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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (221)

Search Parameters:
Keywords = humoral innate immunity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 1238 KiB  
Review
Complement Cascades and Brain Disorders
by Ivana Jovčevska, Alja Videtič Paska and Katarina Kouter
Biomolecules 2025, 15(8), 1179; https://doi.org/10.3390/biom15081179 - 17 Aug 2025
Viewed by 323
Abstract
The complement system is a vital component of innate immunity. Besides its roles in pathogen defense, its significance in neurodevelopment, neurodegeneration, and cancer progression is beginning to be recognized. We performed a comprehensive literature review to summarize the involvement and dysregulation of the [...] Read more.
The complement system is a vital component of innate immunity. Besides its roles in pathogen defense, its significance in neurodevelopment, neurodegeneration, and cancer progression is beginning to be recognized. We performed a comprehensive literature review to summarize the involvement and dysregulation of the complement system in three main CNS-associated conditions: Alzheimer’s disease, schizophrenia, and glioma. In Alzheimer’s disease, activation of the complement system contributes to neuroinflammation, synaptic loss, and neuronal death. In glioblastoma, complement promotes tumor growth, immune evasion, and therapy resistance. In schizophrenia, genetic variations in complement components, particularly C4A, are associated with synaptic pruning abnormalities and disease susceptibility. We conclude that the complement system has a dual role of protector and pathogenic mediator in the central nervous system. While it is critical in neurodegenerative, oncological, and psychiatric disorders, its role is not understood well enough. For therapeutic purposes, targeting the complement system may open new frontiers for therapeutic interventions without disrupting important physiological processes. More research is needed to elucidate the exact roles of the complement and help translate these findings into clinical settings. Full article
Show Figures

Figure 1

16 pages, 875 KiB  
Article
Profile of Selected MicroRNAs as Markers of Sex-Specific Anti-S/RBD Response to COVID-19 mRNA Vaccine in Health Care Workers
by Simona Anticoli, Maria Dorrucci, Elisabetta Iessi, Salvatore Zaffina, Rita Carsetti, Nicoletta Vonesch, Paola Tomao and Anna Ruggieri
Int. J. Mol. Sci. 2025, 26(15), 7636; https://doi.org/10.3390/ijms26157636 - 7 Aug 2025
Viewed by 657
Abstract
Sex-based immunological differences significantly influence the outcome of vaccination, yet the molecular mediators underpinning these differences remain largely elusive. MicroRNAs (miRNAs), key post-transcriptional regulators of gene expression, have emerged as critical modulators of innate and adaptive immune responses. In this study, we investigated [...] Read more.
Sex-based immunological differences significantly influence the outcome of vaccination, yet the molecular mediators underpinning these differences remain largely elusive. MicroRNAs (miRNAs), key post-transcriptional regulators of gene expression, have emerged as critical modulators of innate and adaptive immune responses. In this study, we investigated the expression profile of selected circulating miRNAs as potential biomarkers of sex-specific humoral responses to the mRNA COVID-19 vaccine in a cohort of health care workers. Plasma samples were collected longitudinally at a defined time point (average 71 days) post-vaccination and analyzed using RT-qPCR to quantify a panel of immune-relevant miRNAs. Anti-spike (anti-S) IgG titers were measured by chemiluminescent immunoassays. Our results revealed sex-dependent differences in miRNA expression dynamics, with miR-221-3p and miR-148a-3p significantly overexpressed in vaccinated female HCWs and miR-155-5p overexpressed in vaccinated males. MiR-148a-3p showed a significant association with anti-S/RBD (RBD: receptor binding domain) IgG levels in a sex-specific manner. Bioinformatic analysis for miRNA targets indicated distinct regulatory networks and pathways involved in innate and adaptive immune responses, potentially underlying the differential immune activation observed between males and females. These findings support the utility of circulating miRNAs as minimally invasive biomarkers for monitoring and predicting sex-specific vaccine-induced immune responses and provide mechanistic insights that may inform tailored vaccination strategies. Full article
(This article belongs to the Special Issue Molecular Research on Immune Response to Virus Infection and Vaccines)
Show Figures

Figure 1

24 pages, 7353 KiB  
Article
Characterization and Application of Synergistically Degraded Chitosan in Aquafeeds to Promote Immunity, Antioxidative Status, and Disease Resistance in Nile Tilapia (Oreochromis niloticus)
by Thitirat Rattanawongwiboon, Natthapong Paankhao, Wararut Buncharoen, Nantipa Pansawat, Benchawan Kumwan, Pakapon Meachasompop, Phunsin Kantha, Tanavan Pansiri, Theeranan Tangthong, Sakchai Laksee, Suwinai Paankhao, Kittipong Promsee, Mongkhon Jaroenkittaweewong, Pattra Lertsarawut, Prapansak Srisapoome, Kasinee Hemvichian and Anurak Uchuwittayakul
Polymers 2025, 17(15), 2101; https://doi.org/10.3390/polym17152101 - 31 Jul 2025
Viewed by 595
Abstract
This study investigated the immunonutritional potential of high-molecular-weight (Mw~85 kDa), non-degraded chitosan (NCS) and gamma-radiation-degraded, low-molecular-weight chitosan (RCS) incorporated into aquafeeds for Nile tilapia (Oreochromis niloticus). RCS was produced by γ-irradiation (10 kGy) in the presence of 0.25% (w/ [...] Read more.
This study investigated the immunonutritional potential of high-molecular-weight (Mw~85 kDa), non-degraded chitosan (NCS) and gamma-radiation-degraded, low-molecular-weight chitosan (RCS) incorporated into aquafeeds for Nile tilapia (Oreochromis niloticus). RCS was produced by γ-irradiation (10 kGy) in the presence of 0.25% (w/v) H2O2, yielding low-viscosity, colloidally stable nanoparticles with Mw ranging from 10 to 13 kDa. Five diets were formulated: a control, NCS at 0.50%, and RCS at 0.025%, 0.050%, and 0.075%. No adverse effects on growth were observed, confirming safety. Immune gene expression (e.g., ifng1, nfκb, tnf), antioxidant markers (e.g., reduced MDA, increased GSH and GR), and nonspecific humoral responses (lysozyme, IgM, and bactericidal activity) were significantly enhanced in the NCS-0.50, RCS-0.050, and RCS-0.075 groups. Notably, these benefits were achieved with RCS at 10-fold lower concentrations than NCS. Following challenge with Edwardsiella tarda, fish fed RCS-0.050 and RCS-0.075 diets exhibited the highest survival rates and relative percent survival, highlighting robust activation of innate and adaptive immunity alongside redox defense. These results support the use of low-Mw RCS as a biologically potent, cost-effective alternative to traditional high-Mw chitosan in functional aquafeeds. RCS-0.050 and RCS-0.075 show strong potential as immunonutritional agents to enhance fish health and disease resistance in aquaculture. Full article
(This article belongs to the Special Issue Polysaccharides: Synthesis, Properties and Applications)
Show Figures

Figure 1

12 pages, 1307 KiB  
Article
Protection Against Transplacental Transmission of a Highly Virulent Classical Swine Fever Virus Two Weeks After Single-Dose FlagT4G Vaccination in Pregnant Sows
by Liani Coronado, Àlex Cobos, Adriana Muñoz-Aguilera, Sara Puente-Marin, Gemma Guevara, Cristina Riquelme, Saray Heredia, Manuel V. Borca and Llilianne Ganges
Vaccines 2025, 13(8), 803; https://doi.org/10.3390/vaccines13080803 - 28 Jul 2025
Viewed by 454
Abstract
Background/Objectives: Classical swine fever (CSF) continues to challenge global eradication efforts, particularly in endemic regions, where pregnant sows face heightened risks of vertical transmission following exposure to CSFV. Methods: This study evaluates the early protective efficacy of FlagT4G, a novel live attenuated DIVA-compatible [...] Read more.
Background/Objectives: Classical swine fever (CSF) continues to challenge global eradication efforts, particularly in endemic regions, where pregnant sows face heightened risks of vertical transmission following exposure to CSFV. Methods: This study evaluates the early protective efficacy of FlagT4G, a novel live attenuated DIVA-compatible vaccine. Pregnant sows were vaccinated at mid-gestation and challenged 14 days later with a highly virulent CSFV strain. Results: FlagT4G conferred complete clinical protection, preventing both maternal viremia and transplacental transmission. No CSFV RNA, specific antibodies, or IFN-α were detected in fetal samples from vaccinated animals. In contrast, unvaccinated sows exhibited clinical signs, high viral loads, and widespread fetal infection. Interestingly, early protection was observed even in the absence of strong humoral responses in some vaccinated sows, suggesting a potential role for innate or T-cell-mediated immunity in conferring rapid protection. Conclusions: The demonstrated efficacy of FlagT4G within two weeks of vaccination underscores its feasibility for integration into emergency vaccination programs. Its DIVA compatibility and ability to induce early fetal protection against highly virulent CSFV strains position it as a promising tool for CSF control and eradication strategies. Full article
(This article belongs to the Special Issue Vaccines for Porcine Viruses)
Show Figures

Figure 1

17 pages, 5140 KiB  
Article
Comparative Analysis of Chitosan, Lipid Nanoparticles, and Alum Adjuvants in Recombinant SARS-CoV-2 Vaccine: An Evaluation of Their Immunogenicity and Serological Efficacy
by Majed Ghattas, Garima Dwivedi, Anik Chevrier, Trevor Scobey, Rakan El-Mayta, Melissa D. Mattocks, Dong Wang, Marc Lavertu and Mohamad-Gabriel Alameh
Vaccines 2025, 13(8), 788; https://doi.org/10.3390/vaccines13080788 - 24 Jul 2025
Viewed by 600
Abstract
Background: Chitosan, a family of polysaccharides composed of glucosamine and N-acetyl glucosamine, is a promising adjuvant candidate for eliciting potent immune response. Methods: This study compared the adjuvant effects of chitosan to those of empty lipid nanoparticles (eLNPs) and aluminum hydroxide (alum) following [...] Read more.
Background: Chitosan, a family of polysaccharides composed of glucosamine and N-acetyl glucosamine, is a promising adjuvant candidate for eliciting potent immune response. Methods: This study compared the adjuvant effects of chitosan to those of empty lipid nanoparticles (eLNPs) and aluminum hydroxide (alum) following administration of recombinant SARS-CoV-2 spike immunogen in adult mice. Mice received the adjuvanted recombinant protein vaccine in a prime-boost regimen with four weeks interval. Subsequent analyses included serological assessment of antibody responses, evaluation of T cell activity, immune cell recruitment and cytokine profiles at injection site. Results: Compared to alum, chitosan induced a more balanced Th1/Th2 response, akin to that observed with eLNPs, demonstrating its ability to modulate both the humoral and cellular immune pathways. Chitosan induced a different proinflammatory cytokine (e.g., IL-1⍺, IL-2, IL-6, and IL-7) and chemokine (e.g., Eotaxin, IP-10, MIP-1a) profile compared to eLNPs and alum at the injection site and in the draining lymph nodes. Moreover, chitosan potentiated the recruitment of innate immune cells, with neutrophils accounting for about 40% of the infiltrating cells in the muscle, representing a ~10-fold increase compared to alum and a comparable level to eLNPs. Conclusions: These findings collectively indicate that chitosan has the potential to serve as an effective adjuvant, offering comparable, and potentially superior, properties to those of currently approved adjuvants. Full article
(This article belongs to the Special Issue Advances in Vaccine Adjuvants)
Show Figures

Figure 1

14 pages, 1248 KiB  
Review
The Role of Inflammation in the Pathophysiology of Heart Failure
by Marwan Amara, Ohad Stoler and Edo Y. Birati
Cells 2025, 14(14), 1117; https://doi.org/10.3390/cells14141117 - 21 Jul 2025
Viewed by 906
Abstract
Heart failure (HF), a prevalent global health issue characterized by the heart’s impaired ability to pump or fill blood, affects millions worldwide and continues to pose significant challenges despite advancements in treatment. This review delves into the critical and increasingly recognized role of [...] Read more.
Heart failure (HF), a prevalent global health issue characterized by the heart’s impaired ability to pump or fill blood, affects millions worldwide and continues to pose significant challenges despite advancements in treatment. This review delves into the critical and increasingly recognized role of inflammation in the development and progression of this complex syndrome. While the incidence of HF has seen a decline in some regions due to improved cardiac care, its overall prevalence is rising, particularly among younger adults and those with heart failure with a preserved ejection fraction (HFpEF). Given the persistently high rates of hospitalization and mortality associated with HF, understanding the underlying mechanisms, including the contribution of inflammation, is crucial for identifying novel therapeutic strategies. Inflammation in heart failure is a multifaceted process involving the activation of the immune system, both innate and adaptive, and encompasses various mechanisms such as the release of pro-inflammatory mediators, endothelial dysfunction, and neurohormonal activation. Myocardial damage triggers the innate immune response, while humoral immunity and chronic systemic inflammation, often linked to cardiovascular risk factors and autoimmune diseases, also play significant roles. Notably, heart failure and inflammation have a reciprocal relationship, with HF itself contributing to inflammatory processes within the cardiac tissue and systemically. Understanding these intricate pathways, including the involvement of specific immune cells and molecular mediators, is essential for comprehending the pathogenesis of heart failure and exploring potential therapeutic interventions. The review further examines various inflammatory biomarkers that have been implicated in heart failure, such as cytokines (including TNF-α and IL-1) and C-reactive protein (CRP). While these markers often correlate with the severity and prognosis of HF, clinical trials targeting specific inflammatory mediators have largely yielded disappointing results, highlighting the complexity of the inflammatory response in this context. The exploration of these biomarkers and the challenges encountered in translating anti-inflammatory strategies into effective treatments underscore the need for continued research to unravel the precise role of inflammation across different HF subtypes and to develop more targeted and effective anti-inflammatory therapies. Full article
(This article belongs to the Special Issue Inflammation in Target Organs)
Show Figures

Figure 1

18 pages, 426 KiB  
Article
Exploratory Study on Microbiota and Immune Responses to Short-Term L. paracasei CNCM I-1518 Consumption in Healthy Adults
by Fernando Rivero-Pino, Maria José Castro, Paz Redondo del Río, Eloina Gutierrez, Agustín Mayo-Iscar, Mercedes Nocito and Alfredo Corell
Nutrients 2025, 17(14), 2287; https://doi.org/10.3390/nu17142287 - 10 Jul 2025
Viewed by 790
Abstract
Background/Objectives: The gut microbiota and immune system are interconnected, with targeted nutritional interventions offering potential to modulate immune function. This study aimed to evaluate the short-term immunomodulatory effects of Lacticaseibacillus paracasei subspecies paracasei CNCM I-1518 (L. paracasei CNCM I-1518) in healthy adults. [...] Read more.
Background/Objectives: The gut microbiota and immune system are interconnected, with targeted nutritional interventions offering potential to modulate immune function. This study aimed to evaluate the short-term immunomodulatory effects of Lacticaseibacillus paracasei subspecies paracasei CNCM I-1518 (L. paracasei CNCM I-1518) in healthy adults. Methods: A 15-day dietary intervention was conducted involving healthy adults. Nutritional status, dietary habits, and systemic immune biomarkers were assessed, alongside changes in gut microbiota composition. Results: The results revealed significant effects on both cellular and humoral immunity. Cellular immunity was enhanced through increased circulating B lymphocytes, absolute monocyte counts, and leukocyte numbers, alongside reduced eosinophil levels, potentially mitigating allergic responses. Humoral immunity was improved by elevated serum IgG1, IgG2, and IgG4 levels, enhancing defenses against pathogenic antigens, and increased serum complement proteins C3 and C4, supporting innate immunity. Microbiota analysis showed a reduction in Clostridium and the Clostridium/Escherichia coli ratio, with a notable increase in the Lactobacillus/Clostridium ratio, highlighting the strain’s ability to reshape intestinal bacterial balance. Conclusions: A short-term intake of L. paracasei CNCM I-1518 can simultaneously modulate immune function and gut microbiota composition, supporting its potential as a targeted dietary intervention to promote immune health. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Figure 1

37 pages, 18599 KiB  
Article
Diclofenac Immune-Mediated Hepatitis: Identification of Innate and Adaptive Immune Responses at Clinically Relevant Doses
by Jürgen Borlak and Reinhard Spanel
Int. J. Mol. Sci. 2025, 26(12), 5899; https://doi.org/10.3390/ijms26125899 - 19 Jun 2025
Viewed by 748
Abstract
Diclofenac is an effective medication for pain and inflammation. However, its use has been linked to hepatitis. To gain insight into diclofenac’s ability to cause hepatitis, we investigated the regulation of major effectors of the immune system following daily treatment of minipigs at [...] Read more.
Diclofenac is an effective medication for pain and inflammation. However, its use has been linked to hepatitis. To gain insight into diclofenac’s ability to cause hepatitis, we investigated the regulation of major effectors of the immune system following daily treatment of minipigs at 3 and 15 mg/kg for 28 days. Histopathology evidenced lobular inflammation, and through a combination of immunogenomics and immunopathology, we detected marked innate and adaptive immune responses. We identified 109 significantly regulated genes linked to neutrophil, monocyte, Kupffer cell, and lymphocyte responses and 32 code for cytokine- and interferon-γ-signaling. In support of wound repair, immunopathology evidenced manifest upregulation of macrophage migration inhibitory factor and CD74. Furthermore, the strong expression of IgG and IgM underscored humoral immune responses. Diclofenac caused an activation of the complement system, especially the C1 inhibitor of the classical pathway and C3 with critical functions in liver regeneration. The marked expression of complement factor B and H of the alternate pathway modulated B-cell responses. Likely, the upregulation of factor H protected hepatocytes from injury by limiting complement-mediated damage of inflamed cells. Additionally, diclofenac treatment elicited marked hepatic expression of lysozyme and KLF6. The latter earmarks M1-polarized Kupffer cells. We observed an extraordinary induction of calprotectin/S100A9 and of the monocyte/macrophage CD163 scavenger receptor, and therefore, we detected innate immune sensing of damaged cells. Lastly, we noted an unprecedented induction of the acute phase reactant SAA1 and DEC-205, which recognize apoptotic and necrotic cells. Together, our results offer mechanistic insights into immune-mediated liver injury patterns following diclofenac treatment. Full article
(This article belongs to the Section Molecular Toxicology)
Show Figures

Figure 1

29 pages, 2576 KiB  
Review
A Comprehensive Review of the Mechanisms of Human Q Fever: Pathogenesis and Pathophysiology
by José-Luis Pérez-Arellano, Jose Curbelo and Cristina Carranza-Rodriguez
Pathogens 2025, 14(6), 589; https://doi.org/10.3390/pathogens14060589 - 14 Jun 2025
Viewed by 1546
Abstract
Coxiella burnetii infection has a worldwide distribution, although the incidence and clinical manifestations vary between and within countries. There are the following four basic forms: asymptomatic infection, acute Q fever, chronic Q fever, and post-Q fever fatigue syndrome. The aim of this review [...] Read more.
Coxiella burnetii infection has a worldwide distribution, although the incidence and clinical manifestations vary between and within countries. There are the following four basic forms: asymptomatic infection, acute Q fever, chronic Q fever, and post-Q fever fatigue syndrome. The aim of this review is to provide a comprehensive overview of the important aspects of its pathogenesis and pathophysiology. First, we provide a brief update of the taxonomic aspects, basic structures, and genotypes of C. burnetii necessary for the proper interpretation of the following sections. Routes of infection, different stages of pathogenesis (respiratory entry of C. burnetii; penetration into alveolar macrophages, life cycle, and effects; systemic dissemination), and innate, acquired humoral and cell-mediated immune responses in different forms of infection are described in detail. The pathophysiology and clinical manifestations of Q fever, such as the main mechanisms of injury, in isolation and in combination, are reviewed. The clinical and biological manifestations of the two main forms of Q fever (acute and chronic) are outlined, with a brief definition and mention of the mechanisms of post-Q fever fatigue syndrome. Full article
(This article belongs to the Section Bacterial Pathogens)
Show Figures

Figure 1

13 pages, 504 KiB  
Article
Type 2 Innate Lymphoid Cell (Ilc2)-Deficient Mice Are Transcriptionally Constrained During Nippostrongylus brasiliensis Infection
by Damarius S. Fleming, Fang Liu, Joseph F. Urban and Robert W. Li
Pathogens 2025, 14(6), 571; https://doi.org/10.3390/pathogens14060571 - 7 Jun 2025
Viewed by 597
Abstract
Mouse models serve as a means of examining immune changes when genes of interest are knocked out (KO). One group of immune gene-producing cells that have been identified is type 2 innate lymphoid cells (Ilc2). These cells are involved in the production of [...] Read more.
Mouse models serve as a means of examining immune changes when genes of interest are knocked out (KO). One group of immune gene-producing cells that have been identified is type 2 innate lymphoid cells (Ilc2). These cells are involved in the production of Th2 equivalent immune responses and signal cytokine production during the resolution of Nippostrongylus brasiliensis parasite infection in mice lungs. However, many questions about Ilc2 activity in the gut remain. To study this, retinoic acid receptor (RAR)-related orphan receptor alpha (RORα)-deficient mice were infected with adult N. brasiliensis and arranged into four treatment groups. Ten days post-infection (dpi), mouse ileum tissue was extracted for RNA-Seq. The RORα-deficient mice showed little change in gene expression at 10 dpi (N = 51) when compared to the WT mice at 10 dpi (N = 915), displaying dysregulation within the mouse gut. Based on the results, the gene expression in the gut of Ilc2-deficient mice denoted that the inability to craft Ilc2 cells left the mice unable to mount classical helminth immune responses involving humoral, mast cell, and antibody Th2-driven reactions. Overall, the results showed the importance of Ilc2 in the gut during N. brasiliensis infections and the effect that the lack of these cells had on immunity. Full article
(This article belongs to the Special Issue Immunity and Immunoregulation in Helminth Infections)
Show Figures

Figure 1

9 pages, 511 KiB  
Brief Report
Immunotherapeutic Blockade of CD47 Increases Virus Neutralization Antibodies
by Lamin B. Cham, Thamer A. Hamdan, Hilal Bhat, Bello Sirajo, Murtaza Ali, Khaled Saeed Tabbara, Eman Farid, Mohamed-Ridha Barbouche and Tom Adomati
Vaccines 2025, 13(6), 602; https://doi.org/10.3390/vaccines13060602 - 31 May 2025
Viewed by 804
Abstract
Background/Objectives: CD47 is a cell surface glycoprotein moderately expressed in healthy cells and upregulated in cancer and viral infected cells. CD47’s interaction with signal regulatory protein alpha (SIRPα) inhibits phagocytic cells and its interaction with thrombospondin-1 inhibits T cell response. Experimental evidence has [...] Read more.
Background/Objectives: CD47 is a cell surface glycoprotein moderately expressed in healthy cells and upregulated in cancer and viral infected cells. CD47’s interaction with signal regulatory protein alpha (SIRPα) inhibits phagocytic cells and its interaction with thrombospondin-1 inhibits T cell response. Experimental evidence has revealed that the blockade of CD47 resulted in the increased activation and function of both innate and adaptive immune cells, therefore exerting antitumoral and antiviral effects. Recent studies have shown that the combination of vaccines and immune checkpoint inhibitors could be a promising approach to increasing vaccine immunogenicity. Here, we investigated the vaccinal effect of anti-CD47 antibodies and discussed the possibilities of combining anti-CD47 treatments with vaccines. Methods: Using vesicular stomatitis virus (VSV), a widely used replication-competent vaccine vector, we evaluated the impact of the immunotherapeutic blockade of CD47 on cellular, humoral, and protective immunity. We infected C57BL/6 mice with VSV, treated them with anti-CD47 antibodies or an isotype, and evaluated the total immunoglobulin (Ig), IgG neutralizing antibodies, B cell activation, CD8+ T cell effector function, and survival of the mice. Results: We found that the treatments of anti-CD47 antibodies led to significantly increased Ig and IgG neutralizing antibody levels compared to the isotype treatment. Flow cytometric analysis of B cells revealed no difference in the number of circulating B cells; however, we observed an increased surface expression of CD80 and CD86 in B cells among anti-CD47-treated mice. Further analysis of the impact of CD47 blockade on T immunity revealed a significantly higher percentage of IFN-γ+ CD4 and IFN-γ+ CD8 T cells in anti-CD47-treated mice. Upon infecting mice with a lethal VSV dose, we observed a significantly higher survival rate among the anti-CD47-treated mice compared to control mice. Conclusions: Our results indicate that anti-CD47 treatment induces a stronger cellular and humoral immune response, leading to better protection. As such, immunotherapy by CD47 blockade in combination with vaccines could be a promising approach to improve vaccine efficacy. Full article
(This article belongs to the Section Vaccines against Infectious Diseases)
Show Figures

Figure 1

29 pages, 13121 KiB  
Article
Mechanistic Exploration of Yiqi Zengmian in Regulating the Microenvironment as an Immunopotentiator with the Beijing Bio-Institute of Biological Products Coronavirus Vaccine Based on Transcriptomics and Integrated Serum Pharmacochemistry
by Zeyue Yu, Yudong Wang, Jianhui Sun, Xiaotong Zheng, Liyu Hao, Yurong Deng, Jianliang Li, Zongyuan Li, Zhongchao Shan, Weidong Li, Yuling Qiao, Ruili Huo, Yibai Xiong, Hairu Huo, Hui Li, Longfei Lin, Hanhui Huang, Guimin Liu, Aoao Wang, Hongmei Li and Luqi Huangadd Show full author list remove Hide full author list
Pharmaceuticals 2025, 18(6), 802; https://doi.org/10.3390/ph18060802 - 27 May 2025
Viewed by 723
Abstract
Background: Yiqi Zengmian (YQZM) functions as an immunopotentiator by enhancing both cellular and humoral immunity. However, its pharmacodynamic active constituents, particularly those absorbed into the bloodstream, and mechanism of action remain unclear. This study aimed to investigate the immunopotentiating effects and mechanisms [...] Read more.
Background: Yiqi Zengmian (YQZM) functions as an immunopotentiator by enhancing both cellular and humoral immunity. However, its pharmacodynamic active constituents, particularly those absorbed into the bloodstream, and mechanism of action remain unclear. This study aimed to investigate the immunopotentiating effects and mechanisms of YQZM in mice immunized with the BBIBP-CorV (Beijing Bio-Institute of Biological Products Coronavirus Vaccine). Methods: Serum pharmacochemistry and ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS) were employed to identify bioavailable components of YQZM. The mice received the BBIBP-CorV twice on days 1 and 14, while YQZM was orally administered for 28 days. Neutralization assays and ELISA quantified antigen-specific antibodies (abs), flow cytometry (FC) and intracellular cytokine staining (ICS) were used to assess immune cell populations and their cytokines, and an enzyme-linked immunospot assay (ELISpot) quantified memory T and B cells (MBs and MTs). To identify underlying mechanisms, network pharmacology, RNA sequencing (RNA-Seq), molecular docking, Western blotting (WB), and quantitative reverse transcription PCR (RT-qPCR) were performed. Results: YQZM significantly enhanced antigen-specific antibody titers, immune cell proportions, cytokine levels, and memory lymphocyte functions. UPLC-MS/MS analysis identified 31 bioactive compounds in YQZM. KEGG enrichment analysis based on RNA-Seq and network pharmacology implicated the TLR-JAK-STAT signaling pathway in YQZM’s immune-enhancing effects. WB and RT-PCR validated that YQZM upregulated the expression of critical nodes in the TLR-JAK-STAT signaling pathway. Furthermore, molecular docking indicated that YQZM’s primary active components exhibited strong binding affinity for critical proteins. Conclusions: YQZM effectively enhances vaccine-induced innate and adaptive immunity via a multi-component, multi-target mechanism, among which the TLR-JAK-STAT signaling pathway is a validated molecular target. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Graphical abstract

25 pages, 1538 KiB  
Systematic Review
Effect of Ochratoxin A (OTA) on the Immune System: A Systematic Review
by Yusif Mubarik, Shadrach Tetteh Boyetey, Anastasia Rosebud Aikins and Mohamed Mutocheluh
Toxins 2025, 17(5), 256; https://doi.org/10.3390/toxins17050256 - 20 May 2025
Viewed by 1096
Abstract
Ochratoxin A (OTA) is a mycotoxin with different adverse health effects. The authors conducted a systematic review to evaluate the effects of OTA on the immune system, with more emphasis on its effects on immune system organs, innate and adaptive immunity and related [...] Read more.
Ochratoxin A (OTA) is a mycotoxin with different adverse health effects. The authors conducted a systematic review to evaluate the effects of OTA on the immune system, with more emphasis on its effects on immune system organs, innate and adaptive immunity and related signaling pathways. Studies have demonstrated that exposure to OTA disrupts the functions of immune system organs, resulting in weight loss, histological lesions and a decrease in antibody-secreting cells. There is evidence that OTA impairs epithelial barrier integrity and macrophage function and induces elevated secretion of pro-inflammatory cytokines. In adaptive immunity, OTA regulates T-cell differentiation, particularly Th1 and Th17 subsets, and adversely impacts humoral immunity, ultimately leading to immune suppression. Full article
(This article belongs to the Special Issue Mycotoxins in Food and Feeds: Human Health and Animal Nutrition)
Show Figures

Graphical abstract

21 pages, 3032 KiB  
Review
Bovine Adenoviral Vector-Based Platform for Vaccine Development
by Ekramy E. Sayedahmed, Vivek Gairola, Muralimanohara S. T. Murala and Suresh K. Mittal
Vaccines 2025, 13(5), 494; https://doi.org/10.3390/vaccines13050494 - 3 May 2025
Viewed by 1162
Abstract
Adenoviral (AdV) vector-based vaccines employing the human AdV (HAdV) and chimpanzee AdV (ChAdV) vector platforms played a crucial role in combating the COVID-19 pandemic. However, the widespread use of these platforms, the prevalence of various HAdV types, and the resulting preexisting immunity have [...] Read more.
Adenoviral (AdV) vector-based vaccines employing the human AdV (HAdV) and chimpanzee AdV (ChAdV) vector platforms played a crucial role in combating the COVID-19 pandemic. However, the widespread use of these platforms, the prevalence of various HAdV types, and the resulting preexisting immunity have significantly impacted the vaccines utilizing these vector platforms. Considering these challenges, the bovine AdV type 3 (BAdV-3) vector system has emerged as a versatile and innovative platform for developing next-generation vaccines against infectious diseases. Inherent attributes like a high transduction efficiency, large transgene insertion capacity, broad tissue tropism, and robust induction of innate immunity add significant value to the BAdV vector platform for vaccine design. BAdV-3 vectors effectively elude HAdV-specific preexisting humoral and cellular immune responses. Additionally, BAdV-3 is low in pathogenicity for its host and is anticipated to be safe as a vaccine platform. This systematic review provides an overview of the development of BAdV-3 as a vaccine delivery platform and its application in designing vaccines for infectious agents of human and veterinary importance. Full article
(This article belongs to the Special Issue Innovations in Vaccine Technology)
Show Figures

Figure 1

17 pages, 1693 KiB  
Review
Host Immune Response to Bovine Viral Diarrhea Virus (BVDV): Insights and Strategies for Effective Vaccine Design
by Asamenew Tesfaye Melkamsew, Tesfaye Sisay Tessema and Jan Paeshuyse
Vaccines 2025, 13(5), 456; https://doi.org/10.3390/vaccines13050456 - 25 Apr 2025
Viewed by 1643
Abstract
Bovine viral diarrhea (BVD) is caused by bovine viral diarrhea virus (BVDV), a member of the genus Pestivirus and in the family Flaviviridae. According to some studies, the disease incurs USD 1.5–2.5 billion per year and USD 0.50 to USD 687.80 per cow [...] Read more.
Bovine viral diarrhea (BVD) is caused by bovine viral diarrhea virus (BVDV), a member of the genus Pestivirus and in the family Flaviviridae. According to some studies, the disease incurs USD 1.5–2.5 billion per year and USD 0.50 to USD 687.80 per cow loss in beef and dairy farms, respectively. Using vaccines is among the strategies to prevent the disease. However, complete protection requires vaccines that target both the humoral and cellular immune responses of the adaptive immune system. A comprehensive literature review was made to provide insights into the interaction of BVDV with host immunity, vaccine applications, and the limitation of the currently available vaccines, as well as explore strategies used to advance the vaccines. BVDV causes immunosuppression by interfering with the innate and adaptive immune systems in a manner that is species and biotype-dependent. Interferon production, apoptosis, neutrophil activity, and antigen-processing and presenting cells are significantly affected during the viral infection. Despite maternal antibodies (MatAbs) being crucial to protect calves from early-age infection, a higher level of MatAbs are counterproductive during the immunization of calves. There are numerous inactivated or modified BVDV vaccines, most of which are made of cytopathic BVDV 1 and 2 and the BVDV 1a subgenotypes. Furthermore, subunit, marker, DNA and mRNA vaccines are made predominantly from E2, Erns, and NS3 proteins of the virus in combination with modern adjuvants, although the vaccines have not yet been licensed for use and are in the experimental stage. The existing BVDV vaccines target the humoral immune system, which never gives the full picture of protection without the involvement of the cell-mediated immune system. Several limitations were associated with conventional and next-generation vaccines that reduce BVDV vaccine efficiency. In general, providing complete protection against BVDV is very complex, which requires a multi-pronged approach to study factors affecting vaccine efficacy and strategies needed to improve vaccine efficacy and safety. Full article
(This article belongs to the Special Issue Vaccines and Antibody-Based Therapeutics Against Infectious Disease)
Show Figures

Figure 1

Back to TopTop