Abstract Hemorrhagic fever with renal syndrome (HFRS) occurs throughout Eurasia with considerable morbidity and mortality. Currently, the absence of specific treatments or effective antiviral drugs for hantavirus infection makes developing safe and effective vaccines a high priority. Here, we report the development of three novel nucleic acid vaccine candidates, mRNA, naked DNA, and DNA encapsulated in lipid nanoparticles, encoding the glycoproteins of the Hantaan virus (HTNV). To comprehensively evaluate the potential of candidate HTNV nucleic acid vaccines in preventing HFRS, we focus on evaluating their immunogenicity and efficacy in mice and comparing them with an inactivated vaccine as the benchmark. Our findings reveal that all candidate vaccines activated instant and sustained immune responses, offering comparable in vivo protective efficacy to the inactivated vaccines. Notably, compared to the inactivated vaccine, mRNA vaccine induced stronger virus-specific T-helper 1 cell immune response, while DNA-LNP elicited higher levels of neutralizing antibodies in mice. These results mark a significant step in developing nucleic acid vaccines for HTNV, suggesting that sequential immunization with DNA and mRNA vaccines could further amplify the advantages of nucleic acid vaccines. Subject terms: RNA vaccines, DNA vaccines Introduction Old World hantaviruses are predominantly found in Europe and Asia, where they usually cause hemorrhagic fever with renal syndrome (HFRS), resulting in symptoms including fever, bleeding tendency, and renal impairment^[64]1–[65]4. Rodents are the primary natural hosts of hantaviruses. Human infection is primarily through contact infection or aerosolized excreta inhalation^[66]2,[67]5. HFRS leads to considerable morbidity and mortality and thus represents a significant public health concern. Infections caused by the Hantaan virus (HTNV) strain of HFRS are especially severe, with reported mortality rates ranging from 5% to 15%^[68]6,[69]7. Currently, there are no specific treatment methods or effective antiviral drugs for hantavirus infection^[70]8. Therefore, vaccination is the most viable strategy for disease prevention. In response to HFRS, both China and South Korea have authorized the use of inactivated vaccines developed either from mouse brain or cell cultures^[71]9. These vaccines demonstrate strong immunogenicity and are proven effective in reducing the incidence of HFRS^[72]10. However, existing studies indicate that inactivated vaccines require multiple doses to achieve sufficient protection, and their safety in the elderly and children still requires further validation^[73]11,[74]12. The parallel development of multiple vaccine technologies offers more choices, which is important for enhancing the ability to tackle new pathogens and their variants. Besides, it is believed various vaccine types can meet the specific needs of diverse demographic groups, and respond rapidly to global health emergencies^[75]13. Hantavirus infection has been noted to fluctuate periodically, and recent observations suggest an expansion in the range of its natural hosts^[76]14,[77]15. Consequently, HFRS is garnering increasing attention and underscores the importance of developing new, safe, and effective vaccines against Hantavirus. Nucleic acid vaccines, including DNA and mRNA vaccines, can enter human cells and produce endogenous antigens like viruses. By presenting antigens to the immune system via dual pathways, nucleic acid vaccines activate both cellular and humoral immune responses for enhanced protection^[78]16. And nucleic acid vaccines have become a swift and flexible platform, particularly valuable in emergency response scenarios^[79]17,[80]18. mRNA vaccine represents a significant advancement in nucleic acid vaccine technology^[81]19. With the clinical application of mRNA vaccines in the prevention of COVID-19, mRNA vaccine has demonstrated significant potential in combating various virulent pathogens^[82]20–[83]23. Yet, the efficacy and suitability of mRNA vaccine in addressing HTNV is still an area of uncertainty. Notably, the advancement of mRNA vaccine technology has benefited from pseudouridine modification and the delivery system of lipid nanoparticles (LNP). Recent studies indicate that LNP is crucial in maintaining vaccines at the injection site, allowing for prolonged antigen presentation and immunostimulation of innate immune cells^[84]24. While LNP as effective antigen-delivery systems have been thoroughly exploited in the field of mRNA vaccines, further research is needed to assess their potential for DNA vaccination against various infectious agents. Leveraging the proven potential of nucleic acid vaccine technology in combating viral infections, the pursuit of developing a safe and effective vaccine specifically for Hantavirus is paramount, which would be a valuable addition against this serious viral threat. Hantavirus is classified within the group of single-stranded negative-sense RNA viruses^[85]7. Similar to other members of the Bunyaviridae family, its viral envelope glycoprotein (GP) plays a critical role in the virus’s attachment to and entry into host cells and serves as the primary target for the immune system to recognize and neutralize the virus^[86]25. The envelope GP of Hantavirus is currently reported with multiple neutralizing epitopes which induce the production of neutralizing antibodies to resist viral attacks^[87]26,[88]27. Consequently, the HTNV GP is a preferred candidate for HFRS vaccine development. In the present research, we report the development of three novel candidate nucleic acid vaccines targeting the GP and evaluate the immunogenicity of the candidate vaccines from humoral immune response, cellular immune response, and in vivo protection. Result Construction and identification of candidate vaccines M segment of HTNV 76–118 strain was selected as the target antigen encoding sequence for three nucleic acid vaccines. We successfully constructed the DNA candidate vaccine pVAX1-GP[HTNV] and the vector pGEM-UTR-GP[HTNV] for mRNA in vitro transcription. All relevant vectors were verified by Sanger sequencing and enzyme digestion electrophoresis (Fig. [89]1a). Here, we designed an mRNA platform encoding the full-length HTNV GP protein (Fig. [90]1b). The N-terminus of this protein carries an HTNV GP signal peptide for endoplasmic reticulum translocation and secretion. The desired mRNA molecules were synthesized through in vitro transcription of the pGEM-UTR-GP[HTNV] vector, followed by further capping modification. Microfluidic capillary electrophoresis of RNA revealed a single peak at 4000 nt, indicating its high purity and integrity (Fig. [91]1c). And the dsRNA content in the mRNA-GP[HTNV] vaccine candidate was 0.07%. We developed a vaccine platform based on DNA encapsulated in LNP (DNA-LNP) for in vivo delivery (Fig. [92]1d). DNA (pVAX1-GP[HTNV])-LNP, manufactured using a microfluidic flow-focusing strategy, exhibited an average particle size of 98.83 nanometers (Fig. [93]1e) with an encapsulation efficiency exceeding 70% (Table [94]S4). Transmission electron microscopy (TEM) analysis revealed that DNA-LNP particles exhibited a uniform solid spherical morphology (Fig. [95]1f). mRNA-GP[HTNV] is encapsulated into in vivo-jetRNA, forming mRNA-lipo[jet]. The average particle size of the mRNA-lipo[jet] was 144.57 nanometers (Fig. S[96]2a), with an encapsulation efficiency of 76% (Table [97]S4). TEM shows that these particles are uniformly shaped like solid spheres (Fig. S[98]2b). During the expression of HTNV GP, its N-terminal Gn undergoes autophagy^[99]28,[100]29. Hence, only the expression of Gc could be detected by western blot analysis in transfected cells. Major protein bands containing Gc were detected, with a molecular weight of approximately 61 kDa (Fig. [101]1g and Fig. S[102]3). We confirmed transcription of DNA and transfection of mRNA through qPCR, and the transcription level between Gn and Gc exhibits no substantial differences (Fig. [103]1h). HTNV GP was localized to the cytoplasm in transfected cells, as observed by immunofluorescence microscopy (Fig. [104]1i). Fig. 1. Vaccine design and characterization of the expressed antigens. [105]Fig. 1 [106]Open in a new tab a Plasmids (pVAX1-GP[HTNV], and pGEM-UTR-GP[HTNV]) identified by 1% agarose gel electrophoresis. b Structure of mRNA vaccine expressing the HTNV GP. UTR, untranslated region; SP, signal peptide. c Liquid capillary electropherograms of in vitro-transcribed mRNA. Peaks represent mRNA of HTNV GP (4000 nt) and an indicative marker (20 nt). d Schematic diagram of DNA-LNP, encoding the full-length GP protein of HTNV. e Representative intensity-size graph of HTNV DNA-LNP measured by dynamic light-scattering method. f TEM image of HTNV DNA-LNP. g GP protein expression in HEK293T cells. Cells were transfected with HTNV GP-encoding DNA or mRNA (2 μg/mL), and immunoblotting was performed at 24 h after transfection. The GP protein was detected with a rabbit monoclonal antibody against the flag. h qRT-PCR analyses for HTNV Gn and Gc expression following transfection of HTNV GP-encoding DNA or mRNA (2 μg/mL) into HEK293T cells, β-actin was used as control. i Immunofluorescence assay of GP protein expression in HEK293T cells 24 h post-transfection. GP (red) and DNA (blue). Scale bar, 30 μm. Candidate vaccines induced humoral immune protection in mice We conducted experiments on a group of immunocompetent female BALB/c mice to assess the immunogenicity and effectiveness of candidate vaccines. Following the strategy illustrated in Fig. [107]2a, the mice received two doses of either inactivated vaccine, naked DNA, DNA-LNP, or mRNA-lipo[jet], administered via intramuscular injection. PBS and empty LNP served as control groups. The instant immune responses to each vaccine group were evaluated accordingly. In addition, no local inflammatory or other adverse reactions were observed at the injection sites during the observation period post-immunization. HTNV-specific IgG antibodies in the sera of the immunized mice were quantified using ELISA. Following both prime and boost immunizations, four candidate vaccine groups successfully induced the generation of antigen-specific IgG antibodies. Serum samples from mice immunized with PBS and empty LNP did not exhibit specific IgG detection. Notably, the group receiving the DNA-LNP vaccines demonstrated significantly higher specific IgG titers compared to the inactivated vaccines after the initial immunization (Fig. [108]2b). Fig. 2. Schematic diagram of immunization and evaluation of humoral immune responses. [109]Fig. 2 [110]Open in a new tab a Schematic diagram of immunization, sample collection, and challenge schedule (the prone mouse element was created by Figdraw). b HTNV-specific IgG antibody titers were determined by ELISA (n = 8). The red dashed lines indicate the limit of detection (LOD). The LOD of the HTNV-specific antibody ELISA is 100. c 50% virus-neutralization titers determined using infectious HTNV (n = 6) and the LOD of the HTNV neutralization assay is 10. Data are represented as the mean ± SEM. Statistical differences were analyzed using one-way ANOVA with multiple comparison test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Neutralizing antibody levels serve as a pivotal indicator of HTNV vaccine efficacy. We conducted HTNV-neutralizing antibody tests on sera from mice immunized using a fixed virus (100 plaque forming units (PFUs)) serum dilution method. The results showed that after two doses of vaccine immunization, the PBS group and the empty LNP group induced average neutralizing titers of 1:11 and 1:13, respectively. Meanwhile, the inactivated vaccine, naked DNA, DNA-LNP, and mRNA-lipo[jet] groups induced mean neutralizing titers of 1:79, 1:75, 1:118 and 1:65, respectively. Significantly, the HTNV-neutralizing antibody titers in sera from mice vaccinated with the four candidate vaccine groups were significantly higher than the control group. Furthermore, the DNA-LNP group induced significantly higher neutralizing antibodies than the inactivated vaccine group. Compared to naked DNA, the DNA-LNP group demonstrated a significant increase in neutralizing antibody titers, indicating that LNP-mediated delivery enhances the immune response for the same DNA dosage (Fig. [111]2c). Overall, the sera from mice immunized with the four candidate vaccines effectively neutralized the infectious HTNV and initiated antiviral protective immunity, featured as the DNA-LNP yielding the highest serum neutralizing titers. Candidate vaccines elicited T cell responses in mice T cells play a crucial role in immune protection against hemorrhagic fever viruses. Therefore, we investigated the T cell responses induced by two doses of each candidate vaccine through intramuscular injection and whether these immunizations elicited HTNV-specific T cell immune responses in mice. 24 days after booster vaccination, we characterized the antigen-specific responses of splenocytes in immunized mice. Upon stimulation with a mixture of ten HTNV GP 15-mer peptides, as determined by ELISpot assay, four candidate vaccine groups showed a significant increase in HTNV GP-specific T lymphocytes secreting IFN-γ compared to the control group, indicating the successful induction of antiviral T-helper 1 (Th1) cell-mediated immune responses. Furthermore, naked DNA, DNA-LNP, and mRNA-lipo[jet] vaccines induced significantly higher levels of IFN-γ-secreting T lymphocytes than the inactivated vaccine. DNA-LNP resulted in significantly high IFN-γ levels in the spleen in comparison with the naked DNA vaccine, suggesting that DNA vaccine-induced T cell immune responses were further enhanced under LNP delivery (Fig. [112]3a and Fig. S[113]4a). Furthermore, ELISpot experiments revealed a significant increase in the secretion of interleukin-4 (IL-4) in the splenocytes of mice immunized with inactivated vaccine, DNA-LNP, and mRNA-lipo[jet] vaccines, as compared to mice receiving the control vaccines. And mRNA-lipo[jet] vaccine induced a significantly higher number of IL-4-secreting T lymphocytes compared to the inactivated vaccine (Fig. [114]3b). Our results indicate that naked DNA, DNA-LNP, and mRNA-lipo[jet] vaccines successfully induced Th1-biased, HTNV-specific cellular immune responses. Yet the inactivated vaccine did not exhibit a clear Th1/Th2 bias after two doses of immunization (Fig. [115]3c). We performed single peptide verification on ten GP 15-mer peptides by ELISpot, and the results revealed that stimulation with the 15-mer peptides IALGPYRVQVVYERS, SYCMTGVLIEGKCFV, and KKVMATIDSFQSFNT notably enhanced the IFN-γ secreting T lymphocyte response across all immunization groups (Fig. [116]3d). Additionally, stimulation with KKVMATIDSFQSFNT led to a remarkable increase in the IL-4 secreting T lymphocyte response in all groups. In summary, all ten 15-mer peptides could induce splenocyte responses specific to GP, qualifying as GP-specific dominant peptides for evaluating GP-based HTNV vaccine efficacy. Fig. 3. Analysis of T cells after immunization. [117]Fig. 3 [118]Open in a new tab a HTNV GP-specific IFN-γ spot-forming units (SFCs) determined by ELISpot (n = 8). b HTNV GP-specific IL4 SFCs determined by ELISpot (n = 6). c Scatter plot showing the correlation of HTNV GP-specific IL-4 and IFN-γ secreting cells by ELISpot (n = 6). d IFN-γ and IL-4 SFCs stimulated by single 15-mer peptide of HTNV GP by ELISpot assay after the second immunization and presented as a heatmap of Z scores (n = 8). e Gating strategy to identify cytokine secretion (take the IL-2 release from CD4^+ T cells for example). f IFN-γ, IL-2, IL-4 or Granzyme B release from CD4^+ and CD8^+ T cells by flow cytometry (n ≥ 5). g Gating strategy to identify effector memory T (Tem) cells (CD44^+ CD62L^-) in the spleen (take the CD4^+ Tem cells in splenocytes for example). h HTNV GP-specific CD4^+ (n ≥ 6) and CD8^+ (n ≥ 4) Tem cells in splenocytes were detected by flow cytometry. Data are represented as the mean ± SEM. Statistical differences were analyzed using one-way ANOVA with multiple comparison test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. Further confirmation of Th1-type cell-mediated immune response was obtained by flow cytometry, which detected CD4^+ and CD8^+ T lymphocytes secreting the cytokines IFN-γ or IL-2 in immunized mice following mixed stimulation (Fig. [119]3f). The results showed that splenocytes from mice immunized with the four candidate vaccine groups exhibited a significant increase in the secretion of both IFN-γ and IL-2 by CD4^+ and CD8^+ T cells compared to the control mice. The cellular response patterns for naked DNA and DNA-LNP candidate vaccines were similar, while compared to mice immunized with naked DNA, mice immunized with DNA-LNP produced stronger secretion of IL-2 by CD8^+ T cells (Fig. [120]3f). Furthermore, compared to the control group, mice immunized with inactivated vaccine and mRNA-lipo[jet] showed a significant increase in the IL-4 secretion of CD4^+ and CD8^+ T cells (Fig. [121]3f). Additionally, CD8^+ T cells secreting IL-4 were significantly increased in mice immunized with DNA-LNP when compared to the control group. Compared to mice receiving the control group, mice from four candidate vaccine groups showed a significant increase in granzyme B secretion by CD4^+ and CD8^+ T cells, indicating increased cytotoxic activity. Additionally, CD8^+ T cells secreting granzyme B were significantly increased in the mRNA-lipo[jet] vaccine group compared to the inactivated vaccine group (Fig. [122]3f). The flow cytometry results showed that as compared to the control group, splenocytes from immunized mice in all experimental groups exhibited a significant increase in virus-specific CD4^+ effector memory T (Tem) cells following stimulation with the HTNV GP peptide pool (Fig. [123]3h). Moreover, a significant increase in CD8^+ Tem cells was observed in the inactivated vaccine, naked DNA and mRNA-lipo[jet] vaccine immunized mice compared with PBS immunized mice. Candidate vaccines conferred protective efficacy in vivo To further evaluate in vivo protective efficacy, we assessed the immune-mediated in vivo protective effect using the HTNV 76–118 strain challenge model. On the day of euthanasia, organs (heart, liver, kidney, lung, and spleen) were collected to determine viral loads, revealing HTNV replication in all collected organs. Notably, the levels of viral RNA were significantly reduced in the hearts and livers of all mice immunized with the four candidate vaccines compared to those receiving the control group (Fig. [124]4a). In addition, the spleen viral RNA levels of the DNA-LNP and mRNA-lipo[jet] vaccine groups were significantly lower than those of the control group (Fig. [125]4a). Furthermore, immunohistochemical analysis revealed abundant expression of HTNV NP protein in the heart and liver tissues of mice receiving PBS and LNP, whereas minimal positive cells were detected in the hearts and livers of mice immunized with the candidate vaccines (Fig. [126]4b). These results suggest that two doses of vaccination can prevent HTNV replication in vivo and indicate that three nucleic acid vaccines exhibited in vivo protective efficacy comparable to inactivated vaccines. Based on the viral copy number analysis of the spleen, the groups of DNA-LNP and mRNA-lipo[jet] vaccine induce a better protective immune response. Fig. 4. Candidate vaccines protect mice from HTNV challenge. [127]Fig. 4 [128]Open in a new tab a Viral RNA copies in the main organs (heart, liver, kidney, lung, and spleen) of HTNV-infected mice were determined by qRT-PCR (n = 6). b Immunostaining results for HTNV NP protein in the heart and liver tissues (n = 3). Scale bar, 100 μm. Data are represented as the mean ± SEM. Statistical differences were analyzed using one-way ANOVA with multiple comparison test. *P < 0.05, **P < 0.01, ***P < 0.001. Candidate vaccines induced robust immunoreactive transcriptomes To investigate the potential mechanisms of inactivated vaccine, naked DNA, DNA-LNP, and mRNA-lipo[jet] immunization strategies, we characterized the transcriptome by performing RNA-seq on mouse splenocyte samples after two doses of different vaccine regimens following HTNV peptide stimulation. The pathway enrichment analysis, utilizing both Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) frameworks, revealed significant insights into the immunological responses across all immunization groups. The KEGG-based analysis underscored the enrichment of pathways involved in immune activation, lymphocyte function, Th sub-population differentiation, and the regulation of inflammatory responses, etc. For instance, the Th1 and Th2 cell differentiation pathway increased in the inactivated and mRNA-lipo[jet] vaccines groups compared to the PBS group. The inactivated vaccine, naked DNA, DNA-LNP, and mRNA-lipo[jet] exhibited an increase in the B cell receptor signaling pathway compared to the control group. Additionally, the DNA-LNP group showed an increase in the B cell receptor signaling pathway compared to the naked DNA group (Fig. [129]5a). Concurrently, the GO-based analysis pointed to a pronounced enrichment of pathways associated with innate and adaptive immunity, T cell-mediated cellular immunity together with cytokines secretion, and B cell-mediated humoral immunity. It showed that all four candidate vaccine groups exhibited an increase in T cell activation, cytokine-mediated signaling, cellular response to INF-γ, and B cell proliferation pathway. Furthermore, the inactivated vaccine, DNA-LNP, and mRNA-lipo[jet] groups showed an increase in the cellular response to IL-4 pathway (Fig. [130]5b). Volcano plots highlighted the relative increases in the expression of selected immune-related genes between each two groups of all four candidate vaccines and control (Fig. [131]5c). Immune component analysis of mouse splenocytes was performed using ssGSEA and mMCPcounter algorithms. Clustering analysis based on cell types revealed a distribution pattern as shown in Fig. [132]5d and Fig. S[133]5. Both ssGSEA and mMCPcounter analysis indicated a sequence of PBS, naked DNA, inactivated vaccine, DNA-LNP, and mRNA-lipo[jet], roughly indicating a trend of increasing immune cell activation. In summary, all four candidate vaccine regimens effectively activated the immune response in vivo and induced robust immunoreactive transcriptomes. Fig. 5. Transcriptome analysis for different vaccination strategies. [134]Fig. 5 [135]Open in a new tab a Enriched KEGG immune pathways between each two groups of all four candidate vaccination strategies and control. b Enriched GO immune pathways between each two groups of all four candidate vaccination strategies and control. c Volcano plots of Log2 fold change of the differential expression between each two groups of all four candidate vaccines and control. (log2 fold change cut-off value at ±1, P < 0.05). d Immune component analysis using ssGSEA algorithm. Candidate vaccines elicited long-term immune response in mice To evaluate the long-term efficacy of the immune response, we adhered to the protocol outlined in Fig. [136]6a. Six months after the initial immunization, each group was administered the same dose of booster. The antibody response dynamics demonstrated a significant surge in specific antibody titers by day 14 following each vaccination, reaching their peak levels. Furthermore, all candidate vaccines sustained high antibody titers up to five months after two vaccination doses, indicating prolonged immunogenicity (Fig. [137]6b). After the final immunization, four candidate vaccine groups successfully induced the production of antigen-specific IgG antibodies (Fig. [138]6c). Following the administration of a long-term booster, we conducted HTNV-neutralizing antibody assays on sera collected from immunized mice. The results revealed that after receiving the vaccine booster, the PBS group and the empty LNP group induced average neutralizing titers of 1:9 and 1:7, respectively. The inactivated vaccine, naked DNA, DNA-LNP, and mRNA-lipo[jet] groups generated average neutralizing titers of 1:87, 1:104, 1:113 and 1:89, respectively (Fig. [139]6d). In comparison to the immediate immune response, the average neutralizing antibody titers in the long-term immune response were higher in groups receiving the inactivated vaccine, naked DNA, and mRNA-lipo[jet] boost vaccines. This underscores the importance of long-term booster shots in strengthening and prolonging immune protection. All candidate vaccine groups effectively induced HTNV-neutralizing antibodies, with antibody levels significantly higher than those in mice immunized with PBS or empty LNP controls (Fig. [140]6d). Besides, no significant differences were observed among the experimental groups, indicating that the humoral response of nucleic acid vaccines is not inferior to that of clinically used inactivated vaccines. Fig. 6. Schematic diagram of long-term immunization and evaluation of immune responses. [141]Fig. 6 [142]Open in a new tab a Schematic diagram of immunization, sample collection, and challenge schedule (the prone mouse element was created by Figdraw). b Dynamic HTNV-specific antibody response following vaccination (n ≥ 7) (arrows below the x-axes indicate the day of vaccine injection). c HTNV-specific IgG antibody titers were determined by ELISA (n = 7). The red dashed lines indicate the limit of detection (LOD). The LOD of the HTNV-specific antibody ELISA is 100. d 50% virus-neutralization titers determined using infectious HTNV (n = 6) and the LOD of the HTNV neutralization assay is 10. e Long-term HTNV GP-specific IFN-γ SFCs determined by ELISpot (n = 6). f Long-term HTNV GP-specific IL4 SFCs determined by ELISpot (n = 6). g Scatter plot showing the correlation of long-term HTNV GP-specific IL-4 and IFN-γ secreting cells by ELISpot (n = 6). h IFN-γ and IL-4 SFCs stimulated by single 15-mer peptide of HTNV GP by ELISpot assay after the long-term immunization and presented as a heatmap of Z scores (n = 3). i Long-term HTNV GP-specific CD4^+ (n ≥ 3) and CD8^+ (n ≥ 3) Tem cells in splenocytes were detected by flow cytometry. Data are represented as the mean ± SEM. Statistical differences were analyzed using one-way ANOVA with multiple comparison test. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. To assess the durability of induced T cell responses, we compared the characterization of mice splenocytes three weeks after the final immunization. ELISpot assays revealed a significant increase in virus-specific T lymphocytes secreting IFN-γ in the spleens of immunized mice compared to the control group, which was similar to the instant cellular responses (Fig. [143]6e and Fig. S[144]4b). Additionally, ELISpot experiments demonstrated that, compared to mice receiving the control group, all immunized groups led to significant induction of IL-4 levels in the mice splenocytes, with the inactivated vaccine showing the most significant increase (Fig. [145]6f). Furthermore, T lymphocytes secreting IL-4 in the spleen of mice immunized with naked DNA, DNA-LNP, and mRNA-lipo[jet] vaccines were significantly lower than those in the inactivated vaccine group, suggesting advantage in IL-4 secretion in Th response to the inactivated vaccine under long-term immunity. Our results indicate that naked DNA, DNA-LNP, and mRNA-lipo[jet] vaccines successfully induced predominantly Th1-driven long-lasting HTNV-specific cellular immune responses, while the inactivated vaccine exhibited a Th2-biased response under long-term immunity (Fig. [146]6g). Following long-term immunization, epitope prediction analysis was performed by conducting single peptide validations for ten peptides. The results demonstrated that the 15-mer peptides SYCMTGVLIEGKCFV and KKVMATIDSFQSFNT elicited strong T cell responses across all immunized groups (Fig. [147]6h). Besides, SYCMTGVLIEGKCFV and KKVMATIDSFQSFNT consistently presented as dominant peptides in both instant and long-term responses. Flow cytometry results revealed a significant increase in virus-specific CD4^+ Tem cells in the splenocytes of immunized mice compared to the control group, with no significant difference among the experimental groups. (Fig. [148]6i). In addition, a significant increase in CD8^+ Tem cells was observed in naked DNA and mRNA-lipo[jet] immunized mice compared with PBS immunized mice (Fig. [149]6i). Preliminary safety assessment of candidate vaccines We collected seven organs (heart, liver, kidney, spleen, lung, cerebrum, and gastrocnemius muscle) from the mice after final immunizations, three weeks apart, to preliminarily assess the safety of the vaccines in long-term effects. No significant pathological differences were observed in the tissues between the four candidate vaccine groups and the control groups (Fig. [150]7). These results confirm that immunization with the four candidate vaccines in mice did not result in pathological damage, providing preliminary evidence of vaccine safety. Fig. 7. H&E analyses of major organ tissues after immunization. [151]Fig. 7 [152]Open in a new tab Scale bar, 150 μm (gastrocnemius muscle, heart, liver, and kidney); 300 μm (lung and spleen); and 600 μm (cerebrum). Discussion Since the first isolation of the HTNV in 1978, research into vaccines has been relentless^[153]30–[154]32. The current HFRS vaccines in use are only inactivated vaccines. Additionally, DNA and subunit protein vaccines for HFRS are under study and show promise as effective preventive strategies against Hantavirus infection^[155]33,[156]34. However, global recognition is still pending, necessitating further research and development for safer and more effective vaccines to control HFRS, especially in areas with high incidence rates. Our study focuses on the development of three nucleic acid vaccine candidates (naked DNA, DNA-LNP, and mRNA-lipo[jet]), evaluating their immunogenicity and efficacy in Balb/C mice and comparing them with an inactivated vaccine as a benchmark. Our findings reveal that all candidate vaccines activated both instant and sustained immune responses, offering effective in vivo protection. Notably, the nucleic acid vaccines demonstrated distinct advantages over the inactivated vaccine, mainly reflected in terms of the enhanced humoral immunity observed with the DNA vaccine and the stronger cellular immunity with the mRNA vaccine. Furthermore, the DNA-LNP and mRNA-lipo[jet] vaccines elicited stronger splenocyte cytotoxicity and protective immunity. These results mark a significant step in the development of nucleic acid vaccines for HTNV, suggesting that sequential immunization with DNA and mRNA vaccines could further amplify the advantages of nucleic acid vaccines. In recent years, the development of mRNA vaccine technology has been a transformative breakthrough in vaccine research, offering substantial advantages over traditional methods^[157]35,[158]36. The 2023 Nobel Prize in Physiology or Medicine was awarded to the mRNA vaccine development for its critical significance in COVID-19. Significantly, this research heralds the inaugural creation of an mRNA vaccine targeting the hemorrhagic fever virus, effectively triggering virus-specific humoral and cellular immune responses, and ensuring efficacious protection in vivo. Compared with the current gold standard inactivated vaccine for HFRS, it induced stronger virus-specific Th1-type cellular immune responses, stronger splenocyte cytotoxic activity, and comparable protective immunity in mice. This study not only confirms the effectiveness of mRNA vaccines in preventing HFRS but also indicates their promising potential in combatting other hantavirus infections. Various delivery methods such as electroporation, gene gun, or tail tattoo have shown effectiveness in enhancing the immune response of DNA vaccines in large animals and humans^[159]37–[160]39. Their practical application in widespread human vaccination programs remains a challenge. In contrast, LNP has been extensively explored as an antigen-delivery system^[161]36,[162]40–[163]42. Existing studies on LNP as antigen delivery systems for DNA vaccines have primarily employed cationic lipids, known for their affinity to bind with negatively charged nucleic acids^[164]43–[165]46. However, delivery systems based on cationic lipids exhibit toxicity and immunogenicity both in vitro and in vivo^[166]47. Ionizable cationic lipids, designed to lessen the toxicity of lipid nanoparticles while preserving transfection efficiency, have been primarily applied in mRNA vaccines^[167]17. The exploration of their role in DNA vaccine delivery and the augmentation of immune responses remains limited. Pfeifle et al.’s pioneering work on a Lyme disease DNA vaccine highlights the potential of the DNA-LNP platform for vaccine development^[168]48. However, there was no direct comparison of DNA-LNP with naked DNA vaccines of equivalent copy numbers. In this study, ionizable lipid nanoparticles were used to encapsulate the HTNV DNA vaccine, and for the first time, evaluated against a naked DNA vaccine of the same copy number. While the overall immune response patterns of the DNA-LNP and the naked DNA vaccines were similar, DNA-LNP notably induced a stronger instant cellular response. GO and KEGG pathway enrichment analysis revealed that the DNA-LNP group showed various increased immune-related pathways compared to the naked DNA group. Regarding long-term cellular immune responses, there was an observed increase in virus-specific T lymphocytes secreting IFN-γ in the DNA-LNP group, albeit without a significant statistical difference from the naked DNA group. The advantage of LNP lies in their capacity to prolong the half-life upon entering the body, serving as a delayed-release mechanism and potentially reducing the frequency of immunizations required. Therefore, the instant cellular immune response of DNA-LNP is stronger, and the difference may be evened out after long-term booster immunization. Further research is warranted to investigate the role of LNP in facilitating the nuclear entry of DNA vaccines and to assess the cost-effectiveness of this strategy. Upon injection into muscle tissue, DNA and mRNA vaccines are initially taken up by immune cells at the injection site. DNA vaccines typically require a delivery system to facilitate their entry through cellular and nuclear membranes into the cell nucleus. While cells can also take up naked DNA, this process is much less efficient^[169]49,[170]50. Once inside the cells, DNA vaccines remain longer than mRNA, resulting in prolonged antigen expression and a more enduring immune memory^[171]51. In contrast, mRNA vaccines are typically encapsulated within lipid nanoparticles and enter cells via endocytosis. The mRNA is then released directly into the cytoplasm and quickly translated into the target protein without the need to enter the nucleus. This allows mRNA vaccines to exhibit higher uptake efficiency than DNA vaccines, leading to a quicker and more robust immune response, though their presence within the cells is shorter-lived^[172]52,[173]53. This study represents the first comprehensive evaluation of the immunological efficacy differences among hemorrhagic fever virus DNA vaccines, mRNA vaccines, and traditional inactivated vaccines, revealing the advantages of each candidate nucleic acid vaccine compared to traditional inactivated vaccines. Our findings demonstrate that three candidate nucleic acid vaccines effectively elicited a Th1-dominant, HTNV-specific, long-term cellular immune response, while the inactivated vaccine exhibited a Th2-biased response after long-term immunization. Additionally, either of ELISpot, flow cytometry, and transcriptomic techniques indicated that the mRNA and inactivated vaccine groups prompted notably higher IL-4 secretion levels than the DNA vaccine groups, both instantly or in the long term. This suggests a more robust Th2-type cellular immune response triggered by the mRNA and inactivated vaccines. While inactivated vaccines exhibit a Th2 bias when compared to nucleic acid vaccines, indicating a stronger antibody-mediated immune response, this Th2 bias does not significantly enhance the advantages of inactivated vaccines in humoral immune responses. Our findings demonstrate that three candidate nucleic acid vaccines showed neutralizing antibody titers and in vivo protective efficacy not inferior to those of inactivated vaccines. Therefore, each candidate vaccine could induce effective protective immunity. Moreover, the instant cellular immune response to the inactivated vaccine was less potent compared to the three candidate nucleic acid vaccines, but it showed improvement following a long-term booster, suggesting the necessity of multiple doses of inactivated vaccines for adequate protection. The nucleic acid vaccine exhibited obvious dominant peptides, while the inactivated vaccine did not show consistent dominant peptides. This underscores the advantage of nucleic acid vaccines, which focus immune responses on the target antigen, thereby inducing more specific immune reactions. Furthermore, the research revealed that SYCMTGVLIEGKCFV and KKVMATIDSFQSFNT are key epitopes of GP antigen presentation and can serve as candidate epitopes for a Hantaan virus epitope-based vaccine. Post-translational processing of HTNV GP yields two glycoproteins, Gn and Gc. Both the Gn and Gc proteins contain binding sites for neutralizing antibodies, making the full-length GP a richer source of such sites, including both linear and conformational epitopes^[174]5. Using full-length GP as an immunogen aids in the formation and maintenance of conformational epitopes, which are crucial for inducing neutralizing antibodies^[175]54. Additionally, the full GP offers more T cell epitopes, which help activate the cellular immune response against the virus. However, during the expression of HTNV GP, the Gn terminus induces autophagy and undergoes rapid degradation, leaving only Gc stable within the cells^[176]28,[177]29,[178]55. Given that Gn also contains numerous neutralizing epitopes, concerns arise regarding the balance between uncertain expression levels and immunological efficacy. Our research group previously engineered DNA vaccines targeting the individual Gn and Gc components of HTNV for immunological assessments^[179]56–[180]58. In contrast, our international counterparts developed a DNA vaccine based on the full-length GP, but did not separately assess the immunological effects of Gn and Gc under full-length GP immunization^[181]59,[182]60. We evaluated T cell responses using the dominant epitope peptides screened for Gn and Gc. The performance of these dominant epitopes appears to exhibit no significant differences between Gn and Gc, indicating that Gn and Gc are equivalent in antigen presentation and eliciting T cell responses. Therefore, the non-detection of Gn expression does not compromise its immunogenic efficacy and the use of full-length GP does not raise concerns of response imbalance between Gn and Gc. Our histological evaluation preliminarily confirms the safety of the candidate nucleic acid vaccines. The clinical application of both DNA and mRNA vaccines demonstrates their acceptable tolerability^[183]61–[184]67. The long-term clinical use and trials to date suggest that there are no major safety concerns with these vaccines. Given that the immune response induced by the vaccines may vary depending on the mouse strain and virus type used in the challenge, further research is needed using a variety of mouse strains (e.g., HLA transgenic mice) and diverse viral infection models to confirm our findings. Overall, our research provides promising new insights into the development and application of HFRS vaccines for the effective control of HTNV epidemics. Methods Sequence design The template for the DNA candidate vaccine is a codon-optimized version of the HTNV 76–118 strain M segment, which encodes the HTNV envelope glycoprotein, including Gn and Gc, with a length of 3616 base pairs (Sequence ID [185]Y00386.1). The mRNA vaccine candidate encodes a codon-optimized version of the HTNV 76-118 strain M segment. Besides, it includes 5′ and 3′ untranslated regions (UTR) as well as a poly (A) tail, collectively referred to as the UTR region (Table [186]S1 and Fig. S[187]1). The 5′ UTR and 3′ UTR sequences are derived from homo sapiens hemoglobin subunit beta (Sequence ID: [188]NM_000518.5). The 5′ UTR sequence comprises a regulatory sequence followed by the 5′ UTR sequence of β-globin, while the 3′ UTR sequence consists of the concatenated sequences of two segments from β-globin’s 3′ UTR. Additionally, a 120-nt long poly (A) tail is included to enhance RNA stability and translation efficiency. A 3×Flag tag is appended to the C-terminus of the GP to facilitate subsequent identification of vaccine antigen expression. Cell culture HEK293T cells were cultured in Dulbecco’s modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin at 37 °C in a 5% CO[2] incubator. Construction and identification of the recombinant plasmid We commissioned TsingKe Biotech Ltd to construct the codon-optimized pVAX1-GP plasmid and pUC57-UTR plasmid, and confirmed the accuracy through sequencing and agarose gel electrophoresis. The pGEM-UTR plasmid was obtained by cloning the UTR sequence from pUC57-UTR into the pGEM-11Zf plasmid (Promega). The pGEM-UTR-GP plasmid was obtained by inserting the GP sequence obtained from double enzyme digestion of pVAX1-GP into the pGEM-UTR plasmid. We employed Plasmid Maxi Kits (TIANGEN) for plasmid purification. Subsequently, the recombinant plasmids underwent horizontal electrophoresis on a 1% agarose gel after digesting with the restriction enzymes and were visualized under UV light. Lipid nanoparticle formulation of the DNA Lipid-nanoparticle (LNP) formulations were prepared using a modified procedure, previously employed for siRNA^[189]68,[190]69. In summary, lipids, including ionizable lipid (ALC-0315) (MCE), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) (MCE), cholesterol (MCE) and PEG-lipid (MCE), were dissolved in ethanol at a molar ratio of 55:10:32.5:2.5. The 10 mM lipid mixture was mixed with 10 mM citrate buffer containing DNA at a 1:3 ratio through a mixer. Subsequently, the formulations were transferred to an aqueous buffer system, filtered through a 0.22 μm mesh, and concentrated to the desired concentration via diafiltration using a 50 kD molecular weight cutoff microsep (Millipore) against 30 volumes of PBS. The vaccine candidates were diluted in PBS containing 60 mM sucrose and stored at −20 °C at a 0.5 mg/mL concentration. Characterization of LNP We performed dynamic light scattering measurements using the Malvern Zetasizer Nano-ZS (Malvern). Electron microscopy imaging was conducted with the HT7800 (HITACHI) by depositing the sample onto a porous carbon grid. We utilized the PicoGreen dsDNA Quantification Assay Kit (Solarbio) to assess the encapsulation efficiency of DNA. In essence, DNA-LNP were divided into two groups: one group was subjected to lysis using 2% Triton X-100, while the other remained untreated. Subsequently, the samples were processed with PicoGreen (Solarbio) following the manufacturer’s instructions. The amount of DNA in the samples was then quantified using a microplate reader. Excitation light was set at 480 nm, and emission light at 520 nm. In vitro transcription and purification of RNA The mRNA was produced in vitro through T7 RNA polymerase-mediated transcription, using the plasmid pGEM-UTR-GP linearized by the restriction enzyme Xba I as a template. The DNA template was purified and quantified using spectrophotometry, followed by in vitro transcription using T7 High Yield RNA Transcription kit (Novoprotein), with N1-methylpseudouridine-5’-triphosphate (m1ΨTP) substituting for uridine-5’-triphosphate. The RNA was then purified using lithium chloride, capped enzymatically with cap1 (Novoprotein), and further purified after capping reaction completion. Subsequently, the solution’s concentration was determined, and the integrity of the RNA was assessed through microfluidic capillary electrophoresis. Besides, we employed a double-antibody sandwich NovoFast dsRNA ELISA (Novoprotein) to quantify dsRNA residuals in the mRNA vaccine candidate. Transfection of HEK293T cells HEK293T cells were transfected with plasmid pVAX1-GP, mRNA, and transfection reagent lipofectamine 3000 (Thermo Fisher Scientific) separately, following the manufacturer’s instructions, or transfected with DNA-LNP vaccine candidates. Western blot analysis of transfected cells After 24 h post-transfection, the cell lysates were collected by centrifugation at 1000 × g using RIPA buffer (Beyotime). The collected lysates were then mixed with 5×SDS loading buffer and subjected to SDS-PAGE electrophoresis. The antigen proteins expressed by the cells were detected by western blotting with a flag antibody (Proteintech). Blots were developed with High-sig Western ECL Substrate (Tanon) and imaged with a Fusion FX Imager (Vilber) using the Image Lab software version 6.0. RNA isolation and quantitative real-time PCR The total RNA from transfected 293 T cells was isolated using TRIzol reagent (Sigma). cDNA was prepared using PrimeScript RT Master Mix (TaKaRa), and quantitative real-time PCR (qPCR) was performed using SYBR-green PCR Master Mix (Qiagen). The qPCR experiments were carried out on a Bio-Rad thermal cycler (Bio-Rad Laboratories Inc., CA, USA CFX96). The PCR conditions were as follows: an initial incubation at 50 °C for 30 min, followed by a denaturation step at 95 °C for 5 min. Subsequently, 40 cycles of amplification were performed, each consisting of a 20 s denaturation step at 95 °C and a 1-min annealing/extension step at 55 °C. For the detection of vaccine antigens, we designed two pairs of antigen-specific primers based on the Gn and Gc gene regions. The human beta-actin gene was employed as the reference gene. Gene expression levels were calculated using the comparative Ct method. All primers were synthesized by TsingKe Biotech Ltd (Table [191]S2). Immunofluorescence analysis of eukaryotic protein expression The transfected HEK293T cells were fixed with 4% paraformaldehyde and permeabilized in 0.2% Triton X-100. After blocking with 3% bovine serum albumin-PBS solution to prevent non-specific binding, the cells were incubated overnight at 4 °C with Flag antibody (1:2000, Proteintech). Subsequently, 594-conjugated goat anti-rabbit antibody (1:200, Proteintech) was used as the secondary antibody and incubated in the dark at 37 °C for 2 h. DNA was stained with DAPI (Solarbio). Images were obtained using a confocal microscope (Fluoview FV3000, Olympus, Japan). Animals and immunization Female BALB/c mice (6 weeks old) were procured from the Laboratory Animal Center of the Fourth Military Medical University and were assigned to six groups using a random number generator. All mice were in good health and had never participation in any research previously. They were acclimated for one week prior to immunization. Before all invasive procedures, mice were anesthetized with 2% isoflurane inhalation to ensure they remained pain-free during the operations. Each group received two injections of candidate vaccines, administered into the gastrocnemius muscle under isoflurane anesthesia, on day 0 and day 28, respectively. The six groups of candidate vaccines were as follows: PBS (n = 21), empty lipid nanoparticle (LNP) without encapsulated nucleic acid (n = 21), inactivated vaccine (Inact) (n = 21), endotoxin-free DNA plasmid pVAX1-Gp (n = 21), LNP-encapsulated endotoxin-free DNA-LNP (n = 21), and mRNA-lipo[jet] vaccine (n = 21). The inactivated vaccine, referenced to the manufacturer’s instructions, consisted of 10 μg of inactivated bivalent HFRS vaccine (HANPUWEI) diluted in 0.9% NaCl, with an injection volume of 50 μL. Both DNA and DNA-LNP candidate vaccines contained 30 μg of DNA with an injection volume of 50 μL. The mRNA-lipo[jet] candidate vaccine comprised 10 μg mRNA, which is delivered using in vivo mRNA transfection reagent (Polyplus). The injection volume is 100 μL, administered via intramuscular injection in two separate points. Each group of mice received a final booster vaccination on day 182 with the same dose as the initial two vaccinations. Serum samples were collected from mouse eye blood on days 52 and 206 after the initial immunization to detect neutralizing antibody responses. Additionally, serum samples were collected from mouse tail vein blood on day 0 (before the initial immunization) and on days 7, 14, 21, 28, 35, 42, 49, 56, 63, 70, 84, 98, 182, 196, and 203 after the initial immunization to assess specific antibody responses. Spleen tissues from the immunized mice in each group were collected on days 52 and 206 after the initial immunization for the assessment of cellular immune responses using ELISpot and flow cytometry. After the experiment, all mice were anesthetized with 2% isoflurane inhalation. After confirming adequate anesthesia, trained personnel humanely euthanized the animals by cervical dislocation, following standard procedures. Sera antibody titer evaluation by enzyme-linked immunosorbent assay (ELISA) Determination of the titer of HTNV-specific IgG antibodies was performed by ELISA. In brief, ELISA plates (Costar) were coated with inactivated HTNV virus diluted in sodium carbonate buffer (100 µL/well) and incubated overnight at 4 °C. After coating, the plates were washed once with wash buffer (PBST, PBS with 0.05% Tween 20, 200 µL/well) and then blocked with 1% bovine serum albumin diluted in wash buffer (200 µL/well) for 40 min at 37 °C. Subsequently, mouse sera were serially diluted, starting from 1:100, and 100 µL of each dilution was added to the wells as the primary antibody and incubated for 1 h at 37 °C. The plates were then washed five times and each well received 100 µL of horseradish peroxidase (HRP)-conjugated goat anti-mouse IgG as the secondary antibody (1:2000, Proteintech), followed by an incubation of 30 min at 37 °C. After washing six times, the plates were incubated with TMB substrate (Solarbio) for 15 min at 37 °C. Finally, 50 µL of ELISA stop solution (Solarbio) was added to terminate the reaction, and the absorbance was measured at 450 nm using a microplate reader (TECAN). Serum neutralization test Serum samples were serially diluted in a 2-fold gradient, starting from 1:10 to 1:320. Subsequently, they were mixed with a viral solution at a dose of 100 PFUs, followed by incubation at 37 °C for 1 hour. This mixture was then introduced to a 96-well plate containing a monolayer of Vero E6 cells, which were allowed to adsorb for 2 hours in a cell culture incubator. After discarding the mixture, DMEM containing 1.6% carboxymethyl cellulose and 2% FBS was applied to each well and the plate was incubated at 37 °C in a 5% CO[2] atmosphere for 5 days. The cells were subsequently fixed with 4% paraformaldehyde and permeabilized with 0.5% Triton X-100. A homemade monoclonal antibody, mAb1A8 against HTNV-NP, was added and incubated overnight at 4 °C. Followed by incubation with HRP-conjugated goat anti-mouse antibody (1:2000, Proteintech) at room temperature for 1 hour. Color development was performed using a Maxi-Blue precipitating TMB substrate, and blue plaques were counted after drying. The neutralizing titers were determined and calculated according to the Karber method. Synthesis and preparation of GP peptide The peptide pool used for ELISpot (30 μg/mL of each peptide) and flow cytometry (1 mg/mL of each peptide) consists of 10 sets of 15-mer peptides, all of which were obtained from Shanghai Apeptide Co (Table [192]S3). Enzyme-linked immunospot assay (ELISpot) The ELISpot experiment was conducted following the manufacturer’s protocol to evaluate the cellular immune response in vaccinated mice using the mouse IFN-γ/IL-4 ELISpot kit (BD Pharmingen). In essence, the plates were initially incubated with capture antibodies at 4 °C overnight, followed by blocking with RPMI 1640 medium containing 10% FBS for 2 h. Subsequently, we plated 1,000,000 cells/well and ex vivo reactivated immune mouse splenocytes using either a treatment group consisting of a 15-peptide mixture (each peptide at a final concentration of 0.1 μg/mL, with a total concentration of 2 mg/mL, as detailed in Table [193]S3) or control groups (negative control: RPMI 1640 medium containing 10% FBS; positive control: Concanavalin A, 2 μg/mL (Sigma)). After incubation for 24 h (37 °C, 5% CO[2]), these cells were collected for subsequent transcriptome sequencing as described below. Then the plates were subjected to a washing step, followed by the addition of biotinylated anti-mouse IFN-γ or IL-4 antibodies, and incubation at room temperature for 2 h. After rinsing the plates thoroughly, avidin-horseradish peroxidase and 3-amino-9-ethylcarbazole peroxidase substrate kit were introduced. Following this, an automated ELISpot plate reader (CTL) was employed to read and count the spots on the air-dried plates. Finally, we calculated the number of spot-forming cells per 1 million splenocytes. Cellular immune response by flow cytometry To perform intracellular cytokine staining of T cells from immunized mice, 1,000,000 splenocytes were ex vivo stimulated for 4 h using a cell stimulation cocktail (Invitrogen). After two washes with staining buffer, splenocytes were stained for cell surface antigens using directly labeled antibodies, including CD3 (FITC) (BioLegend), CD4 (PE) (BioLegend), and CD8 (Pacific Blue) (BioLegend). Subsequently, cells were adequately fixed and permeabilized using Cytofix/Cytoperm solution (BD) and incubated in the dark at 4 °C for 20 min. The permeabilized cells were then washed twice and subjected to intracellular cytokine staining using antibodies specific for IL-2, IL-4, IFN-γ, and Granzyme B (APC) (BioLegend) within Perm/Wash buffer (BD). Incubation was carried out in the dark at 4 °C for 30 min. Afterward, cells were washed twice with the staining buffer and subsequently resuspended in the same buffer before being subjected to flow cytometric analysis. Data analysis was conducted using NovoExpress software. Gating strategy to identify cytokine secretion in the T cells is present in Fig. [194]3e (take the IL-2 release from CD4^+ T cells for example). From all events, lymphocytes can be distinguished by their FSC/SSC properties. Single cells are then isolated by their relationship between FSC-H versus FSC-A. From single cells, CD4^+ T cells are gated, from which IL-2^+ CD4^+ T cells can be identified. For the detection of cell surface antigens, 500,000 mouse splenocytes were stimulated with a GP peptide pool (1 mg/mL for each peptide, see Table [195]S3) for 6 h at 37 °C under 5% CO[2]. Following two washes with staining buffer, we applied fluorescently conjugated antibodies, including CD3 (APC-Cyanine7) (BioLegend), CD4 (FITC) (BioLegend), CD8 (FITC) (BioLegend), CD44 (PE) (BD Biosciences), and CD62L (APC) (BioLegend), to label the splenocytes, which were then incubated at 4 °C in the dark for 30 min. After two additional washes with a staining buffer, the samples were analyzed using a flow cytometer. Data analysis was performed using NovoExpress software. Gating strategy to identify effector memory T (Tem) cells in the spleen is present in Fig. [196]3g (take the CD4^+ Tem cells in splenocytes for example). From all events, lymphocytes can be distinguished by their FSC/SSC properties. Single cells are then isolated by their relationship between FSC-H versus FSC-A. From single cells, CD4^+ T cells are gated, from which Tem (CD44^hi CD62L^lo) cells can be identified. Transcriptome analysis We collected 5,000,000 mouse splenocytes from immunized mice in each group, which were stimulated with peptide pools for 24 h. Each sample was derived from three immunized mice and preserved in TRIzol reagent (Sigma). Subsequently, the samples were sent to Seqhealth Ltd (Wuhan, China) for transcriptome analysis. HTNV challenge of mice The challenge model using the HTNV 76–118 strain for viral infection has been extensively described^[197]32,[198]52. Each group of candidate vaccines underwent immunization. Subsequently, on day 53, six mice in each group received the challenge or via intramuscular injection with 5 × 10^5 PFUs of HTNV 76–118 strain per mouse. Five days before the challenge, the mice were relocated from the immunization facility to a biosafety level 3 (BSL-3) animal facility. On the 3rd day post-challenge, all animals were euthanized, and major organs (heart, liver, spleen, lung, and kidney) were collected for subsequent analysis, including viral RNA level determination, histopathological examination, and immunofluorescence staining. All experiments were conducted under BSL-3 conditions and adhered to the international laboratory biosafety guidelines. Quantification of viral RNA in challenged mice tissues After the challenge, we conducted qPCR to detect viral RNA in the major organs of challenged mice. In brief, tissue samples were weighed, and total RNA was extracted using TRIzol reagent (Sigma). We quantified HTNV RNA using the S segment as the target and utilized 18S rRNA as an internal reference gene. All primers were synthesized by TsingKe Biotech Ltd (Table [199]S2). Immunofluorescence staining of challenged mice tissues Tissue samples from each group were collected for paraffin embedding and sectioning. After slide preparation, the tissue sections were subjected to a series of processes including sealing, incubation with the primary antibody, incubation with the secondary antibody, and nuclear staining, as detailed in “Immunofluorescence analysis of eukaryotic protein expression”. The primary antibody was a homemade anti-HTNV-NP mouse antibody, and the secondary antibody employed was a FITC-labeled goat anti-mouse antibody. The results were observed and analyzed using a fluorescence microscope. Hematoxylin and eosin (H&E) staining In terms of histopathology, we collected samples from six major organs and muscle tissue on the 24th day following long-term immunity. These samples were initially fixed using a 4% paraformaldehyde solution, subsequently embedded in paraffin, and then sectioned and stained with hematoxylin and eosin (H&E). The slides are scanned and photographed by a slide scanner (Winmedic). The staining results are certified by more than two pathologists. Statistical analysis All data analyses were conducted using GraphPad Prism 9.0 software. Unless specified, experimental dates are presented as mean ± SEM. Statistical significance among different groups was assessed using one-way ANOVA with multiple comparison test (*P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001). And the data analysts were blinded to the experimental group assignments. Animal ethics statement All animal experiments were conducted strictly under the regulations of the Chinese Regulations of Laboratory Animals and Laboratory Animal-Requirements of Environment and Housing Facilities. The procedures for the care and use of animals were approved by the Animal Ethics Committee of the Fourth Military Medical University (No. FMMU-DWZX-20221201). Supplementary information [200]Supplemental Information^ (2.9MB, pdf) Acknowledgements