Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Immunol., 21 October 2025

Sec. Vaccines and Molecular Therapeutics

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1643855

This article is part of the Research TopicInnovative Adjuvant Strategies: Enhancing Vaccine Efficacy Through Transdisciplinary ApproachesView all 10 articles

Harnessing cytokines: innovative adjuvants for improved veterinary vaccine efficacy

Shihong Yan,*&#x;Shihong Yan1,2*†Huimin Chen,&#x;Huimin Chen1,2†Qiao Liu,Qiao Liu1,2Xinyu Zhang,Xinyu Zhang1,2Qiulu Wei,Qiulu Wei1,2Chenqi Hu,Chenqi Hu1,2Dianfeng ChuDianfeng Chu3Hongxiu Diao,Hongxiu Diao1,2Shasha Liu,Shasha Liu1,2Ji-Long Chen,Ji-Long Chen1,2
  • 1Key Laboratory of Animal pathogen Infection and Immunology of Fujian Province, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
  • 2Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
  • 3YEBIO Bioengineering Co., Ltd, Qingdao, China

Vaccination is one of the most effective methods for controlling animal infectious diseases, and the use of adjuvants plays crucial role in enhancing the immune efficacy of vaccines, particularly in inactivated and subunit vaccines. With the continuous advancement of research in animal immunology and immune mechanisms, our understanding of the functions of cells and cytokines in immune responses has become increasingly comprehensive, laying a solid foundation for the development of novel vaccines and adjuvants. Cytokines are a class of proteins secreted by the animal body that regulate innate and adaptive immune responses through interaction with specific receptors. To date, numerous studies have investigated the potential of using cytokines as adjuvants to enhance the efficacy of veterinary vaccines. This review focuses on cytokines as veterinary vaccine adjuvants, with special attention to the current research progress and mechanisms of cytokines such as interleukins, interferons, chemokines, and colony-stimulating factors. Additionally, examples of the application of cytokine-based adjuvants in combination with veterinary vaccines will be discussed to provide further insights and references for the development of cytokine-based veterinary adjuvants.

1 Introduction

Vaccination is one of the most effective strategies for controlling infectious diseases in livestock and poultry. Adjuvants, as immune enhancers, also play an indispensable role in the immunological control and prevention of animal diseases. Currently, adjuvants are widely used in the preparation of vaccines, particularly in those with weaker immunogenicity such as inactivated vaccines, synthetic peptide vaccines, subunit vaccines, and DNA vaccines (1, 2). Adjuvants can effectively reduce the number of immunizations and the amount of antigens needed, while directing the immune response towards the desired direction (3). Additionally, adjuvants help to overcome antigen competition issues in combined vaccines and improve their efficacy in immunocompromised animals (4, 5). However, the majority of veterinary vaccine adjuvants still rely on traditional adjuvants such as aluminum hydroxide and oil emulsions, which, despite their widespread use, are often associated with side effects like joint pain and muscle discomfort (6). For example, the well-known Freund’s adjuvant can cause severe adverse reactions, leading to local inflammatory lesions, pain, and discomfort. Given these limitations, there is an urgent need to develop safer and more effective new adjuvants to improve vaccine safety and immune efficacy.

Cytokines are soluble proteins produced upon stimulation by immunogens, mitogens, or other factors, and they play critical roles in signal transduction (7). By binding to specific receptors, cytokines can regulate various biological processes, including innate and adaptive immunity, hematopoiesis, cell growth, and tissue repair (8). Recent studies have demonstrated that recombinant cytokines can enhance the host resistance to disease, improve physiological functions, and maintain immune homeostasis. These findings suggest that cytokines have significant potential in enhancing vaccine efficacy and adjuvant activity.

Cytokines encompass a wide variety of molecules, including interleukins, interferons, tumor necrosis factor superfamily, colony-stimulating factors, chemokines, and growth factors. Due to their origin from the animal’s own body, cytokines are efficient, safe, and specific, with clear species specificity, which minimizes the risks of residues and adverse side effects compared to traditional adjuvants. Using cytokines as adjuvants in veterinary vaccines not only significantly enhances vaccine efficacy but also ensures the food safety of livestock and poultry products. Therefore, developing cytokine-based adjuvants is crucial for supporting the sustainable growth of the livestock industry and driving socio-economic progress. With increasing research on cytokines as vaccine adjuvants, diverse delivery methods and carrier systems have become a focus. These strategies play critical roles in cytokine stability, targeting, and immune activation. Figure 1 provides a schematic illustration of these common approaches.

Figure 1
Diagram illustrating cytokine adjuvant development methods. Central red circle labeled “Cytokine Adjuvant” connects to four sections: “Gene Co-expression” using plasmid DNA or viral vectors, “Recombinant Protein” involving direct mixing with vaccine antigens, “Fusion Proteins” combining cytokines with antigens or targeting molecules, and “Nanoparticle Delivery” with cytokines encapsulated in liposomes or polymer nanoparticles. Each section represents a different delivery or expression method.

Figure 1. Delivery methods and carrier systems of cytokines adjuvants in veterinary vaccines.

This paper discusses the current research on cytokine-based vaccine adjuvants, with a focus on the application status and adjuvant mechanisms of major cytokines such as interleukins, interferons, chemokines, and colony-stimulating factors in veterinary vaccine development. This review aims to provide theoretical references and practical guidance for the research and development of cytokine-based adjuvants.

2 Interleukins

Interleukins (IL) were initially described as cytokines produced by leukocytes that regulate interactions among these cells. Today, ILs refer to a family of cytokines with well-characterized molecular structures and biological functions that play critical roles in immune regulation. ILs mediate the transmission of information, activation and regulation of immune cells, as well as the activation, proliferation, and differentiation of T and B cells. They also play essential roles in inflammatory responses. Table 1 presents the key mechanisms of cytokine-related vaccines of interleukin class, the antigens used, and the corresponding supporting literature.

Table 1
www.frontiersin.org

Table 1. Comprehensive overview of cytokine-related vaccines of interleukin class: mechanisms, types, antigens, and references.

2.1 IL-1β

IL-1β directly influences the proliferation and differentiation of CD4 and CD8 T cells, particularly IL-4-producing cells, and also enhances the tissue localization and memory responses of CD8 T cells (21). The production and release of IL-1β are stimulated by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). IL-1β as an adjuvant with a recombinant PRRSV vaccine induce a robust T-cell immune response, increase IFN-γ and IL-4 levels, and elicit higher serum antibody levels (9). This indicates that IL-1β has a dual role in enhancing both cellular and humoral immune responses. Furthermore, IL-1β serves as an effective mucosal vaccine adjuvant based on the capacity of attracting both innate and adaptive immune cells through the induction of chemokines and adhesion molecules (22), specifically inducing tissue-resident memory T cells mediated a rapid clearance of secondary IAV infections in mice, which improves heterosubtypic immunity against influenza A viruses (10).

2.2 IL-2

IL-2 is a cytokine that promotes the growth of bone marrow-derived T lymphocytes and was one of the first cytokines to be characterized at the molecular level (23). IL-2 enhances the growth activity of various cells, particularly the proliferation of CD4+ and CD8+ T lymphocytes (24). Additionally, IL-2 promotes the production of cytokines by natural killer (NK) cells and synergizes with IL-12 to enhance NK cell cytotoxic activity (25). In B cells, IL-2 primarily influences antibody secretion (26). There have been numerous reports on the use of recombinant IL-2 as a vaccine adjuvant (27, 28). Recombinant IL-2 encapsulated in nano-liposomes significantly enhances the levels of foot-and-mouth disease virus (FMDV)-specific antibodies and the concentrations of IFN-γ secreted from spleen cells through Th1 immune response as well as maintain longer periods of time to stimulate T and B cell proliferation and differentiation to improve antibody secretion, which successfully solves the shortcoming of a short half-life of IL-12. Additionally, it increases the proliferative responses of antigen-specific spleen cells, demonstrating its effective adjuvant properties (11). Co-expression of the IL-2 and VP60 genes in a DNA vaccine for rabbit hemorrhagic disease induced higher levels of neutralizing antibodies and IL-4 expression, while reducing tissue damage upon challenge, confirming the effectiveness of IL-2 as an adjuvant (12).

2.3 IL-4

IL-4 is a type I cytokine with a four-α-helix bundle structure that exhibits pleiotropic effects across multiple lineages. While IL-4 is produced by various immune cells, it is primarily secreted by activated CD4+ T cells (29). IL-4 mediates host sensitization and parasitic responses via IgE and induces IgG production, particularly IgG1 in B cells (3032). In humans and mice, IL-4 acts as a T-cell growth factor and promotes Th2 cell differentiation. Studies have shown that recombinant Newcastle disease virus (NDV) expressing chicken IL-4 significantly reduced organ damage and virus shedding in vaccinated chickens compared to wild-type virus, indicating potential antiviral and protective adjuvant effects of IL-4 (13). As a genetic adjuvant, IL-4 co-expressed in a Trichinella spiralis DNA vaccine significantly elevated Th1/Th2 cytokine levels, alleviated intestinal damage, and demonstrated effective adjuvant functionality (14).

2.4 IL-6

IL-6 is a multifunctional pro-inflammatory cytokine with diverse roles in inflammation, immune responses, and hematopoiesis. IL-6 synergizes with transforming growth factor-β (TGF-β) to promote the differentiation of naïve CD4+ T cells, thereby enhancing adaptive immune responses (33). Furthermore, IL-6 promotes the production of IL-21, aids in the differentiation of T follicular helper (Tfh) cells (34) and CD8+ T cells (35), and induces B cell differentiation into plasma cells (36), thereby enhancing antibody production. The co-expression of IL-6 as the molecular adjuvant with FMDV DNA vaccine, induced a higher ratio ofIgG2a/IgG1, higher levels of expression of IFN-γin CD4+ and CD8+ T cells, IL-4 in CD4+ T cells, and in vivo antigen-specific cytotoxic response, which confirm both Th1 and Th2 immune response are activated (37). Both recombinant IL-6 protein and plasmids expressing the IL-6 gene have been used as adjuvants in studies on Japanese flounder (Paralichthys olivaceus). These adjuvants increased antibody levels, enhanced immune responses, reduced bacterial infection-induced damage, and significantly decreased mortality (15). Moreover, vaccination of mice with recombinant rabies virus expressing IL-6 resulted in earlier and higher antibody titers compared to the wild-type virus, demonstrating the potential adjuvant activity of IL-6 in enhancing vaccine immunogenicity (16).

2.5 IL-12

IL-12 is a member of the interleukin-12 (IL-12) family cytokines with an integral effect in activating cellular immune responses in mammals (38). When pathogens infect the host, IL-12 stimulates Th1 cell to release IFN-γ, promoting the Th1 cellular immune response and enhance the host’s property to clear the pathogens. For intracellular pathogens, IL-12 induces macrophages or cytotoxic T lymphocytes (CTLs) to destroy infected cells (39, 40). Macrophages exhibit strengthened activation activities based on regulation of IL-12 and upregulate the production and release of nitric oxide (NO) to further enhance the ability for antigen clearance (41). Multiple functional studies have highlighted IL-12 as a potential vaccine adjuvant with immunomodulatory properties. Co-delivery of an IL-12-expressing plasmid with an NDV F gene DNA vaccine using electroporation has been shown to significantly enhance immune responses in chickens, resulting in higher neutralizing antibody levels, increased lymphocyte proliferation, reduced viral shedding, and complete protection compared to the DNA vaccine alone (17). In addition, co-immunization with an IL-12 eukaryotic expression plasmid and a Toxoplasma gondii multi-epitope vaccine (pcROP8) enhanced the Th1 response and IFN-γ secretion, thereby providing heightened vaccine protection (18).

2.6 IL-15

IL-15 is a critical factor for the development, proliferation, and activation of effector NK cells and CD8+ memory T cells. It plays important roles in NK cell proliferation, cytotoxicity, cytokine production, NK cell-macrophage interactions, and the maintenance of CD4+/CD8+ memory T cell homeostasis (42). IL-15 supports the long-term persistence of CD8+ T cells and effectively extends immune duration, making it a preferred adjuvant for improving immune responses and vaccine longevity (43). The inactivated vaccine is short activities to the immune response, when bovine-derived IL-15 has been used as an adjuvant in guinea pigs immunized with an inactivated FMDV vaccine, the IL-15 adjuvanted vaccine maintained neutralizing antibody levels for up to six months in animals receiving. As well as Compared to animals immunized with the inactivated vaccine alone, those vaccinated with IL-15 adjuvants exhibited stronger Th1 and Th2 immune responses (19).

2.6 IL-18

IL-18, initially identified as an interferon-γ-inducing factor (44), synergizes with IL-12, mitogens, or microbial agents to promote IFN-γ production by T cells and NK cells (4547). IL-18 also induces the expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) in peripheral blood mononuclear cells (PBMCs) (44, 4850) and stimulates IL-13 production (51). When IL-18 plasmids encapsulated in PLGA nanoparticles were used as adjuvants in combination with a foot-and-mouth disease virus DNA vaccine, they induced higher antibody titers and stronger CTL responses in guinea pigs, compensating for the limited cellular immunity often observed with inactivated FMDV vaccines (20).

3 Interferons

Interferons (IFNs) are a large class of cytokines that are critical in activating the immune response of the host. IFNs are categorized into three types: Type I, Type II, and Type III, all of which have the ability to activate antiviral activity by interacting with their respective receptors (52). Type I IFNs (primarily IFN-α, -β, and -ω) participate in viral clearance by inducing immune responses and provide protection against acute viral infections. Type II IFN (IFN-γ), primarily produced by activated NK cells and T cells, plays a pivotal role in both innate and adaptive immunity (53). Type III IFNs (IFN-λ1, -λ2, and -λ3) are associated with antiviral immune responses at epithelial surfaces, with their receptors being most abundantly expressed in cells of epithelial origin (54, 55). Table 2 presents the key mechanisms of cytokine-related vaccines of interferons class, the antigens used, and the corresponding supporting literature.

Table 2
www.frontiersin.org

Table 2. Comprehensive overview of cytokine-related vaccines of interferon class: mechanisms, types, antigens, and references.

3.1 Type I IFNs

Type I IFNs possess immunomodulatory properties and can regulate the activity of other cytokines (62). They enhance the maturation and activation of dendritic cells (56, 63), promote Th1-type immune responses, and activate B cells to facilitate antibody production (64, 65). A combination of recombinant porcine IFN-α protein and inactivated influenza vaccine has been shown to significantly upregulate the expression of immunomodulatory cytokines such as IL-2, IL-6, IL-10, IL-18, and IFN-γ. This combination also significantly increases the transcription of homing factors CCR9 and CCR10, induces a strong mucosal innate immune response, and enhances antibody levels (53). In another study, immunization with a Venezuelan equine encephalitis virus (VEEV) vaccine containing an IFN-α plasmid adjuvant in mice resulted in the robust expression of antiviral proteins and induced specific immunity against VEEV (57).

3.2 Type II IFN

Type II IFN is primarily produced by activated Th cells and NK cells (66). It is a multifunctional homodimeric cytokine, with IFN-γ being its sole member (58, 59). The main biological function of IFN-γ is to induce the expression of various immune factors, thereby enhancing the body’s immune response. Several studies have demonstrated that IFN-γ is an effective adjuvant for veterinary vaccines. For instance, immunization of mice with a recombinant Hyalomma asiaticum rHasCPL protein subunit vaccine combined with an IFN-γ adjuvant increased the production of IFN-γ and IL-4, enhanced antibody levels, and improved the protective efficacy of the vaccine in mice (67). In vitro experiments have shown that the expression of porcine IFN-γ can significantly enhance the pro-inflammatory immune response in cells infected with PRRSV (68). Additionally, in Japanese flounder, the use of an IFN-γ adjuvant with an Edwardsiella tarda subunit vaccine effectively increased survival rates, upregulated the expression of immune-related genes, and enhanced antibody production (69). Furthermore, a study assessing the immunoadjuvant effects of a recombinant poIFN-γ-poGM-CSF fusion protein in an inactivated PRRSV vaccine administered to piglets found that the coadministration of poIFN-γ-linker-poGM-CSF and PRRSV KV significantly increased neutralizing antibody titers, accelerated viral clearance, reduced clinical symptoms, and prevented highly pathogenic PRRSV infection (70). This reinforces the critical role of IFN-γ and its fusion proteins in enhancing vaccine efficacy and providing protection against viral infections in veterinary medicine.

3.3 Type III IFN

Type III IFNs (IFN-λ1, λ2, and λ3) are structurally related to Type I IFNs and the IL-10 family (60), and are also known as IL-29, IL-28a, and IL-28b (61, 71). Their receptor, IL-28Rα, is expressed on a limited range of cells such as macrophages, peripheral blood lymphocytes, conventional dendritic cells, epithelial cells, and plasmacytoid dendritic cells (61, 72). IFN-λ primarily acts on these cell types to regulate antiviral immunity, thus possessing potential as an adjuvant to enhance immune responses. In studies where a Porcine Reproductive and Respiratory Syndrome (PRRS) DNA vaccine expressing IFNλ1 was used to immunize mice, there was an upregulation of serum antibodies and activation of the STAT signaling pathway. This suggests that IFNλ1 can enhance the immune protective effect of PRRSV DNA vaccines (73, 74).

4 Chemokines

Chemokines are a class of cytokines that play a significant role in inducing cell migration and motility, stimulating intracellular signaling pathways (75). They regulate lymphocyte development, activation, and effector functions and play a crucial role in immune surveillance. Many chemokines have been shown to be effective immunological adjuvants, enhancing the protective effects induced by viral, bacterial, and parasitic vaccines (76, 77). They are categorized into four major subclasses based on their conserved cysteine motifs, known as C, CC, CXC, and CX3C (78). Table 3 presents the key mechanisms of cytokine-related vaccines of chemokines class, the antigens used, and the corresponding supporting literature.

Table 3
www.frontiersin.org

Table 3. Comprehensive overview of cytokine-related vaccines of chemokines class: mechanisms, types, antigens, and references.

4.1 CCL4

CCL4, also known as Macrophage Inflammatory Protein-1β (MIP-1β) (79), is effective chemoattractant for CD4+CD25+ T cell populations and is a phenotypic characteristic of regulatory T cells (80). CCL35.2 in crucian carp has the highest identity with mammalian CCL4. Using CCL35.2 plasmid adjuvant in combination with a DNA vaccine to immunize crucian carp can effectively upregulate the mRNA expression of key immune genes IL-1β, IL-2, IFN-γ2, and viperin in Carassius auratus gibelio. It also increases the levels of complement C3, lysozyme, and total superoxide dismutase, significantly enhancing the resistance of crucian carp to Cyprinid herpesvirus 2 (81).

4.2 CCL28

CCL28, also known as Mucosa-Associated Epithelial Chemokine (MEC), has unique immunoregulatory properties in various mucosal areas, attracting IgA and directing their migration to different mucosal sites (82, 83). Many chemokines are effective immune adjuvants in various model systems, enhancing protection induced by viral, bacterial, and parasitic vaccines, and regulating the direction and magnitude of induced immune responses produced by DNA, protein, subunit, or peptide vaccines (77). When used as a virus-like particle (VLP) vaccine adjuvant for influenza vaccines, CCL28 can act as an immune stimulant in a membrane-bound form, eliciting a systemic mucosal immune response and significantly enhancing the host’s cross-protective efficacy against heterologous viruses (84). Furthermore, studies have shown that the addition of a CCL28 adjuvant in H3N2 influenza vaccines can induce a significant increase in IgA levels and hemagglutination inhibition (HI) titers, enhancing long-term cross-protection against H3N2 influenza virus (85). A vaccination strategy that employs the intramuscular co-delivery of CCL27 or CCL28 has been shown to generate strong systemic and local immune responses, leading to the production of long-lived antibodies that neutralize influenza (86).

5 Colony-stimulating factors

Members of the CSF superfamily are involved in the generation of mammalian bone marrow cells, including monocytes, macrophages, dendritic cells, and polymorphonuclear phagocytes such as neutrophils and eosinophils. This family contains three key members: Macrophage Colony-Stimulating Factor (M-CSF or CSF-1), Granulocyte Colony-Stimulating Factor (G-CSF or CSF-3), and Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF or CSF-2). Among these, GM-CSF has been extensively studied for its potential as an adjuvant. Table 4 presents the key mechanisms of cytokine-related vaccines of colony stimulate factor, the antigens used, and the corresponding supporting literature.

Table 4
www.frontiersin.org

Table 4. Comprehensive overview of cytokine-related vaccines of colony stimulate factor: mechanisms, types, antigens, and references.

5.1 GM-CSF

GM-CSF is a hematopoietic growth factor produced by various immune cells (87). It stimulates the proliferation and differentiation of bone marrow progenitor cells into granulocytes and macrophages (88), as well as activating and maintaining mature bone marrow cells (89). GM-CSF responds to immune cell survival, differentiation, and proliferation by inducing various signaling pathways, which is a key step in helping the immune system fight infections (90). GM-CSF levels significantly increase during inflammatory responses (90, 91), and multiple studies have demonstrated that T cell-derived GM-CSF plays an essential role in immune responses against a variety of pathogens, such as Mycobacterium tuberculosis, Epstein-Barr virus, and Human Immunodeficiency Virus and the rabies virus (9296). Based on its pro-inflammatory effects induced by recruiting and activating bone marrow cells (90), recombinant pGM-CSF has been tested as a co-adjuvant with pFLIC protein in conjunction with a Porcine Circovirus (PCV) vaccine to immunize pigs. This approach significantly elevated PCV-specific antibody levels, stimulated CD4+ and CD8+ T cell proliferation, and upregulated the transcription of IL-1, IL-8, and IL-17, indicating a robust enhancement of both humoral and cellular immune responses (97). Similarly, the combination of GM-CSF and APS as a complex immunostimulant in a PRV vaccine model resulted in higher levels of PRV-specific gB and neutralizing antibodies, and concurrently increased the production of cytokines including IL-4, IL-10, IL-2, and IFN-γ (98). These findings indicate that GM-CSF-based adjuvant strategies have promising application prospects.

6 Other cytokines with adjuvant potential

Other cytokines reported to have potential as veterinary vaccine adjuvants include Interferon-Induced Transmembrane Proteins (IFITMs), B-cell Activating Factor (BAFF), α-Galactosylceramide (α-GalCer), Fms-like Tyrosine Kinase 3 Ligand (FLT3-L), and CD40L (Table 5). IFITMs: Transgenic chickens overexpressing IFITM1 can effectively resist H5N1 influenza virus infection by inhibiting viral replication within the body (99). Although most studies on chicken IFITM1 and IFITM3 functionality have been conducted in vitro or in chicken embryos (100), recent findings suggest that recombinant avian-derived antiviral proteins, including cIFITM1, cIFITM3, and cViperin, can serve as effective adjuvants in inactivated H9N2 subtype avian influenza vaccines (101). This highlights the potential of these proteins not only in enhancing viral resistance but also in improving the efficacy of avian influenza vaccines. BAFF: Incorporating membrane-anchored BAFF into Rabies Virus (RABV) virus-like particles (VLPs) induced higher antibody titers compared to inactivated RABV vaccines, demonstrating that BAFF is an effective membrane-anchored molecular adjuvant (102) α-GalCer: When used as an adjuvant with an inactivated H1N1 swine influenza vaccine administered intranasally, α-GalCer enhanced Th1 cytokine (IFN-γ and IL-12) secretion in the lungs, reduced the levels of immunosuppressive cytokines (IL-10 and TGF-β), and decreased lung viral loads (103). FLT3-L: Exogenous FLT3-L addition promoted the proliferation of CD141+ dendritic cells (DCs) and CD1c+ DCs in mouse blood, spleen, and bone marrow, thereby improving antigen presentation capabilities. This indicates its potential to enhance vaccine immunogenicity and promote antigen recognition, although research is currently limited and requires further exploration (104). CD40L: The co-administration of plasmid-expressed CD40L with Montanide™ GEL01 adjuvant enhanced the protective efficacy of a Bovine Herpesvirus-1 (BoHV-1) DNA vaccine. This combination increased the percentage of PBMCs and upregulated the expression of IFN-γ and IL-4 in cattle (105). The co-expression of CD40L and CD205 also receive the similar results (106).

Table 5
www.frontiersin.org

Table 5. Comprehensive overview of cytokine-related vaccines of other cytokines: mechanisms, types, antigens, and references.

7 Conclusion

In modern veterinary vaccine industries, vaccine adjuvants are considered crucial bridges between innate and adaptive immunity. An effective adjuvant can enhance the immunogenicity of a vaccine through various mechanisms, including promoting cytokine production, inducing antibody generation, regulating immune type switching, optimizing surface delivery to immune tissues, and improving the uptake efficiency of antigen-presenting cells. Moreover, the application of nanomaterials holds promise for further enhancing the effects of cytokine adjuvants by optimizing release characteristics and improving bioavailability, thus providing new support for immunological enhancement of vaccines (107, 108). As shown in Figure 2, the potential benefits of cytokines as vaccine adjuvants include different types of cytokines and their corresponding mechanisms, which play a crucial role in enhancing the immunogenicity of vaccines. This review summarizes the research and applications of different cytokines as veterinary vaccine adjuvants.

Figure 2
Circular diagram illustrating the potential benefits of cytokines as adjuvants in vaccines. Sections highlight: chemotactic cytokines enhancing immune cell recruitment, pro-inflammatory promoting activation, regulatory modulating responses, helper enhancing cellular immunity, antibody-producing promoting B cell differentiation, and memory cell-promoting facilitating rapid responses.

Figure 2. Potential benefits of cytokines as adjuvants in in veterinary vaccines.

Interleukin enhances humoral and cellular immunity, induces Th1 and Th2 type responses in key effector cells, thereby prolonging the duration of immunity and improving the actual protective effect of vaccines. Interferon can upregulate the expression of immune regulatory cytokines and activate key signaling pathways such as STAT. On the one hand, it promotes high-level immune responses in mucosal areas, and on the other hand, it synergistically enhances humoral and cellular immunity at the systemic level, activating multifunctional T cell responses. In addition, interferon can stimulate strong pro-inflammatory responses while also avoiding pathological damage by regulating T cells and other mechanisms, endowing vaccines with faster onset speed, wider cross protection range, stronger pathogen clearance ability, and significantly prolonged protection duration. Chemokines can precisely regulate immune cells such as CD4+, CD25+T cells, fully activate immune molecules, induce the expression of specific antiviral proteins, enhance immune response, and increase antibody levels. GM-CSF serves as a bridge between innate and acquired immunity, activating cells to promote the quantity and function of antigen-presenting cells, and significantly increasing pathogen specific antibody levels and enhancing immune protection against pathogens. Due to their natural presence in the host organism, cytokines exhibit good biocompatibility and safe immunomodulatory effects, demonstrating significant potential as candidate adjuvants for vaccines. In recent years, numerous studies have explored the use of cytokines as vaccine adjuvants, clarifying the mechanisms by which they enhance immune responses. These studies indicate that cytokine-based adjuvants can effectively improve the immunogenicity of vaccines. Furthermore, the combination of various cytokines as adjuvants represents a promising strategy and may become a major focus for future adjuvant research and development (109, 110).

With the development of new technologies, the advancement of novel adjuvants is imperative, while their side effects must be continuously monitored to ensure vaccine safety. For low-cost veterinary vaccines, reducing the production cost of new adjuvants is particularly crucial (111). Additionally, research into the combination of nanomaterials and cytokines will further promote the application of novel cytokine-based adjuvants, supporting their feasibility in the veterinary vaccine market. However, to date, no commercialized cytokine-based veterinary vaccine adjuvants have been introduced. Given the frequent occurrence of animal diseases, future research must continuously improve veterinary vaccines and their adjuvants. This will provide new solutions to address the ever-evolving challenges posed by infectious diseases, thus enhancing animal health and the sustainable development of the livestock industry.

Author contributions

SY: Writing – review & editing, Writing – original draft. HC: Writing – original draft, Investigation. QL: Writing – review & editing. XZ: Writing – review & editing. QW: Formal analysis, Writing – review & editing. CH: Writing – review & editing. DC: Writing – review & editing, Project administration. HD: Writing – review & editing. YL: Writing – review & editing, Supervision. J-LC: Writing – review & editing, Supervision.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This paper was supported by the National Natural Science Foundation of China (Grant No. 32302905), the Research Fund of National Key Laboratory of Veterinary Public Health and Safety. (YEBIO Bioengineering Co.,Ltd of Qingdao) (Grant No. 2024SKLVPHS07).

Acknowledgments

It is conducted at the Key Laboratory of Animal Pathogen Infection and Immunology of Fujian Province, and we sincerely acknowledge their support.

Conflict of interest

Authors DC was employed by YEBIO Bioengineering Co., Ltd.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Firdaus FZ, Skwarczynski M, and Toth I. Developments in vaccine adjuvants. Methods Mol Biol. (2022) 2412:145–78. doi: 10.1007/978-1-0716-1892-9_8

PubMed Abstract | Crossref Full Text | Google Scholar

2. Cui Y, Ho M, Hu Y, and Shi Y. Vaccine adjuvants: current status, research and development, licensing, and future opportunities. J materials Chem B. (2024) 12:4118–37. doi: 10.1039/d3tb02861e

PubMed Abstract | Crossref Full Text | Google Scholar

3. Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, et al. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther. (2023) 8:283. doi: 10.1038/s41392-023-01557-7

PubMed Abstract | Crossref Full Text | Google Scholar

4. Akache B, Stark FC, Agbayani G, Renner TM, and McCluskie MJ. Adjuvants: engineering protective immune responses in human and veterinary vaccines. Methods Mol Biol. (2022) 2412:179–231. doi: 10.1007/978-1-0716-1892-9_9

PubMed Abstract | Crossref Full Text | Google Scholar

5. Schijns VEJC, Strioga M, and Ascarateil S. Oil-based emulsion vaccine adjuvants. Curr Protoc Immunol. (2014) 106:2.18.1–2.18.7. doi: 10.1002/0471142735.im0218s106

PubMed Abstract | Crossref Full Text | Google Scholar

6. Burakova Y, Madera R, McVey S, Schlup JR, and Shi J. Adjuvants for animal vaccines. Viral Immunol. (2018) 31:11–22. doi: 10.1089/vim.2017.0049

PubMed Abstract | Crossref Full Text | Google Scholar

7. Boyaka PN and McGhee JR. Cytokines as adjuvants for the induction of mucosal immunity. Adv Drug Delivery Rev. (2001) 51:71–9. doi: 10.1016/s0169-409x(01)00170-3

PubMed Abstract | Crossref Full Text | Google Scholar

8. Rahman T, Das A, Abir MH, Nafiz IH, Mahmud AR, Sarker M, et al. Cytokines and their role as immunotherapeutics and vaccine Adjuvants: The emerging concepts. Cytokine. (2023) 169:156268. doi: 10.1016/j.cyto.2023.156268

PubMed Abstract | Crossref Full Text | Google Scholar

9. Lawson SR, Li Y, Patton JB, Langenhorst RJ, Sun Z, Jiang Z, et al. Interleukin-1β expression by a recombinant porcine reproductive and respiratory syndrome virus. Virus Res. (2012) 163:461–8. doi: 10.1016/j.virusres.2011.11.007

PubMed Abstract | Crossref Full Text | Google Scholar

10. Lapuente D, Storcksdieck Genannt Bonsmann M, Maaske A, Stab V, Heinecke V, Watzstedt K, et al. IL-1β as mucosal vaccine adjuvant: the specific induction of tissue-resident memory T cells improves the heterosubtypic immunity against influenza A viruses. Mucosal Immunol. (2018) 11:1265–78. doi: 10.1038/s41385-018-0017-4

PubMed Abstract | Crossref Full Text | Google Scholar

11. Chen G, Zeng S, Jia H, He X, Fang Y, Jing Z, et al. Adjuvant effect enhancement of porcine interleukin-2 packaged into solid lipid nanoparticles. Res Veterinary Sci. (2014) 96:62–8. doi: 10.1016/j.rvsc.2013.11.017

PubMed Abstract | Crossref Full Text | Google Scholar

12. Deng Z, Geng Y, Wang K, Yu Z, Yang PO, Yang Z, et al. Adjuvant effects of interleukin-2 co-expression with VP60 in an oral vaccine delivered by attenuated Salmonella typhimurium against rabbit hemorrhagic disease. Veterinary Microbiol. (2019) 230:49–55. doi: 10.1016/j.vetmic.2019.01.008

PubMed Abstract | Crossref Full Text | Google Scholar

13. Marcano VC, Susta L, Diel DG, Cardenas-Garcia S, Miller PJ, Afonso CL, et al. Evaluation of chickens infected with a recombinant virulent NDV clone expressing chicken IL4. Microb Pathog. (2021) 159:105116. doi: 10.1016/j.micpath.2021.105116

PubMed Abstract | Crossref Full Text | Google Scholar

14. Xue Y, Zhang B, Huang HB, Li JY, Pan TX, Tang Y, et al. Immunoprotective effects of invasive Lactobacillus plantarum delivered nucleic acid vaccine coexpressing Trichinella spiralis CPF1 and murine interleukin-4. Vet Parasitol. (2021) 298:109556. doi: 10.1016/j.vetpar.2021.109556

PubMed Abstract | Crossref Full Text | Google Scholar

15. Huang P, Cai J, Yu D, Tang J, Lu Y, Wu Z, et al. An IL-6 gene in humphead snapper (Lutjanus sanguineus): Identification, expression analysis and its adjuvant effects on Vibrio harveyi OmpW DNA vaccine. Fish Shellfish Immunol. (2019) 95:546–55. doi: 10.1016/j.fsi.2019.11.013

PubMed Abstract | Crossref Full Text | Google Scholar

16. Luo J, Zhang B, Wu Y, Tian Q, Zhao J, Lyu Z, et al. Expression of interleukin-6 by a recombinant rabies virus enhances its immunogenicity as a potential vaccine. Vaccine. (2017) 35:938–44. doi: 10.1016/j.vaccine.2016.12.069

PubMed Abstract | Crossref Full Text | Google Scholar

17. P X, Y L, J L, B X, Q L, J J, et al. Immune effect of a Newcastle disease virus DNA vaccine with IL-12 as a molecular adjuvant delivered by electroporation. Arch Virol. (2020) 165:1959–68. doi: 10.1007/s00705-020-04669-5

PubMed Abstract | Crossref Full Text | Google Scholar

18. Foroutan M, Barati M, and Ghaffarifar F. Enhancing immune responses by a novel multi-epitope ROP8 DNA vaccine plus interleukin-12 plasmid as a genetic adjuvant against acute Toxoplasma gondii infection in BALB/c mice. Microb Pathog. (2020) 147:104435. doi: 10.1016/j.micpath.2020.104435

PubMed Abstract | Crossref Full Text | Google Scholar

19. Nagaraj V, John L, Bharatiraja S, Dechamma HJ, and Reddy GR. Adjuvantation of inactivated Foot and Mouth Disease Virus vaccine with IL-15 expressing plasmid improves the immune response in Guinea Pigs. Biologicals. (2017) 49:23–7. doi: 10.1016/j.biologicals.2017.07.005

PubMed Abstract | Crossref Full Text | Google Scholar

20. Yang Y, Teng Z, Lu Y, Luo X, Mu S, Ru J, et al. Enhanced immunogenicity of foot and mouth disease DNA vaccine delivered by PLGA nanoparticles combined with cytokine adjuvants. Res Veterinary Sci. (2021) 136:89–96. doi: 10.1016/j.rvsc.2021.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

21. Ben-Sasson SZ, Hogg A, Hu-Li J, Wingfield P, Chen X, Crank M, et al. IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells. J Exp Med. (2013) 210:491–502. doi: 10.1084/jem.20122006

PubMed Abstract | Crossref Full Text | Google Scholar

22. Furuichi K, Wada T, Iwata Y, Kokubo S, Hara A, Yamahana J, et al. Interleukin-1-dependent sequential chemokine expression and inflammatory cell infiltration in ischemia-reperfusion injury. Crit Care Med. (2006) 34:2447–55. doi: 10.1097/01.CCM.0000233878.36340.10

PubMed Abstract | Crossref Full Text | Google Scholar

23. Zhong S, Zhang T, Tang L, and Li Y. Cytokines and chemokines in HBV infection. Front Mol Biosci. (2021) 8:805625. doi: 10.3389/fmolb.2021.805625

PubMed Abstract | Crossref Full Text | Google Scholar

24. Lenardo M, Chan FK-M, Hornung F, McFarland H, Siegel R, Wang J, et al. MATURE T LYMPHOCYTE APOPTOSIS—Immune regulation in a dynamic and unpredictable antigenic environment. Annu Rev Immunol. (1999) 17:221–53. doi: 10.1146/annurev.immunol.17.1.221

PubMed Abstract | Crossref Full Text | Google Scholar

25. McQuaid SL, Loughran ST, Power PA, Maguire P, Szczygiel A, and Johnson PA. Low-dose IL-2 induces CD56bright NK regulation of T cells via NKp44 and NKp46. Clin Exp Immunol. (2020) 200:228–41. doi: 10.1111/cei.13422

PubMed Abstract | Crossref Full Text | Google Scholar

26. Blackman MA, Tigges MA, Minie ME, and Koshland ME. A model system for peptide hormone action in differentiation: Interleukin 2 induces a B lymphoma to transcribe the J chain gene. Cell. (1986) 47:609–17. doi: 10.1016/0092-8674(86)90625-2

PubMed Abstract | Crossref Full Text | Google Scholar

27. Fulcher DA and Wong S. Carboxyfluorescein succinimidyl ester-based proliferative assays for assessment of T cell function in the diagnostic laboratory. Immunol Cell Biol. (1999) 77:559–64. doi: 10.1046/j.1440-1711.1999.00870.x

PubMed Abstract | Crossref Full Text | Google Scholar

28. Li S, Zhao B, Wang F, Wang M, Xie S, Wang S, et al. Yak interferon-alpha loaded solid lipid nanoparticles for controlled release. Res Veterinary Sci. (2010) 88:148–53. doi: 10.1016/j.rvsc.2009.06.010

PubMed Abstract | Crossref Full Text | Google Scholar

29. Iwaszko M, Biały S, and Bogunia-Kubik K. Significance of interleukin (IL)-4 and IL-13 in inflammatory arthritis. Cells. (2021) 10:3000. doi: 10.3390/cells10113000

PubMed Abstract | Crossref Full Text | Google Scholar

30. Isakson PC, Pure E, Vitetta ES, and Krammer PH. T cell-derived B cell differentiation factor(s). Effect on the isotype switch of murine B cells. J Exp Med. (1982) 155:734–48. doi: 10.1084/jem.155.3.734

PubMed Abstract | Crossref Full Text | Google Scholar

31. Howard M, Farrar J, Hilfiker M, Johnson B, Takatsu K, Hamaoka T, et al. Identification of a T cell-derived b cell growth factor distinct from interleukin 2. J Exp Med. (1982) 155:914–23. doi: 10.1084/jem.155.3.914

PubMed Abstract | Crossref Full Text | Google Scholar

32. Maehara T, Koga R, and Nakamura S. Immune dysregulation in immunoglobulin G4-related disease. Jpn Dent Sci Rev. (2023) 59:1–7. doi: 10.1016/j.jdsr.2022.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

33. Tang Y, Ma C, Sun H, Yang S, Yu F, Li X, et al. Serum levels of seven general cytokines in acute brucellosis before and after treatment. Infect Drug Resist. (2021) 14:5501–10. doi: 10.2147/IDR.S341331

PubMed Abstract | Crossref Full Text | Google Scholar

34. Osii RS, Otto TD, Garside P, Ndungu FM, and Brewer JM. The impact of malaria parasites on dendritic cell-T cell interaction. Front Immunol. (2020) 11:1597. doi: 10.3389/fimmu.2020.01597

PubMed Abstract | Crossref Full Text | Google Scholar

35. Bonin-Jacob CM, Almeida-Lugo LZ, Puga MAM, MaChado AP, Padovani CTJ, Noceti MC, et al. IL-6 and IL-10 in the serum and exfoliated cervical cells of patients infected with high-risk human papillomavirus. PloS One. (2021) 16:e0248639. doi: 10.1371/journal.pone.0248639

PubMed Abstract | Crossref Full Text | Google Scholar

36. Kishimoto T. Factors affecting B-cell growth and differentiation. Annu Rev Immunol. (1985) 3:133–57. doi: 10.1146/annurev.iy.03.040185.001025

PubMed Abstract | Crossref Full Text | Google Scholar

37. Su B, Wang J, Wang X, Jin H, Zhao G, Ding Z, et al. The effects of IL-6 and TNF-alpha as molecular adjuvants on immune responses to FMDV and maturation of dendritic cells by DNA vaccination. Vaccine. (2008) 26:5111–22. doi: 10.1016/j.vaccine.2008.03.089

PubMed Abstract | Crossref Full Text | Google Scholar

38. Gerber AN, Abdi K, and Singh NJ. The subunits of IL-12, originating from two distinct cells, can functionally synergize to protect against pathogen dissemination. vivo. Cell Rep. (2021) 37:109816. doi: 10.1016/j.celrep.2021.109816

PubMed Abstract | Crossref Full Text | Google Scholar

39. S M HNC, Cs M, S B, and E A. The role of IL-12 in stimulating NK cells against Toxoplasma gondii infection: a mini-review. Parasitol Res. (2021) 120:2303–9. doi: 10.1007/s00436-021-07204-w

PubMed Abstract | Crossref Full Text | Google Scholar

40. H W GR, Y L, and X L. The role and potential application of IL-12 in the immune regulation of tuberculosis. Int J Mol Sci. (2025) 26:3106. doi: 10.3390/ijms26073106

PubMed Abstract | Crossref Full Text | Google Scholar

41. Hmama Z, Peña-Díaz S, Joseph S, and Av-Gay Y. Immunoevasion and immunosuppression of the macrophage by Mycobacterium tuberculosis. Immunol Rev. (2015) 264:220–32. doi: 10.1111/imr.12268

PubMed Abstract | Crossref Full Text | Google Scholar

42. Ong CY, Abdalkareem EA, and Khoo BY. Functional roles of cytokines in infectious disease associated colorectal carcinogenesis. Mol Biol Rep. (2022) 49:1529–35. doi: 10.1007/s11033-021-07006-4

PubMed Abstract | Crossref Full Text | Google Scholar

43. Bhadra R, Guan H, and Khan IA. Absence of both IL-7 and IL-15 severely impairs the development of CD8 T cell response against Toxoplasma gondii. PloS One. (2010) 5:e10842. doi: 10.1371/journal.pone.0010842

PubMed Abstract | Crossref Full Text | Google Scholar

44. Okamura H, Nagata K, Komatsu T, Tanimoto T, Nukata Y, Tanabe F, et al. A novel costimulatory factor for gamma interferon induction found in the livers of mice causes endotoxic shock. Infection Immun. (1995) 63:3966–72. doi: 10.1128/iai.63.10.3966-3972.1995

PubMed Abstract | Crossref Full Text | Google Scholar

45. Micallef MJ, Ohtsuki T, Kohno K, Tanabe F, Ushio S, Namba M, et al. Interferon-γ-inducing factor enhances T helper 1 cytokine production by stimulated human T cells: synergism with interleukin-12 for interferon-γ production. Eur J Immunol. (1996) 26:1647–51. doi: 10.1002/eji.1830260736

PubMed Abstract | Crossref Full Text | Google Scholar

46. Robinson D, Shibuya K, Mui A, Zonin F, Murphy E, Sana T, et al. IGIF does not drive Th1 development but synergizes with IL-12 for interferon-gamma production and activates IRAK and NFkappaB. Immunity. (1997) 7:571–81. doi: 10.1016/s1074-7613(00)80378-7

PubMed Abstract | Crossref Full Text | Google Scholar

47. Okamura H, Kashiwamura S, Tsutsui H, Yoshimoto T, and Nakanishi K. Regulation of interferon-gamma production by IL-12 and IL-18. Curr Opin Immunol. (1998) 10:259–64. doi: 10.1016/s0952-7915(98)80163-5

PubMed Abstract | Crossref Full Text | Google Scholar

48. Tsutsui H, Nakanishi K, Matsui K, Higashino K, Okamura H, Miyazawa Y, et al. IFN-gamma-inducing factor up-regulates Fas ligand-mediated cytotoxic activity of murine natural killer cell clones. J Immunol. (1996) 157:3967–73. doi: 10.4049/jimmunol.157.9.3967

PubMed Abstract | Crossref Full Text | Google Scholar

49. Dinarello CA, Novick D, Puren AJ, Fantuzzi G, Shapiro L, Muhl H, et al. Overview of interleukin-18: more than an interferon-gamma inducing factor. J Leukoc Biol. (1998) 63:658–64. doi: 10.1002/jlb.63.6.658

Crossref Full Text | Google Scholar

50. Ushio S, Namba M, Okura T, Hattori K, Nukada Y, Akita K, et al. Cloning of the cDNA for human IFN-gamma-inducing factor, expression in Escherichia coli, and studies on the biologic activities of the protein. J Immunol. (1996) 156:4274–9. doi: 10.4049/jimmunol.156.11.4274

PubMed Abstract | Crossref Full Text | Google Scholar

51. Hoshino T, Wiltrout RH, and Young HA. IL-18 is a potent coinducer of IL-13 in NK and T cells: a new potential role for IL-18 in modulating the immune response. J Immunol. (1999) 162:5070–7. doi: 10.4049/jimmunol.162.9.5070

PubMed Abstract | Crossref Full Text | Google Scholar

52. Negishi H, Taniguchi T, and Yanai H. The interferon (IFN) class of cytokines and the IFN regulatory factor (IRF) transcription factor family. Cold Spring Harbor Perspect Biol. (2018) 10:a028423. doi: 10.1101/cshperspect.a028423

PubMed Abstract | Crossref Full Text | Google Scholar

53. Liu L, Fan W, Zhang H, Zhang S, Cui L, Wang M, et al. Interferon as a mucosal adjuvant for an influenza vaccine in pigs. Virologica Sin. (2019) 34:324–33. doi: 10.1007/s12250-019-00102-7

PubMed Abstract | Crossref Full Text | Google Scholar

54. Sommereyns C, Paul S, Staeheli P, and Michiels T. IFN-lambda (IFN-λ) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PloS Pathog. (2008) 4:e1000017. doi: 10.1371/journal.ppat.1000017

PubMed Abstract | Crossref Full Text | Google Scholar

55. Zhou J, Wang Y, Chang Q, Ma P, Hu Y, and Cao X. Type III interferons in viral infection and antiviral immunity. Cell Physiol Biochem. (2018) 51:173–85. doi: 10.1159/000495172

PubMed Abstract | Crossref Full Text | Google Scholar

56. Zhang X, Wang S, Zhu Y, Zhang M, Zhao Y, Yan Z, et al. Double-edged effects of interferons on the regulation of cancer-immunity cycle. Oncoimmunology. (2021) 10:1929005. doi: 10.1080/2162402X.2021.1929005

PubMed Abstract | Crossref Full Text | Google Scholar

57. O’Brien L, Perkins S, Williams A, Eastaugh L, Phelps A, Wu J, et al. Alpha interferon as an adenovirus-vectored vaccine adjuvant and antiviral in Venezuelan equine encephalitis virus infection. J Gen Virol. (2009) 90:874–82. doi: 10.1099/vir.0.006833-0

PubMed Abstract | Crossref Full Text | Google Scholar

58. Gray PW and Goeddel DV. Structure of the human immune interferon gene. Nature. (1982) 298:859–63. doi: 10.1038/298859a0

PubMed Abstract | Crossref Full Text | Google Scholar

59. Farrar MA and Schreiber RD. The molecular cell biology of interferon-gamma and its receptor. Annu Rev Immunol. (1993) 11:571–611. doi: 10.1146/annurev.iy.11.040193.003035

PubMed Abstract | Crossref Full Text | Google Scholar

60. Pestka S, Krause CD, and Walter MR. Interferons, interferon-like cytokines, and their receptors. Immunol Rev. (2004) 202:8–32. doi: 10.1111/j.0105-2896.2004.00204.x

PubMed Abstract | Crossref Full Text | Google Scholar

61. Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, Shah NK, et al. IFN-λs mediate antiviral protection through a distinct class II cytokine receptor complex. Nat Immunol. (2003) 4:69–77. doi: 10.1038/ni875

PubMed Abstract | Crossref Full Text | Google Scholar

62. Biron CA. Interferons alpha and beta as immune regulators–a new look. Immunity. (2001) 14:661–4. doi: 10.1016/s1074-7613(01)00154-6

PubMed Abstract | Crossref Full Text | Google Scholar

63. Fenton SE, Saleiro D, and Platanias LC. Type I and II interferons in the anti-tumor immune response. Cancers (Basel). (2021) 13:1037. doi: 10.3390/cancers13051037

PubMed Abstract | Crossref Full Text | Google Scholar

64. Brown IH. The epidemiology and evolution of influenza viruses in pigs. Vet Microbiol. (2000) 74:29–46. doi: 10.1016/s0378-1135(00)00164-4

PubMed Abstract | Crossref Full Text | Google Scholar

65. R Y, B Z, and D C. Type I interferon-mediated tumor immunity and its role in immunotherapy. Cell Mol Lifesciences : CMLS. (2022) 79:191. doi: 10.1007/s00018-022-04219-z

PubMed Abstract | Crossref Full Text | Google Scholar

66. Kerner G, Rosain J, Guérin A, Al-Khabaz A, Oleaga-Quintas C, Rapaport F, et al. Inherited human IFN-γ deficiency underlies mycobacterial disease. J Clin Invest. (2020) 130:3158–71. doi: 10.1172/JCI135460

PubMed Abstract | Crossref Full Text | Google Scholar

67. Song R, Zhai X, Fan X, Ge T, Li M, Huercha, et al. Recombinant interferon-gamma promotes immunoglobulin G and cytokine memory responses to cathepsin L-like cysteine proteinase of Hyalomma asiaticum and the efficacy of anti-tick. Vet Immunol Immunopathol. (2021) 235:110201. doi: 10.1016/j.vetimm.2021.110201

PubMed Abstract | Crossref Full Text | Google Scholar

68. Charerntantanakul W, Yamkanchoo S, and Kasinrerk W. Plasmids expressing porcine interferon gamma up-regulate pro-inflammatory cytokine and co-stimulatory molecule expression which are suppressed by porcine reproductive and respiratory syndrome virus. Veterinary Immunol Immunopathology. (2013) 153:107–17. doi: 10.1016/j.vetimm.2013.02.013

PubMed Abstract | Crossref Full Text | Google Scholar

69. Wang H, Guo M, Tang X, Xing J, Sheng X, Chi H, et al. Immune adjuvant effects of interferon-gamma (IFN-gamma) of flounder (Paralichthys olivaceus) against Edwardsiella tarda. Dev Comp Immunol. (2021) 123:104159. doi: 10.1016/j.dci.2021.104159

PubMed Abstract | Crossref Full Text | Google Scholar

70. Wang B-L, Zhang S, Liu Y, Zhao Y-H, Wang C-W, Li Y, et al. Enhancing immunogenicity and antiviral protection of inactivated porcine reproductive and respiratory syndrome virus vaccine in piglets. Am J Vet Res. (2024) 85:ajvr.24.02.0025. doi: 10.2460/ajvr.24.02.0025

PubMed Abstract | Crossref Full Text | Google Scholar

71. Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, Whitmore TE, et al. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nat Immunol. (2003) 4:63–8. doi: 10.1038/ni873

PubMed Abstract | Crossref Full Text | Google Scholar

72. Durbin RK, Kotenko SV, and Durbin JE. Interferon induction and function at the mucosal surface. Immunol Rev. (2013) 255:25–39. doi: 10.1111/imr.12101

PubMed Abstract | Crossref Full Text | Google Scholar

73. Du L, Pang F, Yu Z, Xu X, Fan B, Huang K, et al. Assessment of the efficacy of two novel DNA vaccine formulations against highly pathogenic Porcine Reproductive and Respiratory Syndrome Virus. Sci Rep. (2017) 7:41886. doi: 10.1038/srep41886

PubMed Abstract | Crossref Full Text | Google Scholar

74. Österlund P, Veckman V, Sirén J, Klucher KM, Hiscott J, Matikainen S, et al. Gene expression and antiviral activity of alpha/beta interferons and interleukin-29 in virus-infected human myeloid dendritic cells. J Virol. (2005) 79:9608–17. doi: 10.1128/JVI.79.15.9608-9617.2005

PubMed Abstract | Crossref Full Text | Google Scholar

75. Zhang L and LiWang PJ. Chemokine-receptor interactions: solving the puzzle, piece by piece. Structure. (2014) 22:1550–2. doi: 10.1016/j.str.2014.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

76. Smit JJ and Lukacs NW. A closer look at chemokines and their role in asthmatic responses. Eur J Pharmacol. (2006) 533:277–88. doi: 10.1016/j.ejphar.2005.12.064

PubMed Abstract | Crossref Full Text | Google Scholar

77. Mohan T, Zhu W, Wang Y, and Wang B-Z. Applications of chemokines as adjuvants for vaccine immunotherapy. Immunobiology. (2018) 223:477–85. doi: 10.1016/j.imbio.2017.12.001

PubMed Abstract | Crossref Full Text | Google Scholar

78. Zlotnik A and Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity. (2000) 12:121–7. doi: 10.1016/s1074-7613(00)80165-x

PubMed Abstract | Crossref Full Text | Google Scholar

79. Miller M and Mayo K. Chemokines from a structural perspective. Int J Mol Sci. (2017) 18:2088. doi: 10.3390/ijms18102088

PubMed Abstract | Crossref Full Text | Google Scholar

80. Xu H, Xing J, Tang X, Sheng X, and Zhan W. The effects of CCL3, CCL4, CCL19 and CCL21 as molecular adjuvants on the immune response to VAA DNA vaccine in flounder (Paralichthys olivaceus). Dev Comp Immunol. (2020) 103:103492. doi: 10.1016/j.dci.2019.103492

PubMed Abstract | Crossref Full Text | Google Scholar

81. Huo X, Fan C, Ai T, and Su J. The Combination of Molecular Adjuvant CCL35.2 and DNA Vaccine Significantly Enhances the Immune Protection of Carassius auratus gibelio against CyHV-2 Infection. Vaccines. (2020) 8:567. doi: 10.3390/vaccines8040567

PubMed Abstract | Crossref Full Text | Google Scholar

82. He J, Thomas MA, de Anda J, Lee MW, Van Why E, Simpson P, et al. Chemokine CCL28 is a potent therapeutic agent for oropharyngeal candidiasis. Antimicrobial Agents chemotherapy. (2020) 64:e00210-20. doi: 10.1128/AAC.00210-20

PubMed Abstract | Crossref Full Text | Google Scholar

83. Lazarus NH, Kunkel EJ, Johnston B, Wilson E, Youngman KR, and Butcher EC. A common mucosal chemokine (mucosae-associated epithelial chemokine/CCL28) selectively attracts IgA plasmablasts. J Immunol (BaltimoreMd : 1950). (2003) 170:3799–805. doi: 10.4049/jimmunol.170.7.3799

PubMed Abstract | Crossref Full Text | Google Scholar

84. Mohan T, Kim J, Berman Z, Wang S, Compans RW, and Wang B-Z. Co-delivery of GPI-anchored CCL28 and influenza HA in chimeric virus-like particles induces cross-protective immunity against H3N2 viruses. J Controlled Release. (2016) 233:208–19. doi: 10.1016/j.jconrel.2016.05.021

PubMed Abstract | Crossref Full Text | Google Scholar

85. Mohan T, Berman Z, Luo Y, Wang C, Wang S, Compans RW, et al. Chimeric virus-like particles containing influenza HA antigen and GPI-CCL28 induce long-lasting mucosal immunity against H3N2 viruses. Sci Rep. (2017) 7:40226. doi: 10.1038/srep40226

PubMed Abstract | Crossref Full Text | Google Scholar

86. Kutzler MA, Kraynyak KA, Nagle SJ, Parkinson RM, Zharikova D, Chattergoon M, et al. Plasmids encoding the mucosal chemokines CCL27 and CCL28 are effective adjuvants in eliciting antigen-specific immunity. vivo. Gene Ther. (2010) 17:72–82. doi: 10.1038/gt.2009.112

PubMed Abstract | Crossref Full Text | Google Scholar

87. Basheer AS, Abas F, Othman I, and Naidu R. Role of inflammatory mediators, macrophages, and neutrophils in glioma maintenance and progression: mechanistic understanding and potential therapeutic applications. Cancers (Basel). (2021) 13:4226. doi: 10.3390/cancers13164226

PubMed Abstract | Crossref Full Text | Google Scholar

88. Hamilton JA. GM-CSF in inflammation. J Exp Med. (2020) 217:e20190945. doi: 10.1084/jem.20190945

PubMed Abstract | Crossref Full Text | Google Scholar

89. Lin Y-J, Anzaghe M, and Schülke S. Update on the pathomechanism, diagnosis, and treatment options for rheumatoid arthritis. Cells. (2020) 9:880. doi: 10.3390/cells9040880

PubMed Abstract | Crossref Full Text | Google Scholar

90. M P, J M, and S B. Granulocyte macrophage colony-stimulating factor has come of age: From a vaccine adjuvant to antiviral immunotherapy. Cytokine Growth factor Rev. (2021) 59:101–10. doi: 10.1016/j.cytogfr.2021.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

91. Reynaud D. GM-CSF brings (good) memories. Blood. (2024) 143:2683–4. doi: 10.1182/blood.2024024908

PubMed Abstract | Crossref Full Text | Google Scholar

92. Santodonato L, Agostino G D, Nisini R, Mariotti S, Monque DM, Spada M, et al. Monocyte-derived dendritic cells generated after a short-term culture with IFN-α and granulocyte-macrophage colony-stimulating factor stimulate a potent epstein-barr virus-specific CD8+ T cell response. J Immunol. (2003) 170:5195–202. doi: 10.4049/jimmunol.170.10.5195

PubMed Abstract | Crossref Full Text | Google Scholar

93. Barouch DH, Santra S, Tenner-Racz K, Racz P, Kuroda MJ, Schmitz JE, et al. Potent CD4+ T cell responses elicited by a bicistronic HIV-1 DNA vaccine expressing gp120 and GM-CSF. J Immunol. (2002) 168:562–8. doi: 10.4049/jimmunol.168.2.562

PubMed Abstract | Crossref Full Text | Google Scholar

94. Mills KAM, Westermann F, Espinosa V, Rosiek E, Desai JV, Aufiero MA, et al. GM-CSF-mediated epithelial-immune cell cross-talk orchestrates pulmonary immunity to Aspergillus fumigatus. Sci Immunol. (2025) 10:eadr0547. doi: 10.1126/sciimmunol.adr0547

PubMed Abstract | Crossref Full Text | Google Scholar

95. Mwangi W, Brown WC, Splitter GA, Davies CJ, Howard CJ, Hope JC, et al. DNA vaccine construct incorporating intercellular trafficking and intracellular targeting motifs effectively primes and induces memory B- and T-cell responses in outbred animals. Clin Vaccineimmunology : CVI. (2007) 14:304–11. doi: 10.1128/CVI.00363-06

PubMed Abstract | Crossref Full Text | Google Scholar

96. Zhou M, Wang L, Zhou S, Wang Z, Ruan J, Tang L, et al. Recombinant rabies virus expressing dog GM-CSF is an efficacious oral rabies vaccine for dogs. Oncotarget. (2015) 6:38504–16. doi: 10.18632/oncotarget.5904

PubMed Abstract | Crossref Full Text | Google Scholar

97. Guo X, Zheng Q, Jiang X, Wu C, Zhang T, Wang D, et al. The composite biological adjuvants enhance immune response of porcine circovirus type2 vaccine. Veterinary Microbiol. (2019) 228:69–76. doi: 10.1016/j.vetmic.2018.11.015

PubMed Abstract | Crossref Full Text | Google Scholar

98. Chen P, Zhang W, Cui Y, Sun M, Dong X, Li W, et al. Porcine GM-CSF and APS as a novel complex immunostimulant improves the immune effect of pseudorabies inactivated vaccine. Veterinary Microbiol. (2025) 304:110453. doi: 10.1016/j.vetmic.2025.110453

PubMed Abstract | Crossref Full Text | Google Scholar

99. Rohaim MA, Al-Natour MQ, Abdelsabour MA, El NR, Madbouly YM, Ahmed KA, et al. Transgenic chicks expressing interferon-inducible transmembrane protein 1 (IFITM1) restrict highly pathogenic H5N1 influenza viruses. Int J Mol Sci. (2021) 22:8456. doi: 10.3390/ijms22168456

PubMed Abstract | Crossref Full Text | Google Scholar

100. Smith SE, Gibson MS, Wash RS, Ferrara F, Wright E, Temperton N, et al. Chicken interferon-inducible transmembrane protein 3 restricts influenza viruses and lyssaviruses in vitro. J Virol. (2013) 87:12957–66. doi: 10.1128/JVI.01443-13

PubMed Abstract | Crossref Full Text | Google Scholar

101. Yan S, Chen Y, Lin J, Chen H, Hu C, Liu H, et al. Recombinant avian-derived antiviral proteins cIFITM1, cIFITM3, and cViperin as effective adjuvants in inactivated H9N2 subtype avian influenza vaccines. Vet Microbiol. (2024) 298:110277. doi: 10.1016/j.vetmic.2024.110277

PubMed Abstract | Crossref Full Text | Google Scholar

102. Plummer JR and McGettigan JP. Incorporating B cell activating factor (BAFF) into the membrane of rabies virus (RABV) particles improves the speed and magnitude of vaccine-induced antibody responses. PloS Negl Trop Dis. (2019) 13:e0007800. doi: 10.1371/journal.pntd.0007800

PubMed Abstract | Crossref Full Text | Google Scholar

103. Dwivedi V, Manickam C, Dhakal S, Binjawadagi B, Ouyang K, Hiremath J, et al. Adjuvant effects of invariant NKT cell ligand potentiates the innate and adaptive immunity to an inactivated H1N1 swine influenza virus vaccine in pigs. Vet Microbiol. (2016) 186:157–63. doi: 10.1016/j.vetmic.2016.02.028

PubMed Abstract | Crossref Full Text | Google Scholar

104. Ding Y, Wilkinson A, Idris A, Fancke B, O’Keeffe M, Khalil D, et al. FLT3-ligand treatment of humanized mice results in the generation of large numbers of CD141+ and CD1c+ dendritic cells. vivo. J Immunol. (2014) 192:1982–9. doi: 10.4049/jimmunol.1302391

PubMed Abstract | Crossref Full Text | Google Scholar

105. Kornuta CA, Langellotti CA, Bidart JE, Soria I, Quattrocchi V, Gammella M, et al. A plasmid encoding the extracellular domain of CD40 ligand and Montanide GEL01 as adjuvants enhance the immunogenicity and the protection induced by a DNA vaccine against BoHV-1. Vaccine. (2021) 39:1007–17. doi: 10.1016/j.vaccine.2020.11.071

PubMed Abstract | Crossref Full Text | Google Scholar

106. Njongmeta LM, Bray J, Davies CJ, Davis WC, Howard CJ, Hope JC, et al. CD205 antigen targeting combined with dendritic cell recruitment factors and antigen-linked CD40L activation primes and expands significant antigen-specific antibody and CD4(+) T cell responses following DNA vaccination of outbred animals. Vaccine. (2012) 30:1624–35. doi: 10.1016/j.vaccine.2011.12.110

PubMed Abstract | Crossref Full Text | Google Scholar

107. Nazarizadeh A, Staudacher AH, Wittwer NL, Turnbull T, Brown MP, and Kempson I. Aluminium nanoparticles as efficient adjuvants compared to their microparticle counterparts: current progress and perspectives. Int J Mol Sci. (2022) 23:4707. doi: 10.3390/ijms23094707

PubMed Abstract | Crossref Full Text | Google Scholar

108. Wang K, Wang H, and Wang X. Chiral nanomaterials as vaccine adjuvants: a new horizon in immunotherapy. Nanoscale. (2025) 17:1932–5. doi: 10.1039/d4nr03542a

PubMed Abstract | Crossref Full Text | Google Scholar

109. Chrun T, Lacôte S, Urien C, Jouneau L, Barc C, Bouguyon E, et al. A Rift Valley fever virus Gn ectodomain-based DNA vaccine induces a partial protection not improved by APC targeting. NPJ Vaccines. (2018) 3:14. doi: 10.1038/s41541-018-0052-x

PubMed Abstract | Crossref Full Text | Google Scholar

110. Chrun T, Lacôte S, Urien C, Richard CA, Tenbusch M, Aubrey N, et al. A DNA vaccine encoding the gn ectodomain of rift valley fever virus protects mice via a humoral response decreased by DEC205 targeting. Front Immunol. (2019) 10:860. doi: 10.3389/fimmu.2019.00860

PubMed Abstract | Crossref Full Text | Google Scholar

111. Knight-Jones TJD, Edmond K, Gubbins S, and Paton DJ. Veterinary and human vaccine evaluation methods. Proc Biol Sci. (2014) 281:20132839. doi: 10.1098/rspb.2013.2839

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: cytokines, veterinary vaccine, vaccine adjuvant, immune enhancement, vaccination

Citation: Yan S, Chen H, Liu Q, Zhang X, Wei Q, Hu C, Chu D, Diao H, Liu S and Chen J-L (2025) Harnessing cytokines: innovative adjuvants for improved veterinary vaccine efficacy. Front. Immunol. 16:1643855. doi: 10.3389/fimmu.2025.1643855

Received: 09 June 2025; Accepted: 02 October 2025;
Published: 21 October 2025.

Edited by:

Abel A Ramos Vega, National Polytechnic Institute (IPN), Mexico

Reviewed by:

Nikunj Umedbhai Tandel, VNSGU, India
Pengtao Jiao, Chinese Academy of Sciences (CAS), China

Copyright © 2025 Yan, Chen, Liu, Zhang, Wei, Hu, Chu, Diao, Liu and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Shihong Yan, c2hpaG9uZ3lhbjAxMDZAMTYzLmNvbQ==

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.