Macrophages, especially primary macrophages, are challenging to transfect as they specialize in detecting molecules of non-self origin. We describe a protocol that allows highly efficient transfection of primary macrophages with mRNA generated from DNA templates such as plasmids.
Macrophages are phagocytic cells specialized in detecting molecules of non-self origin. To this end, they are equipped with a large array of pattern recognition receptors (PRRs). Unfortunately, this also makes macrophages particularly challenging to transfect as the transfection reagent and the transfected nucleic acids are often recognized by the PRRs as non-self. Therefore, transfection often results in macrophage activation and degradation of the transfected nucleic acids or even in suicide of the macrophages. Here, we describe a protocol that allows highly efficient transfection of murine primary macrophages such as peritoneal macrophages (PM) and bone marrow-derived macrophages (BMDM) with mRNA in vitro transcribed from DNA templates such as plasmids. With this simple protocol, transfection rates of about 50-65% for PM and about 85% for BMDM are achieved without cytotoxicity or immunogenicity observed. We describe in detail the generation of mRNA for transfection from DNA constructs such as plasmids and the transfection procedure.
Macrophages are phagocytic cells that specialize in detecting, ingesting and degrading microbes, apoptotic cells and cellular debris. Moreover, they help to orchestrate immune responses by secreting cytokines and chemokines and by presenting antigens to T cells and B cells. Macrophages also play important roles in numerous other processes, such as wound healing, atherosclerosis, tumorigenesis and obesity.
To be able to detect non-self molecules such as pathogen-associated molecular patterns (PAMPs) and out-of-place molecules such as damage-associated molecular patterns (DAMPs), macrophages are equipped with a large array of pattern recognition receptors (PRR)1. Unfortunately, this also makes macrophages particularly challenging to transfect2 as the transfection reagent3 and the transfected nucleic acids4,5,6,7 often are recognized by the PRRs as non-self. For this reason, transfection of macrophages using chemical or physical methods8 usually results in macrophage activation and degradation of the transfected nucleic acids or even in macrophage suicide via pyroptosis, a form of programmed lytic cell death triggered after recognition of cytosolic PAMPs/DAMPs such as DNA or foreign RNA9. Biological transfection of macrophages using viruses such as adenoviruses or lentiviruses as vectors is often more efficient, yet construction of such viral vectors is time-consuming and requires biosafety level 2 equipment10,11.
Thus, although macrophages are the subject of intensive research, analysis of their functions on the molecular level is hampered because one of the most important tools of molecular biology, the transfection of nucleic acid constructs for exogenous expression of proteins, is hardly applicable. This often forces researchers to use macrophage-like cell lines rather than bona fide macrophages. Applications for nucleic acid construct transfection include expression of mutated or tagged protein versions, overexpression of a specific protein, protein re-expression in a respective knockout background and expression of proteins from other species (e.g., Cre recombinase or guide RNA and Cas9 for targeted gene knockout).
Here, we describe a protocol that allows highly efficient transfection of (usually hard to transfect) primary macrophages, that is murine peritoneal macrophages (PM) and bone marrow-derived macrophages (BMDM) with mRNA generated from DNA templates such as plasmids. Importantly, the in vitro transcribed mRNA generated using this protocol contains the naturally occurring modified nucleosides 5-methyl-CTP and pseudo-UTP that reduce immunogenicity and enhance stability4,6,7,12,13. Moreover, the 5'-ends of the in vitro transcribed mRNA are dephosphorylated by Antarctic phosphatase to prevent recognition by the RIG-I complex14,15. This minimizes innate immune recognition of the in vitro transcribed mRNA. With our easy to perform protocol, transfection rates between 50-65% (peritoneal macrophages (PM)) and 85% (BMDM) are reached while, importantly, there is no cytotoxicity or immunogenicity observed. We describe in detail (i) how the immunologically silenced mRNA for transfection can be generated from DNA constructs such as plasmids and (ii) the transfection procedure itself.
Macrophage isolation from mice was performed in accordance with the Animal Protection Law of Germany in compliance with the Ethics Committee at the University of Cologne.
NOTE: Carry out all steps wearing gloves. Carry out all transfection steps under a laminar flow hood to prevent contamination of the cells. Before working with mRNA, clean all instruments such as pipettes and every surface with 70% ethanol and/or a RNAse-degrading surfactant (Table of Materials). Ensure that all reaction tubes are RNAse-free and sterile. Use only sterile, RNAase-free water for dilutions. Exclusively use pipette tips with filters. Change pipette tips after every pipetting step.
1. Generation of the DNA Template
NOTE: The DNA template for in vitro mRNA transcription using this protocol must contain a T7 promotor sequence to allow docking of the RNA polymerase. If the plasmid containing the DNA sequence of the protein of interest already contains a correctly orientated T7 promotor sequence directly upstream of the sequence of interest, linearization of the plasmid (see step 1.1.) needs to be performed. Otherwise, attach a T7 promotor to the sequence of interest by polymerase chain reaction (PCR, see step 1.2.).
2. mRNA Generation
3. mRNA Purification
4. Macrophage Preparation
5. Transfection of Macrophages with mRNA
We have successfully used this protocol to generate mRNA encoding for FLAG-tagged NEMO and IKKβ variants for transfection of primary macrophages16. The plasmids encoding for FLAG-tagged wild-type (NEMOWT) and C54/347A mutant NEMO (NEMOC54/374A) (see the Table of Materials) already contain a T7 promotor in the correct orientation (Figure 1A). Thus, we only had to linearize the plasmids to generate DNA templates for in vitro transcription. To this end, 10 µg of plasmid DNA were digested with 5 µL Xbal resulting in linearization of the plasmid due to a single cut 3' of the stop codon. Complete linearization of the plasmid DNA was verified by agarose gel electrophoresis (Figure 1B).
The plasmids encoding for FLAG-tagged wild-type (IKKβWT) and S177/181E mutant IKKβ (IKKβS177/181E) do not contain a T7 promotor. Therefore, we have attached a T7 promotor by PCR to generate the DNA templates for in vitro transcription (Figure 2A). Generation of a specific PCR product, i.e., a single product of correct size, was verified by agarose gel electrophoresis (Figure 2B).
After in vitro transcription using the respective DNA templates, we verified generation of a single mRNA product of correct size and poly(A) tailing by agarose gel electrophoresis under denaturing conditions (Figure 3).
To verify that mRNA generated using this protocol does enable the transfection of primary macrophages (see Figure 4A,B for flow cytometric analyses of immunomagnetic enrichment of PM and differentiation status of BMDM), we have generated mRNA encoding for eGFP (Figure 5A-C) and analyzed transfection efficiency by flow cytometry (Figure 6A). 100,000 PM or 50,000 BMDM per well of an untreated microtiter plate were transfected with 50, 100 or 200 ng of eGFP mRNA for 6, 9 or 24 h. In both PM and BMDM, high levels of eGFP expression could be detected at 6 h after transfection. Transfection rate increased with the amount of transfected mRNA and was highest for 200 ng mRNA (Figure 6B,C). For PM, transfection rate reached about 50-65% at 6 to 9 h after transfection (Figure 6B). At 24 h after transfection, transfection rate was substantially lower indicating expiring eGFP expression. Thus, PM should not be transfected overnight. For BMDM, transfection rate reached about 80-85% (Figure 6C). The drop in transfection rate after 24 h was much less pronounced in BMDM. Thus, BMDM can be transfected overnight. In both PM (Figure 6D,E) and BMDM (Figure 6F,G), expression level of eGFP in transfected cells increased in a time- and dose-dependent manner. Importantly, the transfection procedure did not induce lytic or apoptotic cell death as there was no increase in propidium iodide-positive (PI) or annexin V-positive macrophages after transfection (Figure 7A,B).
Transfection efficiency of the mRNAs encoding for FLAG-tagged NEMO or IKKβ variants was analyzed by immunofluorescence microscopy. 300,000 PM per well of a 12-well plate were transfected with 300 ng of respective mRNA for 6 h. Transfection rate was about 60% for NEMO mRNAs and about 55% for IKKβ mRNAs (Figure 8A,B). Thus, their transfection rates were similar to that of eGFP mRNA indicating a general transfection rate of about 55% for PM.
We have also used this protocol to generate mRNA encoding for Cre recombinase. Transfection of 400,000 BMDM from NEMOflox/flox mice17 per well of a 12-well plate with 400 ng of Cre recombinase mRNA resulted in almost complete depletion for NEMO protein after 48 h (Figure 9) indicating highly efficient transfection of the BMDM.
PM and BMDM did not secrete any detectable amounts of IL-1β, IL-6 and TNF after transfection (Figure 10A,B). Moreover, the NF-kB and MAPK signaling pathways were not activated after transfection16 indicating that the transfection procedure does not activate proinflammatory signaling. We also have not observed any functional alterations of transfected macrophages in comparison to untransfected macrophages16.
Figure 1: Linearization of NEMO-encoding plasmids already containing a T7 promotor in the correct orientation. (A) Sequence excerpt of the plasmid encoding for FLAG-tagged NEMOWT or NEMOC54/347A. A T7 promotor in correct orientation and close to the KOZAK sequence and start codon is already present. The T7 promotor region, the coding sequence (CDS) for the FLAG tag and NEMO, the start and stop codons and the restriction site for linearization with XbaI are color-coded. (B) Representative 1% agarose gel showing the plasmids before and after linearization with Xbal; 1 µL of untreated, linearized or purified linearized plasmid DNA was loaded per lane. Please click here to view a larger version of this figure.
Figure 2: T7 promotor attachment to IKKß-encoding plasmids by PCR. (A) Sequence excerpt of the plasmid encoding for FLAG-tagged IKKβWT or IKKβS177/181E. The plasmids do not contain a suitable T7 promotor, which is therefore attached by PCR. Location, orientation and sequence of the forward primer (containing the T7 promotor sequence to be added) and the reverse primer are indicated by the arrows. The T7 promotor region, the CDS for the FLAG tag and IKKβ and the start and stop codons are color-coded. (B) Representative 1% agarose gel verifying generation of a single PCR product of correct size using the primers indicated above. 1 µL of the purified PCR product was loaded per lane. Please click here to view a larger version of this figure.
Figure 3: mRNA synthesis from NEMO- and IKKβ-encoding DNA templates. (A) Representative denaturing 1.2% agarose gel containing 0.7% formaldehyde showing the purified mRNA of NEMO and IKKβ constructs before and after poly(A) tailing. 2 µL of NEMOWT, NEMOC54/347A, IKKβWT and IKKβS177/181E mRNA were loaded per lane. A 32 µL ssRNA ladder was loaded for RNA length determination. Please click here to view a larger version of this figure.
Figure 4: Flow cytometric analyses of the immunomagnetic enrichment of PM and the differentiation status of BMDM. (A) The percentage of F4/80+/CD11b+ PM in the peritoneal lavage before and after immunomagnetic enrichment was analyzed by flow cytometry. (B) Expression of F4/80 and CD11b by BMDM after 6 days of differentiation was verified by flow cytometry. 10,000 or 5,000 cells were counted per sample, respectively. Data are shown as mean ± SEM of n = 3 independent experiments, each. * P < 0.05, ** P < 0.01, *** P < 0.001 and **** P < 0.0001 by Student's t-test. Please click here to view a larger version of this figure.
Figure 5: mRNA synthesis with poly(A) tailing of eGFP. (A) Sequence excerpt of the plasmid encoding for eGFP. The plasmid does not contain a suitable T7 promotor, which is therefore attached by PCR. Location, orientation and sequence of the forward primer (containing the T7 promotor sequence to be added) and the reverse primer are indicated by the arrows. The T7 promotor region, the CDS for eGFP and the start and stop codons are color-coded. (B) Representative 1% agarose gel showing the purified amplicon after PCR using the primers indicated above. 1 µL of the purified PCR product was loaded per lane. (C) Representative denaturing 1.2% agarose gel containing 0.7% formaldehyde showing the purified mRNA of eGFP before and after poly(A) tailing. 2 µL of eGFP mRNA were loaded per lane. A 32 µL ssRNA ladder was loaded for RNA length determination. Please click here to view a larger version of this figure.
Figure 6: Highly efficient transfection of both peritoneal macrophages and BMDM with eGFP mRNA. Macrophages were incubated for 6, 9 or 24 h with 50, 100 or 200 ng of eGFP mRNA complexed to jetMESSENGER or with jetMESSENGER alone (mock) and then analyzed by flow cytometry. (A) Gating strategy used to define the populations of viable macrophages, viable eGFP+ macrophages and PI+ (i.e., dead) macrophages. Representative data for transfection with 200 ng mRNA for 6 h are shown. (B,C) Transfection rates of (B) PM and (C) BMDM were determined by analyzing the percentage of viable eGFP-positive cells by flow cytometry (n = 5-7 and n = 4-5 independent experiments, respectively). (D-G) Expression levels of eGFP were determined by analyzing the mean fluorescence intensity (MFI) of viable (D) PM and (F) BMDM and of the viable and eGFP-positive subpopulation of (E) PM and (G) BMDM (n = 5-7 and n = 4-5 independent experiments, respectively). 10,000 cells were counted per sample. Data are shown as mean ± SEM. n.s. = not significant; * P < 0.05, ** P < 0.01, *** P < 0.001 and **** P < 0.0001 by Student's t-test. Please click here to view a larger version of this figure.
Figure 7: Transfection does not induce lytic or apoptotic cell death. Transfection-induced cell death of (A) PM and (B) BMDM was determined by analyzing the percentage of PI-positive and annexin V-positive cells (n = 5-7 and n = 4-5 independent experiments, respectively). The percentage of dead macrophages present in untransfected samples (some degree of cell death was caused by physical detachment of macrophages from the wells despite usage of non-treated plates) was subtracted from that in the respective transfected samples to only take into account cell death induced by the transfection procedure. The gating strategy used to define the populations of PI+, annexin V+ and PI+/annexin V+ macrophages is shown for a representative data set of PM transfected with 200 ng mRNA for 6 h are shown. Staurosporine (50 µM for 1 h) was used as positive control. 10,000 cells were counted per sample. Data are shown as mean ± SEM. n.d. = not detectable. Please click here to view a larger version of this figure.
Figure 8: Transfection rates using mRNA encoding for NEMO and IKK constructs. Transfection rates using mRNA encoding for (A) NEMO constructs and (B) IKK constructs were quantified by immunofluorescence microscopy (n = 4 independent experiments). Scale bar = 4 µm. Data are shown as mean ± SEM. n.t. = not transfected, n.s. = not significant; * P < 0.05, ** P < 0.01, *** P < 0.001 and **** P < 0.0001 by Student's t-test. Please click here to view a larger version of this figure.
Figure 9: Transfection of NEMOfl/fl BMDM with mRNA encoding for Cre recombinase results in almost complete deficiency for NEMO. BMDM from NEMOfl/fl mice were transfected with mRNA encoding Cre recombinase for 48 h. Deficiency for NEMO protein as a result of Cre-mediated knockout was assessed by western blot using specific antibodies recognizing NEMO or β-actin and quantified by densitometry (n = 5 independent experiments). Data are shown as mean ± SEM. * P < 0.05, ** P < 0.01, *** P < 0.001 and **** P < 0.0001 by Student's t-test. Please click here to view a larger version of this figure.
Figure 10: Transfection does not induce a proinflammatory response. (A) PM and (B) BMDM were transfected with mRNA encoding for NEMOWT or NEMOC54/347A. As positive control, macrophages were infected with Listeria monocytogenes at multiplicity of infection of 1 or stimulated with 5 µg/mL LPS or poly(I:C) for 5 h (PM) or 24 h (BMDM). Secretion of IL-1 β, IL-6, and TNF into the supernatant was quantified by ELISA (n = 3 independent experiments). Data are shown as mean ± SEM. n.t. = not transfected, n.d. = not detectable. Please click here to view a larger version of this figure.
Here we present a protocol for highly efficient transfection of usually hard-to-transfect primary macrophages with in vitro transcribed mRNA. Importantly, transfection of the macrophages using this protocol does not induce cell death or activate proinflammatory signaling indicating that neither the transfection reagent nor the transfected mRNA are recognized as non-self.
The quality of the mRNA is of key importance for successful transfection of macrophages using this protocol. Thus, great care should be taken that the mRNA does not come into contact with RNAses (for example, through contaminated buffers or vials). If in doubt, check for mRNA degradation (for example, by agarose gel electrophoresis). Transfection rate and expression level were already maximal at 6 h after transfection. Therefore, it is possible that the protein of interest is expressed at levels sufficient for analysis at earlier time points after transfection. We have not tested this, though. Furthermore, transfecting larger quantities of mRNA may further increase transfection rate and/or expression level. However, transfecting larger quantities of mRNA may potentially also lead to some degree of immunogenicity or even cytotoxicity. The complete lack thereof is one of the main advantages of this protocol. Thus, if larger quantities of mRNA are to be transfected, immunogenicity and cytotoxicity have to be carefully evaluated.
Expression level clearly correlated with the amount of transfected mRNA. Yet, re-expression of NEMO in NEMO-deficient BMDM resulted in similar NEMO protein expression as in wildtype BMDM16 indicating that mRNA transfection using this protocol does not lead to substantial overexpression of the protein of interest. Thus, this protocol may not be suited for studying the consequences of overexpression of the protein of interest. We rather consider this an advantage, however, as overexpression of a protein often alters its behavior.
The main limitation of mRNA transfection, in general, is that it only results in transient expression of the protein of interest (because the mRNA generated and transfected using this protocol will be subject to normal turnover). PM seem to express the protein of interest for a shorter period of time as compared to BMDM. Thus, in contrast to BMDM, PM should not be transfected overnight. How long a given protein of interest will be expressed will vary (as both the stability of the mRNA and that of the protein will vary). We nevertheless recommend performing the experiment of choice on the same day as the transfection. If expression of the protein of interest is required for longer periods of time, the cells probably can be transfected multiple times (e.g., every day as described in18).
We have successfully used the protocol described here to transfect murine PM and BMDM and mouse embryonic fibroblasts (MEFs)16. In theory, the mRNA generated using this protocol can be used to transfect any given cell type of any mammalian species. The optimal mRNA transfection reagent and content of modified nucleosides may differ for other cell types, though.
Promising future modifications include the inclusion of the 5'- and 3'-UTRs. Most pre-existing plasmids do only contain the CCDS of the protein of interest but not the 5'- and 3'-UTRs. Their inclusion (as described in19) may further enhance mRNA stability and expression. Also, attaching epitope tags to the protein of interest by simply including the sequence of the epitope tag in the forward or reverse primer (for N- and C-terminal epitope tagging, respectively), should be possible.
The authors have nothing to disclose.
This work was supported by the Deutsche Forschungsgemeinschaft (SFB 670).
5-methyl-CTP (100 mM) | Jena Biosience | NU-1138S | stored at -20 °C |
Antarctic phosphatase | New England BioLabs | M0289 | stored at -20 °C |
Antarctic phosphatase reaction buffer (10X) | New England BioLabs | B0289 | stored at -20 °C |
anti-NEMO/IKKγ antibody | Invitrogen | MA1-41046 | stored at -20 °C |
anti-β-actin antibody | Sigma-Aldrich | A2228 | stored at -20 °C |
Petri dishes 92,16 mm with cams | Sarstedt | 821,473 | stored at RT |
CD11b Microbeads mouse and human | Miltenyi Biotec | 130-049-601 | stored at 4 °C |
Cre recombinase + T7-Promotor forward primer | Sigma-Aldrich | 5′-GAAATTAATACGACTCACTATA GGGGCAGCCGCCACCATGTCC AATTTACTGACCGTAC-3´, stored at -20 °C |
|
Cre recombinase + T7-Promotor reverse primer | Sigma-Aldrich | 5′-CTAATCGCCATCTTCCAGCAGG C-3′, stored at -20 °C |
|
DNA purification kit: QIAquick PCR purification Kit | Qiagen | 28104 | stored at RT |
eGFP + T7-Promotor forward primer | Sigma-Aldrich | 5´-GAAATTAATACGACTCACTATA GGGATCCATCGCCACCATGGTG AGCAAGG-3´, stored at -20 °C |
|
eGFP + T7-Promotor reverse primer | Sigma-Aldrich | 5´-TGGTATGGCTGATTA TGATCTAGAGTCG-3´, stored at -20 °C |
|
Fast Digest buffer (10X) | Thermo Scientific | B64 | stored at -20 °C |
FastDigest XbaI | Thermo Scientific | FD0684 | stored at -20 °C |
high-fidelity polymerase with proofreading: Q5 High-Fidelity DNA-Polymerase | New England Biolabs Inc | M0491S | stored at -20 °C |
IKKβ + T7-Promotor forward primer | Sigma-Aldrich | 5′-GAAATTAATACGACTCACTATA GGGTTGATCTACCATGGACTACA AAGACG-3′, stored at -20 °C |
|
IKKβ + T7-Promotor reverse primer | Sigma-Aldrich | 5′-GAGGAAGCGAGAGCT-CCATCTG-3′, stored at -20 °C | |
in vitro mRNA transcription kit: HiScribe T7 ARCA mRNA kit (with polyA tailing) | New England BioLabs | E2060 | stored at -20 °C |
LS Columns | Miltenyi Biotec | 130-042-401 | stored at RT |
MACS MultiStand | Miltenyi Biotec | 130-042-303 | stored at RT |
mRNA transfection buffer and reagent: jetMESSENGER | Polyplus transfection | 409-0001DE | stored at 4 °C |
Mutant IKKβ IKK-2S177/181E plasmid | Addgene | 11105 | stored at -20 °C |
Mutant NEMOC54/347A plasmid | Addgene | 27268 | stored at -20 °C |
pEGFP-N3 plasmid | Addgene | 62043 | stored at -20 °C |
poly(I:C) | Calbiochem | 528906 | stored at -20 °C |
pPGK-Cre plasmid | F. T. Wunderlich, H. Wildner, K. Rajewsky, F. Edenhofer, New variants of inducible Cre recombinase: A novel mutant of Cre-PR fusion protein exhibits enhanced sensitivity and an expanded range of inducibility. Nucleic Acids Res. 29, 47e (2001). stored at -20 °C | ||
pseudo-UTP (100 mM) | Jena Biosience | NU-1139S | stored at -20 °C |
QuadroMACS Separator | Miltenyi Biotec | 130-090-976 | stored at RT |
Rat-anti-mouse CD11b antibody, APC-conjugated | BioLegend | 101212 | stored at 4 °C |
Rat-anti-mouse F4/80 antibody, PE-conjugated | eBioscience | 12-4801-82 | stored at 4 °C |
recombinant M-CSF | Peprotech | 315-02 | stored at -20 °C |
RNA purification kit: MEGAclear transcription clean-up kit | ThermoFisher Scientific | AM1908 | stored at 4 °C |
RNAse-degrading surfactant: RnaseZAP | Sigma-Aldrich | R2020 | stored at RT |
ultrapure LPS from E.coli O111:B4 | Invivogen | stored at -20 °C | |
Wild type IKKβ plasmid | Addgene | 11103 | stored at -20 °C |
Wild type NEMO plasmid | Addgene | 27268 | stored at -20 °C |