Skip to main content

Risk assessment and bioburden evaluation of Agrobacterium tumefaciens-mediated transient protein expression in plants using the CaMV35S promoter

Abstract

Large-scale transient expression of recombinant proteins in plants is increasingly used and requires the multi-liter cultivation of Agrobacterium tumefaciens transformed with an expression vector, which is often cloned in Escherichia coli first. Depending on the promoter, unintentional activity can occur in both bacteria, which could pose a safety risk to the environment and operators if the protein is toxic. To assess the risk associated with transient expression, we first tested expression vectors containing the CaMV35S promoter known to be active in plants and bacteria, along with controls to measure the accumulation of the corresponding recombinant proteins. We found that, in both bacteria, even the stable model protein DsRed accumulated at levels near the detection limit of the sandwich ELISA (3.8 µg L−1). Higher levels were detected in short cultivations (< 12 h) but never exceeded 10 µg L−1. We determined the abundance of A. tumefaciens throughout the process, including infiltration. We detected few bacteria in the clarified extract and found none after blanching. Finally, we combined protein accumulation and bacterial abundance data with the known effects of toxic proteins to estimate critical exposures for operators. We found that unintended toxin production in bacteria is negligible. Furthermore, the intravenous uptake of multiple milliliters of fermentation broth or infiltration suspension would be required to reach acute toxicity even when handling the most toxic products (LD50 ~ 1 ng kg−1). The unintentional uptake of such quantities is unlikely and we therefore regard transient expression as safe in terms of the bacterial handling procedure.

Peer Review reports

Introduction

Plants have been used for the production of valuable small molecules and recombinant proteins since the early 1980s, and this has evolved from a niche topic to a mature technology that now competes with the dominant platforms based on microbial and mammalian cells [1,2,3,4]. Plants have several advantages over cell-based systems, including their inability to support the replication of human viruses, the ease of process scale-up, and short process development times [1, 5, 6]. The latter is often achieved by transient expression, which requires a scalable gene delivery system for each batch of plants [7]. DNA transfer is often mediated by the bacterium Agrobacterium tumefaciens (Rhizobium radiobacter) in species such as Nicotiana benthamiana, tobacco, Arabidopsis and lettuce [8,9,10,11,12,13]. The expression vector is generally cloned in Escherichia coli, so transient expression requires the transformation of E. coli and A. tumefaciens, resulting in two different genetically modified organisms (GMOs) [14]. The use of GMOs requires a formal risk assessment to quantify the hazard posed to personnel and the environment, and the likelihood of harm in practice [5, 15]. Based on this assessment, GMOs are categorized and the precautions required for handling are defined, typically using four biosafety levels (BSLs), where BSL-1 encompasses non-pathogenic GMOs and BSL-2 covers agents with a moderate potential hazard [16,17,18,19]. The BSL rating is linked to the type and quantity of recombinant protein produced in a GMO. For example, E. coli or A. tumefaciens strains carrying a plant expression vector encoding a recombinant toxin may be regarded as BSL-2 by the regulatory authorities because promoters from plant viruses, such as the commonly used cauliflower mosaic virus 35S (CaMV35S) promoter [2, 20, 21], are transcriptionally active in bacteria [22,23,24,25,26,27]. However, the risk assessment routines set out in international guidelines (e.g., the World Health Organization’s Laboratory Biosafety Manual [28]) and supranational laws (e.g., European Union Directive 2000/54/EC [29]) may be interpreted differently at the national level [30]. Accordingly, the same GMO may receive different BSL ratings depending on the country of assessment. This can introduce uncertainty when planning large-scale production, because investment costs for equipment such as filters [31, 32], and especially infrastructure, increase by as much as 25% when BSL-2 is required instead of BSL-1 [33]. Harmonized regulations and classifications would help to resolve this situation [34].

Although plant promoters show some transcriptional activity in bacteria, and A. tumefaciens can persist in transgenic plants grown in greenhouses [35], it is currently unclear how the bacteria spread to the environment from laboratory equipment and surfaces. Also, little is known about the resulting protein accumulation in bacteria and the A. tumefaciens bioburden during transient expression in plants (Fig. 1), especially during infiltration and extraction, which would facilitate evidence-based risk assessment. We therefore set out to quantify recombinant protein accumulation resulting from double enhanced CaMV35S promoter activity in A. tumefaciens and E. coli under representative cultivation conditions compared to bacterial reference promoters. We also monitored the A. tumefaciens bioburden during the preparation and processing of infiltrated plant biomass, including an optional blanching step that facilitates subsequent product purification [36,37,38]. We combined these results with worst-case protein toxicity data and unintended intravenous delivery to personnel in order to derive volumes of process intermediates necessary to reach a hypothetical median lethal dose (LD50). Our results will facilitate the development of safety measures that match actual risks during relevant process steps, taking into account recombinant protein expression levels and activities as well as the colonizing capacity of GMOs, as described, for example, in Annex 1 of the German genetic engineering safety enactment (Gentechnik-Sicherheitsverordnung, GenTSV) [39].

Fig. 1
figure 1

Schematic process flow of A. tumefaciens-mediated transient expression in plants. A Cloning in E. coli, A. tumefaciens pre-culture and fermentation for subsequent infiltration of plants. B Cultivation of wild-type plants, followed by infiltration with the A. tumefaciens suspension and incubation to facilitate transient expression with an optional blanching step before extraction. Downstream processing starts with plant biomass extraction, and includes clarification (e.g., depth filtration), an optional UF/DF step, and purification by chromatography

Materials and methods

Molecular cloning and plant expression vectors

The CaMV35S promoter driving the expression of DsRed, a red fluorescent protein from Discosoma sp., and both chains of a monoclonal IgG1 antibody in the plant expression vector pTRAc [40] was replaced with the β-lactamase (bla) promoter from the Tn3 transposon [41], or the T7 promoter from bacteriophage T7 RNA polymerase [42] (Additional file 1: Table S1) in a restriction-ligation reaction using AscI, EcoRI and NheI (New England BioLabs, Ipswich, USA). Promoter sequences with compatible ends were generated by PCR using specific primers (Additional file 1: Table S2). DNA was purified using a NucleoSpin kit (Machery-Nagel, Düren, Germany) and ligated using T4 DNA ligase (New England BioLabs). The resulting plasmids (Additional file 1: Table S3) were introduced into chemically competent E. coli DH5α cells and electro-competent A. tumefaciens GV3101 cells after purification.

Sources of plant seeds and cells

N. tabacum L. cv Bright Yellow-2 cells were obtained from the Leibniz-Institute DSMZ Deutsche Samlung von Mikroorganismen und Zellkulturen GmbH on the 20th of January 2020 (ID PC 1181). Seeds from N. benthamiana plants were a donation from the RWTH Aachen University in 2006.

Bacterial cell culture and extraction

For E. coli DH5α, we inoculated 0.3 L of sterile lysogeny broth (LB) medium (5 g L−1 yeast extract, 10 g L−1 tryptone, 0.5 g L−1 sodium chloride, supplemented with 100 mg L−1 ampicillin) with 0.3 mL of an overnight pre-culture in a 2-L non-baffled Erlenmeyer flask. The cultures were incubated for up to 48 h at 37 °C, shaking at 180 rpm. We took 50-mL samples after 12, 24 and 36 h for protein analysis.

For A. tumefaciens GV3101:pMP90RK, we inoculated 0.3 L of PAM4 medium (20 g L−1 soy peptone, 0.5 L−1 yeast extract, 5.0 g L−1 fructose, 1.0 g L−1 magnesium sulfate, pH 7.0) [43] or yeast extract broth (YEB) medium (5.0 g L−1 beef extract, 1.0 g L−1 yeast extract, 5.0 g L−1 soy peptone, 5.0 g L−1 sucrose, 0.5 g L−1 magnesium sulfate, pH 7.0), each supplemented with 25 mg L−1 kanamycin, 25 mg L−1 rifampicin and 50 mg L−1 carbenicillin, with 0.3 mL of an overnight pre-culture in a non-baffled 2-L Erlenmeyer flask. The cultures were incubated for up to 100 h at 26 °C, shaking at 160 rpm. Wild-type A. tumefaciens controls were cultured under the same conditions but without antibiotics. We took 50-mL samples after 24, 48 and 72 h to determine the optical density at 600 nm (OD600nm) and the Boltzmann model (Eq. 1) was fitted to the data.

$$f\left( x \right) = \frac{{A_{1} - A_{2} }}{{1 + e^{{\left( {x - x_{0} } \right)/dx}} }} + A_{2}$$
(1)

where A1 is the initial (OD600nm) value, A2 is the final (OD600nm) value, x is the cultivation time, x0 is the curve center, dx is the time constant, and f(x) is the OD600nm as a function of time.

Harvested culture samples were also centrifuged (4000×g, 10 min, 10 °C) and pellets were homogenized on ice using a PowerGen 125 IKA Ultra Turrax homogenizer (Thermo Fisher Scientific, Waltham, USA) in 5 mL NEBExpress Gram-negative bacteria extraction buffer (New England BioLabs). We applied three pulses of 1 min with pauses of 2 min between. Extracts were clarified by centrifugation (4000×g, 10 min, 10 °C) and passage through a 0.2 µm MiniSart syringe filter (Sartorius, Göttingen, Germany) before protein analysis.

Transient expression in differentiated N. benthamiana plants

Seeds were germinated on stone wool blocks soaked with 1.0 g L−1 Ferty 2 Mega fertilizer solution (Planta Düngemittel, Regenstauf) and incubated for ~ 7 days before reducing the number of germ buds to one per block. The stone wool blocks were placed into custom-made plastic trails and incubated in a greenhouse (25/22 °C day night temperature, ~ 14-h photoperiod, ~ 70% relative humidity). Natural light was augmented if necessary with 400-W IP65 or SON-k sodium discharge lamps (Phillips, Amsterdam, Netherlands) [31, 37]. The plants were irrigated with 1.0 g L−1 fertilizer solution for 12 min 2–4 times per day using an ebb-and-flow hydroponics system that removed residual liquid after each watering phase. N. benthamiana plants were infiltrated with A. tumefaciens carrying vectors for the expression of DsRed or antibody M12 at 42–49 days after seeding by vacuum infiltration. Specifically, a bucket containing ~ 5 L of OD600nm = 0.5 bacterial suspension was placed in a desiccator and plants were submerged headlong into the liquid. Then, the pressure was reduced to 5 kPa (50 mbar) using a vacuum pump, held at this level for 2 min and abruptly released. Infiltrated plants were briefly drained of residual infiltration suspension for 1 min and prepared for S2-compliant incubation by wrapping them into translucent plastic film. If necessary, additional bacterial suspension was added to the bucket to maintain a level of ~ 5 L and the suspension was manually agitated for ~ 30 s in between each plant infiltration cycle to prevent sedimentation. The infiltration was conducted at a temperature of ~ 21 °C. Infiltrated plants were harvested after incubation for 5 days at 21 °C and ~ 70% relative humidity and were used for acidic blanching [44].

Acidic blanching of N. benthamiana leaf material

Leaves were blanched in an EKA 3338 heated vessel (Clatronic International, Kempen, Germany) equipped with an MD‐6Z pump (~ 6.0 L min−1) to maintain a constant circulation of 20 L blanching buffer (20 mM trisodium citrate, pH 4.0). The temperature was set to 70 or 80 °C before leaves were submerged in the blanching buffer and carefully agitated to ensure uniform blanching. After blanching for up to 15 min, the leaves were transferred to a bucket filled with cold tap water for ~ 15 s and then carefully dried with paper towels before extraction. The apparent gain in leaf mass due to residual blanching buffer was recorded and used to correct any dilution effect during subsequent protein extraction.

Extraction and filtration

Freshly harvested or blanched N. benthamiana plant material was homogenized using a ProBlend 6 blender (Phillips, Amsterdam, Netherlands) as previously described [45] and a modified extraction buffer (50 mM sodium phosphate, 500 mM sodium chloride, 10 mM sodium bisulfite, pH 6.0). The homogenate pH was immediately adjusted to 8.0 with 0.4 M trisodium phosphate. Coarse particles were removed by passing the homogenate through an acuraLine BP-420-1 bag filter (Fuhr, Klein-Winternheim, Germany) with a 1-µm nominal pore size. Fine particles were removed by depth filtration using dual V700P and V100P layers with nominal pore sizes of 8–20 and 1–3 µm, respectively. Depth filtration was performed at a constant volumetric flux of ~ 1.0 L m−2 min−1 using a Masterflex SE peristaltic pump (Masterflex, Gelsenkirchen, Germany) up to a maximum inlet pressure of 0.2 MPa. The depth filtrate was passed through a Sartopore2 150 sterile filter (Sartorius) with a pore size combination of 0.45 and 0.20 µm.

Bioburden assessment of plant infiltration and plant biomass processing

Process samples (Table 1) were plated on YEB agar (1.5% m v−1) containing the same antibiotics as above and were incubated at 28 °C and ~ 25% relative humidity. Colony forming units (CFU) were counted by visual inspection every 24 h up to 5 days after plating. Specifically, the top and bottom of infiltrated, blanched and wild-type control N. benthamiana leaves were tested by direct contact with a contact area of ~ 0.0056 m2 per leaf (~ 80% of the total area of a 94 mm diameter Petri dish). Up to 250 mL of the blanching buffer (after processing wild-type control or infiltrated leaves) was also passed through a 0.45-µm filter and the filter cake was used in the direct contact test with a contact area of ~ 0.0017 m2. Furthermore, 15-mL samples were taken from wild-type control and infiltrated leaves before and after blanching as well as after bag, depth and sterile filtration. One set of aliquots from these samples was incubated for 15 min in 15-mL reaction tubes to allow dispersed particles to sediment and a second set was centrifuged (15,000×g, 1 min, 21 °C). We then plated 0.1 mL of each supernatant undiluted and also as 1:100 and 1:1000 dilutions in PBS. Finally, air samples during infiltration as well as during blanching were taken at three representative positions 300–600 mm from each device (Additional file 1: Fig. S1) using an exposure time of 15 min. For these samples, we used both selective and antibiotic-free YEB agar plates.

Table 1 Process steps during transient expression in N. benthamiana and monitoring measures to detect A. tumefaciens

All samples were collected across a set of seven independent experiments using up to six replicate plates per sample per batch (including dilutions). Given the log-normal distribution of bacterial population counts on leaf surfaces [46], we used the geometric mean (Eq. 2) and its standard deviation (Eq. 3), which better represent such distributions than their arithmetic counterparts [47], despite some debate about the standard deviation formula [48, 49].

$$\overline{x}^{*} = exp\left( {\frac{1}{n}\mathop \sum \limits_{i = 1}^{n} {\text{log}}\left( {x_{i} } \right)} \right) = \left( {\mathop \prod \limits_{i = 1}^{n} x_{i} } \right)^{\frac{1}{n}}$$
(2)
$$s^{*} = exp\left( {\left[ {\frac{1}{n - 1}\mathop \sum \limits_{i = 1}^{n} \left[ {{\text{log}}\left( {\frac{{x_{i} }}{{\overline{x}^{*} }}} \right)} \right]^{2} } \right]^{\frac{1}{2}} } \right)$$
(3)

where \(\overline{x}^{*}\) is the mean of the log-normal distributed data (i.e., the geometric mean), n is the sample size, x is the value (here CFU count) of observation i, and s* is the geometric standard deviation. CFU counts were converted to an OD600nm using Eq. 4 [50].

$$OD_{{600\,{\text{nm}}}} \left[ - \right] = CFU \left[ {\frac{1}{{{\text{mL}}}}} \right] \times 1.43 \times 10^{ - 9}$$
(4)

For air and direct contact samples, the CFU count per square meter was converted to a volumetric equivalent assuming that a liquid level of ~ 1 mm height (1.0 L m−2) would be needed to re-suspend the bacteria.

Protein expression in plant cell packs

Tobacco (Nicotiana tabacum) Bright Yellow-2 (BY-2) cells from a continuous culture were expanded in 0.5-L shake flasks for 5 days to a cell wet mass of 200 g L−1 in an ISFX-1 shaker (Adolf Kühner, Birsfelden, Switzerland) at 160 rpm and 26 °C, and were used for the preparation of plant cell packs (PCPs) as previously described [51]. Agrobacterium tumefaciens cultures were adjusted to an OD600nm of 0.5 using infiltration buffer (0.5 g L−1 Murashige-Skoog salt M0221, 50.0 g L−1 sucrose, 2.0 g L−1 glucose monohydrate, 0.04 g L−1 acetosyringone, pH 5.6) before application to the PCPs [51]. After incubation in the dark at 26 °C and 80% relative humidity for 4 days, the PCPs were harvested for homogenization in an MM300 bead-mill (Retsch, Haan, Germany). We added three volumes (v m−1) of extraction buffer (20 mM sodium phosphate, 500 mM sodium chloride, 10 mM sodium bisulfite, pH 8.0) followed by three 30-s pulses at 28 Hz. After centrifugation (4000×g, 8 min, 10 °C) the supernatant was analyzed or stored at − 20 °C.

Protein quantification

The samples were analyzed by LDS-PAGE [31] and western blotting [52] as previously described. DsRed was detected using a monoclonal rabbit anti-His6 primary antibody and an alkaline phosphatase (AP)-labeled goat anti-rabbit secondary antibody, whereas M12 was detected using AP-labeled polyclonal goat anti-human heavy and light chain antibodies. DsRed in PCP extracts was quantified by fluorimetry [31].

For sandwich ELISA, the wells of high-binding 96-well ELISA plates (Greiner Bio-One, Kremsmünster, Austria) were coated with 100 µL of the appropriate capture antibody prepared in coating buffer (14.3 mM disodium carbonate, 34.9 mM sodium bicarbonate, pH 9.6) at a concentration of 0.2 mg L−1. After incubation for 18 h at 4 °C, the coating solution was removed and the wells were washed with blocking buffer (50 g L−1 skimmed milk powder in PBST: 137 mM sodium chloride, 2.7 mM potassium chloride, 10 mM disodium hydrogen phosphate, 1.8 mM sodium dihydrogen phosphate, 0.05% (v v−1) Tween-20, pH 7.2) for 1 h at 21 °C. Excess liquid was removed and the coated plates were directly used for ELISA experiments.

Extracts and dilution series of standards (purified target proteins) were prepared in blocking buffer (50 g L−1 skimmed milk powder in PBST) before pipetting 100 µL into the coated wells as technical triplicates or duplicates, respectively. After incubation for 1 h at 21 °C on a shaker at 10 rpm, the liquid was removed and wells were washed five times with 250 µL wash buffer (50 mM Tris, 15 mM sodium chloride, pH 7.2). The detection antibody was prepared in blocking buffer using concentrations as recommended by the manufacturer (1.5–2.5 g L−1) and 100 µL was added to each well. After incubation for 2 h at 21 °C and five wash cycles with 250 µL wash buffer, we added 100 µL 1-Step PNPP solution (Thermo Fisher Scientific) per well and mixed thoroughly by agitation at 10 rpm. The plate was incubated for 30 min at 21 °C before stopping the reaction by adding 50 µL 2.0 M sodium hydroxide to each well and measuring the absorbance at 405 nm in duplicate using a Synergy H1 microplate reader (BioTek, Winooski, USA) at 21 °C. The absorbance of eight standards of DsRed-His6 or M12 IgG1 [31, 38] in the range 0.003–300 mg L−1 was measured in triplicate. Blank-corrected extract samples were log transformed, and a linear log–log curve fit (R2 = 0.98) was used to calculate recombinant protein concentrations. We used a wet mass to dry mass conversion factor of 0.2 [53, 54] combined with an OD600nm to dry mass conversion factor of 0.396 g L−1 (E. coli) or 0.409 g L−1 (A. tumefaciens) [55, 56] to calculate cell mass-specific recombinant protein concentrations.

Results and discussion

CaMV35S promoter activity in bacterial cells

The growth of E. coli and A. tumefaciens strains transformed with pTRAc for DsRed and IgG1 expression (Additional file 1: Table S3) was similar to that of untransformed controls (Fig. 1A, B). E. coli grew significantly faster than A. tumefaciens when comparing the x0 coefficients of a Boltzmann model (Eq. 1) fitted to the growth curve data (two-sided two-sample t-test, p < 0.001), which was anticipated because the doubling times are ~ 0.3 and ~ 3 h, respectively [57, 58]. Whereas all A. tumefaciens cultures reached a similar maximum OD600nm of ~ 6.8 ± 0.5 (± SD, n = 7), the transformed E. coli clones reached a significantly lower maximum OD600nm (p < 0.001) of 6.5 ± 0.8 (± SD, n = 6) after 20 h compared to ~ 7.7 ± 0.1 (± SD, n = 3) for the control (Fig. 2A). This may reflect the plasmid-associated metabolic burden in E. coli [59, 60] because pTRAc vectors have a higher copy number in this species than in A. tumefaciens [61,62,63] (Fig. 2B).

Fig. 2
figure 2

Evaluation of bacterial growth and recombinant protein accumulation. Growth curves of E. coli (A) and A. tumefaciens (B) carrying pTRAc vectors (Additional file 1: Table S3) for the expression of DsRed or IgG1 under control of different promoters. DsRed and IgG1 concentrations in E. coli (C) and A. tumefaciens (D) per cell dry mass were determined by ELISA. Cell-mass specific recombinant protein concentrations were calculated using a dry mass to OD600nm conversion factor of 0.396 g−1 (E. coli) and 0.409 g.−1 (A. tumefaciens) [55, 56] and a wet mass to dry mass conversion factor of 0.2. DsRed and IgG1 concentration in E. coli (E) and A. tumefaciens (F) fermentation broth were quantified by ELISA. The sampling time points were shifted by ± 1.5 h to display significant differences between samples. IgG1 and DsRed accumulation levels in PCPs (G) using different promoters. Western blot of triplicate PCP extracts (H) using plasmids with different promoters for the expression of DsRed (top row) and IgG1 (bottom row). DsRed was detected using a rabbit anti-His6 primary antibody and an alkaline phosphatase (AP)-labeled goat anti-rabbit secondary antibody, whereas IgG1 was detected using an AP-labeled goat anti-human antibody. Full-size blots can be found as Additional file 2: Fig. S2 “Risk_and_bioburden_knödler_et_al_full-size_blots_v2”. CaMV35S—double-enhanced cauliflower mosaic virus 35S promoter with strong activity in plants; bla—β-lactamase promoter with activity in bacteria; T7—bacteriophage T7 promoter with minimal activity in bacteria unless the corresponding polymerase is expressed. Data are means ± SD (n = 3 biological replicates, two-way ANOVA with Bonferroni correction and a significance threshold of α = 0.05; *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001)

The concentrations of DsRed and IgG1 in E. coli and A. tumefaciens extracts were below the western blot detection limit (0.27 mg L−1 for DsRed and 2.60 mg L−1 for IgG1). The more sensitive ELISA revealed concentrations as a proportion of cell dry mass of up to ~ 250 mg kg−1 in E. coli and 12 mg kg−1 in A. tumefaciens (Fig. 2C, D). This corresponded to concentrations below 10 µg L−1 for DsRed and 2 µg L−1 for IgG1 in E. coli fermentation broth or below 3 µg L−1 for both proteins in A. tumefaciens cultures (Fig. 2E, F). Interestingly, and despite their stability [64,65,66], both proteins were only present at relevant levels during the lag and early log phases and were barely detectable after 24 h regardless of the protein or species, corresponding to a consistently low protein concentration in the fermentation broth of < 0.3 µg L−1. Specifically, DsRed was 5–10 times more abundant than the IgG1, probably due to the greater complexity of immunoglobulins and/or the inability of bacteria to introduce sufficient disulfide bonds to ensure correct antibody folding and stability [67, 68]. However, a similar difference in the accumulation of these proteins has also been observed in transgenic plants [69]. We found that the CaMV35S promoter produced the highest levels of recombinant protein in E. coli, but was significantly less active in A. tumefaciens for harvests within 24 h (two-sided two sample t-test, p < 0.032). We speculate that A. tumefaciens may have evolved mechanisms to inhibit transcription initiated from promoters that are active in plants because these bacteria naturally carry plant-specific promoters on their Ti-plasmids [70], and the corresponding proteins are not needed by the bacteria. The leaky activity of plant promoters in A. tumefaciens would therefore cause an unnecessary metabolic burden [71]. The low protein levels we observed were also consistent with previous studies reporting nonspecific but marginal activity of plant promoters in Gram-negative bacteria based on mRNA levels [22, 23]. It is currently unclear whether tightly regulated or inducible plant promoters [72, 73] are inactive in A. tumefaciens and E. coli.

As a control experiment, we also used the same A. tumefaciens strain for transient expression in PCPs [74]. We found that constructs containing the plant-specific CaMV35S promoter produced ~ 90 mg kg−1 and 24 mg kg−1 of DsRed and IgG1, respectively, which was significantly more than the other promoters (Fig. 2G, H, Additional file 2: Fig. S2). This was anticipated based on previous reports [44, 51]. The only non-plant promoter that produced detectable levels of DsRed in PCPs was the T7 promoter.

Agrobacterium tumefaciens bioburden evaluation for a transient expression process using whole plants

We monitored the A. tumefaciens bioburden during the infiltration of N. benthamiana and subsequent incubation, biomass conditioning (blanching), extraction and clarification steps (Table 1). As expected, no A. tumefaciens were found at any process step when non-infiltrated wild-type control plants were handled and processed (Fig. 3). We also found no A. tumefaciens in any of the air samples during infiltration or blanching, and all blanching buffers were also sterile. The abundance of A. tumefaciens on leaf surfaces was ~ 7000 CFU m−2 after infiltration but before blanching. Given a typical N. benthamiana leaf thickness of 0.25 × 10−3 m and a density of 760 kg m−3 [75], this corresponded to an A. tumefaciens concentration of ~ 40 CFU g−1 leaf biomass, which is marginal compared to the 0.3–1.0 × 106 g−1 of total epiphytic bacteria found, for example, on rice leaves [76]. Our observation agreed with earlier data suggesting that bacterial communities on tobacco leaves are stabilized by quorum sensing [77], which should prevent the overgrowth of bacterial communities by pathogenic proteobacteria such as A. tumefaciens [78]. Furthermore, no A. tumefaciens were detected on wild-type or infiltrated leaves after blanching, confirming that this processing method effectively inactivates bacteria before protein extraction. When infiltrated leaves were homogenized without blanching, the A. tumefaciens load was ~ 4 × 108 CFU L−1 of extract. Given our biomass-to-buffer ratio of 1:3, this corresponded to ~ 10 × 106 CFU g−1 fresh leaf mass, which is an order of magnitude more than the concentration calculated above for A. tumefaciens on the surface. Finally, the depth filtrates of extracts from infiltrated (but not blanched) leaves revealed that CFU counts were reduced by two orders of magnitude, and no A. tumefaciens were detected at all in the sterile filtrates. This indicated that downstream processing steps typically used for plant biomass were able to remove the bacteria effectively, and we concluded that the bioburden of A. tumefaciens was small during each process step, especially compared to typical epiphytic bacteria on leaf surfaces.

Fig. 3
figure 3

Bioburden and predicted toxin accumulation in bacteria. A A. tumefaciens bioburden during the infiltration, optional blanching, extraction and clarification of N. benthamiana biomass and in the corresponding process intermediates. The CFU per area (air and surface samples) or per liter of process intermediate was assessed on selective and non-selective (air samples only) YEB agar plates. B Samples as in A but with the CFU [L−1] count converted to an equivalent OD600nm [–] using a conversion factor of 6.99 × 10−13 [L] based on a published correlation [50]. C Correlation between DsRed and IgG1 accumulation in plants and E. coli (Pearson’s r = 0.99, R2 = 0.99) with an average correlation factor (slope) of 0.43. D Correlation between DsRed and IgG1 accumulation in tobacco BY-2 PCPs and A. tumefaciens (Pearson’s r = 0.99, R2 = 0.96) with an average correlation factor of 0.03. DsRed and IgG1 accumulation in E. coli and A. tumefaciens were measured by ELISA (Fig. 2C, D). Data are means ± SD (n = 4–21 data points depending on the sample). CFU—colony forming unit; DM—dry mass; n.a.—not applicable; OD—optical density

Interestingly, the abundance of A. tumefaciens in laboratories (even those working with plants) is low in relation to other bacteria, with a relative frequency of only ~ 0.001 [79]. Indeed, the same is true overall for the class α-proteobacteria, which accounts for < 1% of all bacteria found in such settings. A. tumefaciens is not considered as a laboratory-contaminating reagent introduced by human daily activities, or even a basic environmental bacterium [79]. Furthermore, it is not among the antibiotic-resistant microbes associated with health risks [80]. This bacterium therefore appears to pose little risk to operators in the laboratory even if it is released during transient expression experiments or subsequent processing steps, and is a negligible risk to humans and the environment if it then escapes from the laboratory.

Risk assessment for the transient expression of toxic proteins in plants

The process steps during which operators can be exposed to relevant quantities of potentially harmful bacteria are limited to fermentation, plant infiltration/incubation, extraction, and the subsequent conditioning and clarification steps up to sterile filtration (Fig. 1). Here, we assumed an intermediate process scale of ~ 200 kg plant biomass as previously discussed [7]. The E. coli and A. tumefaciens laboratory strains we used are rated as safety class 1, which means they are not harmful to immunocompetent persons [39]. Accordingly, any potential risk associated with their handling arises from the recombinant proteins they produce due to the residual promoter activity in bacteria described above. The risk is highest for toxic proteins with low median lethal doses (LD50) for intravenous exposure of 0.001–500 µg kg−1 body mass [81,82,83], with the latter being ~ 80.7 kg for an average person in North America as of 2012 [84]. The risk will also increase in line with the protein concentration, which we estimated by first assuming that protein accumulation in plants and nonspecific expression in bacteria are proportional (e.g., due to target protein stability). We therefore fitted linear models for the CaMV35S-driven accumulation of DsRed and M12 in N. benthamiana vs E. coli as well as N. benthamiana vs A. tumefaciens (Fig. 3C, D). We then combined these models with the highest levels reported for the transient expression of a recombinant immunotoxin in tobacco PCPs (to remain consistent with the DsRed and IgG1 expression data) of ~ 40 mg kg−1 (our unpublished data). This allowed us to estimate the expression of such a protein in both bacterial systems, resulting in values of ~ 80 mg kg−1 dry mass in E. coli and ~ 4 mg kg−1 dry mass in A. tumefaciens. Importantly, these values are worst-case scenarios because the correlation with plant-based expression is based on the highest protein accumulation in bacteria, which was only observed at the beginning of cultivation, so the actual toxin concentration when the bacteria are harvested is likely to be lower by one or two orders of magnitude (Fig. 2C–F). Furthermore, the toxin concentration in plants we used to interpolate the corresponding concentration in bacteria was, to our knowledge, the highest reported in PCPs and plant cells so far [21]. Based on these data and our bioburden results, we assessed the risks of an A. tumefaciens-mediated transient expression process in plants (Table 2).

Table 2 Concentrations of toxic recombinant protein in process fluids due to expression in A. tumefaciens and N. benthamiana along with the corresponding hazardous process volumes

We found that, even assuming LD50 values of 1 ng kg−1, the process volumes posing an acute threat to operator health when handling bacteria during cloning or cultivation were either in the same order of magnitude as, or even larger than, the total volume present at that stage, or the critical volumes were so large (several milliliters) that an accidental intravenous exposure is implausible. We concluded that the risk for operators handling such bacteria was low despite some toxin expression in bacteria caused by nonspecific promoter activity.

Blanching inactivated bacteria on the infiltrated leaves (and for all subsequent steps) but had no relevant effect on the concentration of toxic protein because the nonspecific expression in A. tumefaciens was marginal compared to that intentionally triggered in the plant material (Table 2). Accordingly, toxic protein concentrations were dominated by the transient expression in plants at all process steps after infiltration. As we expected, the toxic protein concentration increased substantially following the UF/DF concentration step, suggesting that the purification process is potentially hazardous (Table 2, Fig. 1). Based on our calculation, only the production of highly potent proteins, such as botulinum toxin with an LD50 of ~ 1 ng kg−1 [81], can pose a relevant safety risk during downstream processing. In such cases, the corresponding critical volumes were in the microliter range, especially after the UF/DF step, and accidental intravenous exposure to operators therefore becomes plausible. However, these process steps were GMO-free. Therefore, we concluded that the toxic protein accumulating in the plants would pose a relevant safety risk to both operators and the environment, not the small amounts of protein unintentionally produced in the bacteria.

Conclusions

We detected the nonspecific accumulation of recombinant proteins in E. coli at levels of up to 10 µg L−1 fermentation broth, but this fell to < 1 µg L−1 at the time of cell harvest. The corresponding values were ~ 20-fold lower in A. tumefaciens. The critical volumes of process liquids associated with acute toxicity were several milliliters or more assuming typical OD600nm values of 2–10 and LD50 values as low as 1 ng kg−1 body mass. Accordingly, the unintended intravenous exposure of operators appears unlikely. In contrast, critical volumes during downstream processing, when no GM bacteria are present, were in the microliter range. Furthermore, no airborne distribution of bacteria was detected during infiltration or blanching, indicating that the threat to the environment due to accidental spread from laboratories is negligible. Here, we selected the LPH leader peptide because it facilitates an effective targeting of the secretory pathway where accumulation of the model proteins we used had reported to be highest. Whereas we assume that the accumulation of proteins will be similar when using other targeting signals, we cannot rule out that changes in the signal peptide may also affect expression in bacteria. However, such changes are typically moderate and will not distort the results in a relevant manner. We conclude that nonspecific protein expression in bacteria resulting from residual promoter activity does not pose a relevant risk to experimenters or the environment.

However, some current safety regulations for the risk assessment of GMOs, such as the E. coli and A. tumefaciens strains we used, appear to use binary decisions (i.e., activity vs inactivity of a promoter) instead of quantitative data representing protein accumulation. Accordingly, bacteria containing a vector encoding a toxin under the control of the CaMV35S promoter intended for transient expression in plants would be classified as BSL-2 because residual promoter activity has been shown in the bacteria, even though the data presented here indicate that such activity does not translate into an actual risk for operators or the environment.

Such a binary decision strategy may reduce the workload for regulatory bodies when they assess and rate the risk associated with individual research projects. However, this is likely to reduce scientific innovation, especially in the field of urgently-needed potent new cancer drugs, due to the increased administrative effort and additional infrastructural requirements even for scouting experiments. The application of quantitative rather than qualitative decision criteria for the classification of GMOs would accelerate the development of innovative biopharmaceuticals.

Availability of data and materials

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Abbreviations

ANOVA:

Analysis of variance

AP:

Alkaline phosphatase

bla :

β-Lactamase promoter

BY-2:

Bright Yellow 2

CaMV35S:

Double enhanced cauliflower mosaic virus 35S promoter

CHS:

Petroselinum hortense Chalcone synthase

DSP:

Downstream processing

ELISA:

Enzyme-linked immunosorbent assay

ER:

Endoplasmic reticulum

His6 :

6 × Histidine tag

IgG1:

Immunoglobulin subclass G1

LD50 :

Median lethal dose

LPH:

Signal peptide, murine antibody heavy chain

OD:

Optical density

PCP:

Plant cell pack

POI:

Protein of interest

SD:

Standard deviation

T7:

Bacteriophage T7 RNA polymerase promoter

LDS:

Lithium dodecyl sulfate

UTR:

Untranslated region

YEB:

Yeast extract broth

References

  1. Buyel JF. Plant molecular farming: integration and exploitation of side streams to achieve sustainable biomanufacturing. Front Plant Sci. 2019;9:1893–1893.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Amack SC, Antunes MS. CaMV35S promoter: a plant biology and biotechnology workhorse in the era of synthetic biology. Curr Plant Biol. 2020;24:100179.

    Article  Google Scholar 

  3. Chung YH, et al. Integrating plant molecular farming and materials research for next-generation vaccines. Nat Rev Mater. 2021. https://doi.org/10.1038/s41578-021-00399-5.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Schillberg S, Finnern R. Plant molecular farming for the production of valuable proteins: critical evaluation of achievements and future challenges. J Plant Physiol. 2021;258–259:153359.

    Article  PubMed  Google Scholar 

  5. Commandeur U, Twyman RM, Fischer R. The biosafety of molecular farming in plants. CABI Rev. 2003. https://doi.org/10.1079/cabireview20033177371.

    Article  Google Scholar 

  6. Stoger E, et al. Practical considerations for pharmaceutical antibody production in different crop systems. Mol Breed. 2002;9(3):149–58.

    Article  CAS  Google Scholar 

  7. Spiegel H, et al. Ready-to-use stocks of agrobacterium tumefaciens can simplify process development for the production of recombinant proteins by transient expression in plants. Biotechnol J. 2019;14(10):e1900113.

    Article  PubMed  Google Scholar 

  8. Fischer R, Buyel JF. Molecular farming: the slope of enlightenment. Biotechnol Adv. 2020;40:107519–107519.

    Article  CAS  PubMed  Google Scholar 

  9. Peyret H, Lomonossoff GP. When plant virology met agrobacterium: the rise of the deconstructed clones. Plant Biotechnol J. 2015;13(8):1121–35.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Du D, et al. Infection of embryonic callus with agrobacterium enables high-speed transformation of maize. Int J Mol Sci. 2019;20(2):279.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Hiei Y, Ishida Y, Komari T. Progress of cereal transformation technology mediated by Agrobacterium tumefaciens. Front Plant Sci. 2014. https://doi.org/10.3389/fpls.2014.00628.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Joh LD, et al. High-level transient expression of recombinant protein in lettuce. Biotechnol Bioeng. 2005;91(7):861–71.

    Article  CAS  PubMed  Google Scholar 

  13. Yamamoto T, et al. Improvement of the transient expression system for production of recombinant proteins in plants. Sci Rep. 2018;8(1):4755.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zhang Y, et al. DNA cloning by homologous recombination in Escherichia coli. Nat Biotechnol. 2000;18(12):1314–7.

    Article  CAS  PubMed  Google Scholar 

  15. Hundleby PAC, Sack M, Twyman RM. Biosafety, risk assessment, and regulation of molecular farming. Mol Pharm. 2018. https://doi.org/10.1002/9781118801512.ch13.

    Article  Google Scholar 

  16. Potts PJMJ. Centers for Disease Control and Prevention National Institutes of Health: Biosafety in Microbiological and Biomedical Laboratories. U.S. Department of Health and Human Services, Revised June 2020. 6th Edition.

  17. Richmond JY. The 1, 2, 3's of biosafety levels. Office of Health and Safety: Centers for Disease Control and Prevention. https://www.aphl.org/programs/preparedness/Smallpox/pdf/the-1-2-3s-of-biosafety-levels.pdf. Accessed 8 Apr 2022.

  18. Chosewood L, Wilson D. Biosafety in microbiological and biomedical laboratories, 5th Edition. 2009.

  19. Salerno RM, Gaudioso J. Laboratory biorisk management: biosafety and biosecurity. UK: CRC Press; 2015.

    Book  Google Scholar 

  20. Benfey PN, Chua NH. The cauliflower mosaic virus 35S promoter: combinatorial regulation of transcription in plants. Science. 1990;250(4983):959–66.

    Article  CAS  PubMed  Google Scholar 

  21. Knödler M, Buyel JF. Plant-made immunotoxin building blocks: a roadmap for producing therapeutic antibody-toxin fusions. Biotechnol Adv. 2020;47: 107683.

    Article  PubMed  Google Scholar 

  22. Assaad FF, Signer ER. Cauliflower mosaic virus P35S promoter activity in Escherichia coli. Mol Gen Genet. 1990;223(3):517–20.

    Article  CAS  PubMed  Google Scholar 

  23. Jacob D, et al. Plant-specific promoter sequences carry elements that are recognised by the eubacterial transcription machinery. Transgenic Res. 2002;11(3):291–303.

    Article  CAS  PubMed  Google Scholar 

  24. Jopcik M, Bauer M, Moravcikova J, Boszoradova E, Matusikova I, Libantova J. Plant tissue-specific promoters can drive gene expression in Escherichia coli. Plant Cell Tissue Organ Cult. 2013;113(3):387–96. https://doi.org/10.1007/s11240-012-0278-7.

    Article  CAS  Google Scholar 

  25. Lewin A, et al. Viral promoters can initiate expression of toxin genes introduced into Escherichia coli. BMC Biotechnol. 2005;5(1):19.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lewin A, et al. Gene expression in bacteria directed by plant-specific regulatory sequences. Transgenic Res. 2004;7:403–11.

    Article  Google Scholar 

  27. Antonucci TK, Wen P, Rutter WJ. Eukaryotic promoters drive gene expression in Escherichia coli. J Biol Chem. 1989;264(30):17656–9.

    Article  CAS  PubMed  Google Scholar 

  28. Bennett A, et al. Laboratory biosafety manual fourth edition and associated monographs laboratory design and maintainance. 2020: World Health Organization.

  29. Partiament E. DIRECTIVE 2000/54/EC OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL. Official Journal of the European Communities, 2000: p. L 262/21.

  30. Pastorino B, de Lamballerie X, Charrel R. Biosafety and biosecurity in European containment level 3 laboratories: focus on french recent progress and essential requirements. Front Public Health. 2017. https://doi.org/10.3389/fpubh.2017.00121.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Knödler M, Rühl C, Opdensteinen P, Buyel JF. activated cross-linked agarose for the rapid development of affinity chromatography resins: antibody capture as a case study. J Vis Exp. 2019. https://doi.org/10.3791/59933-v.

    Article  PubMed  Google Scholar 

  32. Menzel S, Holland T, Boes A, Spiegel H, Fischer R, Buyel JF. Downstream processing of a plant-derived malaria transmission-blocking vaccine candidate. Protein Expr Purif. 2018;152:122–30.

    Article  CAS  PubMed  Google Scholar 

  33. DiBerardinis LJ, et al. Guidelines for laboratory design: health, safety, and environmental considerations. In: Guidelines for laboratory design: health, safety, and environmental considerations. Wiley; 2013. p. 196–215.

    Chapter  Google Scholar 

  34. Coelho AC, García Díez J. Biological risks and laboratory-acquired infections: a reality that cannot be ignored in health biotechnology. Front Bioeng Biotechnol. 2015;3:56.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Charity JA, Klimaszewska K. Persistence of Agrobacterium tumefaciens in transformed conifers. Environ Biosaf Res. 2005;4(3):167–77.

    Article  CAS  Google Scholar 

  36. Menzel S, Holland T, Boes A, Spiegel H, Bolzenius J, Fischer R, Buyel JF. Optimized blanching reduces the host cell protein content and substantially enhances the recovery and stability of two plant-derived malaria vaccine candidates. Front Plant Sci. 2016;7(159):1–7.

    Google Scholar 

  37. Knödler M, et al. Seasonal weather changes affect the yield and quality of recombinant proteins produced in transgenic tobacco plants in a greenhouse setting. Front Plant Sci. 2019. https://doi.org/10.3389/fpls.2019.01245.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Rühl C, et al. A linear epitope coupled to DsRed provides an affinity ligand for the capture of monoclonal antibodies. J Chromatogr A. 2018;1571:55–64.

    Article  PubMed  Google Scholar 

  39. Bundesministerium für Ernährung, L.u.V., Bekanntmachung der Liste risikobewerteter Spender-und Empfängerorganismen für gentechnische Arbeiten. 2013.

  40. Regnard GL, et al. High level protein expression in plants through the use of a novel autonomously replicating geminivirus shuttle vector. Plant Biotechnol J. 2010;8(1):38–46.

    Article  CAS  PubMed  Google Scholar 

  41. Braun GB, et al. Urokinase-controlled tumor penetrating peptide. J Control Release. 2016;232:188–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tabor, S., Expression using the T7 RNA polymerase/promoter system. Current Protocols in Molecular Biology, 2001. Chapter 16: p. Unit16.2.

  43. Houdelet M, Galinski A, Holland T, Wenzel K, Schillberg S, Buyel JF. Animal component-free Agrobacterium tumefaciens cultivation media for better GMP-compliance increases biomass yield and pharmaceutical protein expression in Nicotiana benthamiana. Biotechnol J. 2017;12(4):1600721.

    Article  Google Scholar 

  44. Opdensteinen P, Meyer S, Buyel JF. Nicotiana spp. for the expression and purification of functional IgG3 antibodies directed against the staphylococcus aureus alpha toxin. Front Chem Eng. 2021. https://doi.org/10.3389/fceng.2021.737010.

    Article  Google Scholar 

  45. Buyel JF, Fischer POR. Cellulose-based filter aids increase the capacity of depth filters during the downstream processing of plant-derived biopharmaceutical proteins. Biotechnol J. 2015;10(4):584–91.

    Article  CAS  PubMed  Google Scholar 

  46. Hirano SS, et al. Lognormal distribution of epiphytic bacterial populations on leaf surfaces. Appl Environ Microbiol. 1982;44(3):695–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Limpert E, Stahel WA, Abbt M. Log-normal Distributions across the Sciences: Keys and Clues: On the charms of statistics, and how mechanical models resembling gambling machines offer a link to a handy way to characterize log-normal distributions, which can provide deeper insight into variability and probability—normal or log-normal: that is the question. Bioscience. 2001;51(5):341–52.

    Article  Google Scholar 

  48. Bohidar NR. A rebuttal to the “reply.” Drug Dev Ind Pharm. 1993;19(3):397–9.

    Article  CAS  Google Scholar 

  49. Kirkwood TBL. Geometric standard deviation: reply to Bohidar. Drug Dev Ind Pharm. 1993;19(3):395–6.

    Article  CAS  Google Scholar 

  50. Buyel JF, et al. The impact of Pseudomonas syringae type III effectors on transient protein expression in tobacco. Plant Biol. 2015;17(2):484–92.

    Article  CAS  PubMed  Google Scholar 

  51. Gengenbach BB, Opdensteinen P, Buyel JF. Robot cookies: plant cell packs as an automated high-throughput screening platform based on transient expression. Front Bioeng Biotechnol. 2020. https://doi.org/10.3389/fbioe.2020.00393.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Buyel JF, Fischer R. A juice extractor can simplify the downstream processing of plant-derived biopharmaceutical proteins compared to blade-based homogenizers. Process Biochem. 2015;50(5):859–66.

    Article  CAS  Google Scholar 

  53. Button DK, et al. A small, dilute-cytoplasm, high-affinity, novel bacterium isolated by extinction culture and having kinetic constants compatible with growth at ambient concentrations of dissolved nutrients in seawater. Appl Environ Microbiol. 1998;64(11):4467–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Scilipoti S, et al. Oxygen consumption of individual cable bacteria. Sci Adv. 2021;7(7):eabe1870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Myers JA, Curtis BS, Curtis WR. Improving accuracy of cell and chromophore concentration measurements using optical density. BMC Biophys. 2013;6(1):4.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Glazyrina J, et al. High cell density cultivation and recombinant protein production with Escherichia coli in a rocking-motion-type bioreactor. Microb Cell Fact. 2010;9(1):42.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Gibson B, et al. The distribution of bacterial doubling times in the wild. Proc Biol Sci. 1880;2018(285):20180789.

    Google Scholar 

  58. Morton ER, Fuqua C. Laboratory maintenance of Agrobacterium. Current protocols in microbiology, 2012. Chapter 1: p. Unit3D.1.

  59. Rosano GL, Ceccarelli EA. Recombinant protein expression in Escherichia coli: advances and challenges. Front Microbiol. 2014. https://doi.org/10.3389/fmicb.2014.00172.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pasini M, et al. Using promoter libraries to reduce metabolic burden due to plasmid-encoded proteins in recombinant Escherichia coli. New Biotechnol. 2016;33(1):78–90.

    Article  CAS  Google Scholar 

  61. Zhi L, et al. Effect of agrobacterium strain and plasmid copy number on transformation frequency, event quality and usable event quality in an elite maize cultivar. Plant Cell Rep. 2015;34(5):745–54.

    Article  CAS  PubMed  Google Scholar 

  62. Trivedi RN, et al. High-level production of plasmid DNA by Escherichia coli DH5α ΩsacB by introducing inc mutations. Appl Environ Microbiol. 2014;80(23):7154–60.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Platt TG, et al. Ecological dynamics and complex interactions of Agrobacterium megaplasmids. Front Plant Sci. 2014. https://doi.org/10.3389/fpls.2014.00635.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Opdensteinen P, Lobanov A, Buyel JF. A combined pH and temperature precipitation step facilitates the purification of tobacco-derived recombinant proteins that are sensitive to extremes of either parameter. Biotechnol J. 2020. https://doi.org/10.1002/biot.202000340.

    Article  Google Scholar 

  65. Ishikawa T, et al. Influence of pH on heat-induced aggregation and degradation of therapeutic monoclonal antibodies. Biol Pharm Bull. 2010;33(8):1413–7.

    Article  CAS  PubMed  Google Scholar 

  66. Garidel P, et al. Thermal and chemical unfolding of a monoclonal IgG1 antibody: application of the multistate Zimm-Bragg theory. Biophys J. 2020;118(5):1067–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Robinson M-P, et al. Efficient expression of full-length antibodies in the cytoplasm of engineered bacteria. Nat Commun. 2015;6:8072–8072.

    Article  CAS  PubMed  Google Scholar 

  68. Rodriguez C, et al. Efficient antibody assembly in E. coli periplasm by disulfide bond folding factor co-expression and culture optimization. Appl Biochem Biotechnol. 2017;183(2):520–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Buyel JF, Fischer R. Scale-down models to optimize a filter train for the downstream purification of recombinant pharmaceutical proteins produced in tobacco leaves. Biotechnol J. 2014;9(3):415–25.

    Article  CAS  PubMed  Google Scholar 

  70. Herrera-Estrella L, et al. Expression of chimaeric genes transferred into plant cells using a Ti-plasmid-derived vector. Nature. 1983;303(5914):209–13.

    Article  CAS  Google Scholar 

  71. Das A, et al. Promoters of Agrobacterium tumefaciens Ti-plasmid virulence genes. Nucleic Acids Res. 1986;14(3):1355–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kálai K, et al. Comparative study of constitutive and inducible promoters in tobacco. S Afr J Bot. 2008;74(2):313–9.

    Article  Google Scholar 

  73. Denkovskienė E, et al. Inducible expression of agrobacterium virulence gene VirE2 for stringent regulation of T-DNA transfer in plant transient expression systems. Mol Plant Microbe Interact. 2015;28(11):1247–55.

    Article  PubMed  Google Scholar 

  74. Rademacher T, et al. Plant cell packs: a scalable platform for recombinant protein production and metabolic engineering. Plant Biotechnol J. 2019;17:1560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Buyel JF. Numeric simulation can be used to predict heat transfer during the blanching of leaves and intact plants. 2016. Biochem Eng J. https://doi.org/10.1016/j.bej.2016.01.009.

  76. Niwa R, et al. Method for simple and rapid enumeration of total epiphytic bacteria in the washing solution of rice plants. Can J Microbiol. 2011;57(1):62–7.

    Article  CAS  PubMed  Google Scholar 

  77. Lv D, et al. Response of leaf-associated bacterial communities to primary acyl-homoserine lactone in the tobacco phyllosphere. Res Microbiol. 2012;163(2):119–24.

    Article  CAS  PubMed  Google Scholar 

  78. Chaudhry V, et al. Shaping the leaf microbiota: plant–microbe–microbe interactions. J Exp Bot. 2020;72(1):36–56.

    Article  PubMed Central  Google Scholar 

  79. Zhu X, et al. Bacterial contamination screening and interpretation for biological laboratory environments. Med Microecol. 2020;5:100021.

    Article  Google Scholar 

  80. Murray CJ, et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet. 2022;399(10325):629–55.

    Article  CAS  Google Scholar 

  81. Rossetto O, Montecucco C. Tables of toxicity of botulinum and tetanus neurotoxins. Toxins. 2019;11(12):686.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Wong JH, Ng TB. CHAPTER 20: toxins from basidiomycete fungi (mushroom): amatoxins, phallotoxins, and virotoxins. In: Kastin AJ, editor. Handbook of biologically active peptides. Burlington: Academic Press; 2006. p. 131–5.

    Chapter  Google Scholar 

  83. Inns RH, et al. Histochemical demonstration of calcium accumulation in muscle fibres after experimental organophosphate poisoning. Hum Exp Toxicol. 1990;9(4):245–50.

    Article  CAS  PubMed  Google Scholar 

  84. Walpole SC, et al. The weight of nations: an estimation of adult human biomass. BMC Public Health. 2012;12(1):439.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We wish to thank Dr. Richard M. Twyman for editing the manuscript and Patrick Opdensteinen for providing the original IgG1 plant expression plasmid.

Funding

Open access funding provided by University of Natural Resources and Life Sciences Vienna (BOKU). The Funding was provided by Deutsche Forschungsgemeinschaft (Grant No. 331065168), Fraunhofer-Gesellschaft (Grant No. 125-600164), Ministerium für Kultur und Wissenschaft des Landes Nordrhein-Westfalen (Garnt No. 423)

Author information

Authors and Affiliations

Authors

Contributions

MK cloned the constructs for the promoter activity tests and bioburden assays, performed the promoter activity and A. tumefaciens bioburden experiments, analyzed the data, and wrote the manuscript. PWR performed A. tumefaciens bioburden experiments and analyzed the data. JFB planed and designed parts of the experiments, conducted some of the bioburden experiments, analyzed the data, revised the manuscript, and secured funding for the projects. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Johannes Felix Buyel.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors have no conflict of interest to declare.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1

. Table S1: Promoter sequences used to generate the constructs for promoter activity tests. Table S2: Primers used to clone promoter variants. Table S3: Plasmids generated for promoter activity tests in E. coli and A. tumefaciens. Figure S1: Sampling positions for air contamination measurements. Distribution of selective and non-selective plates during the blanching processand infiltration processincluding three individual sampling positions.

Additional file 2

. Figure S2: Evaluation of recombinant protein accumulation in plant cell packs. Full size 2 western blots of triplicate PCP extracts using plasmids with different promotersfor the expression of IgG1and DsRed. DsRed was detected using a rabbit anti-His6 4 primary antibody and an alkaline phosphatase-labeled goat anti-rabbit secondary 5 antibody, whereas IgG1 was detected using an AP-labeled goat anti-human antibody. 6 CaMV35S – double-enhanced cauliflower mosaic virus 35S promoter with strong activity in 7 plants; bla – β-lactamase promoter with activity in bacteria; T7 – bacteriophage T7 promoter 8 with minimal activity in bacteria unless the corresponding polymerase is expressed.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Knödler, M., Reunious, P.W. & Buyel, J.F. Risk assessment and bioburden evaluation of Agrobacterium tumefaciens-mediated transient protein expression in plants using the CaMV35S promoter. BMC Biotechnol 23, 14 (2023). https://doi.org/10.1186/s12896-023-00782-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12896-023-00782-w

Keywords