouse, INSERM, Université Toulouse III - Paul Sabatier (UPS), France
8 figures, 1 table and 4 additional files

Figures

Figure 1 with 1 supplement
Mtb infection upregulates intracellular levels of free SFAs, MUFAs, and upstream PUFAs in host macrophages.

(A) Schematics of SFA and MUFA biosynthetic pathways. PA, palmitic acid; SA, stearic acid; OA, oleic acid; VA, vaccenic acid. (B) Intracellular levels of free SFAs and MUFAs in BMDMs infected with M. bovis BCG (BCG) or M. tuberculosis H37Rv (Mtb) at the same multiplicity of infection (MOI) of 2:1, or treated with LPS or Pam3Csk4 (Pam3), or left untreated (Ctrl) for 24 hr. FA levels were normalized to total DNA content and are shown as fold change relative to Ctrl. (C) Schematics of PUFA biosynthetic pathways. LA, linoleic acid; DGLA, dihomo-γ-LA; AA, arachidonic acid; ALA, α-linolenic acid; DPA, docosapentaenoic acid; DHA, docosahexaenoic acid. (D) Intracellular levels of free PUFAs in BMDMs treated as in (B). All data are means ± SD (n = 3) and are representative of two independent experiments. *p<0.05, **p<0.01, ***p<0.001, unpaired Student’s t-tests.

Figure 1—figure supplement 1
Mtb infection upregulates intracellular levels of total MUFAs and upstream PUFAs in host macrophages.

(A, B) Intracellular levels of total SFAs and MUFAs (A), or total PUFAs (B) in BMDMs treated as in Figure 1B. Data are means ± SD (n = 3) and are representative of two independent experiments. *p<0.05, **p<0.01, ***p<0.001, unpaired Student’s t-tests.

Mtb infection and IFNγ signaling cooperate to stop host PUFA biosynthesis.

(A) Relative mRNA expression of SFA/MUFA biosynthetic enzymes in BMDMs primed with IFNγ before infection with Mtb for the indicated times, as determined by qRT-PCR. (B) SCD activity in BMDMs, as estimated by the ratio of oleic acid (OA) to stearic acid (SA) levels, after 24 hr of infection with Mtb or BCG. (C–D) Relative mRNA expression of biosynthetic enzymes (C) or LXR/SREBP1 target genes (D) in BMDMs treated as in (A), as determined by qRT-PCR. All data are means ± SD (n = 3) and are representative of two independent experiments. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, two-way ANOVA with Dunnett post-hoc multiple comparison tests (A, C, D) and unpaired Student’s t-tests (B).

Figure 3 with 1 supplement
FADS2 inhibition impairs the effector functions of macrophages during Mtb infection.

(A) Activities of PUFA biosynthetic enzymes in resting or IFNγ-primed BMDMs, either left untreated (Ctrl), or infected with Mtb and treated with a FADS2 inhibitor (iFADS2) or vehicle control, as determined by a conversion assay from 6 to 24 hr post infection using the ω6 precursor LA-d11. Enzyme activities were estimated with the ratio of deuterated fatty acid product to substrate levels and are shown as fold change relative to Ctrl. nd, product not detected; n/a, not applicable (substrate and product not detected). (B) Schematics of biosynthetic pathways of arachidonic acid (AA)-derived eicosanoids. COX, cyclooxygenase; LOX, lipoxygenase; PG, prostaglandin; TX, thromboxane; HETE, hydroxyeicosatetraenoic acid; LX, lipoxin. (C, D) Secreted levels of COX- (C) and LOX-derived (D) metabolites of AA by BMDMs either uninfected (Ctrl) or infected with Mtb, and treated with iFADS2 or vehicle control for 24 or 48 hr. Data in (A), (C), and (D) are means ± SD (n = 3), *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, unpaired Student’s t-tests (A) and two-way ANOVA with Dunnett post-hoc multiple comparison tests (C, D). (E, F) Heatmap of mRNA expression levels of inflammatory (E) and antimicrobial (F) genes determined by NanoString analysis of BMDMs treated as in (C) for 6 or 24 hr. Shown are genes that were significantly downregulated by iFADS2 treatment at 6 and/or 24 hr (fold change of at least 1.15 and FDR < 0.05, two-way ANOVA with Benjamini−Hochberg adjustment for multiple comparison). Source data are available in Figure 3—source data 1.

Figure 3—source data 1

Complete list of normalized mRNA levels in BMDMs, either noninfected (NI Ctrl) or infected with Mtb, and treated with iFADS2 (Mtb iFADS2) or vehicle control (Mtb Veh), as determined by NanoString analysis.

https://cdn.elifesciences.org/articles/71946/elife-71946-fig3-data1-v2.xlsx
Figure 3—figure supplement 1
Effects of IFNγ on PUFA biosynthesis by Mtb-infected BMDMs.

(A) Intracellular levels of deuterated ω6 PUFAs in BMDMs treated as in Figure 3A, relative to total deuterated FA levels. (B) Relative mRNA expression of Ptgs2 in BMDMs primed with IFNγ before infection with Mtb, as determined by qRT-PCR. (C) Secreted levels of PGE2 by resting or IFNγ-primed BMDMs, treated with iFADS2 or vehicle control, and infected with Mtb for the indicated time. (D) Proportion of apoptotic or necrotic cells among BMDMs either left untreated (Ctrl), treated with camptothecin, or infected with Mtb and treated with iFADS2 or vehicle control for the indicated time, as estimated by flow cytometry. (E) Relative mRNA expression of Syt7 in Mtb-infected BMDMs treated as in Figure 3C, as determined by qRT-PCR. Data are means ± SD (n = 3), *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001, unpaired Student’s t-tests (A) or two-way ANOVA with Dunnett (B, C) or Bonferroni (D, E) post-hoc multiple comparison tests.

Figure 4 with 1 supplement
Inhibiting FADS2 does not impact Mtb growth in macrophages nor mice.

(A) Intracellular growth of Mtb inside resting or IFNγ-primed BMDMs treated with iFADS2 or with vehicle control, as determined by colony-forming unit (CFU) plating at the indicated days post infection. Data are means ± SD (n = 3) and are representative of two independent experiments. *p<0.05, ***p<0.001, two-way ANOVA with Bonferroni post-hoc multiple comparison tests. (B) Intracellular growth of Mtb inside differentiated THP-1 wild-type (WT) or FADS2 knockout (KO) clones, as determined by CFU plating at the indicated days post infection. Each line represents mean CFUs (n = 3) in one independent THP-1 clone. Data are representative of two independent experiments. (C, D) Growth of Mtb in the lungs (C) and spleen (D) of mice treated with iFADS2 or with vehicle control during 4 weeks, as determined by CFU plating. Data shown are means ± SEM of two pooled independent experiments (Exp 1: n = 3, Exp 2: n = 4–5 mice per time point). (E) Relative mRNA expression of inflammatory and antimicrobial genes in the lungs of mice treated as in (C), as determined by qRT-PCR. Data shown are means ± SEM (Exp 2: n = 4 or 5), *p<0.05, **p<0.01 in a two-way ANOVA with Bonferroni post-hoc multiple comparison tests.

Figure 4—figure supplement 1
Limiting Fads2 gene expression in macrophages.

(A) Relative mRNA expression of Fads2 in BMDMs transfected with nontargeting (siCtrl) or Fads2 siRNA (siFads2) for 48 hr, as determined by qRT-PCR. Data are means ± SD (n = 3) and are representative of two independent experiments. ***p<0.001, unpaired Student’s t-test. (B) Intracellular growth of M. tuberculosis (Mtb) inside BMDMs transfected as in (A), as determined by colony-forming unit (CFU) plating at the indicated days post infection. Data are means ± SD (n = 3) and are representative of at least two independent experiments. (C) Protein expression of FADS2 and GAPDH (loading control) in wild-type (WT) and FADS2 knockout (FADS2 KO) THP-1 clones, as determined by western blot. Source data are available as Figure 4—figure supplement 1—source data 1, Figure 4—figure supplement 1—source data 2, and Figure 4—figure supplement 1—source data 3. (D) Intracellular levels of deuterated ω3 polyunsaturated fatty acids (PUFAs) in THP-1 clones incubated with the ω3 precursor ALA-d14 for 24 hr, relative to total deuterated fatty acids (FAs) levels. Data are means ± SD (n = 2).

Figure 5 with 1 supplement
Mtb efficiently imports ω6 PUFAs through the Mce1 transporter in axenic culture.

(A) Schematics of the click-chemistry approach used to compare the uptake of SFAs, MUFAs and PUFAs by Mtb in axenic culture. (B) Differential kinetics of alkyne-SFA, -MUFA, and -PUFA uptake (all added at a concentration of 20 µM) by Mtb, as estimated by flow cytometry assessment of the geometric mean fluorescence intensities (gMFI) of clicked-FA. Data shown are representative of three independent experiments. (C) Schematics of the composition of the mce1-4 operons and related genes in Mtb genome. (D) Uptake of alkyne-PA by different Mtb strains deficient for the expression of genes belonging to mce1-4 operons, relative to wild-type (WT) Mtb. Data are means ± SD (n = 3) and are representative of two independent experiments, **p<0.01, unpaired Student’s t-tests. (E) Uptake of alkyne-FAs by Mtb Δmce1D and its complemented counterpart (Comp), relative to WT. Data are means ± SD from three independent experiments, *p<0.05, **p<0.01, ***p<0.001, paired t-tests. (F) Relative uptake of alkyne-OA, -LA, and -AA by Mtb WT in the presence of increasing amounts of natural palmitic acid (PA), oleic acid (OA), linoleic acid (LA), or arachidonic acid (AA). Data shown are representative of at least two independent experiments.

Figure 5—figure supplement 1
Inactivating genes of the Mce1 operon impairs Mtb's ability to import PUFAs.

(A) Schematics of the strategy used to generate allelic exchange substrates. Figure 5—figure supplement 1—source data 1 and Figure 5—figure supplement 1—source data 2 show the raw unedited gel. (B) Uptake of alkyne-FAs by the ΔyrbE1A mutant of M. tuberculosis (Mtb), relative to Mtb WT. Data shown are means ± SD (n = 1, 2, or 3 independent experiments).

Figure 6 with 2 supplements
Mtb preferentially internalizes AA in the context of macrophages.

(A) Differential uptake of alkyne-FAs by Mtb isolated from BMDMs infected for 24 or 72 hr, as measured by flow cytometry. Data are means ± SD from two or three independent experiments, *p<0.05, paired t-tests. (B) Distribution of alkyne-AA in Mtb-infected BMDMs at 24 hr post infection, as shown on representative confocal images (green = GFP-expressing Mtb, log scale colormap = clicked AA). Color bar indicates the relative range of pixel intensity (white = high, purple = low, from 0 arbitrary unit to 1). Bar scale = 5 µm. Enlargement of the boxed area in the merged image (bar scale = 1 µm). (C, D) Quantification of the alkyne-FA signal in intracellular bacteria detected based on GFP signal (C) and in whole BMDMs either noninfected (NI) or Mtb-infected (D) using confocal images of BMDMs infected for 24 hr with a GFP-expressing strain of Mtb. Bars show means ± SD, n > 82 for (C) and n > 37 for (D). ns, not significant, ****p<0.0001, unpaired Student’s t-test (C) and one-way ANOVA with Tukey post-hoc multiple comparison tests (D). (E) Uptake of alkyne-FAs by GFP-expressing Mtb Δmce1D and its complemented counterpart (Comp), recovered from BMDMs infected for 24 hr, relative to GFP-expressing Mtb WT, as analyzed by flow cytometry. Data are means ± SD from three independent experiments, *p<0.05, **p<0.01, paired t-tests. (F) Intracellular growth of different Mtb strains inside BMDMs, as determined by colony-forming unit (CFU) plating at the indicated days post infection. (G) Secreted levels of AA metabolites by BMDMs infected with Mtb WT or Δmce1D for 24 or 48 hr. Data in (F) and (G) are means ± SD (n = 3), *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 in a two-way ANOVA with Bonferroni post-hoc multiple comparison tests.

Figure 6—figure supplement 1
Differential uptake of SFAs, MUFAs and PUFAs by intracellular Mtb and host BMDMs.

(A) Schematics of the click-chemistry approach used . (B) Gating strategy for the analysis of FA uptake by a GFP-expressing strain of Mtb isolated from infected BMDMs at 24 hr post infection, and stained by click-chemistry. (C) Differential uptake of alkyne-FAs by Mtb isolated from resting or IFNγ-primed BMDMs infected for 24 hr, as measured by flow cytometry. Data shown are from two or three independent experiments, ns, not significant, *p<0.05, paired t-tests. (D) Quantification of the alkyne-FA signal in intracellular bacteria detected based on GFP signal using confocal images of BMDMs infected for 24 hr with different GFP-expressing Mtb strains. Bars show means ± SD, n > 54. ns, not significant, **p<0.01, ***p<0.001 in a one-way ANOVA with Dunnett post-hoc multiple comparison tests. (E) Relative mRNA expression of pro-inflammatory genes by BMDMs infected with Mtb WT, Δmce1D, or its complemented counterpart (Comp) for 24 hr. Data are means ± SD (n = 3), *p<0.05, **p<0.01, ***p<0.001, in a one-way ANOVA with Tukey post-hoc multiple comparison tests.

Figure 6—figure supplement 2
Relative mRNA expression of PPARγ target genes in BMDMs infected with Mtb for the indicated times, as determined by NanoString analysis.

Data are means ± SD (n = 3). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 in a two-way ANOVA with Bonferroni post-hoc multiple comparison tests.

Author response image 1
Relative mRNA expression of Alox5 in Mtb-infected BMDMs treated as in Figure 3C, as determined by Nanostring analysis.

Data are means +/- SD (n=3), *P<0.05, ****P<0.0001 in a two-way ANOVA with Bonferroni post-hoc multiple comparison tests.

Author response image 2
Relative mRNA expression of inflammatory genes in Mtb-infected BMDMs transfected with siCtrl or siFads2 for 48h (left panel), and infected with Mtb for the indicated periods of time (middle and right panels), as determined by qRT-PCR.

Data are means +/- SD (n=3), *P<0.05, **P<0.01, ***P<0.001 in a two-way ANOVA with Bonferroni post-hoc multiple comparison tests.

Tables

Appendix 1—key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Strain, strain background (Mycobacterium tuberculosis)M. tuberculosisLaleh Majlessi, Institut Pasteur, ParisH37RvAnimal passaged
Strain, strain background (M. tuberculosis)M. tuberculosis DsRedMarino et al., 2015H37Rv DsRedAnimal passaged
Strain, strain background (Mycobacterium bovis)BCG PasteurRoland Brosch, Institut Pasteur, Paris1173P2
Strain, strain background (Mus musculus), maleC57BL/6JCharles River LaboratoriesJAX stock no: 000664;RRID: IMSR_JAX:000664
Cell line (Homo sapiens)THP-1ATCCATCC TIB-202;RRID:CVCL_0006
Strain, strain background (Escherichia coli)HB101PromegaCat# L2011Recombinant cloning and subcloning strain
Gene (M. tuberculosis)lucAATCC27294Rv3723
Gene (M. tuberculosis)mce1DATCC27294Rv0172
Gene (M. tuberculosis)mceGATCC27294Rv0655
Gene (M. tuberculosis)omamBATCC27294Rv0200
Gene (M. tuberculosis)yrbE1AATCC27294Rv0167
Gene (M. tuberculosis)yrbE2AATCC27294Rv0587
Gene (M. tuberculosis)yrbE3AATCC27294Rv1964
Gene (M. tuberculosis)yrbE4AATCC27294Rv3501
Genetic reagent (M. tuberculosis)ΔlucAThis study, available from the corresponding authorΔlucA::kmChromosomal deletion of Rv3723 (lucA) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)Δmce1DThis study, available from the corresponding authorΔmce1D::kmChromosomal deletion of Rv0172 (mce1D) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)ΔmceGThis study, available from the corresponding authorΔmceG::kmChromosomal deletion of Rv0655 (mceG) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)ΔomamBThis study, available from the corresponding authorΔomamB::kmChromosomal deletion of Rv0200 (omamB) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)ΔyrbE1AThis study, available from the corresponding authorΔyrbE1A::kmChromosomal deletion of Rv0167 (yrbE1A) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)ΔyrbE2AThis study, available from the corresponding authorΔyrbE2A::kmChromosomal deletion of Rv0587 (yrbE2A) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)ΔyrbE3AThis study, available from the corresponding authorΔyrbE3A::kmChromosomal deletion of Rv1964 (yrbE3A) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)ΔyrbE4AThis study, available from the corresponding authorΔyrbE4A::kmChromosomal deletion of Rv3501 (yrbE4A) and insertion of an antibiotic resistance marker by double crossover recombination
Genetic reagent (M. tuberculosis)Δmce1D CompThis study, available from the corresponding authorΔmce1D::km::yrbE1 to rv0178WT copy of the yrbE1 to rv0178 operon integrated at AttB
Recombinant DNA reagentPX458 (plasmid)Addgene(Ran et al., 2013)pSpCas9(BB)–2A-GFP (pX458)
Recombinant DNA reagentpET26b (plasmid)Sigma-AldrichpET26b(+) - Novagen
Recombinant DNA reagentpJV53H (plasmid)This study, available from C. GuilhotpJV53HpJV53 (van Kessel and Hatfull, 2007) with hygromycin resistance gene
Recombinant DNA reagentpMV361 (plasmid)Stover et al., 1993pMV361AttB integrating M. tuberculosis plasmid
Recombinant DNA reagentpMVZ621 (plasmid)Didier Zerbib, Toulouse Biotechnology Institute, FrancepMVZ621pMV261 carrying a Zeocin resistance gene
Recombinant DNA reagentpWM430 (plasmid)This studypWM430pMV361 with insertion of a zeocin cassette between the XbaI and Eco147i restriction sites
Recombinant DNA reagentpWM431 (plasmid)This studypWM431pWM430 with a fragment going from gene yrbE1A to gene rv0178 of M. tuberculosis H37Rv
Recombinant DNA reagentpWM251 (plasmid)This studypWM251pMIP12 (Le Dantec et al., 2001) carrying the streptomycin resistance cassette from pHP45Ω (Prentki and Krisch, 1984) and the gfp under the control of the pBlaF* promotor
Recombinant DNA reagentRv165 (cosmid)Brosch et al., 1998Rv165 cosmidCosmid carrying a large fragment of the H37Rv genome covering the Mce1 region.
Sequence-based reagentPrimers used to generate AES and Mtb mutantsMerckPCR primersSupplementary file 2
Sequence-based reagentEurofins GenomicsqPCR primersSupplementary file 3
Sequence-based reagentFADS2_ex2_FEurofins GenomicsPCR primer5′-GCACATTTCCAGTGCCAAGG-3′
Sequence-based reagentFADS2_ex2_REurofins GenomicsPCR primer5′-GGAGAGAGGAGACGCCACTA-3′
Sequence-based reagentGuide RNA targeting the exon 2 of FADS2Eurofins GenomicsOligonucleotide5′-GCACCCTGACCTGGAATTCGT-3′
Transfected construct (M. musculus)ON-TARGETplus siRNAs (SMARTpool)Dharmacon/Horizon DiscoveriesNon-targeting: D-001810-10Mouse Fads2:L-049816-01
AntibodyAnti-FADS2 (rabbit polyclonal)Thermo Fisher ScientificPA576611; RRID:AB_2720338(1:1000) dilution
AntibodyAnti-GAPDH (rabbit monoclonal)Cell Signaling Technology2118; RRID:AB_561053(1:1000) dilution
AntibodyGoat anti-rabbit IgG (HRP conjugate)Santa CruzSC-2004;RRID:AB_631746(1:1000) dilution
Chemical compound, drugSC-26196Cayman Chemical10792
Chemical compound, drugUltrapure LPS from E. coliEnzo Life SciencesALX-581-013Serotype 055:B5
Chemical compound, drugPam3Csk4Invivogentlrl-pms
Chemical compound, drugLipofectamine RNAiMAXThermo Fisher Scientific13778075
Commercial assay or kitVenorGeM Advance Mycoplasma detection kitMinerva Biolabs11-7024
Commercial assay or kitHuman Monocyte Nucleofector KitLonzaVPA-1007
Commercial assay or kitClick-iT Plus Alexa Fluor-647 Picolyl Azide kitThermo Fisher ScientificC10643
Commercial assay or kitQuant-iT dsDNA Assay Kit, broad rangeThermo Fisher ScientificQ33130
Commercial assay or kitApoptosis/ Necrosis Assay KitAbcamab176750
Commercial assay or kitHigh-Capacity cDNA Reverse Transcription KitThermo Fisher Scientific4368814
Commercial assay or kitPower SYBR Green PCR Master MixThermo Fisher Scientific4367659
Commercial assay or kitmiRNeasy Mini KitQIAGEN217004
Commercial assay or kitCloneJET PCR Cloning KitThermo Fisher ScientificK1231
Commercial assay or kitAmpliTaq Gold 360 master mixThermo Fisher Scientific4398876
Peptide, recombinant proteinDreamTaq Green polymeraseThermo Fisher ScientificEP0711
Peptide, recombinant proteinT4 DNA ligaseThermo Fisher ScientificEL0011
Peptide, recombinant proteinPrimeSTAR GXL DNA PolymeraseTakara BioR050B
Software, algorithmGraphPad PrismGraphPad Prism (https://graphpad.com)RRID:SCR_015807
Software, algorithmZen Imaging softwareZeissRRID:SCR_013672
Software, algorithmIcy opensource platformhttp://www.icy.bioimageanalysis.org de Chaumont et al., 2012RRID:SCR_010587
Software, algorithmFlowJo softwarehttps://www.flowjo.com/RRID:SCR_008520
Software, algorithmnSolver analysis softwarehttp://www.nanostring.com/products/nSolverRRID:SCR_003420
Other4–12% NuPAGE Bis-Tris gelsThermo Fisher ScientificNP0322BOX
OtheriBlot 2 gel transfer Stacks Nitrocellulose systemThermo Fisher ScientificIB23001
OtherProlong Diamond Antifade MountantThermo Fisher ScientificP36961
OtherDAPI stainSigma-AldrichD9542Used at 1 µg/mL

Additional files

Supplementary file 1

NanoString nCounter Custom CodeSet.

https://cdn.elifesciences.org/articles/71946/elife-71946-supp1-v2.docx
Supplementary file 2

Primers used to generate the various allelic exchange substrates (AES) and M. tuberculosis (Mtb) mutants.

DraIII and Van91I restriction sites introduced into the primers are indicated in red and green, respectively.

https://cdn.elifesciences.org/articles/71946/elife-71946-supp2-v2.docx
Supplementary file 3

qPCR primers sequences.

https://cdn.elifesciences.org/articles/71946/elife-71946-supp3-v2.docx
Transparent reporting form
https://cdn.elifesciences.org/articles/71946/elife-71946-transrepform1-v2.pdf

Download links

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

Downloads (link to download the article as PDF)

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

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

  1. Thomas Laval
  2. Laura Pedró-Cos
  3. Wladimir Malaga
  4. Laure Guenin-Macé
  5. Alexandre Pawlik
  6. Véronique Mayau
  7. Hanane Yahia-Cherbal
  8. Océane Delos
  9. Wafa Frigui
  10. Justine Bertrand-Michel
  11. Christophe Guilhot
  12. Caroline Demangel
(2021)
De novo synthesized polyunsaturated fatty acids operate as both host immunomodulators and nutrients for Mycobacterium tuberculosis
eLife 10:e71946.
https://doi.org/10.7554/eLife.71946