Abstract
Human Vγ2Vδ2 T cells monitor isoprenoid metabolism by recognizing foreign (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP), a metabolite in the 2-C-methyl-D-erythritol-4-phosphate pathway used by most eubacteria and apicomplexan parasites, and self isopentenyl pyrophosphate, a metabolite in the mevalonate pathway used by humans. Whereas microbial infections elicit prolonged expansion of memory Vγ2Vδ2 T cells, immunization with prenyl pyrophosphates or aminobisphosphonates elicit short-term Vγ2Vδ2 expansion with rapid anergy and deletion upon subsequent immunizations. We hypothesized that a live, attenuated bacterial vaccine that overproduces HMBPP would elicit long lasting Vγ2Vδ2 T cell immunity by mimicking a natural infection. Therefore, we metabolically engineered the avirulent aroA− Salmonella enterica serovar Typhimurium SL7207 strain by deleting the gene for LytB (the downstream enzyme from HMBPP) and functionally complementing for this loss with genes encoding mevalonate pathway enzymes. LytB− Salmonella SL7207 had high HMBPP levels, infected human cells as efficiently as the wild-type bacteria, and stimulated large ex vivo expansions of Vγ2Vδ2 T cells from human donors. Importantly, vaccination of a rhesus monkey with live lytB− Salmonella SL7207 stimulated a prolonged expansion of Vγ2Vδ2 T cells without significant side effects or anergy induction. These studies provide proof-of-principle that metabolic engineering can be used to derive live bacterial vaccines that boost Vγ2Vδ2 T cell immunity. Similar engineering of metabolic pathways to produce lipid Ags or B vitamin metabolite Ags could be used to derive live bacterial vaccine for other unconventional T cells that recognize nonpeptide Ags.
Keywords: human, gamma delta T cell, Vγ2Vδ2 T cells, prenyl pyrophosphates, isopentenyl pyrophosphate, attenuated live bacterial vaccines, S. enterica serovar Typhimurium
Introduction
Unconventional, innate-like T cells, such as γδ T cells, αβ invariant natural killer T cells (iNKT), and mucosal-associated invariant αβ T cells, have unique roles that bridge innate and adaptive immunity by responding to nonpeptide stimulators/Ags or stress molecules in an MHC-unrestricted manner. The major subset of human γδ T cells use Vγ2Vδ2 TCRs (also termed Vγ9Vδ2 TCRs) to preferentially respond to the foreign microbial isoprenoid metabolite, HMBPP, which is produced in the 2-C-methyl-D-erythritol-4-phosphate (MEP) pathway (1, 2). They also are stimulated by the self-metabolite, IPP, produced in the mevalonate pathway (3). Aminobisphosphonates (4, 5) and alkylamines (6) also stimulate Vγ2Vδ2 T cells indirectly because they block farnesyl diphosphate synthase leading to the accumulation of its IPP substrate (7–10). Stimulation by all of these compounds requires the butyrophilin 3A1 Ig superfamily protein that may serve as a sensor for prenyl pyrophosphates (11–14).
Vγ2Vδ2 T cells expand to large numbers in the blood (up to 55% of T cells) with a variety of infections (reviewed in Ref.15) and can accumulate at peripheral sites (16, 17). HMBPP stimulates γδ T cells to produce Th1 cytokines and other inflammatory cytokines and chemokines, to kill infected cells, and to produce growth factors to repair mucosal surfaces (18). Because Vγ2Vδ2 T cell responses are not MHC restricted, can be elicited in all individuals regardless of their MHC haplotype, occur rapidly, and are present at birth (reviewed in Ref.15), they play an important role in T cell responses to bacterial and parasitic infections in humans and other primates.
In addition to their role in microbial immunity, human Vγ2Vδ2 T cells can also be used to treat malignancies. Activated Vγ2Vδ2 T cells can use their TCR and NK receptors to recognize and kill a variety of tumor cells, irrespective of their tissue origin, MHC expression, or MHC haplotype. Because many Vγ2Vδ2 T cells express the CD16 Fc receptor, they also kill tumor cells sensitized by antitumor Abs (19). Human Vγ2Vδ2 T cells provide immunity for non-Hodgkin’s lymphoma and other tumors in preclinical humanized SCID mouse models (20–23). In clinical trials, synthetic vaccines (bromohydrin pyrophosphate or aminobisphosphonates with IL-2) that stimulate Vγ2Vδ2 T cells resulted in complete or partial remissions for some patients with lymphoma (24) and stabilized disease in patients with metastatic prostate cancer (25) without major toxicities or autoimmunity. However, although initially effective, these vaccines result in anergy and deletion of Vγ2Vδ2 T cells after a few immunizations (26–28). In contrast, Vγ2Vδ2 T cell responsiveness is preserved after mycobacterial (29) or listeria (30) infections with reinfection resulting in earlier and more vigorous Vγ2Vδ2 T cell expansion.
Metabolic engineering of bacteria has focused on altering bacteria for drug or chemical synthesis or for the generation of alternative fuels (31–33). Directed changes in bacterial metabolism are made by modifying specific biochemical reactions or by introducing new ones to improve pathway efficiency or to produce new compounds. We reasoned that metabolic engineering could be used to develop a bacterial vaccine that overproduces HMBPP to stimulate for Vγ2Vδ2 T cells and that avoids the anergy and deletion that can occur with direct immunization with prenyl pyrophosphates or aminobisphosphonates. The bacteria would provide the necessary adjuvants to activate innate immunity and Ags to stimulate CD4 αβ T cells to provide help to Vγ2Vδ2 T cells.
Attenuated Salmonella bacteria have been used as live vaccines to prevent human typhoid fever caused by systemic infection with Salmonella enterica serovar Typhi (34, 35), fowl typhoid caused by S. enterica serovar Gallinarum, and infection of egg-laying hens by S. enterica serovars Typhimurium and Enteritidis. However, the vaccines for human typhoid fever are only 50–80% protective (36) and there are no vaccines to prevent human nontyphoidal salmonellosis typically caused by S. enterica serovars Typhimurium and Enteritidis. Nontyphoidal salmonellosis is a major cause of bacterial gastroenteritis in both developed and underdeveloped nations and is estimated to have caused 93.8 million cases and 155,000 deaths worldwide in 2006 (37). While generally restricted to the gastrointestinal tract, in AIDS patients and other immunocompromised patients these infections can be invasive, resulting in bacteremia and in death in 20–25% of African cases (38, 39). Thus, development of Salmonella vaccines targeting the Typhimurium serovar could help prevent these deaths.
In this study, we provide proof-of-principle that metabolic engineering can be used to develop bacterial vaccines by engineering a vaccine strain of S. enterica serovar Typhimurium to overproduce HMBPP, the major Vγ2Vδ2 T cell stimulator. The engineered bacteria expand human Vγ2Vδ2 T cells from PBMC upon ex vivo culture and monkey Vγ2Vδ2 T cells upon in vivo immunization.
Materials and Methods
Bacterial strains and plasmids
The attenuated vaccine strain, aroA− S. enterica serovar Typhimurium SL7207 (also termed 2337-65) hisG46 DEL407 (aroA544::TN10, Tc-s) was used for this study. SL7207 was derived from strain SL3261 by extensive deletion of the aroA gene for 5-enolpyruvylshikimate 3-phosphate synthase in aromatic amino acid and folic acid biosynthesis which renders the bacteria auxotrophic for para-aminobenzoic acid and 2,3 dihydroxybenzoic acid, two compounds that are not normally produced by mammals. To complement for the loss of the MEP pathway in this bacteria, the mevalonate pathway gene cluster from Streptomyces sp. strain CL190 was used (40, 41). This cluster contains all six genes for the mevalonate pathway enzymes (3-hydroxy-3-methylglutaryl-CoA reductase, 3-hydroxy-3-methylglutaryl-CoA synthase, isopentenyl diphosphate isomerase, mevalonate kinase, mevalonate diphosphate decarboxylase, phosphomevalonate kinase) required for the production of IPP and dimethylallyl pyrophosphate (diphosphate) (DMAPP). The high-copy-number pTMV19 kanomycin resistance gene (kmr) plasmid was derived by inserting the Streptomyces gene cluster from the pUMV19 plasmid (41) into the cloning site of the pTTQ18 plasmid and a kanamycin resistance gene into the ampicillin resistance gene. This is a high-copy-number plasmid based on a pUC plasmid (42). The low-copy-number pMMV19kmr plasmid was derived by inserting the Streptomyces gene cluster into the cloning site and a kanamycin resistance gene into the ampicillin resistance gene of the pMW118 plasmid. This is a low-copy-number plasmid (fewer than 5 copies per bacteria) based on the pSC101 plasmid (43).
Metabolic engineering of aroA− S. enterica serovar Typhimurium SL7207 by deletion of the lytB gene and complementation by mevalonate pathway genes
The lytB gene was deleted from aroA− S. enterica serovar Typhimurium SL7207 by homologous recombination using the “one-step inactivation” technique with the λ Red system (44). As detailed in Supplemental Fig. 1, aroA− Salmonella were transformed by electroporation (2.5 kV, 25 μF, 200 ohms, Gene Pulser II with Pulse Controller Plus, Bio-Rad, Hercules, CA) with either the pTMV19kmr or the pMMV19kmr plasmid containing the genes for mevalonate pathway enzymes. Plasmid retention was selected for with kanamycin (25 μg/ml). Next, the temperature sensitive pKD46 plasmid containing the λ Red locus was introduced into the bacteria and plasmid retention selected for with ampicillin (100 μg/ml) and incubation at 30°C. The resulting transformants were then grown for 2 d at 30°C with fosmidomycin (12.5 μg/ml) to block the MEP pathway. This switches isoprenoid synthesis to the introduced mevalonate pathway and greatly improves recovery of deletion mutants. On the third day, the bacteria were diluted to 0.1 OD at 600 nm (OD600) and grown with fosmidomycin and L-arabinose (10 mM) (to induce the recombination enzymes) until reaching an OD600 of 0.4–0.6, at which time they were either used for electroporation or frozen. A PCR product targeting the lytB gene was produced by using primers composed of sequences flanking the lytB gene (50 bp) and sequences from the pKD3 chloramphenicol resistance gene (21 bp). The sequence of the lytB forward primer was 5′-cactttgatattgaagtgctggaaatcgatccggcgctggaggcataaacatgtgtaggctggagctgcttc-3′ and the lytB reverse primer was 5′-taagatctggtcaacgtagcgtcatcaggcagatttctgaattgtgaaatccatatgaatatcctccttagt-3′. The resulting 1.1-kB DNA fragment was introduced by electroporation into aroA− Salmonella containing pTMV19kmr or pMMV19kmr and pKD46. After incubation for 2 h, deletion mutants were selected by spreading on Luria-Bertani (LB) agar plates containing chloramphenicol (10–25 μg/ml) and incubating at 30° C. Chloramphenicol-resistant deletion mutants were then grown at 37°C to eliminate the temperature sensitive pKD46 plasmid.
Preparation of bacterial sonicates and supernatants and assaying the bioactivity of Salmonella bacteria for human Vγ2Vδ2 T cells
To quantitate the bioactivity of Salmonella bacterial mutants for Vγ2Vδ2 T cells, bacterial clones were grown in 100 ml LB media in baffled 500 ml Erlenmeyer flasks to late stationary phase (~24 h) at 37°C in an Innova 4400 shaker (New Brunswick Scientific, Enfield, CT) oscillating at 225 revolutions/min. Bacteria were harvested and washed twice with Milli-Q H2O. The bacteria were then suspended in 10 ml of Milli-Q H2O and continuously probe sonicated for 10 min on ice at the 4.5 setting using a microtip probe (Q700 Sonicator, Qsonica, Newtown, CT). The bacterial sonicates and culture supernatants were heated in a boiling water bath for 5 min, cooled on ice for 5 min, centrifuged at 16,000 × g for 30 min at 4°C, filter sterilized with a 0.22 μm filter, and, in some cases, size fractionated by ultrafiltration (3000 Da cut-off units; Pall, Port Washington, NY) for the pass-through fraction. The samples were stored at −80°C until testing. To assay bioactivity, 5–10 × 104 12G12 Vγ2Vδ2 T cells were cultured with 5–10 × 104 mitomycin C-treated Va-2 cells (a transformed human fibrosarcoma cell line) as antigen presenting cells in 200 μl of complete medium (RPMI 1640 with 8% FBS, L-glutamine, sodium pyruvate, nonessential MEM amino acids, essential MEM amino acids, HEPES, and 2-ME) in round bottom 96-well plates. Culture supernatants and bacterial sonicates were added to the cultures at half-log dilutions and incubated at 37°C. The cultures were pulsed with 1 μCi of [3H]-thymidine after 24 h and harvested 16–24 h later (45). To quantitate bioactivity, the dilution of the bacterial supernatant or sonicate that stimulated half-maximal proliferation was determined and the reciprocal of this dilution gave the units of bioactivity (45). To control for variations in this assay (~3-fold), monoethyl pyrophosphate and HMBPP were used as positive controls to standardize the assay. One unit per milliliter of bioactivity corresponds to an HMBPP concentration of 31.6 pM (31.6 fmol/ml) or an IPP concentration of 3 μM (3 nmol/ml).
Growth and morphology of Salmonella vaccine bacteria
Wild-type aroA− S. enterica serovar Typhimurium SL7207 bacteria and lytB deletion mutants were cultured overnight and then diluted to 0.05 OD600 in 100 ml LB broth and cultured for 96 h with periodic measurement of absorbance. To assess morphology, bacteria were grown overnight on LB plates at 37°C. Then a colony was resuspended in H2O, spread on a slide, and Gram stained (BD Biosciences, Franklin Lakes, NJ).
Invasiveness and intracellular survival of Salmonella bacteria in human epithelial cells
To assess the ability of Salmonella bacterial mutants to invade and survive in human epithelial cells, Salmonella bacteria were cultured with HeLa tumor cells (derived from cervical tissue and positive for keratin) and intracellular bacterial numbers determined at 2 and 24 h postinfection in a standard gentamicin survival assay (46). For this assay, HeLa cells were grown in 24-well plates for 1 day prior to infection. Salmonella bacteria that had been grown to midlog exponential phase were added at a 10:1 multiplicity of infection (MOI). For the 2-h assay, the cells were washed at 2 h and intracellular bacteria recovered by lysing the cells with 0.1% Triton X-100. For the 24-h assay, the cells were washed with PBS 2 h after infection and incubated in media supplemented with high dose gentamicin (100 μg/ml) for an additional 2 h, washed again, and further incubated for 20 h with media supplemented with low-dose gentamicin (25 μg/ml) to kill any remaining extracellular bacteria. At 24 h postinfection, the cells were lysed as above. Intracellular bacterial numbers were assessed by plating the cell lysate on LB agar plates to determine CFUs. The 2-h incubation period assesses bacterial invasiveness whereas the 24-h incubation period assesses bacterial intracellular survival.
Ex vivo expansion of human Vγ2Vδ2 T cells stimulated by live Salmonella bacteria
For ex vivo expansion of Vγ2Vδ2 T cells stimulated by live Salmonella bacteria, PBMCs were isolated from the blood or leukopacs of normal human donors by Ficoll-Hypaque density centrifugation. 1 × 105 PBMCs in 0.2 ml of supplemented RPMI 1640 media (termed P-media (47)) were added to 96-well round-bottom wells and infected with Salmonella bacteria at an MOI of 1:1. After 6 h of infection, the cells were washed, treated with high-dose gentamicin (100 μg/ml) for 2 h, followed by low-dose gentamicin (25 μg/ml) for the remainder of the culture period. The cells were then incubated at 37°C with 5% CO2. On day 3, 2 nM (100 IU/ml) IL-2 was added and the cells harvested on day 9. For the Transwell assay, 1–3 × 106 bacteria were added in 0.1 ml RPMI 1640 medium to the inner wells of a Transwell plate (Corning Costar, Kennebunk, ME). The inner well was separated from the outer well by a 0.4 μm membrane. 2 × 106 PBMCs were added to the outer well in 0.9 ml of medium. After 4 h, the inner wells were removed. On day 3, 2 nM IL-2 was added and the media changed every 3 d. On day 9, the cells were harvested, washed, and γδ and Vγ2Vδ2 T cells enumerated by flow cytometric analysis using an LSR II flow cytometer and CellQuest or FACSDiva software (BD Biosciences, San Jose, CA). PBMCs were stained with anti-Cδ (anti-TCRδ1 [5A6.E9]), anti-Vδ1 (TS8.2), anti-Vδ2 (15D for rhesus monkey cells and B6 for human cells), or anti-Vγ2 mAbs (7A5) (all from Thermo Scientific, Rockford, IL except for B6 from BD Biosciences), followed by PE-goat F(ab′)2 anti-mouse IgG (H+L) secondary Ab (BioSource International, Camarillo, CA). After washing, residual Ig binding by the secondary antiserum was blocked by incubation with 5% normal mouse serum for 15 min at room temperature. Cells were then stained with FITC-anti-CD3ε (SP34; BD Biosciences). To determine T cell memory subsets, the cells were additionally reacted with allophycocyanin-anti-CD27 (O323, eBioscience, San Diego, CA) and PE-Cy7-anti-CD28 (CD28.2; eBioscience). The mouse IgG1κ mAb P3 (BD Biosciences) was used in the unconjugated form or in the FITC-, allophycocyanin-, or PE-Cy7-conjugated forms as a control mAb because all of the mAbs were IgG1 except for the anti-CD4 and anti-CD8β mAbs. Live cells were distinguished from dead cells by staining with Hoechst 33258 or propidium iodide. Adult blood donors were enrolled with written informed consent in accordance with the requirements of the University of Iowa and Veterans Affairs Health Care System Institutional Review Board as well as the guidelines of the World Medical Association’s Declaration of Helsinki.
Immunization of rhesus monkeys with live Salmonella bacteria
Female rhesus monkeys of Chinese origin were obtained from the National Institutes of Health specific-pathogen-free breeding colony (Corpus Christi, TX) for use in these studies (Covance Research Products, Denver, PA). The monkeys were free of infection with retroviruses, herpes B virus, and Shigella and Salmonella spp. All animals were maintained and used in accordance with protocols approved by the Institutional Animal Care and Use Committee of the Iowa City Veterans Health Care System. To prepare Salmonella vaccines for immunization, the bacteria were cultured in LB media overnight, diluted the next day, and grown to midlog phase. The bacteria were then harvested, washed twice with PBS, and resuspended in PBS to give the desired concentration of bacteria based on the absorbance at 600 nm. The bacterial dose given was verified by determining CFUs in the bacterial suspensions. For intranasal immunization of monkeys, the monkeys were anesthetized and 50–100 μl bacterial suspension instilled in each nostril. The intranasal route was used because it provides efficient infection via the nasal associated lymphoid tissue without the potential problem of incomplete neutralization of stomach acid. Wild-type aroA− S. enterica serovar Typhimurium strain SL7207 was used at 1 × 107 bacteria, lytB− aroA− S. enterica SL7207 pTMV19K-C7 was used at 3 × 109 bacteria, and lytB− aroA− S. enterica SL7207 pMMV19K-C22 was used at 3 × 1010 bacteria. Prior to immunization, baseline γδ T cell levels of each monkey were determined by flow cytometry. After immunization, blood was drawn twice a week and γδ T cells were assessed by staining with the mAbs used for monkeys (listed above) followed by flow cytometric analysis as detailed above. To determine the levels of monkey γδ T cells in the blood, 2–10 μl of various anti-γδ TCR mAbs (listed above) were mixed with 100 μl of heparinized blood, incubated for 30 min at room temperature, washed, and then reacted with PE-goat F(ab′)2 anti-mouse IgG (H+L) secondary Ab for 30 min. After washing, residual Ig binding by the secondary antiserum was blocked by incubation with 5% normal mouse serum for 15 min at room temperature. The cells were then washed and reacted with FITC-anti-CD3 mAb for 30 min at room temperature. RBCs were then lysed by resuspending in 0.5 ml of OptiLyse C buffer (Beckman Coulter, Fullerton, CA) for 10 min at 4°C. The cells were then washed, stained with either Hoechst 33258 or propidium iodide to identify dead cells, and then analyzed by flow cytometry. To assess memory Vδ2 T cell subsets, 100 μl of blood was reacted with the 15D anti-Vδ2 mAb followed by PE-goat F(ab′)2 anti-mouse IgG (H+L) secondary Ab and then blocked with normal mouse serum. The cells were then reacted with FITC-anti-CD3, allophycocyanin-anti-CD27, and PE-Cy7-anti-CD28 for 60 min at room temperature, washed, and RBCs lysed by resuspending in 0.5 ml of OptiLyse C buffer (Beckman Coulter) for 10 min at 4°C. CD4 and CD8β αβ T cells were measured by staining with FITC-anti-CD3 mAb followed by either PE-anti-CD4 (M-T477; BD Biosciences) or PE-anti-CD8β (2ST8.5H7; Beckman Coulter) mAbs.
Statistical methods
The nonparametric Mann-Whitney U-test was used for all statistical comparisons because of the potential non-Gaussian distribution of the data. Statistical analyses were done using the Prism program (version 4.0c) with p values of <0.01 considered statistically significant.
Results
Metabolically engineering lytB− aroA− S. enterica serovar Typhimurium SL7207 to overproduce HMBPP
There are two pathways for the synthesis of the IPP and DMAPP isoprenoid precursors (Fig. 1). Most eubacteria and all apicomplexan parasites use the MEP pathway to synthesize isoprenoid metabolites whereas all mammals use the mevalonate pathway. S. enterica, a Gram-negative bacterium in the order Enterobacteriales, uses the MEP pathway. The major stimulator of Vγ2Vδ2 T cells in microbes using the MEP pathway is HMBPP. HMBPP is produced by GcpE (also termed HMBPP synthase or IspG) and then converted to IPP and DMAPP by LytB (also termed HMBPP reductase or IspH). To derive vaccine bacteria that overproduce HMBPP, we deleted the downstream lytB gene causing HMBPP to accumulate. To functionally complement for this lethal mutation in the MEP pathway, genes encoding enzymes for the mevalonate isoprenoid synthetic pathway from Streptomyces sp. strain CL190 were introduced into the bacteria (Fig. 1). The mevalonate pathway genes were cloned into a high-copy-number plasmid based on the pUC plasmid. Additionally, because high levels of certain isoprenoid metabolites (e.g., 3-hydroxy-3-methylglutaryl-CoA [HMG-CoA]) are toxic to bacteria (48–50), we also cloned the mevalonate pathway genes into a low-copy-number plasmid (51) to reduce the levels of the mevalonate enzymes and their metabolites. Using this approach (detailed in Supplemental Fig. 1), we generated lytB deletion mutants in attenuated aroA− S. enterica serovar Typhimurium strain SL7207. A key step in deriving mutants was to grow the bacteria with fosmidomycin after transforming the bacteria with plasmids containing the mevalonate pathway. Fosmidomycin inhibits 1-deoxy-D-xylulose 5-phosphate reductase (52) in the MEP pathway. Blocking the MEP pathway switches bacterial isoprenoid synthesis to the introduced mevalonate pathway avoiding an abrupt shift upon deletion of lytB.
Deletion of lytB greatly increases the bioactivity for Vγ2Vδ2 T cells of aroA− S. enterica serovar Typhimurium SL7207 bacteria
Attenuated aroA− S. enterica serovar Typhimurium strain SL7207 bacteria had low levels of bioactivity for Vγ2Vδ2 T cells (Fig. 2A), similar to or lower than those measured in the W3110 laboratory strain of Escherichia coli (1). Deletion of the lytB gene in Salmonella greatly increased bioactivity for Vγ2Vδ2 T cells compared with the wild-type bacteria (Fig. 2A). Mutants with lytB deletion that were complemented with the mevalonate pathway cloned into the high-copy-number plasmid, (representative pTMV19K mutants are shown out of 19 mutants, Fig. 2A, left panel) produced high levels of bioactivity compared with the wild-type parent bacteria (mean values for the wild type bacteria [mean of seven lots] and for the 19 pTMV19K mutants are shown in the middle panel of Fig. 2A). Bacteria complemented with the low-copy-number plasmid (pMMV19K C10 and C22, Fig. 2A, right panel) also produced high levels of bioactivity. Similar to mycobacteria (53) and E. coli (1), a significant proportion of the bioactivity produced by the lytB deletion mutants was present in the supernatant (open portion of bars in Fig. 2). The levels of bioactivity in the pMMV19K C22 strain peaked in early stationary phase and decreased thereafter (Fig. 2C, right panels) whereas the bioactivity in W3110 Escherichia coli increased throughout growth and peaked in that late stationary phase of growth (Fig. 2B, left panels). The levels of HMBPP varied between different pTMV19K colonies and between different lots of the same colony likely reflecting differences in the bacterial growth phase, culture conditions, and perhaps epigenetic differences. This level of variation in bioactivity was also noted for E. coli (data not shown).
Normal growth and morphology of lytB deletion mutants of aroA− Salmonella SL7207 complemented with the pMMV19K low-copy-number plasmid but not with the pTMV19K high-copy-number plasmid
The introduction of the mevalonate pathway into E. coli can inhibit its growth due to the overproduction of HMG-CoA (49, 50). This metabolite is not normally present in E. coli and inhibits fatty acid biosynthesis, leading to generalized membrane stress (49). To assess the effect of lytB deletion and introduction of the mevalonate pathway on the Salmonella vaccine bacteria, their growth was compared with the wild-type parent. Although all mutant bacteria grew at similar rates to the wild-type parent bacteria during the logarithmic growth phase, the lytB deletion mutants complemented with the pTMV19K high-copy-number plasmid entered stationary phase at a lower density than the wild-type parent (~1.3–1.4 OD600 for mutants versus ~1.6 for the parent) (Fig. 3A, left panel). In contrast, one of the lytB deletion mutants (pMMV19K-C22) complemented with the pMMV19K low-copy-number plasmid showed identical growth as the wild-type parent while two other pMMV19K clones grew to slightly lower densities (Fig. 3A, right panel). The differences in the growth of the pTMV19K clones were reflected in the morphology of the bacteria. LytB− aroA− Salmonella SL7207 mutants complemented with the pTMV19K high-copy-number plasmid exhibited an elongated morphology with some bacteria >5 μm in length (Fig. 3B, middle panels) as compared to the short rod shape of the wild-type parent which were all <2.5 μm (Fig. 3B, left panel). LytB− aroA− S. enterica mutants complemented with the mevalonate pathway on the pMMV19K low-copy-number plasmid exhibited a similar morphology to the wild-type parent and were all <2.5 μm in length (Fig. 3B, right panel). Therefore, complementation of lytB deletion with the pTMV19K high-copy-number plasmid resulted in early entry in stationary phase and abnormal morphology whereas complementation with the pMMV19K low-copy-number plasmid resulted in mutants with similar growth and morphology to the parent strain.
Normal invasiveness and persistence in human HeLa cells of lytB− aroA− S. enterica serovar Typhimurium bacteria complemented with the pMMV19K low-copy-number plasmid but not with the pTMV19K high-copy-number plasmid
The ability of Salmonella bacteria to invade and persist or proliferate in human cells is a major determinant of their infectivity and virulence. To assess the effect of deleting lytB on this ability, we tested Salmonella mutants for their invasiveness and persistence in human epithelial HeLa tumor cells using the standard gentamicin resistance assay (46). Invasion by lytB− aroA− S. enterica serovar Typhimurium bacteria complemented with the high-copy-number plasmid was reduced 96% compared with wild-type aroA− S. enterica bacteria (Fig. 4, left upper panel). In contrast, invasion by the lytB− aroA− Salmonella bacteria complemented with the low-copy-number plasmid (pMMV19K-C1 and pMMV19K-C10) was identical to the wild-type parent (Fig. 4, right upper panel). Although lytB− aroA− Salmonella pTMV19K bacteria complemented with the high-copy-number plasmid invaded poorly, they were able to persist within HeLa cells (reduced 75% compared with wild-type bacteria) (Fig. 4, left lower panel) similar to lytB− aroA− Salmonella complemented with the low-copy-number plasmid or the wild-type parent (Fig. 4, right lower panel). Consistent with an earlier report (46), there was minimal proliferation of Salmonella serovar Typhimurium bacteria in HeLa cells. Thus, by using a low-copy-number plasmid, we were able to retain normal invasiveness and persistence of lytB− aroA− S. enterica serovar Typhimurium bacteria in human cells.
Ex vivo expansion of Vγ2Vδ2 T cells from human donors upon exposure to live lytB− aroA− Salmonella bacteria
To assess the ability of the Salmonella mutants to stimulate human Vγ2Vδ2 T cells in vivo, we exposed human PBMCs to live vaccine bacteria and measured the ex vivo expansion of Vγ2Vδ2 T cells. Direct infection of human PBMCs by the lytB− aroA− Salmonella bacteria, pMMV19k-C22 (complemented with the low-copy-number plasmid) stimulated expansion of Vγ2Vδ2 T cells (Fig. 5) similar to that observed with direct stimulation with HMBPP or zoledronate. Because many bacteria, including Salmonella, secrete HMBPP (Fig. 2 and Refs. 1, 53, 54), we tested the ability of the lytB− aroA− Salmonella bacteria to stimulate Vγ2Vδ2 T cells without direct contact by culturing the bacteria in the upper well of a Transwell where they were separated from the PBMCs by a 0.4 μm membrane. Despite this barrier, the lytB− aroA− Salmonella bacteria stimulated Vγ2Vδ2 T cell expansion albeit to a lesser degree than those in direct contract (Fig. 6). Thus, the vaccine bacteria were able to efficiently stimulate Vγ2Vδ2 T cell expansion ex vivo suggesting that they would be able to stimulate Vγ2Vδ2 T cells upon in vivo immunization.
Immunization of rhesus monkeys with lytB− aroA− Salmonella bacteria complemented with the pMMV19K low-copy-number plasmid expands Vγ2Vδ2 T cells while maintaining early memory subsets
To test the effectiveness of the lytB− aroA− Salmonella bacteria in vivo, we used rhesus monkeys as an animal model for human immunization. γδ T cells that can be stimulated by prenyl pyrophosphates are not present in mice, rats, and other small animals precluding their use. In contrast, rhesus monkeys and other nonhuman primates have reactivity to HMBPP, alkylamines, and aminobisphosphonates identical to humans (55). This reactivity is consistent with the high sequence conservation of their Vγ2Vδ2 TCRs with human Vγ2Vδ2 TCRs (55, 56). To assess the effectiveness of immunization, two parameters were followed: the percentage of Vγ2Vδ2 T cells among total CD3 T cells before and after immunization, and the proportions of the different memory Vγ2Vδ2 T cell subsets.
Memory Vγ2Vδ2 T cells can be divided into early central memory, CD27− early central memory, intermediate effector memory, and CD45RA+ late effector memory subsets based on their expression of CD28 and CD27 (C. Jin and C.T. Morita, unpublished observations and Ref. 57) similar to CD8 αβ T cells (58). Each subset has distinctive functional and migratory properties with the proportions of the subsets varying between individuals (C. Jin and C.T. Morita, unpublished observations and Ref. 57). Adoptive transfer of early central memory CD8 αβ T cells has shown that these cells are the most effective at establishing T cell memory on transfer (59) and are more effective than late effector memory T cells at mediating tumor immunity (60, 61). This is presumably due to the ability of early central memory T cells to proliferate allowing their amplification and persistence. For this reason, we followed the proportion of early central memory Vγ2Vδ2 T cells to assess the effect of immunization with Salmonella bacteria on these immune cells.
Immunization with the attenuated aroA− S. enterica serovar Typhimurium strain SL7207 parent bacteria was studied first. Unlike infection with pathogenic Salmonella (62, 63), immunization with the attenuated aroA− Salmonella bacteria did not stimulate significant expansion of Vγ2Vδ2 T cells (Fig. 7A, left panel) nor did it alter the proportions of the Vγ2Vδ2 T cell memory subsets (Fig. 7A, right panel). Because aroA− Salmonella bacteria did not stimulate Vγ2Vδ2 T cells in vivo, we next assessed the ability of lytB− aroA− Salmonella pTMV19K-C7 (complemented with a high-copy-number plasmid) to stimulate Vγ2Vδ2 T cell immunity. Although pTMV19K-C7 bacteria do not efficiently infect mammalian cells, they do produce high levels of HMBPP that could stimulate Vγ2Vδ2 T cell responses. To determine which effect predominated, a rhesus monkey was immunized with pTMV19K-C7 bacteria. Immunization did not increase Vγ2Vδ2 T cells (Fig. 7B, left panel) nor were there any changes in the proportion of Vγ2Vδ2 T cell memory subsets (Fig. 7B, right panel).
Given that the SL7207 and pTMV19K-C7 bacteria had minimal effects, lytB− aroA− Salmonella pMMV19K-C22 bacteria complemented with the low-copy-number plasmid were tested. Immunization with pMMV19K-C22 bacteria resulted in modest expansions of Vγ2Vδ2 T cells in two out of three monkeys. In monkey RQ5234, Vγ2Vδ2 T cells increased to 5.6% (2.3-fold) on primary immunization and to 4.6% of total T cells on secondary immunization (Fig. 7C, left upper panels). There were no significant changes in the memory subsets that accompanied these responses (Fig. 7C, right upper panels). Elevated numbers of Vγ2Vδ2 T cells persisted until the second immunization (96 days after the first immunization). A second monkey, RQ7125 (Fig. 7C, left middle panels), did not respond on primary immunization, but did respond on secondary immunization to 2.4%. Again, there were no significant changes in the Vγ2Vδ2 T cell memory subsets (Fig. 7C, right middle panels). A third monkey, RQ5264, did not respond to either immunization. Although the initial Vγ2Vδ2 T cell response by monkey RQ5234 did not have associated increases in the level of Vδ1 T cells, the Vγ2Vδ2 T cell responses to secondary immunization for both monkey RQ5234 and RQ7125 did have slight increases (Fig. 7C, left upper and middle panels). Moreover, total γδ T cell levels also increased significantly and these increases were more than the sum of the Vδ1 and Vδ2 T cells suggesting that γδ T cells expressing alternative V genes to Vδ1 and Vδ2 were present. The broad nature of the response suggests that production of T cell growth cytokines (IL-2 and IL-15) might have contributed to the response. Unlike immunization with bromohydrin pyrophosphate (BrHPP, Phosphostim) (64, 65) or zoledronate (Ref. 28 and data not shown), anergy was not observed after immunization with Salmonella pMMV19K-C22 bacteria as evidenced by the expansion of Vγ2Vδ2 T cells in response to HMBPP on in vitro stimulation of PBMCs from the three monkeys (Supplemental Fig. 2). In summary, complementation of lytB deletion with mevalonate pathway genes cloned into a low-copy-number plasmid preserved invasiveness and allowed for the in vivo stimulation of Vγ2Vδ2 T cells, albeit weakly. Metabolic engineering of attenuated aroA− Salmonella bacteria to overproduce HMBPP restored their ability to stimulate expansion of Vγ2Vδ2 T cells.
Discussion
Development of vaccines targeting Vγ2Vδ2 and other unconventional T cells has been hampered by the rapid development of T cell anergy and T cell deletion when the stimulators/Ags are directly administered. In this study, we have used metabolic engineering to derive a live Salmonella vaccine for Vγ2Vδ2 T cells. Depletion of the lytB gene resulted in the accumulation of its substrate, HMBPP, to high levels compared with the low levels found in the parent vaccine strain. Because this mutation is lethal, the loss of lytB was complemented with a plasmid expressing mevalonate pathway enzymes from Streptomyces. It was crucial to control the levels of the enzymes to minimize the production of toxic intermediates such as HMG-CoA, that caused early entry into stationary phase and that inhibited bacterial invasiveness for human cells. LytB− aroA− Salmonella complemented with the mevalonate pathway on a low-copy-number plasmid expanded human Vγ2Vδ2 T cells ex vivo. When tested in rhesus monkeys, immunization with the vaccine expanded Vγ2Vδ2 T cells with elevated levels of these cells persisting for at least 3 mo in one monkey. The levels of expansion were of lower magnitude compared to those noted during Salmonella-induced gastroenteritis in humans (10.8 ± 8.5% and 13.6 ± 9.0% in Refs. (62, and 63), respectively).
Immunization with live bacterial vaccines for Vγ2Vδ2 T cells has significant advantages over immunization with prenyl pyrophosphates or aminobisphosphonates. Although immunization with these compounds in conjunction with IL-2 can result in very high numbers of Vγ2Vδ2 T cells, these increases are generally transient (lasting <10–15 d) and responsiveness to further immunization wanes rapidly in most patients and in nonhuman primate models. For example, in patients with solid tumors, immunization with BrHPP without IL-2 gave no response. BrHPP immunization with IL-2 resulted in dose-dependent expansion of Vγ2Vδ2 T cells of up to 60-fold (27). However, the next two immunizations (administered at 21-d intervals) gave only 2- to 3-fold expansions followed by 0- to 1.6-fold expansions (27). Similar results were obtained with BrHPP/IL-2 immunization of cynomolgus monkeys (at 28- or 56-d intervals). There were large expansions on the first immunization with Vγ2Vδ2 T cells constituting up to 50–60% of circulating T cells followed by rapid loss of responsiveness such that the responses to the fourth immunization were <8% (64).
Aminobisphosphonates show a similar loss of responsiveness with repeated immunizations. In seven out of seven breast cancer patients repeatedly immununized with zoledronate there were either minimal increases or decreases in Vγ2Vδ2 T cells (66). Decreases in Vγ2Vδ2 T cells from baseline were observed in four out of the seven patients such that by the third zoledronate/IL-2 treatment, Vγ2Vδ2 T cell numbers were 20–40% of the starting levels (66). Similarly, a patient with renal cell carcinoma exhibited an initial expansion of Vγ2Vδ2 T cells to 10% but responsiveness waned such there were no further responses and Vγ2Vδ2 T cells levels decreased to 2% by the fourth immunization (26). In our studies in rhesus monkeys, the initial zoledronate/IL-2 immunization resulted in increases in Vγ2Vδ2 T cell numbers of 15.4- to 43.1-fold whereas the second immunization given 2 mo later resulted in expansions of only 1.2- to 8.3-fold from baseline. Immunization with HMBPP/IL-2 after a prolonged rest period (35 weeks) did little to restore responsiveness with expansions of 3- to 10-fold (data not shown). Thus, the loss of Vγ2Vδ2 T cell responsiveness with direct immunization with stimulating compounds is a major problem limiting the potential effectiveness of these cells in cancer immunotherapy.
Given these results, we pursued a different approach and have now developed a live bacterial vaccine that stimulates Vγ2Vδ2 T cells. We predicted that such a vaccine might be significantly better at stimulating Vγ2Vδ2 T cell immunity because it mimics the natural response of Vγ2Vδ2 T cells to infections. Vγ2Vδ2 T cells expand to large number in response to a number of different microbial infections (reviewed in Ref. 15). For example, in infections with Salmonella, γδ T cells increase from 4.5% to an average of 27.1% of total T cells in patients with typhoid/paratyphoid fever or to 12.35% for patients with serovars causing gastroenteritis (Fig. 8) (62, 63). Similar expansions are noted with infections with Listeria monocytogenes (67, 68) and with Brucella melitensis after ingestion of contaminated milk (69). Moreover, during tularemia infection (due to Francisella tularensis), circulating Vγ2Vδ2 T cells increase as early as 1 wk with levels 2 wks postinfection between 22 and 50% of the circulating T cells (70). These increases persist for >1 y (70). Similar expansions to bacterial infections are also noted in nonhuman primates. In collaborative studies performed by the Z. W. Chen lab, we found strong evidence that Vγ2Vδ2 T cells mount adaptive immune responses to mycobacterial infections. Whereas the initial infection of rhesus monkeys with Mycobacterium bovis bacille Calmette-Guérin (BCG) leads to Vγ2Vδ2 T cell expansions that peak on day 29, reinfection leads to earlier responses that peak on day 19 with higher numbers of Vγ2Vδ2 T cells (29). Similar recall responses are also observed with i.v. infection with attenuated L. monocytogenes (30). These results suggest that bacterial infections lead to the development of Vγ2Vδ2 T cell memory responses. The development of CD4 αβ T cell immunity to provide T cell help, the conversion of the remaining naive Vγ2Vδ2 T cells to memory Vγ2Vδ2 T cells, and the activation of the innate immune system probably accounts for the boost in the Vγ2Vδ2 T cell response.
In contrast to pathogenic bacterial infections, vaccination of humans with live attenuated bacterial vaccines in clinical use has not been found to expand Vγ2Vδ2 T cells. No expansion of circulating Vγ2Vδ2 T cells was found on immunization with F. tularensis LVS to prevent tularemia (70) and only slight increases in some monkeys on immunization with BCG to prevent tuberculosis (71) whereas virulent F. tularensis (70) and Mycobacterium tuberculosis (16, 29, 72) do expand Vγ2Vδ2 T cells on infection. Vγ2Vδ2 T cell expansions are also observed in rhesus monkeys in response to i.v. infection with attenuated L. monocytogenes ΔactA prf* but this vaccine is not licensed for human use and the i.v. route would not be amenable for mass immunizations (30). The effectiveness of Listeria may reflect the fact that attenuation in this strain is due to alterations in virulence rather than in metabolism and that the bacteria were given i.v., causing bacteremia. Although intradermal BCG immunization did not greatly expand circulating Vγ2Vδ2 T cells, it partially stimulates Vγ2Vδ2 T cell immunity as evidenced by increases in the in vitro responsiveness of blood Vγ2Vδ2 T cells to mycobacteria (71, 73, 74).
The inability of the licensed attenuated bacterial vaccines to expand Vγ2Vδ2 T cells likely reflects the fact that both F. tularensis LVS (70) and BCG (75) produce lower levels of stimulatory compounds (e.g., HMBPP) than do the comparable pathogenic bacteria. Similarly, we found that bioactivity for Vγ2Vδ2 T cells was very low in the attenuated aroA− S. enterica serovar Typhimurium SL7207 vaccine strain used in the present study (Fig. 2). Because we and others have found that the deletion of the lytB gene in E. coli greatly increases HMBPP levels (1, 2, 76), we decided to delete the lytB gene from Salmonella in an attempt to develop an attenuated bacterial vaccine that retained its ability to expand Vγ2Vδ2 T cells. The ability of lytB− aroA− Salmonella bacteria to expand Vγ2Vδ2 T cells in rhesus monkeys upon immunization demonstrates that the success of this strategy.
The use of attenuated Salmonella as the basis for this new Vγ2Vδ2 T cell vaccine was guided by the success of present Salmonella vaccines. Live Salmonella vaccines have been successfully developed for use in humans as well as cattle, chickens, and other farm animals. A variety of different attenuation mutations affecting pathogenicity or metabolism have been made to develop Salmonella as a platform to allow immunization against a number of different protein Ags to vaccinate against other bacteria, viruses, and cancer Ags (77–80). In this study, we used S. enterica serovar Typhimurium with an aroA deletion mutation that is deficient in the synthesis of aromatic amino acids and folic acid (81). Salmonella serovar Typhimurium that have deletions in aromatic acid biosynthesis alone or with other deletions had prolonged fecal shedding when given to some volunteers (82) (although not in rhesus monkeys (83)) whereas serovar Typhi caused silent bacteremia but only short periods of fecal shedding (84). Thus, for the development of a clinical Vγ2Vδ2 T cell vaccine, the lytB mutation might need to be made in a more attenuated Salmonella strain than SL7207.
An additional advantage of using a metabolically engineered Salmonella bacteria with a lytB deletion is that variants that lose production of HMBPP are unlikely to develop. The MEP pathway is essential in Salmonella as evidenced by the fact that lethal point mutations were found in all of the MEP pathway enzymes, including lytB (85), when S. enterica bacteria that had been transformed with a synthetic operon containing downstream yeast mevalonate pathway enzymes were chemically mutagenized. Additionally, we and others showed that deletion of any of the MEP pathway enzymes including lytB is lethal for the closely related species, E. coli (1, 86, 87). Importantly, although the loss of the first two enzymes, Dxs (also required for thiamine and pyrodoxine synthesis) and Dxr, can be rescued by mutations in other enzymes (88, 89), no compensatory mutations have been described for lytB deletion or for the deletion of ygbP, ychB, ygbB, and gcpE (90, 91). Moreover, there do not appear to be any alternative pathways for the production of IPP and DMAPP from earlier MEP pathway intermediates or from other metabolites, nor are there alternative enzymes with lytB enzymatic function that can circumvent the requirement for the lytB gene (91). Thus, our complementation of the lytB deletion with the mevalonate pathway is a form of balanced lethal mutation. However, instead of providing an essential gene with a protein Ag on an introduced plasmid, we are providing an essential metabolic pathway. Because the lytB mutation is a complete deletion of the gene, spontaneous reversion is not possible and, as detailed above, no compensatory mechanisms to circumvent the loss of lytB appear to exist.
Isoprenoid compounds (also termed terpenoids) are among the largest and most diverse group of natural organic compounds. Despite their variety and complexity, all of these compounds are derived from IPP and DMAPP precursors. Given the importance of many of these compounds as drugs, plant products, and fuels, efforts have been made to alter isoprenoid metabolism in bacteria by the introduction of new enzymes or other modifications to increase the overall metabolic flux through the isoprenoid pathway (33, 48, 92) and to allow for the synthesis of complex isoprenoid compounds (48, 93). However, to our knowledge, this is the first report using metabolic engineering to develop a vaccine for unconventional T cells.
Altering isoprenoid metabolism by the introduction of mevalonate pathway enzymes can lead to the overproduction of toxic metabolic intermediates. Isoprenoid metabolism is normally tightly regulated to avoid this but regulation can be lost with the introduction of heterologous enzymes as was noted in this study. When the mevalonate pathway was introduced into Salmonella using the pTMV19k high-copy-number plasmid, the bacteria entered stationary phase early and exhibited an abnormal elongated morphology. It also led to their inability to efficiently invade human cells and to expand monkey Vγ2Vδ2 T cells upon in vivo immunization. These defects are likely due to the overproduction of HMG-CoA given that the accumulation of this metabolite was found to correlate with decreased growth in E. coli engineered by the introduction of yeast mevalonate pathway genes (50). Overproduction of HMG-CoA inhibits fatty acid biosynthesis leading to generalized membrane stress and upregulation of bacterial stress response genes (49). We were able to overcome toxicity by using the low-copy-number pMW118 plasmid that is limited to fewer than five copies per bacteria. The similar use of low-copy-number plasmids allowed cloning of a mevalonate gene cluster from Streptomyces griseolosporeus (94) and improved isoprenoid product yields (51). Thus, regulation of isoprenoid biosynthesis is critical to restore sufficient virulence to the lytB− aroA− Salmonella for effective immunization.
What are the potential uses of a live S. enterica vaccine that stimulates Vγ2Vδ2 T cells? One use would be to stimulate Vγ2Vδ2 T cells for immunotherapy of tumors. A live Salmonella vaccine could be given to immunocompetent patients with cancer or as adjuvant therapy for patients after treatment or be used to expand Vγ2Vδ2 T cells ex vivo for use in adoptive immunotherapy. Finally, Salmonella preferentially accumulate in tumors in experiments in mice and is tumoricidal (95, 96), so therapy with attenuated Salmonella with deletion of lipid A to avoid septic shock (97) has been proposed. Although insufficient tumor colonization was noted in clinical trial (98), recent vaccines have engineered Salmonella to express antitumor Abs to target them directly to tumors (99). Similar treatment of a lytB mutant Salmonella to localize it to tumors could be used to attract adoptively transferred Vγ2Vδ2 T cells to tumors and activate them.
A second use would be to prime Vγ2Vδ2 T cell immunity in infants and adults to increase protection from bacterial and protozoan infections. Vγ2Vδ2 T cells play an important role in primate immunity to bacteria and apicomplexan parasites. Once primed, Vγ2Vδ2 T cells have the unique ability to mount memory responses to bacteria and protozoa unrelated to previous infections except for the production of the HMBPP metabolite in the MEP isoprenoid pathway. Vγ2Vδ2 T cells likely play important role in containing infections in their early phases thereby gaining time for the establishment of microbe-specific αβ T cell and B cell responses to provide sterilizing immunity. For example, in tuberculosis in nonhuman primates, stimulation of Vγ2Vδ2 T cells by an HMBPP analog decreased M. tuberculosis numbers in the lung by ~75–90% with less severe lung damage (100). In in vitro experiments, Vγ2Vδ2 T cells decrease intracellular numbers of M. tuberculosis (100, 101), M. bovis BCG (102, 103), L. monocytogenes (30), and Brucella suis (104, 105). Control of bacterial numbers can be through several mechanisms including perforin- (100), granzyme A- (103), or Fas/Fas ligand- (106) mediated lysis of the infected cell followed by direct killing of the bacteria by granulolysin (100, 101), through activation of infected monocytes by TNF-α (103) and IFN-γ (104), or by the production of antibacterial peptides such as the cathelicidin, LL-37 (105) or elafin (107). Besides reducing intracellular bacterial replication and killing infected cells, Vγ2Vδ2 T cells can also traffic to infected organs and release an array of inflammatory cytokines and chemokines while also producing growth factors to speed healing of epithelial and endothelial damage (reviewed in Refs. 15, 108, 109).
At birth in the U.S.A., we found that the Vγ2Vδ2 T cell subset is a minor subset making up only 9.4% of γδ T cells with as many Vγ2Vδ1 and Vγ1Vδ2 T cell as Vγ2Vδ2 T cells (110). Vδ1 T cells predominate by a two to one margin. Moreover, the γδ T cell population only constitutes 1.7% of total T cells. In contrast, in adults the Vγ2Vδ2 T cell population constitutes 51% of γδ T cells and γδ T cells constitute 4.8% of total T cells (110). This increase in γδ T cells is almost entirely due to the expansion of Vγ2Vδ2 T cells to ~4.5% of total T cells between ages 1 and 10 y, leading to their predominance among γδ T cells in adults (111). This expansion of Vγ2Vδ2 T cells is not present in every individual and is not inherited but is driven by environmental factors (111). Thus, identical twins can differ and there was no evidence in families for inheritance of the relative proportions of γδ V gene subsets (111). Sequence analysis of Vγ2Vδ2 TCRs (112) provides support for the influence of environmental factors because expansion of cord blood Vγ2Vδ2 T cells by stimulation with an aminobisphosphonate leads to the enrichment for adult-like Vγ2 chains supporting selection for efficient prenyl pyrophosphate stimulation of Vγ2Vδ2 T cells (113). Studies of rhesus monkeys provide further evidence that environmental factors shape the normal γδ T cell V gene repertoire. Unlike human adults but like human infants, adolescent and adult rhesus monkeys raised in captivity in specific pathogen-free colonies do not have Vγ2Vδ2 T cells as the predominant γδ T cell subset. Instead, Vδ1 T cells are the major subset constituting 32% of total γδ T cells whereas Vγ2Vδ2 T cells constitute only 24% (55). Thus, without proper environmental cues, normal development of Vγ2Vδ2 T cell immunity does not occur. A live Salmonella vaccine could be used to prime Vγ2Vδ2 T cell immunity in young infants rather than waiting for a natural infection that might cause illness or death. This might be especially useful in underdeveloped nations where bacterial gastroenteritis, tuberculosis, and malaria (caused by HMBPP-producing apicomplexan protozoa) are major causes of infant morbidity and mortality.
Although increases in Vγ2Vδ2 T cell levels were noted after immunization with Salmonella pMMV19K-C22 vaccine bacteria, these increases were relatively modest compared to the high levels that occur with certain infections (reviewed in Ref. 15). However, the Vγ2Vδ2 T cell levels achieved by one monkey (6.3% in monkey RQ5234) reached the mean levels noted in human infants when Vγ2Vδ2 T cells are expanding (111) suggesting that the vaccine response may be sufficient to prime Vγ2Vδ2 T cell immunity. One potential drawback was the variability in the response with one monkey not responding. This may reflect natural variability in the response given that even infections with virulent serovar Typhimurium bacteria do not uniformly induce Vγ2Vδ2 T cell responses (Fig. 8). Moreover, the levels found in the monkeys were similar to the values reported for patients with infections with other pathogenic S. enterica serovars that cause gastroenteritis (Fig. 8) (62, 63). Of 28 patients with gastroenteritis, 13 (46%) had γδ T cells <10% of total T cells with mean values of 12.4%. In contrast, systemic infection with serovars Typhi and Paratyphi had uniformly higher levels of γδ T cells (mean of 27.1%) (Fig. 8). Because serovar Typhi bacteria only infect great apes (Hominidae family), we chose to use an attenuated Typhimurium strain to allow direct testing in lower primates and virulence testing in mice. However, based on the results presented in the present study and the differences in Vγ2Vδ2 T cells in patients infected with different Salmonella serovars, it is likely that a lytB deletion mutant in an attenuated Typhi serovar strain would induce higher Vγ2Vδ2 T cell expansions. This will be the focus of our future vaccine efforts because the responses induced by the pMMV19K-C22 serovar Typhimurium vaccine are likely to be too variable to be of significant benefit. Our findings further suggest that systemic bacterial and protozoan parasite infections are the most effective at expanding Vγ2Vδ2 T cells compared to local infections in the mucosa or skin. This is consistent with the presence of resting Vγ2Vδ2 T cells primarily in central lymphoid organs and in the blood rather than being concentrated in the intestinal mucosa or skin (114). Thus, initial activation of Vγ2Vδ2 T cells likely occurs in the spleen and lymph nodes. After expansion, Vγ2Vδ2 T cells can then migrate in high numbers to peripheral sites to provide local immunity (16, 29, 30).
In conclusion, genetic engineering of vaccine bacteria has focused on the deletion of virulence genes, on the deletion of genes important in bacterial metabolism to decrease virulence and/or survival, or on the introduction of heterologous proteins or protein fragments to induce immune responses to other microbes or to cancer cells. However, we now show that genetic engineering can also be used to alter the metabolism of vaccine bacteria to increase production of nonpeptide compounds that are stimulators/Ags for unconventional T cells. A similar approach could be used to develop vaccines for other unconventional T cells. αβ iNKT cells recognize specific bacterial lipids presented by CD1d that are produced by Sphingomonas spp. (115) and Borrelia burgdorferi (116). The enzymes required to produce a stimulatory glycolipid could be introduced into attenuated Salmonella to allow them to stimulate iNKT cells. Metabolic engineering of B vitamin synthetic pathways could similarly be performed to increase the production of the metabolites (117, 118) that are recognized by αβ mucosal-associated invariant T cells (119, 120), thereby increasing the ability of Salmonella vaccine bacteria to stimulate mucosal-associated invariant T cell immunity. Altering the polysaccharides of vaccine bacteria could also be used to stimulate antibody formation against unique microbial carbohydrate structures (121). Finally, introduction of pathways involved in the synthesis of TLR ligands or ligands for other innate receptors could be used to increase the immunogenicity of the bacteria without increasing their virulence. Altering the metabolism of bacteria to produce nonpeptide stimulators/Ags for T cells or nonpeptide ligands for innate receptors could be a powerful approach for developing vaccine to stimulate innate T lymphocyte immunity without inducing anergy.
Supplementary Material
Acknowledgments
This work was supported by Department of Veterans Affairs (Veterans Health Administration, Office of Research and Development, Biomedical Laboratory Research and Development) Grant 1 I01 BX000972-01A1 (to C.T.M.), National Cancer Institute Grants CA097274 (University of Iowa/Mayo Clinic Lymphoma Specialized Program of Research Excellence) and P30CA086862 (Core Support) (to C.T.M.), National Institute of Allergy and Infectious Diseases Grants AI057160 (Midwest Regional Center of Excellence for Biodefense and Emerging Infectious Diseases Research) (to C.T.M. and B.D.J.) and 2PO1 AI044464-2 (to B.D.J.), and Bill and Melinda Gates Foundation Exploration Grant (to C.T.M.). M.H.N. was supported in part by the Higher Committee for Education Development in Iraq and National Institutes of Health Grant 5T32 AI007485. The content of this manuscript are solely the responsibility of the authors and do not necessarily represent the official views of the granting agencies.
We thank Zhimei Fang and Amy Raker for technical assistance. We thank Dr. Toshirou Hara for providing additional data on γδ T cell levels in Salmonella patients. We thank Dr. Roy Curtiss III for helpful discussion.
Abbreviations used in this article
- BCG
bacillus Calmette-Guérin
- BrHPP
bromohydrin pyrophosphate
- DMAPP
dimethylallyl pyrophosphate (diphosphate)
- HMBPP
(E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (diphosphate)
- HMG-CoA
3-hydroxy-3-methylglutaryl-CoA
- iNKT
invariant NKT
- IPP
isopentenyl pyrophosphate (diphosphate)
- kmr
kanomycin resistance gene
- LB
Luria-Bertani
- MEP
2-C-methyl-D-erythritol-4 phosphate
- MOI
multiplicity of infection
- OD600
OD at 600 nm
Footnotes
The online version of this article contains supplemental material.
Disclosures
C.T.M. and B.D.J. are co-inventors of US Patent 8,012,466 on the development of live bacterial vaccines for activating γδ T cells. The other authors declare no financial or commercial conflict of interest.
References
- 1.Puan KJ, Jin C, Wang H, Sarikonda G, Raker AM, Lee HK, Samuelson MI, Märker-Hermann E, Pasa-Tolic L, Nieves E, Giner JL, Kuzuyama T, Morita CT. Preferential recognition of a microbial metabolite by human Vγ2Vδ2 T cells. Int Immunol. 2007;19:657–673. doi: 10.1093/intimm/dxm031. [DOI] [PubMed] [Google Scholar]
- 2.Hintz M, Reichenberg A, Altincicek B, Bahr U, Gschwind RM, Kollas AK, Beck E, Wiesner J, Eberl M, Jomaa H. Identification of (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate as a major activator for human γδ T cells in Escherichia coli. FEBS Lett. 2001;509:317–322. doi: 10.1016/s0014-5793(01)03191-x. [DOI] [PubMed] [Google Scholar]
- 3.Tanaka Y, Morita CT, Tanaka Y, Nieves E, Brenner MB, Bloom BR. Natural and synthetic non-peptide antigens recognized by human γδ T cells. Nature. 1995;375:155–158. doi: 10.1038/375155a0. [DOI] [PubMed] [Google Scholar]
- 4.Kunzmann V, Bauer E, Wilhelm M. γ/δ T-cell stimulation by pamidronate. N Engl J Med. 1999;340:737–738. doi: 10.1056/NEJM199903043400914. [DOI] [PubMed] [Google Scholar]
- 5.Sanders JM, Ghosh S, Chan JMW, Meints G, Wang H, Raker AM, Song Y, Colantino A, Burzynska A, Kafarski P, Morita CT, Oldfield E. Quantitative structure-activity relationships for γδ T cell activation by bisphosphonates. J Med Chem. 2004;47:375–384. doi: 10.1021/jm0303709. [DOI] [PubMed] [Google Scholar]
- 6.Bukowski JF, Morita CT, Brenner MB. Human γδ T cells recognize alkylamines derived from microbes, edible plants, and tea: implications for innate immunity. Immunity. 1999;11:57–65. doi: 10.1016/s1074-7613(00)80081-3. [DOI] [PubMed] [Google Scholar]
- 7.Gober HJ, Kistowska M, Angman L, Jenö P, Mori L, De Libero G. Human T cell receptor γδ cells recognize endogenous mevalonate metabolites in tumor cells. J Exp Med. 2003;197:163–168. doi: 10.1084/jem.20021500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Thompson K, Rogers MJ. Statins prevent bisphosphonate-induced γ,δ-T-cell proliferation and activation in vitro. J Bone Miner Res. 2004;19:278–288. doi: 10.1359/JBMR.0301230. [DOI] [PubMed] [Google Scholar]
- 9.Thompson K, Rojas-Navea J, Rogers MJ. Alkylamines cause Vγ9Vδ2 T-cell activation and proliferation by inhibiting the mevalonate pathway. Blood. 2006;107:651–654. doi: 10.1182/blood-2005-03-1025. [DOI] [PubMed] [Google Scholar]
- 10.Wang H, Sarikonda G, Puan KJ, Tanaka Y, Feng J, Giner JL, Cao R, Mönkkönen J, Oldfield E, Morita CT. Indirect stimulation of human Vγ2Vδ2 T cells through alterations in isoprenoid metabolism. J Immunol. 2011;187:5099–5113. doi: 10.4049/jimmunol.1002697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wang H, Henry O, Distefano MD, Wang YC, Räikkönen J, Mönkkönen J, Tanaka Y, Morita CT. Butyrophilin 3A1 plays an essential role in prenyl pyrophosphate stimulation of human Vγ2Vδ2 T cells. J Immunol. 2013;191:1029–1042. doi: 10.4049/jimmunol.1300658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Palakodeti A, Sandstrom A, Sundaresan L, Harly C, Nedellec S, Olive D, Scotet E, Bonneville M, Adams EJ. The molecular basis for modulation of human Vγ9Vδ2 T cell responses by CD277/butyrophilin-3 (BTN3A)-specific antibodies. J Biol Chem. 2012;287:32780–32790. doi: 10.1074/jbc.M112.384354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Harly C, Guillaume Y, Nedellec S, Peigné CM, Mönkkönen H, Mönkkönen J, Li J, Kuball J, Adams EJ, Netzer S, Déchanet-Merville J, Léger A, Herrmann T, Breathnach R, Olive D, Bonneville M, Scotet E. Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human γδ T-cell subset. Blood. 2012;120:2269–2279. doi: 10.1182/blood-2012-05-430470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Vavassori S, Kumar A, Wan GS, Ramanjaneyulu GS, Cavallari M, El Daker S, Beddoe T, Theodossis A, Williams NK, Gostick E, Price DA, Soudamini DU, Voon KK, Olivo M, Rossjohn J, Mori L, De Libero G. Butyrophilin 3A1 binds phosphorylated antigens and stimulates human γδ T cells. Nat Immunol. 2013;14:908–916. doi: 10.1038/ni.2665. [DOI] [PubMed] [Google Scholar]
- 15.Morita CT, Jin C, Sarikonda G, Wang H. Nonpeptide antigens, presentation mechanisms, and immunological memory of human Vγ2Vδ2 T cells: discriminating friend from foe through the recognition of prenyl pyrophosphate antigens. Immunol Rev. 2007;215:59–76. doi: 10.1111/j.1600-065X.2006.00479.x. [DOI] [PubMed] [Google Scholar]
- 16.Huang D, Shen Y, Qiu L, Chen CY, Shen L, Estep J, Hunt R, Vasconcelos D, Du G, Aye P, Lackner AA, Larsen MH, Jacobs WR, Jr, Haynes BF, Letvin NL, Chen ZW. Immune distribution and localization of phosphoantigen-specific Vγ2Vδ2 T cells in lymphoid and nonlymphoid tissues in Mycobacterium tuberculosis infection. Infect Immun. 2008;76:426–436. doi: 10.1128/IAI.01008-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ali Z, Shao L, Halliday L, Reichenberg A, Hintz M, Jomaa H, Chen ZW. Prolonged (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate-driven antimicrobial and cytotoxic responses of pulmonary and systemic Vγ2Vδ2 T cells in macaques. J Immunol. 2007;179:8287–8296. doi: 10.4049/jimmunol.179.12.8287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Workalemahu G, Foerster M, Kroegel C. Expression and synthesis of fibroblast growth factor-9 in human γδ T-lymphocytes. Response to isopentenyl pyrophosphate and TGF-β1/IL-15. J Leukoc Biol. 2004;75:657–663. doi: 10.1189/jlb.0902471. [DOI] [PubMed] [Google Scholar]
- 19.Gertner-Dardenne J, Bonnafous C, Bezombes C, Capietto AH, Scaglione V, Ingoure S, Cendron D, Gross E, Lepage JF, Quillet-Mary A, Ysebaert L, Laurent G, Sicard H, Fournié JJ. Bromohydrin pyrophosphate enhances antibody-dependent cell-mediated cytotoxicity induced by therapeutic antibodies. Blood. 2009;113:4875–4884. doi: 10.1182/blood-2008-08-172296. [DOI] [PubMed] [Google Scholar]
- 20.Malkovska V, Cigel F, Storer BE. Human T cells in hu-PBL-SCID mice proliferate in response to Daudi lymphoma and confer anti-tumour immunity. Clin Exp Immunol. 1994;96:158–165. doi: 10.1111/j.1365-2249.1994.tb06246.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kabelitz D, Wesch D, Pitters E, Zöller M. Characterization of tumor reactivity of human Vγ9Vδ2 γδ T cells in vitro and in SCID mice in vivo. J Immunol. 2004;173:6767–6776. doi: 10.4049/jimmunol.173.11.6767. [DOI] [PubMed] [Google Scholar]
- 22.Benzaïd I, Mönkkönen H, Stresing V, Bonnelye E, Green J, Mönkkönen J, Touraine JL, Clézardin P. High phosphoantigen levels in bisphosphonate-treated human breast tumors promote Vγ9Vδ2 T-cell chemotaxis and cytotoxicity in vivo. Cancer Res. 2011;71:4562–4572. doi: 10.1158/0008-5472.CAN-10-3862. [DOI] [PubMed] [Google Scholar]
- 23.D’Asaro M, La Mendola C, Di Liberto D, Orlando V, Todaro M, Spina M, Guggino G, Meraviglia S, Caccamo N, Messina A, Salerno A, Di Raimondo F, Vigneri P, Stassi G, Fournié JJ, Dieli F. Vγ9Vδ2 T lymphocytes efficiently recognize and kill zoledronate-sensitized, imatinib-sensitive, and imatinib-resistant chronic myelogenous leukemia cells. J Immunol. 2010;184:3260–3268. doi: 10.4049/jimmunol.0903454. [DOI] [PubMed] [Google Scholar]
- 24.Wilhelm M, Kunzmann V, Eckstein S, Reimer P, Weissinger F, Ruediger T, Tony H-P. γδ T cells for immune therapy of patients with lymphoid malignancies. Blood. 2003;102:200–206. doi: 10.1182/blood-2002-12-3665. [DOI] [PubMed] [Google Scholar]
- 25.Dieli F, Vermijlen D, Fulfaro F, Caccamo N, Meraviglia S, Cicero G, Roberts A, Buccheri S, D’Asaro M, Gebbia N, Salerno A, Eberl M, Hayday AC. Targeting human γδ T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res. 2007;67:7450–7457. doi: 10.1158/0008-5472.CAN-07-0199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lang JM, Kaikobad MR, Wallace M, Staab MJ, Horvath DL, Wilding G, Liu G, Eickhoff JC, McNeel DG, Malkovsky M. Pilot trial of interleukin-2 and zoledronic acid to augment γδ T cells as treatment for patients with refractory renal cell carcinoma. Cancer Immunol Immunother. 2011;60:1447–1460. doi: 10.1007/s00262-011-1049-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bennouna J, Levy V, Sicard H, Senellart H, Audrain M, Hiret S, Rolland F, Bruzzoni-Giovanelli H, Rimbert M, Galéa C, Tiollier J, Calvo F. Phase I study of bromohydrin pyrophosphate (BrHPP, IPH 1101), a Vγ9Vδ2 T lymphocyte agonist in patients with solid tumors. Cancer Immunol Immunother. 2010;59:1521–1530. doi: 10.1007/s00262-010-0879-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sugie T, Murata-Hirai K, Iwasaki M, Morita CT, Li W, Okamura H, Minato N, Toi M, Tanaka Y. Zoledronic acid-induced expansion of γδ T cells from early-stage breast cancer patients: effect of IL-18 on helper NK cells. Cancer Immunol Immunother. 2013;62:677–687. doi: 10.1007/s00262-012-1368-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shen Y, Zhou D, Qiu L, Lai X, Simon M, Shen L, Kou Z, Wang Q, Jiang L, Estep J, Hunt R, Clagett M, Sehgal PK, Li Y, Zeng X, Morita CT, Brenner MB, Letvin NL, Chen ZW. Adaptive immune response of Vγ2Vδ2+ T cells during mycobacterial infections. Science. 2002;295:2255–2258. doi: 10.1126/science.1068819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ryan-Payseur B, Frencher J, Shen L, Chen CY, Huang D, Chen ZW. Multieffector-functional immune responses of HMBPP-specific Vγ2Vδ2 T cells in nonhuman primates inoculated with Listeria monocytogenes ΔactA prfA*. J Immunol. 2012;189:1285–1293. doi: 10.4049/jimmunol.1200641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Keasling JD. Synthetic biology and the development of tools for metabolic engineering. Metab Eng. 2012;14:189–195. doi: 10.1016/j.ymben.2012.01.004. [DOI] [PubMed] [Google Scholar]
- 32.Immethun CM, Hoynes-O’Connor AG, Balassy A, Moon TS. Microbial production of isoprenoids enabled by synthetic biology. Front Microbiol. 2013;4:Article 75. doi: 10.3389/fmicb.2013.00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ajikumar PK, Tyo K, Carlsen S, Mucha O, Phon TH, Stephanopoulos G. Terpenoids: opportunities for biosynthesis of natural product drugs using engineered microorganisms. Mol Pharm. 2008;5:167–190. doi: 10.1021/mp700151b. [DOI] [PubMed] [Google Scholar]
- 34.Cryz SJ, Jr, Furer E, Levine MM. Effectiveness of oral, attenuated live Salmonella typhi Ty 21a vaccine in controlled field trials. Schweiz Med Wochenschr. 1988;118:467–470. [PubMed] [Google Scholar]
- 35.Wahdan MH, Sérié C, Cerisier Y, Sallam S, Germanier R. A controlled field trial of live Salmonella typhi strain Ty 21a oral vaccine against typhoid: three-year results. J Infect Dis. 1982;145:292–295. doi: 10.1093/infdis/145.3.292. [DOI] [PubMed] [Google Scholar]
- 36.Anwar E, Goldberg E, Fraser A, Acosta CJ, Paul M, Leibovici L. Vaccines for preventing typhoid fever. Cochrane Database Syst Rev. 2014;1:CD001261. doi: 10.1002/14651858.CD001261.pub3. [DOI] [PubMed] [Google Scholar]
- 37.Majowicz SE, Musto J, Scallan E, Angulo FJ, Kirk M, O’Brien SJ, Jones TF, Fazil A, Hoekstra RM International Collaboration on Enteric Disease ‘Burden of Illness’ Studies. . The global burden of nontyphoidal Salmonella gastroenteritis. Clin Infect Dis. 2010;50:882–889. doi: 10.1086/650733. [DOI] [PubMed] [Google Scholar]
- 38.MacLennan CA, Levine MM. Invasive nontyphoidal Salmonella disease in Africa: current status. Expert Rev Anti Infect Ther. 2013;11:443–446. doi: 10.1586/eri.13.27. [DOI] [PubMed] [Google Scholar]
- 39.Feasey NA, Dougan G, Kingsley RA, Heyderman RS, Gordon MA. Invasive non-typhoidal salmonella disease: an emerging and neglected tropical disease in Africa. Lancet. 2012;379:2489–2499. doi: 10.1016/S0140-6736(11)61752-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Takagi M, Kuzuyama T, Takahashi S, Seto H. A gene cluster for the mevalonate pathway from Streptomyces sp. strain CL190. J Bacteriol. 2000;182:4153–4157. doi: 10.1128/jb.182.15.4153-4157.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kaneda K, Kuzuyama T, Takagi M, Hayakawa Y, Seto H. An unusual isopentenyl diphosphate isomerase found in the mevalonate pathway gene cluster from Streptomyces sp. strain CL190. Proc Natl Acad Sci USA. 2001;98:932–937. doi: 10.1073/pnas.020472198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Stark MJR. Multicopy expression vectors carrying the lac repressor gene for regulated high-level expression of genes in Escherichia coli. Gene. 1987;51:255–267. doi: 10.1016/0378-1119(87)90314-3. [DOI] [PubMed] [Google Scholar]
- 43.Cohen SN, Chang AC, Boyer HW, Helling RB. Construction of biologically functional bacterial plasmids in vitro. Proc Natl Acad Sci USA. 1973;70:3240–3244. doi: 10.1073/pnas.70.11.3240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Datsenko KA, Wanner BL. One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci USA. 2000;97:6640–6645. doi: 10.1073/pnas.120163297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Puan KJ, Wang H, Dairi T, Kuzuyama T, Morita CT. fldA is an essential gene required in the 2-C-methyl-D-erythritol 4-phosphate pathway for isoprenoid biosynthesis. FEBS Lett. 2005;579:3802–3806. doi: 10.1016/j.febslet.2005.05.047. [DOI] [PubMed] [Google Scholar]
- 46.Elsinghorst EA. Measurement of invasion by gentamicin resistance. Methods Enzymol. 1994;236:405–420. doi: 10.1016/0076-6879(94)36030-8. [DOI] [PubMed] [Google Scholar]
- 47.Sarikonda G, Wang H, Puan KJ, Liu XH, Lee HK, Song Y, Distefano MD, Oldfield E, Prestwich GD, Morita CT. Photoaffinity antigens for human γδ T cells. J Immunol. 2008;181:7738–7750. doi: 10.4049/jimmunol.181.11.7738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Martin VJ, Pitera DJ, Withers ST, Newman JD, Keasling JD. Engineering a mevalonate pathway in Escherichia coli for production of terpenoids. Nat Biotechnol. 2003;21:796–802. doi: 10.1038/nbt833. [DOI] [PubMed] [Google Scholar]
- 49.Kizer L, Pitera DJ, Pfleger BF, Keasling JD. Application of functional genomics to pathway optimization for increased isoprenoid production. Appl Environ Microbiol. 2008;74:3229–3241. doi: 10.1128/AEM.02750-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pitera DJ, Paddon CJ, Newman JD, Keasling JD. Balancing a heterologous mevalonate pathway for improved isoprenoid production in Escherichia coli. Metab Eng. 2007;9:193–207. doi: 10.1016/j.ymben.2006.11.002. [DOI] [PubMed] [Google Scholar]
- 51.Jones KL, Kim SW, Keasling JD. Low-copy plasmids can perform as well as or better than high-copy plasmids for metabolic engineering of bacteria. Metab Eng. 2000;2:328–338. doi: 10.1006/mben.2000.0161. [DOI] [PubMed] [Google Scholar]
- 52.Steinbacher S, Kaiser J, Eisenreich W, Huber R, Bacher A, Rohdich F. Structural basis of fosmidomycin action revealed by the complex with 2-C-methyl-D-erythritol 4-phosphate synthase (IspC). Implications for the catalytic mechanism and anti-malaria drug development. J Biol Chem. 2003;278:18401–18407. doi: 10.1074/jbc.M300993200. [DOI] [PubMed] [Google Scholar]
- 53.Morita CT, Beckman EM, Bukowski JF, Tanaka Y, Band H, Bloom BR, Golan DE, Brenner MB. Direct presentation of nonpeptide prenyl pyrophosphate antigens to human γδ T cells. Immunity. 1995;3:495–507. doi: 10.1016/1074-7613(95)90178-7. [DOI] [PubMed] [Google Scholar]
- 54.Poquet Y, Constant P, Halary F, Peyrat MA, Gilleron M, Davodeau F, Bonneville M, Fournié JJ. A novel nucleotide-containing antigen for human blood γδ T lymphocytes. Eur J Immunol. 1996;26:2344–2349. doi: 10.1002/eji.1830261011. [DOI] [PubMed] [Google Scholar]
- 55.Wang H, Lee HK, Bukowski JF, Li H, Mariuzza RA, Chen ZW, Nam KH, Morita CT. Conservation of nonpeptide antigen recognition by rhesus monkey Vγ2Vδ2 T cells. J Immunol. 2003;170:3696–3706. doi: 10.4049/jimmunol.170.7.3696. [DOI] [PubMed] [Google Scholar]
- 56.Wang H, Fang Z, Morita CT. Vγ2Vδ2 T cell receptor recognition of prenyl pyrophosphates is dependent on all CDRs. J Immunol. 2010;184:6209–6222. doi: 10.4049/jimmunol.1000231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Puan KJ, Low JSH, Tan TWK, Wee JTS, Tan EH, Fong KW, Chua ET, Jin C, Giner JL, Morita CT, Goh CHK, Hui KM. Phenotypic and functional alterations of Vγ2Vδ2 T cell subsets in patients with active nasopharyngeal carcinoma. Cancer Immunol Immunother. 2009;58:1095–1107. doi: 10.1007/s00262-008-0629-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Appay V, van Lier RAW, Sallusto F, Roederer M. Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytometry A. 2008;73:975–983. doi: 10.1002/cyto.a.20643. [DOI] [PubMed] [Google Scholar]
- 59.Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305. doi: 10.1172/JCI32103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Klebanoff CA, Gattinoni L, Torabi-Parizi P, Kerstann K, Cardones AR, Finkelstein SE, Palmer DC, Antony PA, Hwang ST, Rosenberg SA, Waldmann TA, Restifo NP. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc Natl Acad Sci USA. 2005;102:9571–9576. doi: 10.1073/pnas.0503726102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Klebanoff CA, Gattinoni L, Palmer DC, Muranski P, Ji Y, Hinrichs CS, Borman ZA, Kerkar SP, Scott CD, Finkelstein SE, Rosenberg SA, Restifo NP. Determinants of successful CD8+ T-cell adoptive immunotherapy for large established tumors in mice. Clin Cancer Res. 2011;17:5343–5352. doi: 10.1158/1078-0432.CCR-11-0503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hara T, Mizuno Y, Takaki K, Takada H, Akeda H, Aoki T, Nagata M, Ueda K, Matsuzaki G, Yoshikai Y, Nomoto K. Predominant activation and expansion of Vγ9-bearing γδ T cells in vivo as well as in vitro in Salmonella infection. J Clin Invest. 1992;90:204–210. doi: 10.1172/JCI115837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hoshina T, Kusuhara K, Saito M, Mizuno Y, Hara T. NKRP1A+ γδ and αβ T cells are preferentially induced in patients with Salmonella infection. Hum Immunol. 2012;73:623–628. doi: 10.1016/j.humimm.2012.04.006. [DOI] [PubMed] [Google Scholar]
- 64.Sicard H, Ingoure S, Luciani B, Serraz C, Fournié JJ, Bonneville M, Tiollier J, Romagné F. In vivo immunomanipulation of Vγ9Vδ2 T cells with a synthetic phosphoantigen in a preclinical nonhuman primate model. J Immunol. 2005;175:5471–5480. doi: 10.4049/jimmunol.175.8.5471. [DOI] [PubMed] [Google Scholar]
- 65.Laurent G. A phase I/II open label study of IPH1101 (with low dose of IL-2) in combination with rituximab re-treatment in patients with follicular lymphoma. European Cancer Organization/European Society for Medical Oncology Congress; Berlin, Germany. September 20–24.2009. [Google Scholar]
- 66.Meraviglia S, Eberl M, Vermijlen D, Todaro M, Buccheri S, Cicero G, La Mendola C, Guggino G, D’Asaro M, Orlando V, Scarpa F, Roberts A, Caccamo N, Stassi G, Dieli F, Hayday AC. In vivo manipulation of Vγ9Vδ2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients. Clin Exp Immunol. 2010;161:290–297. doi: 10.1111/j.1365-2249.2010.04167.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Jouen-Beades F, Paris E, Dieulois C, Lemeland JF, Barre-Dezelus V, Marret S, Humbert G, Leroy J, Tron F. In vivo and in vitro activation and expansion of γδ T cells during Listeria monocytogenes infection in humans. Infect Immun. 1997;65:4267–4272. doi: 10.1128/iai.65.10.4267-4272.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bertotto A, Spinozzi F, Gerli R, Bassotti G, Forenza N, Vagliasindi C, Vaccaro R. Peripheral blood γδ T cells in human listeriosis. Acta Paediatr. 1995;84:1434–1435. doi: 10.1111/j.1651-2227.1995.tb13584.x. [DOI] [PubMed] [Google Scholar]
- 69.Bertotto A, Gerli R, Spinozzi F, Muscat C, Scalise F, Castellucci G, Sposito M, Candio F, Vaccaro R. Lymphocytes bearing the γδ T cell receptor in acute Brucella melitensis infection. Eur J Immunol. 1993;23:1177–1180. doi: 10.1002/eji.1830230531. [DOI] [PubMed] [Google Scholar]
- 70.Poquet Y, Kroca M, Halary F, Stenmark S, Peyrat MA, Bonneville M, Fournié JJ, Sjöstedt A. Expansion of Vγ9Vδ2 T cells is triggered by Francisella tularensis-derived phosphoantigens in tularemia but not after tularemia vaccination. Infect Immun. 1998;66:2107–2114. doi: 10.1128/iai.66.5.2107-2114.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Cairo C, Hebbeler AM, Propp N, Bryant JL, Colizzi V, Pauza CD. Innate-like γδ T cell responses to mycobacterium Bacille Calmette-Guerin using the public Vγ2 repertoire in Macaca fascicularis. Tuberculosis (Edinb) 2007;87:373–383. doi: 10.1016/j.tube.2006.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Balbi B, Valle MT, Oddera S, Giunti D, Manca F, Rossi GA, Allegra L. T-lymphocytes with γδ+ Vδ2+ antigen receptors are present in increased proportions in a fraction of patients with tuberculosis or with sarcoidosis. Am Rev Respir Dis. 1993;148:1685–1690. doi: 10.1164/ajrccm/148.6_Pt_1.1685. [DOI] [PubMed] [Google Scholar]
- 73.Hoft DF, Brown RM, Roodman ST. Bacille Calmette-Guérin vaccination enhances human γδ T cell responsiveness to mycobacteria suggestive of a memory-like phenotype. J Immunol. 1998;161:1045–1054. [PubMed] [Google Scholar]
- 74.Mazzola TN, Da Silva MTN, Moreno YMF, Lima SCBS, Carniel EF, Morcillo AM, Antonio MARGM, Zanolli ML, Aranha Netto A, Blotta MH, Raw I, Vilela MMS. Robust γδ+ T cell expansion in infants immunized at birth with BCG vaccine. Vaccine. 2007;25:6313–6320. doi: 10.1016/j.vaccine.2007.06.039. [DOI] [PubMed] [Google Scholar]
- 75.Constant P, Poquet Y, Peyrat MA, Davodeau F, Bonneville M, Fournié JJ. The antituberculous Mycobacterium bovis BCG vaccine is an attenuated mycobacterial producer of phosphorylated nonpeptidic antigens for human γδ T cells. Infect Immun. 1995;63:4628–4633. doi: 10.1128/iai.63.12.4628-4633.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Eberl M, Altincicek B, Kollas AK, Sanderbrand S, Bahr U, Reichenberg A, Beck E, Foster D, Wiesner J, Hintz M, Jomaa H. Accumulation of a potent γδ T-cell stimulator after deletion of the lytB gene in Escherichia coli. Immunology. 2002;106:200–211. doi: 10.1046/j.1365-2567.2002.01414.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Wang S, Kong Q, Curtiss R., III New technologies in developing recombinant attenuated Salmonella vaccine vectors. Microb Pathog. 2013;58:17–28. doi: 10.1016/j.micpath.2012.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kong W, Clark-Curtiss J, Curtiss R., III Utilizing Salmonella for antigen delivery: the aims and benefits of bacterial delivered vaccination. Expert Rev Vaccines. 2013;12:345–347. doi: 10.1586/erv.13.7. [DOI] [PubMed] [Google Scholar]
- 79.Tennant SM, Wang JY, Galen JE, Simon R, Pasetti MF, Gat O, Levine MM. Engineering and preclinical evaluation of attenuated nontyphoidal Salmonella strains serving as live oral vaccines and as reagent strains. Infect Immun. 2011;79:4175–4185. doi: 10.1128/IAI.05278-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Galen JE, Wang JY, Chinchilla M, Vindurampulle C, Vogel JE, Levy H, Blackwelder WC, Pasetti MF, Levine MM. A new generation of stable, nonantibiotic, low-copy-number plasmids improves immune responses to foreign antigens in Salmonella enterica serovar Typhi live vectors. Infect Immun. 2010;78:337–347. doi: 10.1128/IAI.00916-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hoiseth SK, Stocker BA. Aromatic-dependent Salmonella typhimurium are non-virulent and effective as live vaccines. Nature. 1981;291:238–239. doi: 10.1038/291238a0. [DOI] [PubMed] [Google Scholar]
- 82.Hindle Z, Chatfield SN, Phillimore J, Bentley M, Johnson J, Cosgrove CA, Ghaem-Maghami M, Sexton A, Khan M, Brennan FR, Everest P, Wu T, Pickard D, Holden DW, Dougan G, Griffin GE, House D, Santangelo JD, Khan SA, Shea JE, Feldman RG, Lewis DJM. Characterization of Salmonella enterica derivatives harboring defined aroC and Salmonella pathogenicity island 2 type III secretion system (ssaV) mutations by immunization of healthy volunteers. Infect Immun. 2002;70:3457–3467. doi: 10.1128/IAI.70.7.3457-3467.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Steger KK, Pauza CD. Immunization of Macaca mulatta with aroA attenuated Salmonella typhimurium expressing the SIVp27 antigen. J Med Primatol. 1997;26:44–50. doi: 10.1111/j.1600-0684.1997.tb00318.x. [DOI] [PubMed] [Google Scholar]
- 84.Tacket CO, Hone DM, Curtiss R, III, Kelly SM, Losonsky G, Guers L, Harris AM, Edelman R, Levine MM. Comparison of the safety and immunogenicity of ΔaroC ΔaroD and Δcya Δcrp Salmonella typhi strains in adult volunteers. Infect Immun. 1992;60:536–541. doi: 10.1128/iai.60.2.536-541.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cornish RM, Roth JR, Poulter CD. Lethal mutations in the isoprenoid pathway of Salmonella enterica. J Bacteriol. 2006;188:1444–1450. doi: 10.1128/JB.188.4.1444-1450.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Altincicek B, Kollas A, Eberl M, Wiesner J, Sanderbrand S, Hintz M, Beck E, Jomaa H. LytB, a novel gene of the 2-C-methyl-D-erythritol 4-phosphate pathway of isoprenoid biosynthesis in Escherichia coli. FEBS Lett. 2001;499:37–40. doi: 10.1016/s0014-5793(01)02516-9. [DOI] [PubMed] [Google Scholar]
- 87.McAteer S, Coulson A, McLennan N, Masters M. The lytB gene of Escherichia coli is essential and specifies a product needed for isoprenoid biosynthesis. J Bacteriol. 2001;183:7403–7407. doi: 10.1128/JB.183.24.7403-7407.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Sauret-Güeto S, Urós EM, Ibáñez E, Boronat A, Rodríguez-Concepción M. A mutant pyruvate dehydrogenase E1 subunit allows survival of Escherichia coli strains defective in 1-deoxy-D-xylulose 5-phosphate synthase. FEBS Lett. 2006;580:736–740. doi: 10.1016/j.febslet.2005.12.092. [DOI] [PubMed] [Google Scholar]
- 89.Perez-Gil J, Uros EM, Sauret-Güeto S, Lois LM, Kirby J, Nishimoto M, Baidoo EE, Keasling JD, Boronat A, Rodriguez-Concepcion M. Mutations in Escherichia coli aceE and ribB genes allow survival of strains defective in the first step of the isoprenoid biosynthesis pathway. PLoS One. 2012;7:e43775. doi: 10.1371/journal.pone.0043775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sauret-Güeto S, Ramos-Valdivia A, Ibáñez E, Boronat A, Rodríguez-Concepción M. Identification of lethal mutations in Escherichia coli genes encoding enzymes of the methylerythritol phosphate pathway. Biochem Biophys Res Commun. 2003;307:408–415. doi: 10.1016/s0006-291x(03)01211-7. [DOI] [PubMed] [Google Scholar]
- 91.Pérez-Gil J, Rodríguez-Concepción M. Metabolic plasticity for isoprenoid biosynthesis in bacteria. Biochem J. 2013;452:19–25. doi: 10.1042/BJ20121899. [DOI] [PubMed] [Google Scholar]
- 92.Yoon SH, Lee SH, Das A, Ryu HK, Jang HJ, Kim JY, Oh DK, Keasling JD, Kim SW. Combinatorial expression of bacterial whole mevalonate pathway for the production of β-carotene in E. coli. J Biotechnol. 2009;140:218–226. doi: 10.1016/j.jbiotec.2009.01.008. [DOI] [PubMed] [Google Scholar]
- 93.Ajikumar PK, Xiao WH, Tyo KEJ, Wang Y, Simeon F, Leonard E, Mucha O, Phon TH, Pfeifer B, Stephanopoulos G. Isoprenoid pathway optimization for Taxol precursor overproduction in Escherichia coli. Science. 2010;330:70–74. doi: 10.1126/science.1191652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hamano Y, Dairi T, Yamamoto M, Kawasaki T, Kaneda K, Kuzuyama T, Itoh N, Seto H. Cloning of a gene cluster encoding enzymes responsible for the mevalonate pathway from a terpenoid-antibiotic-producing Streptomyces strain. Biosci Biotechnol Biochem. 2001;65:1627–1635. doi: 10.1271/bbb.65.1627. [DOI] [PubMed] [Google Scholar]
- 95.Wall DM, Srikanth CV, McCormick BA. Targeting tumors with Salmonella Typhimurium - potential for therapy. Oncotarget. 2010;1:721–728. doi: 10.18632/oncotarget.206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hoffman RM. Bugging tumors. Cancer Discov. 2012;2:588–590. doi: 10.1158/2159-8290.CD-12-0227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Low KB, Ittensohn M, Le T, Platt J, Sodi S, Amoss M, Ash O, Carmichael E, Chakraborty A, Fischer J, Lin SL, Luo X, Miller SI, Zheng L, King I, Pawelek JM, Bermudes D. Lipid A mutant Salmonella with suppressed virulence and TNFα induction retain tumor-targeting in vivo. Nat Biotechnol. 1999;17:37–41. doi: 10.1038/5205. [DOI] [PubMed] [Google Scholar]
- 98.Chorobik P, Czaplicki D, Ossysek K, Bereta J. Salmonella and cancer: from pathogens to therapeutics. Acta Biochim Pol. 2013;60:285–297. [PubMed] [Google Scholar]
- 99.Massa PE, Paniccia A, Monegal A, de Marco A, Rescigno M. Salmonella engineered to express CD20-targeting antibodies and a drug-converting enzyme can eradicate human lymphomas. Blood. 2013;122:705–714. doi: 10.1182/blood-2012-12-474098. [DOI] [PubMed] [Google Scholar]
- 100.Chen CY, Yao S, Huang D, Wei H, Sicard H, Zeng G, Jomaa H, Larsen MH, Jacobs WR, Jr, Wang R, Letvin N, Shen Y, Qiu L, Shen L, Chen ZW. Phosphoantigen/IL2 expansion and differentiation of Vγ2Vδ2 T cells increase resistance to tuberculosis in nonhuman primates. PLoS Pathog. 2013;9:e1003501. doi: 10.1371/journal.ppat.1003501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Dieli F, Troye-Blomberg M, Ivanyi J, Fournié JJ, Krensky AM, Bonneville M, Peyrat MA, Caccamo N, Sireci G, Salerno A. Granulysin-dependent killing of intracellular and extracellular Mycobacterium tuberculosis by Vγ9/Vδ2 T lymphocytes. J Infect Dis. 2001;184:1082–1085. doi: 10.1086/323600. [DOI] [PubMed] [Google Scholar]
- 102.Spencer CT, Abate G, Blazevic A, Hoft DF. Only a subset of phosphoantigen-responsive γ9δ2 T cells mediate protective tuberculosis immunity. J Immunol. 2008;181:4471–4484. doi: 10.4049/jimmunol.181.7.4471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Spencer CT, Abate G, Sakala IG, Xia M, Truscott SM, Eickhoff CS, Linn R, Blazevic A, Metkar SS, Peng G, Froelich CJ, Hoft DF. Granzyme A produced by γ9δ2 T cells induces human macrophages to inhibit growth of an intracellular pathogen. PLoS Pathog. 2013;9:e1003119. doi: 10.1371/journal.ppat.1003119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ottones F, Dornand J, Naroeni A, Liautard JP, Favero J. Vγ9Vδ2 T cells impair intracellular multiplication of Brucella suis in autologous monocytes through soluble factor release and contact-dependent cytotoxic effect. J Immunol. 2000;165:7133–7139. doi: 10.4049/jimmunol.165.12.7133. [DOI] [PubMed] [Google Scholar]
- 105.Dudal S, Turriere C, Bessoles S, Fontes P, Sanchez F, Liautard J, Liautard JP, Lafont V. Release of LL-37 by activated human Vγ9Vδ2 T cells: a microbicidal weapon against Brucella suis. J Immunol. 2006;177:5533–5539. doi: 10.4049/jimmunol.177.8.5533. [DOI] [PubMed] [Google Scholar]
- 106.Oliaro J, Dudal S, Liautard J, Andrault JB, Liautard JP, Lafont V. Vγ9Vδ2 T cells use a combination of mechanisms to limit the spread of the pathogenic bacteria Brucella. J Leukoc Biol. 2005;77:652–660. doi: 10.1189/jlb.0704433. [DOI] [PubMed] [Google Scholar]
- 107.Marischen L, Wesch D, Schröder JM, Wiedow O, Kabelitz D. Human γδ T cells produce the protease inhibitor and antimicrobial peptide elafin. Scand J Immunol. 2009;70:547–552. doi: 10.1111/j.1365-3083.2009.02337.x. [DOI] [PubMed] [Google Scholar]
- 108.Chen ZW. Multifunctional immune responses of HMBPP-specific Vγ2Vδ2 T cells in M. tuberculosis and other infections. Cell Mol Immunol. 2013;10:58–64. doi: 10.1038/cmi.2012.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chen ZW. Immune biology of Ag-specific γδ T cells in infections. Cell Mol Life Sci. 2011;68:2409–2417. doi: 10.1007/s00018-011-0703-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Morita CT, Parker CM, Brenner MB, Band H. TCR usage and functional capabilities of human γδ T cells at birth. J Immunol. 1994;153:3979–3988. [PubMed] [Google Scholar]
- 111.Parker CM, Groh V, Band H, Porcelli SA, Morita C, Fabbi M, Glass D, Strominger JL, Brenner MB. Evidence for extrathymic changes in the T cell receptor γ/δ repertoire. J Exp Med. 1990;171:1597–1612. doi: 10.1084/jem.171.5.1597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Cairo C, Sagnia B, Cappelli G, Colizzi V, Leke RG, Leke RJ, Pauza CD. Human cord blood γδ T cells expressing public Vγ2 chains dominate the response to bisphosphonate plus interleukin-15. Immunology. 2013;138:346–360. doi: 10.1111/imm.12039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Davodeau F, Peyrat MA, Hallet MM, Houde I, Vie H, Bonneville M. Peripheral selection of antigen receptor junctional features in a major human γδ subset. Eur J Immunol. 1993;23:804–808. doi: 10.1002/eji.1830230405. [DOI] [PubMed] [Google Scholar]
- 114.Groh V, Porcelli S, Fabbi M, Lanier LL, Picker LJ, Anderson T, Warnke RA, Bhan AK, Strominger JL, Brenner MB. Human lymphocytes bearing T cell receptor γ/δ are phenotypically diverse and evenly distributed throughout the lymphoid system. J Exp Med. 1989;169:1277–1294. doi: 10.1084/jem.169.4.1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kinjo Y, Wu D, Kim G, Xing GW, Poles MA, Ho DD, Tsuji M, Kawahara K, Wong CH, Kronenberg M. Recognition of bacterial glycosphingolipids by natural killer T cells. Nature. 2005;434:520–525. doi: 10.1038/nature03407. [DOI] [PubMed] [Google Scholar]
- 116.Kinjo Y, Tupin E, Wu D, Fujio M, Garcia-Navarro R, Benhnia MR, Zajonc DM, Ben-Menachem G, Ainge GD, Painter GF, Khurana A, Hoebe K, Behar SM, Beutler B, Wilson IA, Tsuji M, Sellati TJ, Wong CH, Kronenberg M. Natural killer T cells recognize diacylglycerol antigens from pathogenic bacteria. Nat Immunol. 2006;7:978–986. doi: 10.1038/ni1380. [DOI] [PubMed] [Google Scholar]
- 117.Kjer-Nielsen L, Patel O, Corbett AJ, Le Nours J, Meehan B, Liu L, Bhati M, Chen Z, Kostenko L, Reantragoon R, Williamson NA, Purcell AW, Dudek NL, McConville MJ, O’Hair RAJ, Khairallah GN, Godfrey DI, Fairlie DP, Rossjohn J, McCluskey J. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature. 2012;491:717–723. doi: 10.1038/nature11605. [DOI] [PubMed] [Google Scholar]
- 118.Corbett AJ, Eckle SBG, Birkinshaw RW, Liu L, Patel O, Mahony J, Chen Z, Reantragoon R, Meehan B, Cao H, Williamson NA, Strugnell RA, Van Sinderen D, Mak JYW, Fairlie DP, Kjer-Nielsen L, Rossjohn J, McCluskey J. T-cell activation by transitory neo-antigens derived from distinct microbial pathways. Nature. 2014;509:361–365. doi: 10.1038/nature13160. [DOI] [PubMed] [Google Scholar]
- 119.López-Sagaseta J, Dulberger CL, Crooks JE, Parks CD, Luoma AM, McFedries A, Van Rhijn I, Saghatelian A, Adams EJ. The molecular basis for Mucosal-Associated Invariant T cell recognition of MR1 proteins. Proc Natl Acad Sci USA. 2013;110:E1771–1778. doi: 10.1073/pnas.1222678110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Patel O, Kjer-Nielsen L, Le Nours J, Eckle SBG, Birkinshaw R, Beddoe T, Corbett AJ, Liu L, Miles JJ, Meehan B, Reantragoon R, Sandoval-Romero ML, Sullivan LC, Brooks AG, Chen Z, Fairlie DP, McCluskey J, Rossjohn J. Recognition of vitamin B metabolites by mucosal-associated invariant T cells. Nat Commun. 2013;4:2142. doi: 10.1038/ncomms3142. [DOI] [PubMed] [Google Scholar]
- 121.Yi W, Liu X, Li Y, Li J, Xia C, Zhou G, Zhang W, Zhao W, Chen X, Wang PG. Remodeling bacterial polysaccharides by metabolic pathway engineering. Proc Natl Acad Sci USA. 2009;106:4207–4212. doi: 10.1073/pnas.0812432106. [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.