Health
Vaccine-induced protection against SARS-CoV-2 requires IFN-γ-driven cellular immune response
BNT162b2 induces robust cellular responses in B lymphocyte-deficient mice
To evaluate the cellular responses in μMT mice, we collected spleen tissues from vaccinated and unvaccinated C57BL/6 J wild type (WT) and μMT mice. The splenocytes were then stimulated with spike protein of SARS-CoV-2 (Fig. 1a). We first quantified antibodies titer against SARS-CoV-2 spike with ELISA and confirmed that antibody response was absent in BNT162b2-vaccinated μMT mice (Fig. 1b). Our flow cytometry analysis demonstrated that 13.2% of CD4+ and 18.5% of CD8+ T memory cells in spleen tissue of μMT mice immunized with BNT162b2 secreted IFN-γ upon stimulating with spike proteins of Alpha variant (Fig. 1c). In parallel, spike protein of Omicron BA.1 induced IFN-γ production in 17.3% of CD4+ and 16.1% of CD8+ T memory cells, suggesting robust induction of cellular responses upon stimulation of spike proteins from different SARS-CoV-2 variants (Fig. 1d). Importantly, the mean percentage of CD4+ and CD8+ T memory cells producing IFN-γ is higher in vaccinated μMT mice than in WT mice (Fig. 1c, d), suggesting a higher level of activation of cellular responses in the vaccinated μMT mice when compared to WT mice. In contrast, IL-4 expression, which reflects the differentiation of B cells into plasma cells13, was not stimulated by the spike proteins in either WT or μMT mice (Fig. 1e, f). Besides, it is reported that CD8+ T cells in B cell-deficient mice were skewed more toward effector phenotype during viral infection14,15. Interestingly, our data revealed that stimulation of spike protein of SARS-CoV-2 could also induce the skewed differentiation of CD8+ T cells into highly cytotoxic terminal effector cells in the spleen of vaccinated µMT mice (Fig. S1a). The proportion of long-lived CD8+ T memory cell precursors is, however, dramatically reduced in vaccinated µMT mice compared to that in vaccinated WT mice (Fig. S1a). Taken together, our results demonstrated that robust cellular immunity was elicited in μMT mice immunized with BNT162b2 in the absence of humoral immunity.
BNT162b2 elicits significant protective immunity against SARS-CoV-2 Alpha and Omicron BA.1 in B lymphocyte-deficient mice
To assess the requirement of antibody-mediated immunity in the protection against SARS-CoV-2, we vaccinated C57BL/6 J WT and μMT mice with the BNT162b2 mRNA vaccine and infected the vaccinated mice with SARS-CoV-2 Alpha or Omicron BA.1 at 21 days post boost (Fig. 2a). We found that BNT162b2 induced protective immunity against SARS-CoV-2 Alpha and Omicron BA.1 variant in both WT and μMT C57BL/6 J mice. BNT162b2 vaccination significantly inhibited Alpha and Omicron BA.1 replication in the lung tissues and nasal turbinate (NT) tissues of both WT and μMT mice at 2 dpi or 4 dpi (Fig. 2b, c). Despite the lack of B cells, BNT162b2 vaccination reduced Alpha replication in the lungs of μMT mice by 11.5-fold (P = 0.0274) and 11.6-fold (P = 0.0151) on 2 and 4 dpi, respectively (Fig. 2b). Similarly, BNT162b2 vaccination reduced Omicron BA.1 replication in the lungs of μMT mice by 3.1-fold (P = ns) and 50-fold (P = 0.0188) on 2 and 4 dpi, respectively (Fig. 2c).
SARS-CoV-2 pathogenesis is associated with the accumulation of proinflammatory myeloid cells within the lung tissue in mouse model16,17. To identify correlates of vaccine-induced immunity in C57BL/6 J mice, we assessed the histopathological changes in the lung tissues of vaccinated and unvaccinated mice after SARS-CoV-2 Alpha and Omicron BA.1 challenge. For mice infected with Alpha, lungs in vaccinated WT mice had the least severe pulmonary pathology with relatively intact structure at 2 dpi or 4 dpi (Fig. 2d). More interstitial pneumonia and immune cell influx were observed in unvaccinated WT mice. Similarly, reduced in vivo pathology was detected in vaccinated μMT mice when compared with the unvaccinated ones. For mice infected with Omicron BA.1, the pathology of virus-induced lung tissues was milder than that of counterparts infected with Alpha, in keeping with recent findings18 (Fig. 2d, e). In addition, the degree of inflammatory cell infiltration in the lungs of vaccinated μMT mice was lower than that observed in the lungs of un-vaccinated μMT mice (Fig. 2d, e). Together, these findings indicated that BNT162b2 vaccination is capable of inducing protective immunity in μMT mice in the absence of the antibody response, suggesting cellular immunity may serve to protect against SARS-CoV-2 infection.
Cytokines are key coordinators of the cellular immune response and essential to the activation of T cells. We next evaluated the impact of BNT162b2 vaccination on the cytokine milieu in mice infected with SARS-CoV-2 Alpha (Fig. 3a, b). Cytokine profiling of blood samples from mice challenged with Alpha revealed that IFN-γ, CXCL1, MCP-1 and IP-10 protein concentrations were significantly higher in vaccinated μMT mice than those in the unvaccinated mice (Fig. 3c). In particular, Alpha challenge drove a 253-fold (p < 0.0001) increase of IFN-γ secretion in vaccinated μMT mice when compared to un-vaccinated μMT mice (Fig. 3c), suggesting strong activation of the cellular immunity upon viral challenge in vaccinated μMT mice. On the contrary, BNT162b2 vaccination is associated with a marked decrease of these key cytokines in WT mice when compared with unvaccinated ones (Fig. 3c).
We next measured the cytokine levels in lung and NT tissue of vaccinated and unvaccinated mice upon Alpha or Omicron BA.1 infection by quantitative reverse transcription PCR (RT-qPCR). In line with the data in blood samples, BNT162b2 vaccination increased the expression of IFN-γ, MCP-1, and IP-10 in lung and NT tissues of μMT mice challenged with Alpha or Omicron BA.1 (Fig. 3d–i).
The K18-hACE2 mouse model has been frequently used to assess SARS-CoV-2 pathogenesis12,19. To evaluate the impact of B cell depletion in the K18-hACE2 mice, we obtained K18-hACE2 μMT mice by breeding K18-hACE2 mice with μMT mice. In the K18-hACE2/μMT mice, BNT162b2 vaccination similarly offered protection against Alpha and Omicron BA.1 infection. Alpha and Omicron BA.1 replication in the lung and NT tissues were lower in vaccinated K18-hACE2/μMT mice when compared with un-vaccinated ones (Fig. 4b, c). In line with the results in C57BL/6 J model, we observed ameliorated lung pathology in vaccinated WT or μMT mice when compared with unvaccinated ones (Fig. 4d, e). For cytokine profiling, we quantified IFN-γ, MCP-1, and IP-10 expression in lung tissue and NT in the mice by RT-qPCR. We found that BNT162b2 vaccination increased the expression of IFN-γ, MCP-1, and IP-10 in lung and NT tissues of K18-hACE2 μMT mice challenged with Alpha or Omicron BA.1 (Fig. 4f-k), similar to what we observed in C57BL/6 J μMT mice (Fig. 3d–i). Taken together, these findings indicate that BNT162b2 elicits significant protective immunity against SARS-CoV-2 Alpha and Omicron BA.1 in B lymphocyte-deficient mice.
T cell-mediated immunity is highly activated in B lymphocyte-deficient mice upon SARS-CoV-2 infection
To characterize the vaccine-induced protective immunity in μMT mice, we performed bulk RNA-seq of WT and µMT C57BL/6 J mice lung and NT tissues with or without vaccination on 2 dpi (Fig. 5a). Gene functional enrichment analysis based on the significantly differential expressed genes showed prominent immune characters between different conditions (Fig. 5b). We found that BNT162b2 vaccination boosted both humoral and cellular immunity in both NT and lung tissues in WT mice (Fig. 5b). In contrast, due to the lack of mature B cells, only genes involved in T cell activation and T cell receptor signaling pathways were significantly upregulated in vaccinated µMT mice comparing to unvaccinated µMT mice, suggesting that cellular immunity can be effectively activated by BNT162b2 vaccination in B cell-deficient mice (Fig. 5b). Cell type functional enrichment also indicated that the function of CD8 + T cell is highly enriched in vaccinated µMT mice when compared to the unvaccinated µMT mice (Fig. S2). According to the differential gene expression analysis, we found that T cell-related genes/pathways were significantly upregulated in the vaccinated µMT mice in both NT and lung tissue when compared to unvaccinated µMT mice (Fig. 5c, d). Among these genes, integral membrane glycoprotein CD8a serves as a coreceptor of class I MHC molecular in CD8+ T cells, and cell adhesion molecular CD6 that regulates T cell responses was both upregulated in NT and lung tissues (Fig. 5c, d). Besides, the cxcr6 gene, which controls the localization of resident memory T lymphocytes to different compartments of the lung and maintains airway resident memory T lymphocytes20, was also significantly upregulated in vaccinated µMT mice (Fig. 5c, d).
IFN-γ facilitates vaccine-induced immunity in B lymphocyte-deficient mice
As mentioned earlier, IFN-γ secretion was substantially upregulated in serum, NT, and lung tissues of vaccinated μMT mice. To further evaluate the role of IFN-γ in the vaccine-induced protective immunity in μMT mice, we injected IFN-γ depleting antibody into vaccinated WT and μMT mice before Omicron BA.1 challenge (Fig. 6a). Our results demonstrated that the IFN-γ depleting antibody significantly increased Omicron BA.1 replication by 19.3-fold (P = 0.0029) in the lung of vaccinated μMT mice. In contrast, the IFN-γ depleting antibody did not modulate Omicron BA.1 replication in the lungs of vaccinated WT mice (0.7-fold, P = NS) (Fig. 6b). In the NT, the IFN-γ depleting antibody has a trend to promote virus replication in vaccinated WT and μMT mice, albeit no statistical significance was reached (Fig. 6b). In keeping with these observations, treatment of IFN-γ depleting antibody resulted in more severe lung pathology in vaccinated μMT mice while no significant change in lung pathology was observed in vaccinated WT mice when IFN-γ depleting antibody was administered (Fig. 6c). A recent study reported that supplementation of IFN-γ reverses the age-dependent COVID-19 phenotype in mice21. Therefore, we next treated naïve μMT mice daily with 10 μg IFN-γ from −1 to 1 d.p.i. to investigate whether administration of IFN-γ could alleviate the disease severity upon SARS-CoV-2 infection. Supplementation of IFN-γ resulted in lower viral load and milder lung pathology in NT and lung tissues of naïve μMT mice than PBS control group when infected with SARS-CoV-2 Alpha (Fig. 6e, f). Together, our results indicate that IFN-γ contributes to the vaccine-induced cellular immunity that offers protection against SARS-CoV-2 in B lymphocyte-deficient mice.
CD4+ and CD8+ T cells are required for the vaccine-induced clearance of SARS-CoV-2 in NT and lung tissue in B lymphocyte-deficient mice
As previously mentioned, CD4+ and CD8+ T memory cells in vaccinated μMT mice produce IFN-γ upon spike protein stimulation (Fig. 1c, d). We then investigated if CD4+ and CD8+ T cells contribute to the vaccine-induced cellular immunity in μMT mice by using CD4-, CD8-, or CD4/CD8-depleting antibodies. In lung tissues, CD4-, CD8-, and CD4/CD8-depleting antibodies increased Omicron BA.1 replication by 10-fold (P = NS), 10-fold (P = NS), and 42-fold (P = 0.0038), respectively (Fig. 7b). In the NT tissues, CD4-, CD8-, and CD4/CD8-depleting antibodies increased Omicron BA.1 replication by 0.6-fold (P = NS), 15.6-fold (P = NS), and 7.6-fold (P = 0.028), respectively (Fig. 7b). In keeping with the virus replication findings, simultaneous depletion of both CD4+ and CD8+ T cells resulted in the most severe lung pathology in vaccinated μMT mice among all evaluated groups (Fig. 7c). To further dissect the role of CD4+ and CD8+ T cells, we characterized tissue-resident memory (TRM) and effector cells in lung tissues of vaccinated μMT mice by flow cytometry analysis. Consistent with the bulk RNA-Seq data, our results demonstrated that 26-fold (P = 0.015) higher percentage of CD44+ CD69+ CD8+ T cells were found in lung tissues of vaccinated μMT mice than in unvaccinated ones after SARS-CoV-2 Alpha infection (Fig. 7e). In addition, the percentage of CXCR6+ lung tissue-resident memory (TRM) CD8+ T cells was also significantly higher (67.6-fold, P = 0.0007) in vaccinated μMT mice (Fig. 7e, S7a). More IFN-γ+ and Granzyme B+ CD8+ T cells were detected in vaccinated μMT mice (5.5-fold, P = 0.0159 and 8.3-fold, p < 0.0001, respectively) when compared with unvaccinated μMT mice, suggesting that robust cellular immunity was induced in vaccinated μMT mice upon SARS-CoV-2 challenge (Fig. 7f, g). Likewise, the percentage of CD44+ CD69+ CD4+, CXCR6+ CD4+, and IFN-γ+ CD4+ T cells were also higher in vaccinated μMT mice than unvaccinated mice, but less significant differences were found (Fig. 7e–g, S7a, b). Taken together, these data indicate that while both CD4+ and CD8+ T cells are required for optimal vaccine-induced protection against SARS-CoV-2 in lung tissues of μMT mice, CD8+ T cells play a more predominant role than CD4+ T cells in the NT of μMT mice.
BNT162b2 induces protective immunity against SARS-CoV-2 Omicron BA.5.2 in B lymphocyte-deficient mice
SARS-CoV-2 Omicron BA.5, which has substantially higher immune escape capacity than BA.1, are the dominant strain in many countries as of January 202322,23,24. To test whether BNT162b2 could induce protective immunity against SARS-CoV-2 Omicron BA.5.2 in μMT mice, we infected vaccinated C57BL/6 J and K18-hACE2 mice with SARS-CoV-2 Omicron BA.5.2 and evaluated the viral replication in these mice. The quantitative RT-PCR showed that the viral load in lung tissues of vaccinated μMT mice is 34-fold (P = 0.0283) and 188-fold (P = NS) lower than C57BL/6 J and K18-hACE2 unvaccinated μMT mice at 2 d.p.i., respectively (Fig. 8b). Although much lower viral load was detected in NT of both C57BL/6 J and K18-hACE2 vaccinated mice at 2 and 4 d.p.i., the statistical difference was not seen between the vaccinated and unvaccinated groups (Fig. 8b). In line with the viral replication, the lung pathology was also alleviated in vaccinated mice (Fig. 8c). Consistent with the therapeutic effect of IFN-γ in naïve μMT mice against SARS-CoV-2 Alpha, supplementation of IFN-γ could also mitigate the viral replication and lung pathology in naïve μMT mice upon Omicron BA.5.2 challenge (Fig. 8d, e).
Sources 2/ https://www.nature.com/articles/s41467-023-39096-y The mention sources can contact us to remove/changing this article |
What Are The Main Benefits Of Comparing Car Insurance Quotes Online
LOS ANGELES, CA / ACCESSWIRE / June 24, 2020, / Compare-autoinsurance.Org has launched a new blog post that presents the main benefits of comparing multiple car insurance quotes. For more info and free online quotes, please visit https://compare-autoinsurance.Org/the-advantages-of-comparing-prices-with-car-insurance-quotes-online/ The modern society has numerous technological advantages. One important advantage is the speed at which information is sent and received. With the help of the internet, the shopping habits of many persons have drastically changed. The car insurance industry hasn't remained untouched by these changes. On the internet, drivers can compare insurance prices and find out which sellers have the best offers. View photos The advantages of comparing online car insurance quotes are the following: Online quotes can be obtained from anywhere and at any time. Unlike physical insurance agencies, websites don't have a specific schedule and they are available at any time. Drivers that have busy working schedules, can compare quotes from anywhere and at any time, even at midnight. Multiple choices. Almost all insurance providers, no matter if they are well-known brands or just local insurers, have an online presence. Online quotes will allow policyholders the chance to discover multiple insurance companies and check their prices. Drivers are no longer required to get quotes from just a few known insurance companies. Also, local and regional insurers can provide lower insurance rates for the same services. Accurate insurance estimates. Online quotes can only be accurate if the customers provide accurate and real info about their car models and driving history. Lying about past driving incidents can make the price estimates to be lower, but when dealing with an insurance company lying to them is useless. Usually, insurance companies will do research about a potential customer before granting him coverage. Online quotes can be sorted easily. Although drivers are recommended to not choose a policy just based on its price, drivers can easily sort quotes by insurance price. Using brokerage websites will allow drivers to get quotes from multiple insurers, thus making the comparison faster and easier. For additional info, money-saving tips, and free car insurance quotes, visit https://compare-autoinsurance.Org/ Compare-autoinsurance.Org is an online provider of life, home, health, and auto insurance quotes. This website is unique because it does not simply stick to one kind of insurance provider, but brings the clients the best deals from many different online insurance carriers. In this way, clients have access to offers from multiple carriers all in one place: this website. On this site, customers have access to quotes for insurance plans from various agencies, such as local or nationwide agencies, brand names insurance companies, etc. "Online quotes can easily help drivers obtain better car insurance deals. All they have to do is to complete an online form with accurate and real info, then compare prices", said Russell Rabichev, Marketing Director of Internet Marketing Company. CONTACT: Company Name: Internet Marketing CompanyPerson for contact Name: Gurgu CPhone Number: (818) 359-3898Email: [email protected]: https://compare-autoinsurance.Org/ SOURCE: Compare-autoinsurance.Org View source version on accesswire.Com:https://www.Accesswire.Com/595055/What-Are-The-Main-Benefits-Of-Comparing-Car-Insurance-Quotes-Online View photos
to request, modification Contact us at Here or [email protected]