Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (424)

Search Parameters:
Keywords = virus-specific CD8 T cell

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 2547 KiB  
Article
A Host Cell Vector Model for Analyzing Viral Protective Antigens and Host Immunity
by Sun-Min Ahn, Jin-Ha Song, Seung-Eun Son, Ho-Won Kim, Gun Kim, Seung-Min Hong, Kang-Seuk Choi and Hyuk-Joon Kwon
Int. J. Mol. Sci. 2025, 26(15), 7492; https://doi.org/10.3390/ijms26157492 (registering DOI) - 2 Aug 2025
Viewed by 218
Abstract
Avian influenza A viruses (IAVs) pose a persistent threat to the poultry industry, causing substantial economic losses. Although traditional vaccines have helped reduce the disease burden, they typically rely on multivalent antigens, emphasize humoral immunity, and require intensive production. This study aimed to [...] Read more.
Avian influenza A viruses (IAVs) pose a persistent threat to the poultry industry, causing substantial economic losses. Although traditional vaccines have helped reduce the disease burden, they typically rely on multivalent antigens, emphasize humoral immunity, and require intensive production. This study aimed to establish a genetically matched host–cell system to evaluate antigen-specific immune responses and identify conserved CD8+ T cell epitopes in avian influenza viruses. To this end, we developed an MHC class I genotype (B21)-matched host (Lohmann VALO SPF chicken) and cell vector (DF-1 cell line) model. DF-1 cells were engineered to express the hemagglutinin (HA) gene of clade 2.3.4.4b H5N1 either transiently or stably, and to stably express the matrix 1 (M1) and nucleoprotein (NP) genes of A/chicken/South Korea/SL20/2020 (H9N2, Y280-lineage). Following prime-boost immunization with HA-expressing DF-1 cells, only live cells induced strong hemagglutination inhibition (HI) and virus-neutralizing (VN) antibody titers in haplotype-matched chickens. Importantly, immunization with DF-1 cells transiently expressing NP induced stronger IFN-γ production than those expressing M1, demonstrating the platform’s potential for differentiating antigen-specific cellular responses. CD8+ T cell epitope mapping by mass spectrometry identified one distinct MHC class I-bound peptide from each of the HA-, M1-, and NP-expressing DF-1 cell lines. Notably, the identified HA epitope was conserved in 97.6% of H5-subtype IAVs, and the NP epitope in 98.5% of pan-subtype IAVs. These findings highlight the platform’s utility for antigen dissection and rational vaccine design. While limited by MHC compatibility, this approach enables identification of naturally presented epitopes and provides insight into conserved, functionally constrained viral targets. Full article
(This article belongs to the Special Issue Molecular Research on Immune Response to Virus Infection and Vaccines)
Show Figures

Graphical abstract

18 pages, 2125 KiB  
Article
A Replication-Defective Myxoma Virus Inducing Pro-Inflammatory Responses as Monotherapy and an Adjuvant to Chemo- and DC Immuno-Therapy for Ovarian Cancer
by Martin J. Cannon and Jia Liu
Viruses 2025, 17(8), 1058; https://doi.org/10.3390/v17081058 - 29 Jul 2025
Viewed by 342
Abstract
Myxoma virus (MYXV), a rabbit-specific poxvirus and non-pathogenic in humans and mice, is an excellent candidate oncolytic virus for cancer therapy. MYXV also has immunotherapeutic benefits. In ovarian cancer (OC), immunosuppressive tumor-associated macrophages (TAMs) are key to inhibiting antitumor immunity while hindering therapeutic [...] Read more.
Myxoma virus (MYXV), a rabbit-specific poxvirus and non-pathogenic in humans and mice, is an excellent candidate oncolytic virus for cancer therapy. MYXV also has immunotherapeutic benefits. In ovarian cancer (OC), immunosuppressive tumor-associated macrophages (TAMs) are key to inhibiting antitumor immunity while hindering therapeutic benefit by chemotherapy and dendritic cell (DC) vaccine. Because MYXV favors binding/entry of macrophages/monocytes, we examined the therapeutic potential of MYXV against TAMs. We found previously that a replication-defective MYXV with targeted deletion of an essential gene, M062R, designated ΔM062R MYXV, activated both the host DNA sensing pathway and the SAMD9 pathway. Treatment with ΔM062R confers therapeutic benefit comparable to that of wild-type replicating MYXV in preclinical models. Here we found that ΔM062R MYXV, when integrated with cisplatin and DC immunotherapy, further improved treatment benefit, likely through promoting tumor antigen-specific T cell function. Moreover, we also tested ΔM062R MYXV in targeting human immunosuppressive TAMs from OC patient ascites in a co-culture system. We found that ΔM062R treatment subverted the immunosuppressive properties of TAMs and elevated the avidity of cytokine production in tumor antigen-specific CD4+ T cells. Overall, ΔM062R presents a promising immunotherapeutic platform as a beneficial adjuvant to chemotherapy and DC vaccine. Full article
(This article belongs to the Special Issue Women in Virology 2025)
Show Figures

Figure 1

19 pages, 14428 KiB  
Article
Bivalent Oral Vaccine Using Attenuated Salmonella Gallinarum Delivering HA and NA-M2e Confers Dual Protection Against H9N2 Avian Influenza and Fowl Typhoid in Chickens
by Muhammad Bakhsh, Amal Senevirathne, Jamal Riaz, Jun Kwon, Ram Prasad Aganja, Jaime C. Cabarles, Sang-Ik Oh and John Hwa Lee
Vaccines 2025, 13(8), 790; https://doi.org/10.3390/vaccines13080790 - 25 Jul 2025
Viewed by 374
Abstract
Background: Fowl typhoid (FT), a septicemic infection caused by Salmonella Gallinarum (SG), and H9N2 avian influenza are two economically important diseases that significantly affect the global poultry industry. Methods: We exploited the live attenuated Salmonella Gallinarum (SG) mutant JOL3062 (SG: ∆lon [...] Read more.
Background: Fowl typhoid (FT), a septicemic infection caused by Salmonella Gallinarum (SG), and H9N2 avian influenza are two economically important diseases that significantly affect the global poultry industry. Methods: We exploited the live attenuated Salmonella Gallinarum (SG) mutant JOL3062 (SG: ∆lonpagLasd) as a delivery system for H9N2 antigens to induce an immunoprotective response against both H9N2 and FT. To enhance immune protection against H9N2, a prokaryotic and eukaryotic dual expression plasmid, pJHL270, was employed. The hemagglutinin (HA) consensus sequence from South Korean avian influenza A virus (AIV) was cloned under the Ptrc promoter for prokaryotic expression, and the B cell epitope of neuraminidase (NA) linked with matrix protein 2 (M2e) was placed for eukaryotic expression. In vitro and in vivo expressions of the H9N2 antigens were validated by qRT-PCR and Western blot, respectively. Results: Oral immunization with JOL3121 induced a significant increase in SG and H9N2-specific serum IgY and cloacal swab IgA antibodies, confirming humoral and mucosal immune responses. Furthermore, FACS analysis showed increased CD4+ and CD8+ T cell populations. On day 28 post-immunization, there was a substantial rise in the hemagglutination inhibition titer in the immunized birds, demonstrating neutralization capabilities of immunization. Both IFN-γ and IL-4 demonstrated a significant increase, indicating a balance of Th1 and Th2 responses. Intranasal challenge with the H9N2 Y280 strain resulted in minimal to no clinical signs with significantly lower lung viral titer in the JOL3121 group. Upon SG wildtype challenge, the immunized birds in the JOL3121 group yielded 20% mortality, while 80% mortality was recorded in the PBS control group. Additionally, bacterial load in the spleen and liver was significantly lower in the immunized birds. Conclusions: The current vaccine model, designed with a host-specific pathogen, SG, delivers a robust immune boost that could enhance dual protection against FT and H9N2 infection, both being significant diseases in poultry, as well as ensure public health. Full article
(This article belongs to the Special Issue Development of Vaccines Against Bacterial Infections)
Show Figures

Graphical abstract

15 pages, 1064 KiB  
Article
Overexpression of Interleukin-17 Modulates Responses to Marek’s Disease Virus Infection and Tumor Formation in Chickens
by Nitish Boodhoo, Katherine Blake, Fatemeh Fazel, Janan Shoja Doost and Shayan Sharif
Viruses 2025, 17(7), 1009; https://doi.org/10.3390/v17071009 - 18 Jul 2025
Viewed by 359
Abstract
Marek’s Disease Virus (MDV) is a highly contagious pathogen in chickens, resulting in immunosuppression and T-cell lymphomas. Understanding the role of host cytokines in MDV pathogenesis is crucial for developing effective interventions. This study investigated the in vivo effects of overexpressing avian interleukin-17 [...] Read more.
Marek’s Disease Virus (MDV) is a highly contagious pathogen in chickens, resulting in immunosuppression and T-cell lymphomas. Understanding the role of host cytokines in MDV pathogenesis is crucial for developing effective interventions. This study investigated the in vivo effects of overexpressing avian interleukin-17 (IL-17) in Marek’s disease virus infection model and its impact on T-cell populations. We utilized a recombinant pCDNA3.1 plasmid that expresses IL-17 at days 4 and 10 post-MDV infection in chickens. Our findings demonstrate that IL-17 overexpression significantly enhanced MDV replication. However, treatment with the plasmid expressing IL-17 led to a reduction in MD disease severity. Additionally, IL-17 treatment markedly altered the frequency of CD4+ and CD8α+ αβ T-cells. Specifically, at 21-dpi, there was an increase in CD3+ CD8α+ αβ T cells and a decrease in CD3+ CD4+ αβ T-cells within the spleen of chickens treated with the plasmid expressing IL-17. These modulatory effects suggest a possible mechanism by which IL-17 facilitates immune system cell activation and enhances viral persistence. This study underscores the pivotal role of IL-17 in MDV infection dynamics and offers. Full article
(This article belongs to the Special Issue Marek's Disease Virus)
Show Figures

Figure 1

29 pages, 5028 KiB  
Article
Moloney Murine Leukemia Virus-like Nanoparticles Pseudo-Typed with SARS-CoV-2 RBD for Vaccination Against COVID-19
by Bernhard Kratzer, Pia Gattinger, Peter A. Tauber, Mirjam Schaar, Al Nasar Ahmed Sehgal, Armin Kraus, Doris Trapin, Rudolf Valenta and Winfried F. Pickl
Int. J. Mol. Sci. 2025, 26(13), 6462; https://doi.org/10.3390/ijms26136462 - 4 Jul 2025
Viewed by 592
Abstract
Virus-like nanoparticles (VNPs) based on Moloney murine leukemia virus represent a well-established platform for the expression of heterologous molecules such as cytokines, cytokine receptors, peptide MHC (pMHC) and major allergens, but their application for inducing protective anti-viral immunity has remained understudied as of [...] Read more.
Virus-like nanoparticles (VNPs) based on Moloney murine leukemia virus represent a well-established platform for the expression of heterologous molecules such as cytokines, cytokine receptors, peptide MHC (pMHC) and major allergens, but their application for inducing protective anti-viral immunity has remained understudied as of yet. Here, we variably fused the wildtype SARS-CoV-2 spike, its receptor-binding domain (RBD) and nucleocapsid (NC) to the minimal CD16b-GPI anchor acceptor sequence for expression on the surface of VNP. Moreover, a CD16b-GPI-anchored single-chain version of IL-12 was tested for its adjuvanticity. VNPs expressing RBD::CD16b-GPI alone or in combination with IL-12::CD16b-GPI were used to immunize BALB/c mice intramuscularly and subsequently to investigate virus-specific humoral and cellular immune responses. CD16b-GPI-anchored viral molecules and IL-12-GPI were well-expressed on HEK-293T-producer cells and purified VNPs. After the immunization of mice with VNPs, RBD-specific antibodies were only induced with RBD-expressing VNPs, but not with empty control VNPs or VNPs solely expressing IL-12. Mice immunized with RBD VNPs produced RBD-specific IgM, IgG2a and IgG1 after the first immunization, whereas RBD-specific IgA only appeared after a booster immunization. Protein/peptide microarray and ELISA analyses confirmed exclusive IgG reactivity with folded but not unfolded RBD and showed no specific IgG reactivity with linear RBD peptides. Notably, booster injections gradually increased long-term IgG antibody avidity as measured by ELISA. Interestingly, the final immunization with RBD–Omicron VNPs mainly enhanced preexisting RBD Wuhan Hu-1-specific antibodies. Furthermore, the induced antibodies significantly neutralized SARS-CoV-2 and specifically enhanced cellular cytotoxicity (ADCC) against RBD protein-expressing target cells. In summary, VNPs expressing viral proteins, even in the absence of adjuvants, efficiently induce functional SARS-CoV-2-specific antibodies of all three major classes, making this technology very interesting for future vaccine development and boosting strategies with low reactogenicity. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

17 pages, 9016 KiB  
Article
GCRV-II Triggers B and T Lymphocyte Apoptosis via Mitochondrial ROS Pathway
by Jie Wang, Wen-Jing Dong, Chang-Song Wu, Tian-Tian Tian, Xu-Jie Zhang and Yong-An Zhang
Viruses 2025, 17(7), 930; https://doi.org/10.3390/v17070930 - 30 Jun 2025
Viewed by 290
Abstract
Grass carp reovirus (GCRV), particularly the highly prevalent genotype II (GCRV-II), is known to infect peripheral blood leukocytes (PBLs) of grass carp. However, it is unclear whether GCRV-II can induce apoptosis in bystander lymphocytes within infected PBLs. Here, we have shown that GCRV-II [...] Read more.
Grass carp reovirus (GCRV), particularly the highly prevalent genotype II (GCRV-II), is known to infect peripheral blood leukocytes (PBLs) of grass carp. However, it is unclear whether GCRV-II can induce apoptosis in bystander lymphocytes within infected PBLs. Here, we have shown that GCRV-II infection induces apoptosis via the mitochondria-dependent caspase-3 pathway in infected PBLs. GCRV-II infection was also found to induce a significant increase in reactive oxygen species (ROS) accumulation in leukocytes and lymphocytes, accompanied by increased apoptosis in IgM+ B and CD4+ T lymphocyte subsets. Further studies have demonstrated that the targeted inhibition of mitochondrial ROS production can effectively attenuate apoptosis in neighboring B and T lymphocytes within infected PBLs, suggesting that GCRV-II-induced pro-apoptotic effects on bystander lymphocytes largely require the involvement of the mitochondrial-dependent ROS pathway. Taken together, our study reveals the underlying mechanism by which GCRV-II induces apoptosis in bystander B and T lymphocytes through ROS production, providing new insights into understanding the virus-induced pro-apoptotic mechanism in specific immune cells and a potential strategy for viral immune escape. Full article
(This article belongs to the Section Animal Viruses)
Show Figures

Figure 1

18 pages, 2983 KiB  
Article
IRF4 Mediates Immune Evasion to Facilitate EBV Transformation
by Ling Wang, Culton R. Hensley, Jahan Rifat, Adam D. Walker, Katharine Ning, Jonathan P. Moorman, Zhi Q. Yao and Shunbin Ning
Viruses 2025, 17(7), 885; https://doi.org/10.3390/v17070885 - 24 Jun 2025
Viewed by 479
Abstract
The lymphocyte-specific transcription factor interferon regulatory factor 4 (IRF4) is a key player in immune evasion in cancers, with the complex mechanism(s) being barely understood. In this study, we have focused on the role of IRF4 in regulating T cell functions through its [...] Read more.
The lymphocyte-specific transcription factor interferon regulatory factor 4 (IRF4) is a key player in immune evasion in cancers, with the complex mechanism(s) being barely understood. In this study, we have focused on the role of IRF4 in regulating T cell functions through its transcriptional regulation of programmed death 1 (PD1) and its ligand PD1 ligand 1 (PD-L1), which were identified as IRF4 transcriptional targets in multi-omics analysis. We have shown that IRF4 transcriptionally regulates both PD1 and PD-L1, promoting immune suppression in the context of Epstein–Barr virus (EBV) infection. Co-culturing EBV+ JiJoye lymphoma cells with CD4+ T cells or with peripheral blood mononuclear cells (PBMCs) downregulates CD4+ T cell functions, but the depletion of IRF4 in EBV+ JiJoye lymphoma cells reduces PD1 and PD-L1 expression, and partially restores CD4+ T cell functions. Moreover, CD4+ T cell depletion from PBMCs enhances EBV transformation, and EBV has a greater efficiency in transforming PBMCs from HIV patients with impaired CD4+ T cell functions. These findings support the role of IRF4 in immune evasion by upregulating PD1/PD-L1 during EBV transformation, and that functional CD4+ T cells are essential for limiting EBV transformation. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

20 pages, 3479 KiB  
Article
Age-Related Impairment of Innate and Adaptive Immune Responses Exacerbates Herpes Simplex Viral Infection
by Ruchi Srivastava, Sweta Karan, Yassir Lekbach, Afshana Quadiri, Ava Tohidian, Chhaya Maurya, Sarah Xue Le Ng, Reilly Chow, America Garcia, Anshu Agrawal, Hawa Vahed, Aziz A. Chentoufi and Lbachir BenMohamed
Pathogens 2025, 14(7), 624; https://doi.org/10.3390/pathogens14070624 - 23 Jun 2025
Viewed by 497
Abstract
Immune function declines with age, leading to increased vulnerability of the elderly to viral infectious pathogens. The mechanisms by which aging negatively impacts the innate and adaptive immune system, leading to enhanced susceptibility to respiratory viral pathogens, remain incompletely understood. In the present [...] Read more.
Immune function declines with age, leading to increased vulnerability of the elderly to viral infectious pathogens. The mechanisms by which aging negatively impacts the innate and adaptive immune system, leading to enhanced susceptibility to respiratory viral pathogens, remain incompletely understood. In the present study, we utilized a mouse model of infection with herpes simplex virus type 1 (HSV-1), a virus that can infect the lungs and lead to pneumonia, a rare but serious health concern in the elderly. Following intranasal inoculation of young (6 weeks), adult (36 weeks), and aged mice (68 weeks) with HSV-1 (KOS strain) we: (i) compared the local and systemic immune responses to infection in young, adult, and aged mice, and (ii) correlated the level and type of immune responses to protection against HSV-1 infection and disease. Compared to young and adult mice, aged mice displayed: (i) increased activation of epithelial cells with a decreased expression of TLR3; (ii) increased activation of dendritic cells with increased expression of MHC-I, MHC-II, and CD80/86; (iii) decreased production of type-I interferons; (iv) delayed production of anti-inflammatory cytokines and chemokines in the lungs; and (v) impairment frequencies of functional HSV-specific CD107+IFN-γ+CD8+ T cells associated with the increased incidence of viral infection and disease. These findings suggest that age-related impairments in innate and adaptive immune responses may exacerbate respiratory viral infections and disease in the elderly. Full article
Show Figures

Figure 1

14 pages, 1700 KiB  
Article
Delayed Viral Clearance Accompanied by Early Impaired Humoral and Virus-Specific T-Cell Response in Patients with Coronavirus Disease 2019 and Interstitial Lung Disease
by Jiaying Zhong, Juan Li, Rui Wei, Bingpeng Guo, Tingting Cui, Peiyu Huang, Zhongfang Wang, Qun Luo and Qian Han
Vaccines 2025, 13(6), 655; https://doi.org/10.3390/vaccines13060655 - 19 Jun 2025
Viewed by 491
Abstract
Objectives: Patients with interstitial lung disease (ILD) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are at high risk of severe coronavirus disease 2019. It is unclear whether anti-viral cellular and humoral immunity is impacted in patients with ILD in the presence [...] Read more.
Objectives: Patients with interstitial lung disease (ILD) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are at high risk of severe coronavirus disease 2019. It is unclear whether anti-viral cellular and humoral immunity is impacted in patients with ILD in the presence of immune disorders and immunosuppressive therapy. This results in poor control of viral infections following SARS-CoV-2 infection. We aimed to highlight the clinical management of patients with ILD with regard to the adjustment of anti-inflammatory therapy during SARS-CoV-2 infection. Methods: We compared viral clearance, antibody levels, and T-cell immune response between healthy controls and patients with connective tissue disease-related ILD (CTD-ILD) or interstitial pneumonia with autoimmune features (IPAF). Results: Patients with ILD exhibited a higher viral load than the control group (1.58 × 106 vs. 2.37 × 103 copies/mL, p = 0.018), as well as a significantly lower level of neutralizing antibodies against the wild-type (WT) virus (7.01 vs. 625.6, p < 0.0001) and Omicron BA.5 (7.19 vs. 128.4, p < 0.001). Similarly, a lower virus-specific T-cell (VST) immune response was observed 14 days post-symptom onset in the ILD group (CD4+ VSTs: 0.018 vs. 0.082, p = 0.005; CD8+ VSTs: 0.0008 vs. 0.047, p = 0.004). The ILD group had no other heightened inflammatory biomarkers compared with the control group. Conclusions: Our study provides novel evidence of the underlying interaction between virus clearance and host immune status and sheds light on the clinical management of patients with ILD with regard to the adjustment of anti-inflammatory therapy during SARS-CoV-2 infection. Full article
(This article belongs to the Section COVID-19 Vaccines and Vaccination)
Show Figures

Figure 1

18 pages, 2111 KiB  
Article
Consensus Sequences for Gag and Pol Introduced into HIV-1 Clade B Laboratory Strains Differentially Influence the Impact of Point Mutations Associated with Immune Escape and with Drug Resistance on Viral Replicative Capacity
by Sven Breitschwerdt, Benedikt Grandel, Benedikt Asbach, Franziska Winter, Todd Allen, Ralf Wagner, Bernd Salzberger and Arne Schneidewind
Viruses 2025, 17(6), 842; https://doi.org/10.3390/v17060842 - 12 Jun 2025
Viewed by 673
Abstract
Viral evasion from effective human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T-cell responses and from antiretroviral therapy through viral sequence variation is frequently accompanied by a loss in viral fitness. The impact of sequence variations on replication capacity in vitro was mostly studied [...] Read more.
Viral evasion from effective human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T-cell responses and from antiretroviral therapy through viral sequence variation is frequently accompanied by a loss in viral fitness. The impact of sequence variations on replication capacity in vitro was mostly studied by introducing single mutations into a specific clonal strain such as NL4-3. How the specific viral backbone itself impacts replicative fitness remains elusive. To test for a potential effect of the viral backbone, we constructed HIV-1 clade B clones with consensus sequences for gag and/or pol and evaluated the infectivity of viral variants harboring well-defined cytotoxic T-lymphocyte (CTL) escape mutations or drug resistance mutations within this backbone or the clonal NL4-3 strain. Viral variants with consensus sequences were replication-competent in vitro, although at lower rates than the NL4-3 virus. Introduction of the dominant CTL escape mutation R264K into the newly constructed viruses or into NL4-3 led to a dramatic reduction in infection rates. In contrast to the NL4-3 backbone, the combination of R264K with its compensatory mutation S173A on the consensus backbone led to higher infection rates as compared to the same virus in the absence of R264K and S173A. Furthermore, 2 out of 10 drug resistance mutations in pol led to opposing effects, with an increase in infection rates on the consensus gag/pol backbone and a reduction on NL4-3. Therefore, the effect of the respective viral backbone on infectivity observed in vitro might constitute an additional factor to explain differential kinetics of mutational evasion from immune and pharmaceutical pressure. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

14 pages, 1489 KiB  
Article
Orally Dissolving Film-Based Influenza Vaccines Confer Superior Protection Compared to the Oral Administration of Inactivated Influenza Virus
by Keon-Woong Yoon, Jie Mao, Gi-Deok Eom, Su In Heo, Ki Back Chu, Mi Suk Lee and Fu-Shi Quan
Vaccines 2025, 13(6), 600; https://doi.org/10.3390/vaccines13060600 - 31 May 2025
Viewed by 635
Abstract
Background: Self-administered orally dissolving films (ODFs) encapsulating inactivated influenza vaccines represent an effective strategy for stimulating mucosal immunity. While this vaccination method offers several advantages over conventional influenza vaccines, a comparative efficacy study remains lacking. Methods: Female BALB/c mice were immunized [...] Read more.
Background: Self-administered orally dissolving films (ODFs) encapsulating inactivated influenza vaccines represent an effective strategy for stimulating mucosal immunity. While this vaccination method offers several advantages over conventional influenza vaccines, a comparative efficacy study remains lacking. Methods: Female BALB/c mice were immunized with inactivated A/PR/8/34 (H1N1) either via orogastric inoculation or through the oral mucosal delivery using pullulan and trehalose-based ODF vaccines. Each group received equivalent antigen doses across three immunizations. Humoral responses and antibody functionality were assessed using sera collected post-immunization. After lethal viral challenge, other immunological and virological parameters were determined in corresponding tissues. Body weight and survival were monitored over a 14-day period after challenge. Results: ODF vaccination elicited significantly higher virus-specific IgA levels, HAI titers, and neutralizing antibody activity than oral gavage. After the viral challenge, ODF-immunized mice exhibited stronger IgG and IgA responses in respiratory tissues, increased antibody-secreting cells in lungs and spleen, and elevated germinal center B cells and CD8+ T cell responses. Both vaccination methods reduced lung pro-inflammatory cytokines and provided full protection against lethal challenge; however, the ODF group showed lower cytokine levels, better weight maintenance, and reduced viral loads. Conclusions: ODF vaccination elicits more robust systemic and mucosal immune responses than oral vaccination and may serve as a promising alternative method of influenza vaccine delivery. Full article
(This article belongs to the Special Issue Virus Pandemics and Vaccinations)
Show Figures

Figure 1

22 pages, 3973 KiB  
Article
Dysregulated Adaptive Immune Responses to SARS-CoV-2 in Immunocompromised Individuals
by Núria Mayola Danés, Demi Brownlie, Rebecca Folkman, Anna Nordlander, Kim Blom, Renata Varnaite, Julia Niessl, Oskar Karlsson Lindsjö, Sandra Söderholm, Mira Akber, Puran Chen, Marcus Buggert, Andreas Bråve, Jonas Klingström, Piotr Nowak, Nicole Marquardt, Klara Sondén, Ola Blennow and Sara Gredmark-Russ
Microorganisms 2025, 13(5), 1077; https://doi.org/10.3390/microorganisms13051077 - 6 May 2025
Viewed by 612
Abstract
The SARS-CoV-2 virus poses a significant risk to immunocompromised patients, who display weakened immunity and reduced seroconversion following infection and vaccination. In this study, we recruited 19 hospitalized patients with immune disorders (ImCo) and 4 immunocompetent controls (ICC) with COVID-19. We evaluated their [...] Read more.
The SARS-CoV-2 virus poses a significant risk to immunocompromised patients, who display weakened immunity and reduced seroconversion following infection and vaccination. In this study, we recruited 19 hospitalized patients with immune disorders (ImCo) and 4 immunocompetent controls (ICC) with COVID-19. We evaluated their serological, humoral, and cellular immune responses at <30 days and >90 days post-symptom onset. ICC patients showed robust B and T cell responses against SARS-CoV-2, indicated by detectable antibody levels, memory antibody-secreting cells (mASCs) towards the spike protein and spike-specific CD4+ and CD8+ T cells. ImCo patients showed impaired immune responses, with lower levels of B cell responses. Further subdivision of the ImCo patients demonstrates that solid organ transplant (SOT) patients generated B cell responses similar to ICC patients, whereas the other ImCo patients, including patients with hematological malignancies and anti-CD20 therapy, did not. Absolute T cell numbers and spike-specific CD4+ and CD8+ T cell responses were low in the ImCo patients at <30 days but increased at later time points. Our findings suggest that even when B cell responses were reduced, patients could present a T cell response, suggesting a more successful line of passive immunization for immunocompromised individuals focusing on boosting T cell responses. Full article
(This article belongs to the Special Issue Immune Modulation to SARS-CoV-2 Vaccination and Infection)
Show Figures

Graphical abstract

16 pages, 1435 KiB  
Article
Programmed Cell Death-1 Expression in T-Cell Subsets in Chickens Infected with Marek’s Disease Virus
by Jumpei Sato, Yoshinosuke Motai, Shunsuke Yamagami, Shwe Yee Win, Fumiya Horio, Hikaru Saeki, Naoya Maekawa, Tomohiro Okagawa, Satoru Konnai, Kazuhiko Ohashi and Shiro Murata
Pathogens 2025, 14(5), 431; https://doi.org/10.3390/pathogens14050431 - 29 Apr 2025
Cited by 1 | Viewed by 659
Abstract
Marek’s disease virus (MDV) causes Marek’s disease (MD) in chickens, characterized by malignant lymphomas and immunosuppression. Sporadic MD outbreaks continue to occur even among vaccinated flocks in certain regions due to the increased virulence of the field strains. However, the mechanisms of tumorigenesis [...] Read more.
Marek’s disease virus (MDV) causes Marek’s disease (MD) in chickens, characterized by malignant lymphomas and immunosuppression. Sporadic MD outbreaks continue to occur even among vaccinated flocks in certain regions due to the increased virulence of the field strains. However, the mechanisms of tumorigenesis and immunosuppression caused by MDV remain to be fully elucidated. We previously reported that the mRNA expression of programmed cell death 1 (PD-1), an immune checkpoint molecule, was increased in tumor lesions caused by MDV, and its expression was positively correlated with the mRNA expression of Meq, an MDV-specific oncogene. In this study, we characterized PD-1-expressing T-cell subsets in the spleen and tissues of chickens that developed tumors to investigate the association between PD-1 expression and immunosuppression. Flow cytometric analysis revealed that the proportion of PD-1-expressing CD4+ T-cells, which are targets of MDV tumorigenesis, increased in the spleen and tumor tissues of chickens with MD. The proportion of PD-1+ CD4+ T-cells was higher in Meq-expressing cells than in those that were not. In the spleens of chickens with MD, the proportions of PD-1-expressing cells were increased in CD8+ and γδ T-cells, which play pivotal roles in defense against MD pathogenesis, relative to those of spleens from uninfected chickens. Moreover, the proportion of PD-1+ CD8+ T-cells expressing interferon (IFN)-γ did not increase in the spleen of chickens with MD. Additionally, no difference in the proportion of IFN-γ+ γδ T-cells expressing and not expressing PD-1 was observed in the spleens of chickens with MD, although the proportion of IFN-γ+ γδ T-cells expressing PD-1 in the spleens of uninfected chickens was higher. The function of PD-1-expressing CD8+ and γδ T-cells in chickens may be impaired after developing MD, which may cause MDV-induced immunosuppression. Full article
(This article belongs to the Section Viral Pathogens)
Show Figures

Figure 1

19 pages, 2856 KiB  
Article
Dysfunctional Senescent Herpes Simplex Virus-Specific CD57+CD8+ T Cells Are Associated with Symptomatic Recurrent Ocular Herpes in Humans
by Aziz A. Chentoufi, Arif A. Khan, Ruchi Srivastava, Sweta Karan, Yassir Lekbach, Hawa Vahed and Lbachir BenMohamed
Viruses 2025, 17(5), 606; https://doi.org/10.3390/v17050606 - 24 Apr 2025
Viewed by 744
Abstract
Herpes simplex virus (HSV)-specific CD8+ T cells protect mice from herpes infection and disease. However, the phenotype and function of HSV-specific CD8+ T cells that play a key role in the “natural” protection seen in HSV-1-seropositive healthy asymptomatic (ASYMP) patients (who [...] Read more.
Herpes simplex virus (HSV)-specific CD8+ T cells protect mice from herpes infection and disease. However, the phenotype and function of HSV-specific CD8+ T cells that play a key role in the “natural” protection seen in HSV-1-seropositive healthy asymptomatic (ASYMP) patients (who have never had clinical herpes disease) remain to be determined. We previously reported that symptomatic (SYMP) patients (who have frequent bouts of recurrent ocular herpes disease) had more undifferentiated and dysfunctional HSV-specific CD8+ T cells. In contrast, healthy ASYMP individuals maintained a significantly higher proportion of differentiated polyfunctional CD8+ T cells. Here, we report that HSV-specific CD8+ T cells from 10 SYMP patients, but not HSV-specific CD8+ T cells from 10 ASYMP patients, have phenotypic and functional characteristics of cellular senescence, including: (i) high frequency of senescent (CD57+) and exhausted (PD-1+) CD8+ T cells; (ii) late terminally differentiated (KLRG1+), non-proliferating CD8+ T cells; (iii) HSV-specific CD8+ T cells which decreased in number over time and were not homeostatically maintained, as indicated by a reduction in the number of CD127+CD8+ T cells; (iv) loss of the co-stimulatory molecule CD28 on HSV-specific CD8+ T cells; and (v) decreased production of effector molecules (granzyme B and perforin) by HSV-specific CD8+ T cells. Our findings provide insights into the role of senescence in HSV-specific CD8+ T cells in susceptibility to recurrent herpes and have implications for T-cell-based immunotherapeutic strategies against recurrent herpes in humans. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

33 pages, 1847 KiB  
Review
Immunological Control of Herpes Simplex Virus Type 1 Infection: A Non-Thermal Plasma-Based Approach
by Julia Sutter, Jennifer L. Hope, Brian Wigdahl, Vandana Miller and Fred C. Krebs
Viruses 2025, 17(5), 600; https://doi.org/10.3390/v17050600 - 23 Apr 2025
Viewed by 1444
Abstract
Herpes simplex virus type 1 (HSV-1) causes a lifelong infection due to latency established in the trigeminal ganglia, which is the source of recurrent outbreaks of cold sores. The lifelong persistence of HSV-1 is further facilitated by the lack of cure strategies, unsuccessful [...] Read more.
Herpes simplex virus type 1 (HSV-1) causes a lifelong infection due to latency established in the trigeminal ganglia, which is the source of recurrent outbreaks of cold sores. The lifelong persistence of HSV-1 is further facilitated by the lack of cure strategies, unsuccessful vaccine development, and the inability of the host immune system to clear HSV-1. Despite the inefficiencies of the immune system, the course of HSV-1 infection remains under strict immunological control. Specifically, HSV-1 is controlled by a CD8+ T cell response that is cytotoxic to HSV-1-infected cells, restricts acute infection, and uses noncytolytic mechanisms to suppress reactivation in the TG. When this CD8+ T cell response is disrupted, reactivation of latent HSV-1 occurs. With antiviral therapies unable to cure HSV-1 and prophylactic vaccine strategies failing to stimulate a protective response, we propose non-thermal plasma (NTP) as a potential therapy effective against recurrent HSV-1 infection. We have demonstrated that NTP, when applied directly to HSV-1-infected cells, has antiviral effects and stimulates cellular stress and immunomodulatory responses. We further propose that the direct effects of NTP will lead to long-lasting indirect effects such as reduced viral seeding into the TG and enhanced HSV-1-specific CD8+ T cell responses that exert greater immune control over HSV-1 infection. Full article
(This article belongs to the Special Issue Herpesviruses and Associated Diseases)
Show Figures

Figure 1

Back to TopTop