Skip to main content

Natural and induced immune responses in oral cavity and saliva

Abstract

This review comprehensively explores the intricate immune responses within the oral cavity, emphasizing the pivotal role of saliva in maintaining both oral and systemic health. Saliva, a complex biofluid, functions as a dynamic barrier against pathogens, housing diverse cellular components including epithelial cells, neutrophils, monocytes, dendritic cells, and lymphocytes, which collectively contribute to robust innate and adaptive immune responses. It acts as a physical and immunological barrier, providing the first line of defense against pathogens. The multifaceted protective mechanisms of salivary proteins, cytokines, and immunoglobulins, particularly secretory IgA (SIgA), are elucidated. We explore the natural and induced immune responses in saliva, focusing on its cellular and molecular composition. In addition to saliva, we highlight the significance of a serum-like fluid, the gingival crevicular fluid (GCF), in periodontal health and disease, and its potential as a diagnostic tool. Additionally, the review delves into the impact of diseases such as periodontitis, oral cancer, type 2 diabetes, and lupus on salivary immune responses, highlighting the potential of saliva as a non-invasive diagnostic tool for both oral and systemic conditions. We describe how oral tissue and the biofluid responds to diseases, including considerations to periodontal tissue health and in disease periodontitis. By examining the interplay between oral and systemic health through the oral-systemic axis, this review underscores the significance of salivary immune mechanisms in overall well-being and disease pathogenesis, emphasizing the importance of salivary mechanisms across the body.

Peer Review reports

Background

The oral mucosa, the lining of the oral cavity, stands as a critical interface between the body’s internal environment and the external world. It serves as a dynamic barrier, constantly challenged by a diverse array of microbes, food particles, and environmental insults. Saliva, a complex biofluid secreted by the salivary glands, plays a pivotal role in maintaining the integrity of this barrier and orchestrating the intricate balance between host defense and microbial tolerance.

Saliva’s composition is a testament to its multifaceted functions. Beyond its primary constituents of water and electrolytes, saliva contains a rich repertoire of proteins, enzymes, mucins, and immunoglobulins that contribute to its lubricating, digestive, and antimicrobial properties. The constant flow of saliva not only facilitates the mechanical clearance of debris and microbes but also regulates the oral pH, buffering against the acidic byproducts of bacterial metabolism. This buffering capacity is essential for maintaining a healthy oral microbiome and preventing the demineralization of tooth enamel, a precursor to dental caries.

The oral cavity harbors a diverse and dynamic microbial community, collectively known as the oral microbiome. This intricate ecosystem plays a crucial role in both oral and systemic health. A balanced microbiome contributes to digestion, nutrient metabolism, and colonization resistance against opportunistic pathogens. However, disruptions in the microbial balance, often triggered by dietary habits, oral hygiene practices, or systemic diseases, can lead to dysbiosis. This imbalance can manifest as oral infections, periodontal disease, and even systemic complications.

Saliva serves as a critical mediator of host-microbial interactions in the oral cavity. Its array of antimicrobial peptides, such as lysozyme, lactoferrin, and histatins, provides a first line of defense against invading pathogens. Additionally, salivary immunoglobulins, particularly secretory IgA (SIgA), play a crucial role in neutralizing toxins, agglutinating bacteria, and preventing their adherence to mucosal surfaces. The interplay between these salivary components and the oral microbiome is essential for maintaining a healthy equilibrium. Clearly, the mechanisms by which saliva responds to disease are not well known in periodontitis, oral cancer, mucositis as illustrative examples. We will analyze the salivary changes associated with these conditions and discuss the potential of saliva as a diagnostic tool. Additionally, we explore the interplay between oral and systemic health by examining the oral-systemic axes.

We review intricate immune mechanisms within saliva, exploring its diverse cellular and molecular constituents and their roles in maintaining oral homeostasis and combating disease. We will examine the various cell types found in saliva, including epithelial cells, neutrophils, monocytes, dendritic cells, and lymphocytes, and discuss their individual contributions to both innate and adaptive immunity. We will further elucidate the protective functions of salivary proteins, cytokines, and immunoglobulins, emphasizing their interactions with the oral microbiome and their responses to various disease states.

By providing a comprehensive overview of the natural and induced immune responses in the oral cavity and saliva, this review aims to shed light on the critical role of saliva in maintaining oral and systemic health. Understanding the molecular mechanisms for developing innovative strategies to prevent and treat oral diseases, harness the diagnostic potential of saliva, and ultimately improve human health.

Saliva composition and function is regulated by immune cells

Saliva and the epithelial layer

Saliva acts as the lubrication of the mouth and interacts with every surface of the oral cavity. The mucous membrane is what lines the surfaces of the oral cavity and must fulfill a variety of different functions, from acting as an immune and physical barrier to maintain homeostasis. To properly function, the mucosal tissue has a high epithelial cell turnover which causes layers of epithelial cells to constantly shed into saliva. This constant shedding is what makes epithelial cells the primary type of cell found in saliva. Stratified squamous epithelial cells make up about 55–60% of all cell content found in saliva [1,2,3]. However, the number of epithelial cells in the host’s saliva can vary based on the host’s demographics [3]. The majority of epithelial cells found in saliva are non-keratinous, consisting of about 80% of the total epithelial cell content. This fraction accounts for the many non-keratinous surfaces within the oral cavity such as the inner cheeks and the floor of the mouth. The remaining content of the epithelial population is shed by the keratinous surfaces of the hard palate and parts of the gingiva. Lastly, about 1% of epithelial cell content are characterized as intermediate cells found below the superficial layer of the epithelium [2].

The main function of these cells is to prevent the initial stages of invasion of the oral tissues and thereby offer protection at a systemic level from invading pathogens. As the epithelium is shed, an average of about 100 individual bacterial organisms are removed from the surface of the oral cavity per epithelial cell [4]. Once in saliva, the pathogens attached to epithelial cells can be eliminated by gastric acid, after swallowing, or by the broad innate immune system within saliva [5]. In this way, saliva acts as the medium or vehicle in which pathogens attached to epithelial cells can be expelled or eliminated from the oral cavity.

Predominant innate immune cells protect oral mucosa

Polymorphonuclear neutrophils

Polymorphonuclear neutrophils (PMNs) are the second most common cell type, and the most abundant leukocyte in saliva acting as the primary cell of the innate immune system within the oral cavity. Neutrophils account for about 30–40% of the total cell count in saliva and make up about 95–98% of the total leukocyte population [1, 6]. Since saliva acts as a medium during the first step of eliminating pathogens and preventing infections, neutrophils act as the first point of contact and swiftly produce an immune response. In the presence of pathogens, neutrophils can undergo various morphological changes to induce an innate immune response such as phagocytosis, degranulation, reactive oxygen species (ROS) production, and neutrophil extracellular traps (NETs). An ex vivo study found that in healthy participants the oral neutrophil population holds a larger percentage of late stage apoptotic/necrotic neutrophils than those found in venous blood at ~ 50% in comparison to ~ 10% of total neutrophil population, respectively. In the same study the isolated oral neutrophils were found to be far more active in comparison to neutrophils in circulating blood when stimulated by F. nucleatum, producing higher expression values for CD11b, CD63, CD66b, and ROS levels [7]. This suggests oral neutrophils are far more prepared to produce a rapid and robust inflammatory response in the presence of a pathogen which may lead to a stronger antimicrobial effect accompanied with tissue damage. However, their inflammatory responses can become dysregulated leading to pathogenic effects as seen with rheumatoid arthritis and systemic lupus erythematosus [8].

In healthy participants, flow cytometry and computational methods have determined sub-populations of neutrophils showing varying sizes and granularity, along with differing expression levels of CD markers, ROS production, and NET formation [9]. Two major and distinct populations of neutrophils were found in healthy saliva where they primarily differ in level of immune activity. The less active population has reduced expression levels for CD55 and CD63, but an increase in CD170 and CD16 when compared to the more active neutrophil population. The more active sub-population is responsible for most of the phagocytic, ROS production, and NET formation produced by saliva in the healthy oral cavity [10]. However, other studies have been able to determine 8 sub-populations using scRNAseq, showing high variation in the neutrophil population to account for many of the possible pathogens and immune system needs that may exist in saliva. In diseases, such as chronic periodontitis, there is a larger sub-population of inflammatory neutrophils, showing elevated levels of CD63, CD66, CD10, CD64, CD55, CD11b, and CD18, than in healthy counterparts. In addition, NETosis markers citrullinated histone H3 (CitH3) and myeloperoxidase (MPO) are found to be elevated in this population of neutrophils. Baseline measurements of ROS production in disease-state neutrophils are significantly higher than both sub-populations of neutrophils found in healthy. This overproduction of ROS and higher citH3 and MPO are signs of an over-inflammatory and dysregulated neutrophil activity that leads to excessive tissue damage [11, 12]. However, in an in vitro study inducing ROS production with phorbol myristate acetate (PMA), the more active population of neutrophils in healthy produce higher levels of ROS than in disease [10]. This could be an indication that neutrophil ROS production in the disease state could become exhausted and further disease progression or increase the susceptibility of the oral cavity to new pathogens.

Monocytes

Within saliva, CD14+ monocytes account for about 1.3% of the total leukocyte population [1, 6]. Monocytes and their differentiated populations, such as macrophages and dendritic cells, originate from areas in the oral cavity with high populations of these cell types such as the salivary glands. The population of macrophages can be separated by their activated state termed as M1 and M2 where the shape, function, and some surface markers are unique to each state [13]. However, it is worth noting that these two profiles of macrophages are an oversimplification of the actual activated macrophage population. Within the subset of M1 or M2 macrophages there can exist vast differences and overlapping properties of their immunophenotype [14]. Nonetheless, these two categorizations of macrophages are used to determine whether the tissue environment leans toward a pro- or anti-inflammatory macrophage profile. The activation of macrophages into either the pro-inflammatory M1 state or anti-inflammatory M2 state depends on the location and environment of where the macrophage resides. M1 macrophages are considered to be “classically activated” by offering increased phagocytic activity, greater antigen-presenting capacity, and increased release of proinflammatory cytokines such as TNF-α and IL- 6 [15]. An in vitro study found that sterile human saliva influenced the activation of murine macrophages into an M1 state by detecting IL- 12 and IL- 6 expression [16]. This supports the idea that saliva promotes an environment that is pro-inflammatory to create an innate immune response that can handle constant exposure to pathogens. In cases of chronic periodontitis, the ratio of M1/M2 is much higher than what is found in healthy participants [17, 18]. This indicates that M1 macrophages may play a role in either the pathogenesis or the development of periodontitis.

On the other hand, M2 macrophages are involved in producing an anti-inflammatory response that also positively influences tissue repair. For example, one murine study found that bone formation is dependent on the production of Cystatin C by M2 macrophages [19]. Another murine study found that injecting induced M2 macrophages into sites of inflammation caused by periodontitis actually suppressed both the excessive immune response and inhibited the differentiation of osteoclasts [20]. However, it is worth noting that the majority of the studies involving the use of M2 macrophages to dampen the immune response in periodontitis are done in mice. Nonetheless, the importance of the ratio between M1/M2 macrophages is apparent, and further research in this area is required.

Oral dendritic cells (DCs)

DCs play an important role in bridging the innate response to the adaptive immune response. Their role falls into recognizing circulating antigen to produce an immune response that activates the downstream adaptive immune system. Dendritic cells can be organized into two classes based on their function and location before and after maturation. Conventional DCs are characterized by their function of patrolling tissue environments for foreign antigens. When reaching an antigen, these DCs will migrate to draining lymph nodes to undergo a maturation process where pathogen-derived peptides are presented to CD4+ and CD8+ T cells. On the other hand, the second class of DCs are considered unconventional as they circulate the bloodstream and enter inflamed tissues after infection [21]. Once found in tissue, unconventional DCs will activate T cells by the production of interferon (IFN).

Although not found in high numbers in saliva, Langerhans Cells (LCs) play an important role in responding to circulating pathogens found in saliva. LCs are part of conventional DCs that are characterized by residing on the epithelial layer of skin or mucosal lining to respond to circulating pathogens, migrate to nearby lymph nodes, and then induce an adaptive immune response. Although LCs are found in most tissues with an epithelial layer such as the skin and intestinal mucosa, oral LCs have a unique profile that differentiates these from other LCs. For example, oral LCs have higher expression levels for IgG (FcγRIII/CD16 and FcγRI/CD64) receptors which may imply these cells respond to pathogens coated with IgG antibodies much faster [22]. This finding also implies oral LCs have enhanced phagocytic capabilities to capture and process antigens and become antigen-presenting cells (APCs) thereby increasing the chances to induce an adaptive immune response. Increased IgG receptors could be explained by the need to recognize pathogens in an environment like the oral cavity, where cells are constantly exposed to these foreign pathogens. In addition to these receptors, oral LCs also have increased expression levels of major histocompatibility complex (MHC) I and II along with costimulatory CD40, CD80/B7.1, and CD86/B7.2 when compared to other LCs [22]. The high expression of both MHCs indicate that oral LCs are far better equipped to form into APCs and activate both CD4+ and CD8+ T cells. However, the higher expression of the three costimulatory molecules could indicate that oral LCs will tend to favor a CD4+ T cell environment. In an environment like the oral cavity that depends on B cell activity for a strong humoral response against pathogens floating in saliva, it would make sense for oral LCs to favor CD4+ T cells. Ultimately, dendritic cells, such as Langerhans cells of the oral mucosal lining, indirectly help saliva produce a strong innate immune response by favoring an adaptive immune cell environment that produces high levels of immunoglobulins that are free floating in saliva.

The adaptive immune responses in saliva

Lymphocytes

T and B cells make up the adaptive immune system and respond with high specificity and sustained protection [23]. However, to achieve such a high level of specificity the adaptive immune system must first undergo a primary adaptive response from a first infection. During a first infection, the innate immune system interacts with a pathogen that eventually leads to the formation of APCs that will migrate to the nearest lymph node [24]. Within the lymph nodes, naive T cells will activate through the recognition of antigenic material presented by APCs and mature into CD4+ or CD8+ T cells that in-turn begin the adaptive immune response and return to the site of infection. Since the adaptive immune response largely depends on antigen uptake and processing, lymphocytes have a slower reaction time to a pathogen than the immune cells of the innate immune system during a primary infection. This may explain the large differences in cell populations observed between innate immune cells and lymphocytes within mucosal barriers serving as the first line of defense, like the oral cavity. The population of lymphocytes can be generally categorized into CD45+CD3+ T cells and CD45+CD20+ B cells and flow cytometry studies have found cells at about 0.8% and 0.5% of the total leukocyte population, respectively [6]. The population of lymphocytes can be further subset based on their function. The activation of CD8+ T cells, also known as Cytotoxic T cells (CTLs), begins the cell-mediated immune response to target infected cells and induce apoptosis. However, their role in oral disease progression is shown to be minimal when compared to other lymphocytes[cite study here]. Although these cells may not play a large role in the immune response of the oral cavity, some studies have shown CD8+ T cells contribute towards the suppression of both inflammatory responses and bone loss by osteoclastogenesis [25, 26]. On the other hand, CD4+ T cells, also known as T helper cells (Th-cells), drive the downstream process to activate the humoral response and begin the production of high-affinity and pathogen-specific antibodies. However, the subsets of Th cells can have drastic differences in their role and function and are primarily differentiated based on the surrounding environment. Some of the important Th cells in saliva, or the oral cavity as a whole, include Th1, Th2, and Treg cells. Th1 cells are involved in the bone loss characterized in chronic periodontitis by stimulating osteoclasts to reabsorb bone matter [27]. Th2 and Treg cells have a stronger role in the regulation of the immune response and its inflammatory effects in the affected tissue including the inhibition of bone loss in periodontitis [28].

B cell activation at the presence of an antigen begins the humoral response responsible for the high specificity and affinity of the adaptive immune system towards a recognized and targeted pathogen. Naive B cells can activate to become memory B cells and plasma cells which will lead to the production of antibodies. B cell activation, in the presence of APCs and mediated by Th cells, induces the proliferation and differentiation into plasma cells. In turn, these short-lived plasma cells respond by producing antibodies, such as IgA, that are secreted outside of the tissue and into saliva [29]. On the other hand, memory B cells do not immediately produce some response, but instead act as the reservoir of humoral responses to the same pathogen by proliferating into plasma cells in subsequent infections. In cases of chronic or aggressive periodontitis, the saliva levels of the B cell activating factor (BAFF) become elevated when compared to the healthy control [30]. This could be an indication that B cells play some type of role in the progression of periodontitis. Additionally, B cells also play a prominent role in autoimmune diseases such as Sjogren’s syndrome. Part of the characterization of primary Sjogren’s syndrome is the increase of CD27+ memory B cells into the salivary glands where downstream effects eventually lead to the production of autoantibodies thereby damaging the surrounding tissue [31]. In healthy tissue, B cells exhibit tolerance towards auto-antigens, but if this tolerance is exacerbated autoimmune diseases can arise. Although the level of lymphocytes is small in saliva and the oral cavity, these cells play an important role in providing an immune response with high specificity and affinity to invading pathogens and any subsequent infections. Additionally, changes in the population ratios of these cells could serve as strong indicators for disease in the oral cavity.

The oral immune system tolerance

The healthy oral cavity is constantly introduced to foreign antigens, and yet the immune system can tailor its response to address insults from pathogenic microorganisms while remaining tolerant to non-pathogenic bacteria and food. This is, in part, explained by the complex interplay and communication between oral epithelial cells (OECs), tolerogenic DCs, and Treg CD4+ cells found in the oral cavity. Based on the integrity of the cell barrier, epithelial cells will send molecular signals to circulating DCs to either maintain them in a tolerogenic state or induce a proinflammatory response [32]. An in vitro study found that OECs will signal DCs to maintain a tolerogenic state and decrease the frequency of CD80 and CD86 used to activate the downstream proliferation of T cells. Additionally, in the presence of OECs, DCs were found to have decreased production of inflammatory cytokines such as TNF-α and IL- 12. This same study suggests that OECs can dampen CD4+ T cell activity when in close proximity, as seen by the suppression of IFN-γ and TNF-α when CD4+ T cells are incubated with OECs [33]. In a subsequent study by the same researchers, they found that OEC’s also directly inhibit inflammatory T cell activity even after activation through DC’s or anti-CD3/CD28. The research group determined that T cell expression of inflammatory markers, T-bet and IFNγ, was largely controlled by blocking the synthesis of prostaglandin E2 (PGE2) or its binding ability to EP2/EP4 receptors [34]. In addition, other immune cell interactions using CD40/CD40L, CD58/CD2, and PD-L1/PD- 1 were not involved in mediating T cell activity. These findings suggest that PGE2 is the primary chemical mediator between OEC’s and T cells for the suppression of inflammatory activity, but only in a healthy oral environment. In a later section of this review, we further discuss the role of molecules like PGE2 in periodontitis and how the detection of a viral insult blocks T cell suppression by OEC. These findings highlight the complex communication between OECs, immune cells, and the local microbial environment.

DCs also play a role in properly responding to the external stimuli through the detection of pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) using pattern-recognition receptors (PPRs). In the presence of PAMPs, dendritic cells, as such other innate immune cells, can recognize molecular signatures of pathogens and bacteria. This mechanism allows dendritic cells to recognize local tissue damage and adequately respond to external stimuli. As previously stated, as part of their response, DCs are involved in the activation of T cells of a specific phenotype. In the presence of non-harmful antigens, DCs can activate the pathway to produce mature T reg cells which in turn limit the proliferation of inflammatory T cells. Studies have found that the ligation of Toll-like receptors (TLRs) in dendritic cells, such as TLR4 in oral Langerhan cells, after coming in contact with commensal bacteria will induce an anti-inflammatory environment. In this scenario, LCs release IL- 10 while also inducing the production of IL- 10, TGF-β1, FOXP3, IFN-γ, and IL- 2 in T cells [35]. Additionally, another paper found that DCs of the oral cavity, after stimulation with an antigen such as ovalbumin, will induce CD4+ T cells with anti-inflammatory markers such as IL- 10 and IFN-γ [36]. In the same study, researchers were able to show an increase in ovalbumin specific Th1 cells after multiple rounds of sublingual immunotherapy [36]. Ultimately, DCs have a unique role of properly responding to circulating stimuli, whether pathogenic or not, to determine what type of signal should be sent to downstream effectors of the immune system.

Research on the topic of immune cell tolerance has to be studied more in the oral mucosa since this plays a pivotal role in ensuring an adequate defense against new foreign organisms constantly introduced in the mouth. However, mechanisms of immune tolerance can also be observed and studied in other mucosal tissue where host cells constantly interact with microbes. For example, epithelial cells of the gut will signal to circulating immune cells about a pathogenic insult to elicit an immune response [37]. Another study found that in the presence of Salmonella, intestinal epithelial cells had higher levels of gene expression involved in anti-bacterial pathways [38]. Research on the tolerance of the immune system and its mediators should be an area of research where more focus should be placed. This could help answer questions as to why patients have different levels of an immune response when introduced to the same pathogenic insult.

Salivary immune molecules

Oral immunoglobulins

Antibodies, also known as immunoglobulins (Ig), are a method of protection utilized by our immune systems to eliminate foreign pathogens. Immunoglobulins can vary in type, structure, and function. While all these antibodies are produced by B cells differentiated into plasma cells, some can take different forms in different parts of the body to carry out various protective and immune activities. The most studied and abundant immunoglobulin isotype in our bodies is immunoglobulin G (IgG), which is produced, primarily, in the secondary immune response to pathogens and can activate the complement system [39]. With 4 subclasses, IgG provides a wide range of protection from within the systemic immune pathways, as IgG makes up around 80% of the immunoglobulins found circulating in the bloodstream. Immunoglobulin M (IgM) has a pentameric structure, making it the largest of the immunoglobulins, and is mostly produced in the primary immune response to infectious agents and antigens [39], allowing for an initial response while other antibodies are produced over time. In mucosal surfaces, immunoglobulin A (IgA) is the predominant isotype at 90% (Fig. 1). It is locally produced in a dimeric composition from plasma cells and, as it is secreted through the epithelium into the mucosa, gains a protective secretory component (SC) to create its known form, secretory IgA (SIgA). The added SC contributes to stability of dimeric IgA, as monomeric forms are connected with a heavy J chain, as well as contributing to its increased ability to competitively inhibit binding of pathogens [40]. SIgA comes into two forms, SIgA1 and SIgA2, which differ slightly in structure where SIgA2 has a shorter hinge, resulting in a difference in planarity [41]. The most relevant consequence of this structural difference is predicted to be differences in antigen interaction at the Fab regions, with SIgA1 binding to various antigens in a variety of orientations and SIgA2’s more rigid, planar structure allows for its binding on antigens that are on relatively fixed surfaces [42]. In saliva, SIgA contributes to more than 80% of the salivary IgA pool and concentrations from unstimulated whole saliva stand at ~ 0.19 mg/ml, compared to serum where IgA concentration is ~ 2.2 mg/ml [43]. SIgA2 is, overall, found to be at the same concentrations, if not greater, than SIgA1 although, the levels of SIgA1 versus SIgA2 vary across secretion and mucosal sites, indicating selective advantages to one subclass for specific immune mechanisms [42]. Its prevalence at these mucosal sites has much to do with SIgA’s immune response potential and the natural defense mechanisms the antibody has, due to its structure.

Fig. 1
figure 1

Salivary and Mucosal Barriers and Responses. Free-floating cells and proteins found within saliva will prevent microbes from disrupting the mucosal epithelial layer. At the moment of infection or cell damage within the epithelial layer, the targeted cell will send signals out to dendritic cells that will then migrate to the lymph nodes and mature in the form of APCs. These APC’s will then activate subsets of T cells (CD8+ or CD4+) responsible for the activation of the humoral or cellular response which can either alleviate infections or exacerbate inflammation

Secretory IgA in saliva

The secretory IgA is largely utilized in physically protective ways such as “coating, cross-linking, agglutination, and enchained growth of mucosal antigens” [44]. The cross-linking capabilities are important in fluids like saliva because it allows the SIgA to act as a physical barrier before pathogens can even reach the tissues of the body. Specifically secreted from epithelial cells within the salivary glands, thymic stromal lymphopoietin (TSLP), aids in host defense due to its antimicrobial properties. Existing in a long form (lfTSL) and a short form (sfTSLP), researchers have found that both forms have antimicrobial functions, likely due to its C-terminal that can disrupt cell membranes. In healthy control participants, sfTSLP is dominant and produced constitutively, with detection of lfTSLP significantly increasing over sfTSLP in inflammatory conditions [45, 46]. A variety of cytokines with a variety of functions, from immune cell signaling to specific microbe clearing abilities, are present in saliva and all contribute to the oral cavity’s overall immune abilities. A method called agglutination is when IgA will bind to pathogenic particles, or antigens, using both of its binding sites, effectively clumping particles together, which prevents adherence to tissue surfaces, such as oral epithelium or teeth, and creates a large target for immune cells to come and destroy. This all happens without any signaling involved and is a product of the structure and binding abilities of the isotype. SIgA can also bind to any pathogenic toxins or enzymes that are released from a pathogen, effectively neutralizing those secreted toxins. These physical means of eliminating pathogenic threats by cross-linking or binding is known as immune exclusion as the IgA is not actively setting out to eliminate/kill pathogens, but rather preventing them from operating within our bodies. However, SIgA is not alone in immune defense and can also help present antigens to immune cells to communicate which particles are pathogenic and should be targeted by fellow immune cells. SIgA has specific effector mediators that exist on many different immune cells such as macrophages, neutrophils, eosinophils, and monocytes that, when the mediators are activated, can trigger elimination processes in the forms of phagocytosis, cytokine release, or antibody dependent cell-mediated cytotoxicity [41]. The FcαRI receptor found on various immune effector cells is where SIgA will bind to signal inflammatory cytokines or inflammatory degranulation. Pathogen recognition by SIgA will alter the structure of the immunoglobulin to increase binding to the FcαRI host cell receptor [47]. Considering the duality of the defense mechanism methodology, SIgA would fall into the innate and acquired immune system categories, producing a robust physical barrier defense as well as recruiting and directing immune cells to carry out their own defensive mechanisms.

The role of SIgA healthy vs. disease

With SIgA as the first barrier within the oral cavity, it stands as a primary physical line of defense against pathogens entering the bloodstream where serum IgA is found to have a monomeric composition and is associated more with signaling inflammatory reactions [48]. There are observed differences in the glycosylation of these different IgAs [43], illustrating that they circulate within two distinct immune pathways and can use them to detect different kinds of diseases, mainly those where pathogens infiltrate via mucosal pathways. Salivary IgA cannot be used as a proxy for serum IgA, however, it can be used distinctly to identify infection and disease where serum IgA is less impactful in immune response. As a response to pathogen invasion in the oral cavity, or disease originated from the oral or other bodily areas, SIgA production increases to combat these foreign invaders, compared to a healthy person, from which there is no need for an elevated immune response. An evaluation of IgA from saliva compared to serum, from patients who were infected with SARS-CoV- 2, illustrated a higher salivary IgA response in acute infection phase compared to healthy levels [49]. Another study had examined IgA levels from saliva in patients infected with SARS-CoV- 2 compared to non-infected patients, finding a three-fold higher response in infected patients (13.8 AU/mL) against healthy (4.2 AU/mL) [50]. These studies were able to utilize enzyme-linked immunosorbent assays (ELISAs) for a highly customizable and specific method of measuring IgA. In these studies, SARS-CoV- 2 specific antigens were used, to which IgA showed high binding affinity, proving the efficacy of IgA against pathogenic particles. Bio-fluids other than saliva can be and were evaluated. While blood holds a higher concentration of antibodies, saliva is partial to IgA dominance and can elicit a more specific defense. In paired serum and saliva samples, patients in the convalescent phase of COVID- 19 had a more robust response to SARS-CoV- 2 S1, S2, and Nucleoprotein in their saliva compared to their paired serum (p < 0.0001, p = 0.0036, and p = 0.0009, respectively) [49]. This antibody specificity and difference is indicative of the functional pathways in which salivary IgA operates.

The cytokine responses in oral fluids

Cytokines are signaling proteins that play an important role when mounting any kind of immune response. Secreted mainly by immune cells, cytokines of all different types and functions direct cellular immune response and regulate inflammation. Immune cells such as macrophages, neutrophils, lymphocytes, and others release cytokines as well as cells in various tissues throughout the body, such as epithelial cells and cells within connective tissues. This allows for high interconnectivity throughout the body with cytokines being able to bind locally, affecting cells at an infection or damage site, and distally, traveling through the bloodstream to bind to a specific target receptor. Specific kinds of cytokines can have different functions; chemokines are responsible for directing immune cells towards sites of infection, interferons signal cells to put up cellular defenses against pathogen invasion, tumor necrosis factors (TNF) help to regulate inflammation and can signal to immune cells that kill tumor cells, interleukins stimulate communication and coordination between WBCs, and colony-stimulating factors signal hematopoietic stem cells to develop into specific immune cell types.

These mediators play a significant role in regulating the inflammatory response to invaders and tissue damage. Inflammation is an immune system defense mechanism that initiates immune cell recruitment to the site of invasion, induces fever, and increases vascular permeability. Regulating and knowing when to inhibit inflammation is just as important as stimulating the immune system because, if left unchecked, inflammatory mechanisms can result in tissue damage. The potential for cell signaling is almost limitless. With so many different groups and factors, various origins and target effector cells, differing and overlapping functions, beneficial inflammation walks a very fine line. As signaling markers, cytokines, and other markers, are in constant communication to either produce a higher inflammatory response or to inhibit induction and start tissue repair. Some cytokines can fall into either a pro-inflammatory or anti-inflammatory category, or both. The cytokine families of interleukin- 1 (IL- 1) and interleukin- 6 (IL- 6) can be categorized as pro-inflammatory, while IL- 1 receptor antagonist (IL- 1RA), IL- 4, and IL- 10 can be characterized as anti-inflammatory, suppressing and dampening inflammation signaling and overall immune response [51]. In inflammation or diseased states, detection of cytokines often increases as the immune system mounts a defense and these signaling molecules begin to differentiate T cells, thus involving a cascade of increased inflammation and recruitment of immune cells to sites of infection. Cytokines are major factors in modulating this response and when it reaches the point of chronic inflammation or prolonged disease, the balance shifts to where inflammatory inhibitors are no longer being produced to dampen or counteract the inflammation damage.

Functional signaling proteins in saliva

The pathways involving these signaling molecules are complex and interconnected but are being increasingly studied in research to see specific mechanisms of inflammation when different diseases or infections manifest. As T lymphocytes play a major role in adaptive immunity, the types of Th cells that cytokines release from and act upon can be examined at heightened specificity. For example, the lymphocytes that develop into Th1 cells produce vast amounts of inflammatory cytokines such as IL- 2, IFN-γ, and TNF-α, lending to a cell-mediated immune response while Th2 cells produce IL- 4, IL- 5, IL- 10, and IL- 13, lending to B lymphocyte antibody responses [52, 53]. These maturation pathways are regulated by IL- 6 and IL- 4 [53, 54], creating a somewhat circular feedback loop that is carefully mediated by the anti-inflammatory mechanisms of IL- 10 and IL- 13. The tenuous cyclic relationship involving all these factors becomes faulty and imbalanced in cases of chronic inflammation. There are various immunoassays to test for the presence and concentrations of a variety of different cytokines, 45 of which can be found categorized and briefly summarized in Table 1. Many studies evaluate cytokine levels in infectious diseases (Human Herpesvirus- 6 and Epstein Barr Virus [55]), oral diseases (caries [56], gingival inflammation [57], and periodontitis [58]), and autoimmune diseases (Sjogren’s Syndrome [59]) to find, in many cases, increased concentrations of cytokines in diseased patients over controls. This increased detection in diseased patients can have diagnostic potential for some diseases, but, conversely, there seems to be a lack of association between increased cytokines and specific symptoms. The complexity of the pathway mechanisms requires more specific research for diagnostic power, but there is definitive evidence for the importance of cytokines in understanding how diseases are progressed and how the threshold from acute infection or disease could lead to something more chronic or autoimmune.

Table 1 Cytokines and their general main functions for immunity and inflammation. List of cytokines tested from saliva based on cellular mechanism pathways influenced on or by the cytokine. Pathways include Th1/Th2 cellular maturation and effects, Th9/Th17/Th22/Treg cellular maturation and effects, inflammatory cytokines, chemokines, and growth factors. Includes descriptions of their general functions for immunity and inflammation. Many of these analytes are detectable in saliva, especially when testing in diseased patients

Salivary cytokines

Pathways of inflammation are controlled by salivary cytokines and their imbalance causing an excessive response, potentially leading to tissue damage, opening possibilities of therapeutics to target these imbalanced pro-inflammatory cytokines. Looking into salivary cytokines reveals interesting relationships between pro- and anti-inflammatory cytokines, as well as other signaling factors, in systemic diseases such as HIV or tuberculosis, oral and dental diseases such as periodontitis or gingivitis, and oral and neck cancers [61]. Periodontitis is an inflammatory disease of the periodontal oral region and is initiated and progressed by pathogenic oral microbiota changes and the resulting immune response [117]. Similar to SIgA, cytokines can be produced locally from oral epithelial cells and resident immune cells [77], marking distinct pathways from blood cytokines. While there is still communication and potential cross-signaling to and from the blood, which suggests some relation between cytokines, local cytokine production in the oral cavity leads to differences in recorded cytokine levels between blood and saliva. In saliva, compared to blood, there is a greater amount of secreted cytokines in response to various diseases or pathogenic invasions. A study comparing blood vs. saliva cytokines in older adults found high correlation between plasma IFN-γ and salivary TNF-α, IL- 12p70, IL- 2, and IL- 10 [77], illustrating some methods of cytokine communication/signaling.

Lactoferrin

Lactoferrin (Lf) is an antibacterial and antiviral glycosylated protein that is secreted by exocrine glands and neutrophils in sites of infection or inflammation [118]. Depending on the saturation of iron, Lf can either be regarded as apo-lactoferrin or holo-lactoferrin when devoid of iron or fully saturated with iron, respectively. However, Lf has many anti-bacterial and anti-viral properties that are independent from its ability to sequester iron ions. For example, human Lf was found to inhibit the short term growth of P. gingivalis and P. intermedia by either apo- or holo-lactoferrin suggesting that the mechanisms at play don’t involve iron and the chelating ability of Lf [119]. These properties can be explained by two mechanistic pathways. The first increases the permeability of the bacterial cell membrane causing the release of cell contents and directly impeding bacterial function [120]. The second mechanism of Lf will detach external nutrients needed for bacterial survival from the bacterial wall through a process called hydrolysis [120]. Additionally, some of the antiviral properties of Lf are independent from its ability to chelate iron such as its ability to prevent the adsorption of HCMV [121]. Nonetheless, many of its functions as a key player in the innate immune system are dependent on its ability to bind to ions of irons. Lactoferrin competes with bacteria’s ability to accept free floating iron needed for growth or the production of biofilm. If the affinity for iron is greater in Lf than the bacterial receptors for capturing iron then Lf will produce bacteriostatic properties [122]. Lf also plays a role in promoting an immune response. Lactoferrin can increase the activity of natural killer (NK) cells, as well as promoting phagocytosis in neutrophils and the activation of macrophages [123]. Lactoferrin can also control the production of proinflammatory cytokines, TNF-α, IL- 6, and IL- 1β, depending on the local environment [123]. Due to the antibacterial and antiviral properties of lactoferrin, many papers have found success in the oral administration of Lf to prevent the progression of an infection [124, 125].

Mucins

Mucins are another important salivary protein that gives saliva its important function in aiding the innate immune response. Multiple mucins exist in the body, but the mucins found within the oral cavity are MUC5B, MUC7, MUC19, MUC1, and MUC4 and can have various functions such as giving a gel-like consistency to saliva, antimicrobial properties, and aiding in cell signal transduction [126]. Mucins that give saliva its gel-like consistency create a physical barrier that microbes must first pass before reaching any mucosal tissue. Streptococcus sanguinis, S. sobrinus, and S. oralis have all shown to bind to MUC7 showing there are receptors within mucins that may interact with microbes [127]. Additionally, MUC7 was shown to have direct antifungal properties against C. albicans and tested at various conditions to model the variation in pH, salt, and temperature of the mouth [128]. These findings could indicate that mucins may have a role in aggregating bacteria for easy clearance by swallowing or coughing while preventing microbes from coming in contact with the epithelial wall. However, many pathogens have ways of dealing with the mucus layer alone to gain access to tissue [129]. Therefore, mucins must also serve as a vehicle for other antimicrobial proteins and effectors of the innate immune system to deal with microbes. MUC7 and MUC5B are two mucins that are heavily involved in enhancing the antimicrobial properties of saliva. For example, MUC5B has shown to interact with antibacterial and antiviral proteins to prolong their availability within saliva by protecting these proteins from proteolytic degradation [130]. These findings show the importance of mucins to act as the delivery system in saliva.

Defensins

The family of defensins are another set of proteins in saliva with antimicrobial and antiviral properties. These small peptides are cationic with multiple cysteine residues forming three types of defensins (α, β, and γ) based on the pattern of the peptide chain and their disulfide bridges [131]. α-defensin- 1 (human neutrophil peptide 1) is stored in granules of neutrophils and secreted during neutrophilic apoptotic processes for both bactericidal properties and the regulation of inflammatory responses by other immune cells [132, 133]. Additionally, α-defensins have been found to have antiviral properties by inactivating HSV types 1 and 2, cytomegalovirus, vesicular stomatitis virus, and influenza virus A [134]. In SARS-CoV, defensins such as α-HD5 and HD6 can interfere with ACE2 and prevent 2019-nCoV from binding to the receptor making defensins very popular as a treatment option [135]. However, beta-defensins (hBDs) are the primary effectors of the innate immune response within saliva. hBDs are produced by epithelial cells in the oral cavity as a response to bacteria or inflammatory cytokines. The antimicrobial function of hBDs can be described by their unique ability to quickly kill bacteria by intercalating into the outer membrane to create holes in which contents of the cell are expelled [136]. hBD- 3 has antimicrobial function against both negative-gram and positive-gram bacteria, while hBD- 1 and hBD- 2 only have functions against negative-gram bacteria. Additionally, hBD- 1 and hBD- 3 have shown to have antifungal properties against C. albicans in vitro [137]. Within saliva, hBD- 1 and hBD- 2 can be found in saliva at concentrations of 150 ng/mL and have shown to synergistically work with other antimicrobial peptides [138].

Histatins

Histatins are another family of salivary peptides that primarily reside within saliva while exhibiting antimicrobial properties. Histatin- 1, histatin- 3, and histatin- 5 are the primary histatins that are found within human parotid saliva at concentrations of 2.5 µM, 1 µM, and 4 µM, respectively [139]. Histatin- 5 is produced from histatin- 3 after post-translational modifications and is considered to be the most chemically active of the three. The mode of antimicrobial action for these histatins has been largely disputed on whether this peptide directly alters the cell membrane or its uptake disrupts cellular metabolism subsequently leading to cell rupture [140]. Nonetheless, this protein is capable of causing the destabilization of cell membranes, based on its electrostatic charge, leading to cell death. Antimicrobial interactions between histatins can be observed in gram-positive and gram-negative bacteria along with yeast, fungi and parasites [139]. Such an example is observed in C. albicans where the cell walls appear fragile after cells are treated with histatin- 5 [139]. However, it is worth noting that the rate at which histatin- 5 kills off C. albicans is much slower than when compared to other antimicrobial proteins. Another paper found that Histatin- 5 could be advantageously used as a therapeutic against oral candidosis in immunocompromised patients [141]. In addition to histatin’s role in preventing infections, these peptides are also capable of playing a role in wound healing. Histatins, particularly histatin- 1, have been found to induce the migration of epithelial cells to seal off wounds in vitro, not just within the oral mucosa but also in skin [142]. Another report found that the use of histatin- 5 also promotes epithelial cell migration and MAPK/ERK pathways in vitro and corneal wound healing in murine models [143]. Lastly, due to histatin’s role in antimicrobial and wound healing properties this protein is of great interest as a biomarker and therapeutic. One study found that the level of histatin- 5 can be used as a biomarker in the progression of childhood caries [144]. This has the potential to improve caries treatments and inhibit the progression of severe caries. Histatin- 5 has also been proposed as a promising candidate for a therapeutic gel that treats oral mucositis in patients undergoing cancer treatment with results showing improved tissue health along other benefits [145]. Histatins play an important role in maintaining the barrier of the oral cavity by preventing tissue infections while promoting a healing tissue environment.

Lysozymes

Another salivary protein with promising diagnostic use are lysozyme proteins that play a pivotal role in the defense against pathogens. Lysosomes are polypeptides of 130 amino acids and a positive charge found at concentrations that range between 0.5 and 4.0 ng/mL [146]. These proteins are produced by cells capable of undergoing phagocytosis such as macrophages and neutrophils [147, 148]. Due to the cationic structure of lysozymes, these proteins are capable of inserting themselves into negatively charged bacterial walls to form pores that disrupt the stability of the cell membrane. In addition, through a different mechanism, lysozymes can also hydrolyze the peptidoglycan cell membrane, extracellularly exposed in gram-positive bacteria, which contributes to the destabilization of the bacterial membrane leading to antimicrobial effects [149]. However, research on mechanisms of antibiotic resistant bacteria have found that some Gram-positive bacteria engulfed by phagocytic macrophages can evade autolysis through lysozymes that induce the β-lactam resistant L-form [150]. This illustrates the complex relationship between agents of the innate oral immune system and the changing landscape of pathogens. It is also worth noting that lysozymes offer some protection against viral pathogens, such as SARS-CoV- 2, by reducing the cell entry capabilities of the virus, although the exact mechanisms are still unknown [151]. Research has also shown that lysozyme concentration in the oral cavity changes as the innate immune system becomes overloaded and, therefore, can be used as a diagnostic tool for the progression of caries, cancer, and hypertension among other diseases [152,153,154,155]. Additionally, depletion of lysozyme levels was found to have a strong correlation with common microbes, such as Streptococcus, Prevotella, Haemophilus and Veillonella, that form a dysregulated microbiota possibly caused by an exhausted and inflammatory innate immune response [156].

Defense mechanisms of the gingival barrier

Gingival crevicular fluid (GCF) in periodontal health and disease

In addition to saliva, gingival crevicular fluid (GCF) is an exudate that flows through the epithelial cells of the gingival sulcus, a concave space surrounding each tooth [157]. This fluid plays a critical role in periodontal health and disease, facilitating the transport of inflammatory mediators, immune cells, bacterial antigens, and enzymes of both host and bacterial origin. The GCF is closely tied to the subgingival biofilm, a dynamic microbial community that thrives in the anaerobic environment of the sulcus, protected from the cleaning action of saliva [158].

GCF composition and role in inflammation

The composition of GCF includes various biological markers, such as cytokines, enzymes (e.g., MMPs, collagenases), immune cells (primarily neutrophils), and inorganic ions. These components offer insights into the inflammatory status and the healing potential of the periodontium. In particular, the GCF serves as a valuable tool for assessing periodontal disease activity, as its composition reflects the tissue breakdown and immune responses within the periodontal pocket [159, 160].

GCF has emerged as a promising tool in the diagnosis of periodontal disease due to its ability to provide specific samples of biomarkers of inflammation and bone resorption directly at the site of the lesion. Among the most common biomarkers evaluated in GCF are IL- 1β, IL- 6, IL- 8, TNF-α, MMP- 8, MMP- 9, MMP- 13 and CRP, which are associated with active periodontal tissue destruction [161].

GCF, as a biopsy fluid sample, allows for more accurate diagnoses of the type and severity of periodontal disease, facilitating early detection of inflammation and potential bone destruction. Collecting GCF can help clinicians identify active inflamed sites, as well as predict disease progression by identifying early signs of periodontitis. This approach is especially valuable in the current context, where efforts are being made to optimize resources and make more rational and cost-effective treatment decisions.

Diagnostic value of GCF in periodontal disease

The flow of GCF and its composition can provide valuable diagnostic information regarding periodontal disease activity. Research has demonstrated a strong correlation between protease activity (such as collagenase) and clinical parameters, like pocket depth, which are commonly used to assess the severity of periodontal disease. As a non-invasive biomarker, GCF offers a promising avenue for monitoring disease progression and evaluating the effectiveness of periodontal treatments. In conclusion, GCF is a complex fluid that plays a crucial role in periodontal immunity and disease. Its composition reflects both the microbial and host responses in the periodontal pocket, making it a valuable tool in the diagnosis and management of periodontal disease. Future research into the molecular and cellular components of GCF will likely continue to expand its clinical applications [157].

Host-microbial challenge and the gingival epithelium barrier

Tooth eruption is the unique condition in which a hard tissue perforates the soft tissues, resulting in a disruption of epithelial continuity, jeopardizing the internal environment [162]. These gingival tissues are primarily designed to protect the host against oral microbial challenges [163]. The first tissue barrier against oral microbial invasion is the epithelial lining of gingival tissues. Basically, three different epithelial structures may be distinguished: the gingival epithelium (GE), the sulcular epithelium (SE) and the junctional epithelium (JE). GE and SE are derived from oral epithelium and JE is embryologically formed by the combining of the outer cells of enamel reduced epithelium and the basal cells of the oral epithelium after tooth eruption [162,163,164,165].

The GE is derived from the oral epithelium and is classified as a keratinized stratified squamous epithelium, containing all cell layers: basal (stratum basale or germinativum), prickle (stratum spinosum), granular (stratum granulosum) and keratinized (stratum corneum). This structure acts as a physical and immunological barrier against microbial challenge through different mechanisms, including increased proliferation rate, cell signaling, cell differentiation, apoptosis, and tissue homeostasis [162, 163], In addition to its role as a physical barrier, GE has a key role in the pathogenesis of periodontal diseases, since it represents one of the first defense mechanisms against microbial challenge. Keratinocytes are the major cell component of GE, but other cell types exist, including melanocytes, Langerhans, and Merkel’s cells [162,163,164].

It was a common thought that GE acted as a physical barrier to the invasion by microorganisms. However, more recent findings suggested that the host-microbe interaction is able to induce the synthesis of cytokines, adhesion molecules, growth factors, chemokines, and matrix metalloproteinases by GE cells. Under physiological conditions, GE allows fluxes of solutes and nutrients, collection of antigens and surveillance by mucosal immune cells [166].

Biofilms can modulate the epithelial cellular immune response based on their properties and composition [166]. Some keystone pathogens and pathobionts such as Porphyromonas gingivalis (P. gingivalis) can trigger innate and adaptive immune responses and disrupt the homeostatic state. The dysbiosis of the periodontal microbiota implies changes in the relative abundance of bacterial species compared to health, leading to alterations in the host-microbe interaction that result in inflammation and periodontal tissue destruction [167]. Once the epithelial barrier integrity is compromised by the biofilm, oral microorganisms may invade periodontal tissues and trigger an inflammatory response. Since that, cell-to-cell connections in the GE may be considered as a critical part of the innate immune response to resist oral microbiome challenge [166].

GE cells are predominantly keratinocytes, where progenitor lineage resides in the basal layer. As the keratinocyte moves from the basal to superficial layers, substantial changes in cell morphology and structure are observed, including the flattening of the cell and its nucleus, which can disappear (orthokeratinized epithelium) or be entrapped into the flattened cell and keratin layer (parakeratinized epithelium) [162,163,164].

A great number of different types of cell-to-cell junctions are observed in GE. Besides a 4 times greater number of desmosomes than in JE, tight (TJ), adherens (AJ) and gap (GJ) junctions are seen in GE Different signaling pathways that regulate cell differentiation, proliferation, and polarization in GE are coordinated by claudin and occludin, components of the TJ, and don’t exist in JE or SE [166].

This epithelial barrier can be disrupted by oral microbes in different manners, including cleavage of endothelial cells, degradation of cellular signaling molecules, and inactivation of cellular functions related to healing, regeneration, and homeostasis of periodontal tissues. P. gingivalis was shown to interfere with the cell-to-matrix and cell-to-cell adhesion of oral keratinocytes, reducing its adhesion to the extracellular matrix and altering its morphology and motility. When challenged with P. gingivalis gingipains, cultured oral keratinocytes showed proteolysis of focal contact components, such as focal adhesion kinase, catenins and adhesion signaling molecules [168] P. gingivalis fimbriae type II enhance bacterial adhesion and invasion of epithelial cells, degrade focal adhesion kinase, inhibit cellular migration and induce the serum release of IL- 1β and IL- 6 [169].

In addition, the modulation of epithelial cells signal transduction pathways by P. gingivalis can affect gene encodings. IL- 8 is down-regulated by P. gingivalis even in the presence of other stimulatory organisms, such as F. nucleatum. In health, IL- 8 forms a gradient of expression that directs neutrophils toward sites of infection, protecting the host against bacteria and neutrophil-mediated tissue degradation. Low expression of IL- 8 is important to maintain gingival health. Inhibition of IL- 8 accumulation by P. gingivalis can impair innate host defense at microbe-epithelium interface and the host could no longer be able to detect bacteria and direct neutrophils for their removal [169].

Immunity from gingival cells

GE cells present TLRs, transmembrane proteins that recognize molecular structures classified as pathogen associated molecular patterns or PAMPs. TLRs play a central role in the recognition of invading pathogens and the subsequent activation of inflammatory and immune responses. The TLR family harbors an extracellular leucine-rich repeat (LRR) domain and a cytoplasmic domain similar to IL- 1 receptor (IL- 1R). When stimulated, TLR recruits IL- 1R-associated protein kinases, activating nuclear factor-kappa B and mitogen-activated protein kinases. The response to TLR ligands varies, suggesting different signaling pathways [170]. In the intestinal epithelium, TLR1-TLR6 and TLR10 are expressed on the cell surface for recognition of extracellular microorganisms and ligands, while TLR3 and TLR7-TLR9 are intracellularly localized in the cytosolic endosomal compartment, binding microorganisms and ligands which passed the membrane of the host cell [171]. In the GE, TLR2 is highly expressed in the basal layer, and to a lesser extent, in the more superficial cell layers [166]. A mouse GE cell line (GE1) constitutively expresses TLR4 and TLR7. The stimulation of GE1 cells with CL075 induced cytokine, chemokine and antimicrobial peptide expressions and differed from the stimulation with LPS, with higher mRNA levels of IFN-β, CXCL10, and β-defensina 14 (analogous of human BD3), lower levels of TNF, CCL5, CCL11, CCL20, CXCL2, and CX3 CL1 [172].

In addition to TLRs, GE expresses NOD1 and NOD2, which are nucleotide-binding oligomerization domain receptors that bind to peptidoglycans present in bacterial cell walls [173]. TLR 2, TLR4, NOD1 and NOD2 are expressed in normal oral epithelium and gingival tissues [166, 173, 174]. These structures are also expressed in oral, tongue, salivary gland, pharyngeal, esophageal, intestinal, cervical, breast, lung and kidney epithelial cells, in addition to TLR3 and TLR7 [166, 173]. When challenged in vitro with bacterial components, GE cells increase the expression of TLR2, TLR4, NOD1 and NOD2, without significantly affecting cytokines secretion, although PRPGs and beta-defensin 2 were significantly upregulated [173,174,175]. The higher expression of TLR2 and TLR4 is associated with the release of inflammatory mediators and worsening of periodontal tissues [173, 176,177,178,179].

Human β-defensins

Are a group of small antimicrobial cysteine-rich peptides against Gram-positive and Gram-negative bacteria, fungi, and viruses [180]. Two subfamilies are described: α-defensins (6 subtypes) and β-defensins (4 subtypes) [180,181,182,183]. Human β-defensins are expressed in gingiva, oral mucosa, GE keratinocytes, parotid gland, saliva, buccal mucosa, tongue, and GCF [138, 184,185,186]. While the expression of HβD- 1 is constitutional, HβD- 2 expression is stimulated by IL- 1 and LPS, suggesting that they play an important role in innate immune defenses against oral microorganisms [138, 184]. HβD- 3 was detected in 88% of gingival biopsies obtained from healthy and chronic periodontitis patients, confined to the GE. In health, HβD- 3 was detected more frequently in the basal layer when compared to diseased subjects (53% vs. 18%), while in chronic periodontitis HβD- 3 expression occurred from the basal to spinous layer (82%). In healthy tissues from diseased subjects, HβD- 3 expression was noticed in the basal and deep spinous layers, while in pocket areas the expression was found in the superficial spinous layer. In healthy and diseased patients, HβD- 3 was expressed by GE keratinocytes, Langerhans and Merkel cells. These findings suggested the role of HβD- 3 in periodontal homeostasis through its antimicrobial activity and adaptive immune responses [187].

Disruption of the GE barrier may also be the result of excessive neutrophil elastase (NE) function. Although neutrophils are primarily involved in host responses against bacterial challenges, excessive activity may lead to the destruction of human tissues. NE is positively correlated to GCF flow rate and clinical attachment loss in periodontitis patients, but its underlying mechanisms remain unclear. For that reason, Hiyoshi et al. (2022) investigated how NE induces periodontitis severity and the role of NE inhibition in periodontitis treatment in ligature-induced periodontitis in mice. A greater neutrophil recruitment and NE activity was seen in the animals along with bone loss. The administration of an NE inhibitor significantly decreased NE activity in periodontal tissues, downregulated proinflammatory cytokines, and limited bone loss. NE cleaves cell adhesion molecules desmoglein 1, occludin and E-cadherin and induced exfoliation of epithelial keratinous layer in a 3-D in vitro model, suggesting that NE induces the disruption of the gingival epithelial barrier and bacterial invasion of periodontal tissues, contributing to a worsening of periodontitis [188].

Periodontal diseases

Periodontitis is a chronic inflammatory disease characterized by microbe-host response interaction that results in loss of periodontal attachment through the activation of host-derived proteinases that enable the loss of marginal periodontal ligament fibers, apical migration of JE, and biofilm along the root surface [189]. Biofilm is required but not sufficient to induce periodontitis since it is the host inflammatory response to this microbial challenge that results in the loss of periodontal tissues [167, 190]. The initiation and progression of periodontitis depend on disruption of homeostasis by ecological changes in the microbiome induced by inflammation triggered by host responses to bacterial challenge in an attempt to contain it within the gingival sulcus, once the inflammation has initiated [189].

Severe periodontitis affects approximately 19% of people greater than 15 years of age, representing more than 1 billion cases worldwide, according to WHO. Prevalence across country income groups is similar, although the highest case numbers are seen in lower-middle-income countries and the lowest in low-income countries. It almost equally affects men and women, starting in adolescence, peaking at 55 years of age, and remaining high with aging [191].

Advanced periodontitis can result in tooth loss, severely compromising oral function and aesthetics, with a negative impact on the quality of life. In addition, severe periodontitis is associated with systemic diseases and conditions, such as cardiovascular diseases, diabetes, rheumatoid arthritis, lupus, and chronic pulmonary obstructive disease, among others. The pathogenic mechanisms underlying these associations may be explained by the direct migration of bacteria and its products to sites distant from the oral cavity via either hematogenous routes (direct mechanisms), increased blood levels of inflammatory mediators, or even mimetics (indirect mechanisms) [192].

The transition from health to disease involves profound alterations in the biofilm ecology, with the conversion of a symbiotic to a dysbiotic biofilm in a pathogenic mechanism named polymicrobial synergy and dysbiosis [167, 193]. This implies a change in the relative abundance of individual components of the microbial community compared to health, ultimately mediating destructive inflammation and bone loss [193, 194]. This could be related to microbial challenges and the activation of the innate and adaptive immune system and its role in host-microbial interactions.

Besides the role of keystone pathogens, homeostasis can be disrupted by congenital or acquired immunodeficiencies, aging, systemic diseases such as diabetes and obesity, environmental factors, such as smoking and diet, and epigenetic modifications in response to environmental changes [167]. The accumulation of dental plaque at gingival margins results in the development of gingivitis, which is a reversible inflammatory lesion that affects the gingival connective tissues. The resolution of gingival inflammation is achieved by personal and professional mechanical plaque control. Not all cases of gingivitis progress to periodontitis, but a periodontitis lesion develops from a previous gingival inflammatory lesion. If untreated, periodontitis lesions may lead to tooth loss.

Some research has shown that oral fluids can be used to assess the level or progression of periodontitis. A study investigated the levels of cytokines (IL- 1, IL- 2, IL- 4, IL- 6, IFN-γ and TNF-α) in GCF and saliva of 40 patients with aggressive periodontitis before and after treatment compared to 40 healthy volunteers [195]. The results showed increased pocket depth in AP before treatment. The concentrations of cytokines in both GCF and saliva were significantly higher in AP than in healthy patients and decreased after treatment. There was a positive association between GCF cytokine levels and clinical periodontal parameters, as well as a satisfactory reliability of cytokines in saliva and GCF. Similarly, 16 s rDNA measuring the microbiota of saliva from aggressive and chronic periodontitis against a healthy control, found a close relationship between the abundance of P. gingivalis and periodontitis [196]. These two studies suggest that measuring cytokines and microbiota in saliva may be considered an easy and noninvasive method for monitoring periodontal disease activity.

Pathogenesis of periodontal disease

Classically, the pathogenesis of periodontal disease is described in four stages after initiation of plaque accumulation: initial (2–4 days after plaque formation), early (4–7 days after plaque formation), established (12–21 days after plaque formation), and advanced lesion [197]. The initial lesion is characterized by neutrophil recruitment and emigration through JE and a discrete inflammatory infiltrate localized immediately beneath the JE occupying 5–10% of connective tissue. This inflammatory infiltrate is mainly composed of monocytes/macrophages, lymphocytes, and neutrophils. This lesion is subclinical and can be observed only at the histopathological level. The only discrete clinical signs are the slight increase in GCF flow rate. Streptococcus are the main bacterial components of this stage and they are known to secrete a number of enzymes that increase SE and JE permeability. Lipoteichoic acid and peptidoglycans are components of the cell wall of these early colonizers which are capable of activating complement via the alternative pathway, inducing the production of C3a and C5a, anaphylatoxins that flow back to the tissues and establish a gradient from the gingival sulcus to the tissues, where they can induce the release of vasoactive amines from mast cells, increasing vascular permeability. Mast cells also release other cytokines, such as TNF-α, stimulating endothelial cells to express adhesion molecules, and recruiting neutrophils to the gingival tissues. Simultaneously, bacterial-released substances and C5a attract neutrophils to the JE. Within the gingival sulcus, neutrophils release their lysosomal contents (abortive phagocytosis) that return to the tissues and degrade connective tissue. PMNs also release neutrophil extracellular traps (NETs) during pathogen-induced cell death (NETosis), which represents one of the first lines of host defense against microbial challenge. NETs are released by both dead and live PMNs and are associated with severe tissue damage. Additionally, many proinflammatory substances, such as LPS, IL- 8, TNF-α, and Streptococcus M protein, induce NET formation. Other cell types are able to secrete extracellular traps, as well, especially mastocytes. These cells release mast cell extracellular traps which limit the entrance of bacteria and its vesicles into the tissues, also contributing to localized tissue destruction. PMNs also secrete other cytokines, such as IL- 1 and its antagonist receptor (IL- 1RA) and IL- 17, which induces the production of IL- 8 by SE cells. IL- 8 is a strong chemoattractant for PMNs and stimulates NET formation. IL- 17 has a protective role in periodontal disease, maintaining the PMN barrier in the gingival sulcus [198].

The early lesion is characterized by increased neutrophil emigration and initial lateral proliferation of JE cells, approximately 4–7 days after initial plaque accumulation. A larger inflammatory infiltrate is observed at the gingival connective tissue, along with vascular proliferation, dilated vessels, and an increase in vessel permeability and collagen loss. Therefore, the GCF flow rate dramatically increases. Similarly, there is an increase in the permeability of SE and JE, allowing the entrance of bacterial products into gingival tissues. At this stage, there is a shift from PMN-predominant inflammatory infiltrate to lymphocytes and macrophage prevalence. Clinically, it is possible to notice early signs of gingival inflammation. In the established stage, a marked increase in the lateral proliferation of JE and neutrophil emigration, along with a greatly increased leukocytic inflammatory infiltrate composed by 10–30% of plasma cells is observed. Clinically, there are clear signs of chronic gingival inflammation, characterized by bleeding on probing, swallowing, and redness of the gingival margin [197, 198].

Around 12–21 days after initial plaque accumulation, the lesion becomes clinically evident. At 21 days, lymphocytes are 70% of the inflammatory infiltrate and a 4-fold increase in the number of PMNs within JE occurs. PMNs and plasma cells represent less than 10% of the total inflammatory infiltrate. TNF-α and IL- 17 are secreted from mast cells and PMNs undergoing NETosis, leading to an increase in endothelial cell leukocyte adhesion molecule- 1 (ELAM- 1) and intercellular adhesion molecule- 1 (ICAM- 1), resulting in an increase in the secretion of IL- 8 by epithelial cells, attracting neutrophils to the JE and forming a barrier against bacterial invasion. Degradation of collagen is estimated to be of the order of 60–70%. A delayed-type hypersensitivity-like reaction develops, with the formation of perivascular lymphocyte-macrophages inflammatory infiltrate, consisting of 2 CD4+:1 CD8+ T cells, dendritic APCs, and macrophages. Activated T cells and SE cells express high levels of human leukocyte antigen HLA-DR and HAL-DQ. Langerhans cells existent in the GE uptake soluble antigens entering the tissues, presenting it to lymph nodes and activating T cells, which turn back to the tissues and, together with macrophages, control the entrance of invading microorganisms and reestablish the homeostasis with biofilm [198].

In cases in which gingivitis progresses to periodontitis, the gingival inflammation can exert pressure for the development of a dysbiotic and inflammophilic microbiota, including members that can subvert or evade the immune system. The severity of periodontal lesion may depend on host-related parameters that influence the host’s immune, inflammatory, and regenerative response [167]. The advanced lesion corresponds to the advanced stage, in which the inflammatory infiltrate is mostly composed of plasma cells occupying > 50% of the gingival connective tissue volume. An apical proliferation and migration of the JE along with detachment and ulceration of the pocket epithelium are observed [197] In the presence of generalized, moderate, periodontal pockets, the ulcerated epithelium is estimated to correspond to the size of the palm (approximately 50 to 75 cm2) [199], constituting an important risk factor for the development of systemic diseases.

The role of gingival innate and adaptive immune responses

The innate and adaptive immune responses to microbial challenges are therefore the key for destructive periodontal diseases. The first point to be highlighted is the massive accumulation of neutrophils within the JE, pocket epithelium, and gingival connective tissue [167, 190, 197]. Neutrophils are vital to maintain periodontal health and any impairment in its function, such as in cyclic neutropenia or any other condition, results in rupture of the tissue homeostasis [167]. Periodontal tissue destruction is mediated by the release of enzymes, such as matrix metalloproteinases, and other substances such as cytokines and ROS [200], activation of osteoclastic bone resorption [201], chemotaxis for IL- 17-producing CD4+ Th17 cells by the production of CCL2 and CCL20 [202], activating T cells and leading to bone loss. B and T cells are the major source of membrane bound and secreted Receptor Activator of Nuclear Factor-kappa B Ligand (RANKL) involved in periodontal bone loss. Th1 has a protective role in periodontal lesions through the release of IFN and IL- 12 cytokines that promote cell-mediated immunity and inhibit osteoclastogenesis [203, 204], although in some instances Th1 cells may also activate bone loss by expressing RANKL [203]. Th1 cells predominate in stable lesions, while Th2 predominates in progressive lesions characterized by a B cell dominant inflammatory response [204].

Increased deposition of immune complexes and complement fragments in diseased gingiva suggests the role of plasma/B cell antibodies in inflammatory responses [167]. Indeed, greater deposition of immune complexes was found in gingival tissue samples of systemic lupus erythematosus (SLE) and periodontitis than in periodontitis-only patients, suggesting the role of inflammation in the pathogenesis of the two conditions [205]. In addition, B cells produce inflammatory cytokines and MMPs that can further contribute to tissue damage [204].

The attachment of JE cells to the enamel surface and to the underlying connective tissue is stronger than the cell-cell attachment, while the few desmosomes and wide intercellular spaces between JE cells allow the gingival crevicular fluid (GCF) and inflammatory and immune cells to transmigrate, protecting the body against microbial invasion [206]. In the course of periodontitis, there is a conversion of JE to pocket epithelium. Detachment of JE from the tooth surface seems to occur after the increasing degree of inflammation and transmigration of neutrophils, eventually leading to focal disintegration of JE [206]. Bacteria can also contribute to the detachment of JE and the formation of the pocket epithelium. P. gingivalis gingipains are capable of degrading components of cell-cell junctional complexes and cleaving intercellular adhesion molecule- 1 (CD54) on oral epithelial cells, resulting in disruption of the interaction of neutrophils and epithelial cells [207]. Apically, the pocket epithelium is continuous with a reduced JE, which proliferates apically as the pocket deepens, maintaining the integrity of the gingival sulcus [206].

Dental plaque represents a continuous challenge to the innate immune system. In healthy sites, this challenge is beneficial, while in diseased sites there is an imbalance between host defenses and bacterial challenges. Due to the specific characteristics of JE, a highly orchestrated expression of mediators is released in the periodontal tissues, including e-selectin, ICAMs, and IL- 8, which facilitate the transit of neutrophils from the gingival connective tissue to the gingival crevice [190, 208]. Other innate immune mediators expressed in periodontal tissues are β-defensins (βD) 1, βD2, βD3 [184, 187, 190], CD14 and lipopolysaccharide-binding protein (LBP) [190]. LBP, produced by gingival epithelial cells and the liver as an acute phase response marker [190], and LBP mRNA are more highly expressed in healthy than diseased sites [209]. As previously described [190], gingival tissues express TLRs, especially TLR2 and TLR4 in the development of periodontal diseases. Il- 1β was demonstrated in periodontal tissues of germ-free mice.

Many cytokines associated with inflammation are found in GCF of periodontitis patients at higher levels than in healthy ones, including IL- 1β, TNF, and prostaglandin E2 (PGE2). In addition, soluble and membrane-bound CD14 and LBP are also present in higher concentrations in GCF from diseased than healthy sites [190].

Considering that, host responses are primarily involved in the destruction of periodontal tissues in the course of the disease. The main mechanism explaining the couple effect of osteoblasts and osteoclasts is the RANKL: Osteoprotegerin (OPG) ratio. High OPG levels prevent the RANK-RANKL interaction and binding of RANKL to osteoclast precursors, impeding bone resorption. OPG levels are regulated by bone morphogenetic proteins (BMPs) and the synthesis of RANKL is induced by pro-inflammatory cytokines, such as TNF and IL- 1, which suggests that the RANKL: OPG ratio is increased in the presence of gingival inflammation, resulting in bone loss [190]. Ultimately research demonstrates that local tissues are not affected in isolation, but rather an inflammatory process impacts systemic tissues across the human body.

Oral pathogens and their interactions with the immune system

The oral cavity hosts a wide diversity of microorganisms, including bacteria, viruses, and fungi, which coexist in balance with the host under healthy conditions [210]. However, environmental, immunological, or behavioral changes can lead to uncontrolled growth of these pathogens, triggering inflammatory processes and diseases. Among the most studied bacteria are Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, and Streptococcus mutans, all of which are associated with conditions such as periodontitis and dental caries. P. gingivalis, for example, has the ability to modulate the host’s immune response, subverting defense mechanisms to favor its own survival and contributing to chronic inflammation [211, 212].

Saliva does not have a resident microbiota, but resident oral microorganisms can be found in saliva, originating from teeth, oral mucosa and tongue, mainly. These microorganisms are essential to maintain homeostasis, since they prevent colonization by exogenous microorganisms, downregulating proinflammatory responses and converting nitrate to nitrite, contributing to antimicrobial activities, mucus production and vasodilation, which could potentially lower blood pressure. The composition of the acquired pellicle, which is formed short after the tooth enters in contact with saliva, determines the pattern of microbial colonization [213].

Based on that, since periodontitis arises from the complex interaction between subgingival microbiota composition and the host immune response, an interplay between periodontal diseases and other systemic diseases exist. For instance, in SLE patients, periodontal bacterial diversity was reduced, with an enrichment of specific periodontal pathogens dominating the microbial environment. The connectivity of F. nucleatum with other bacterial species and cytokines was higher in individuals with SLE compared to control subjects. A. gerencseriae, C. ochracea, and T. forsythia exhibited greater connectivity in both control and SLE-A individuals, whereas S. oralis and P. nigrescens showed increased connectivity in SLE-A subjects. Additionally, S. noxia was more prevalent in the control group, while C. gingivalis displayed no connectivity in control individuals. These findings suggest that subgingival bacterial species associated with SLE influence systemic host cytokine patterns, impacting overall health [214].

In addition to bacteria, viral infections also play a significant role in oral health. Herpes simplex virus type 1 (HSV- 1) is one of the most prevalent, causing recurrent lesions in the oral mucosa and impacting local immunity [215]. Epstein-Barr virus (EBV) and cytomegalovirus (CMV) can also be present in the oral cavity, especially in immunocompromised individuals, where they contribute to immune dysfunction and the worsening of periodontal diseases [216]. These viral infections can alter both innate and adaptive immune responses, promoting a microenvironment favorable for viral persistence and chronic inflammation. Furthermore, evidence suggests that the interaction between these viruses and the immune system can trigger complex mechanisms of immune modulation, favoring both the chronicity of infections and the progression of oral inflammatory condition [216].

Fungi, especially Candida albicans, represent another group of relevant pathogens. Although C. albicans is part of the commensal oral microbiota, changes in host immunity can lead to oral candidiasis. The interaction between the microbiota and the immune system is crucial in controlling C. albicans infections [217]. The microbiota not only protects against fungal overgrowth but also regulates the immune response, particularly the function of cells like neutrophils and dendritic cells. Deficiency in IL- 17 is an example of how failures in the immune response increase susceptibility to fungal infections [218, 219]. New therapeutic approaches, such as the use of probiotics and immunomodulatory molecules, which can restore immune protection, are being explored [217].

Recently, advances in omics approaches, such as metagenomics, metaproteomics, and metabolomics, have allowed for a more comprehensive understanding of oral microbial communities and the endogenous and exogenous factors influencing their composition. Growing evidence supports the notion that various internal and external factors cause dysbiosis of the oral microbiota, contributing significantly to oral and systemic diseases. Current studies still focus on bacteria and C. albicans, with identifying the mechanisms of communication and regulation within and between the microbiome, virome, and host immune system being a challenge. Promising therapeutic strategies for oral diseases are under development, but few investigations on their effects on oral dysbiosis have produced consistent results [210].

This section details the main oral pathogens, addressing their characteristics, pathogenicity mechanisms and impact on periodontal health, summarized in Table 2.

Table 2 List of most common pathogens that affect the oral cavity, including the associated oral manifestations and cellular targets

Pathogenic bacteria

The bacterial microbiota is the main factor involved in periodontitis. Some specific pathogens are closely associated with the progression of periodontal disease.

Porphyromonas gingivalis

One of the best known and most widely studied periodontal pathogens. P. gingivalis is a gram-negative anaerobic bacterium that can invade periodontal tissues, leading to the destruction of collagen fibers and alveolar bone by inducing high levels of proinflammatory cytokines, such as IL- 1𝛽 and IL- 6 by periph-eral CD4 + T helper cells [220]. It produces enzymes such as gingipain, which interfere with the host’s immune response and aid in the colonization of the periodontal environment [233].

Tannerella forsythia

Also an anaerobic gram-negative bacterium, it is frequently found in individuals with severe periodontitis. It plays an important role in periodontal inflammation, causing destruction of periodontal tissue and interacting with other bacteria to form a pathogenic biofilm [224]. More recently, Tannerella forsythia, like Porphyromonas gingivalis, was found to be associated with an increased risk of esophageal cancer [234]. Although the exact mechanisms are unknown, it is thought Tannerella forsythia induces an inflammatory response characterized by IL- 1 and TNF-α, which can help the development of tumors through COX- 2. However, more research is required to know exactly how Tannerella forsythia contributes to tumor progression.

Treponema denticola

An anaerobic spirochete bacterium that is associated with aggressive periodontal diseases. T. denticola is capable of forming biofilms and producing proteases that disintegrate gingival tissue [225].

Fusobacterium nucleatum

This gram-negative, anaerobic bacterium is known for its ability to interact with other bacterial species to form complex biofilms and contribute to the chronic inflammation associated with periodontitis [222]. Additionally, this bacteria has been implicated in some cancers. A study on the gene expression profile of human tumor tissues found that the level of Fusobacterium nucleatum was associated with a suppressive immune tumor environment characterized by CD8+ T cell depletion and enrichment of FoxP3+ regulatory T cells [235].

Aggregatibacter actinomycetemcomitans

A facultatively anaerobic gram-negative bacterium, often associated with aggressive periodontitis, especially in young people. It produces leukotoxins that induce leukocyte apoptosis and help the bacteria escape the immune response. Its presence is linked to rapid bone and periodontal attachment destruction [236].

Prevotella intermedia

A strict anaerobic gram-negative bacterium, adapted to the subgingival environment. Commonly associated with necrotizing ulcerative gingivitis (NUG) and necrotizing ulcerative periodontitis (NUP), severe inflammatory conditions. It produces proteolytic enzymes that degrade the extracellular matrix of healthy cells and human IgG, facilitating the destruction of periodontal tissue while evading some immune responses [223, 237].

Pathogenic viruses

Although periodontal diseases are predominantly bacterial, viruses also play an important role, especially in situations where immune function is compromised, such as in autoimmune diseases.

Herpes simplex virus (HSV)

Although HSV is most often associated with oral lesions, it can also aggravate periodontal inflammatory conditions, especially in immunocompromised patients. HSV type 1 is commonly found in lesions on the gingiva and oral mucosa, causing primary herpetic gingivostomatitis, recurrent ulcers, and exacerbation of periodontal inflammation. HSV type 2, although more commonly associated with genital infections, can be found in the oral cavity and has been implicated in the pathogenesis of oral ulcers and increased severity of periodontitis in immunosuppressed individuals [226].

Cytomegalovirus (CMV)

It is frequently present in patients with severe periodontal diseases. CMV may participate in the development of periodontitis by causing macrophages and T cells to release osteoclast-inducing IL- 1β and TNF-α. Also, gingival fibroblasts infected with cytomegalovirus exhibit diminished production of collagens I and III and enhanced generation of MMPss 1 and 2. However, although biologically plausible, the extent to which cytomegalovirus participates in the destruction of the human periodontium is still a matter of research [228].

Epstein-Barr virus (EBV)

Also detected in inflamed periodontal tissues, EBV can contribute to the chronic inflammatory response and is involved in increasing the virulence of other bacterial periodontal infections. Persistent EBV facilitates viral evasion and exacerbates the infection. Increased viral activity leads to heightened local immune and inflammatory responses, worsening the inflammatory condition, promoting B cell and plasma cell infiltration, and advancing epithelial tissue degradation. EBV-induced impairment of host defenses results in bacterial superinfection, culminating in destructive periodontitis [238].

Human immunodeficiency virus (HIV)

HIV is not directly a periodontal pathogen, but its presence profoundly alters the immune response, making individuals more susceptible to periodontal infections. Patients with HIV have a higher prevalence of severe periodontal diseases, such as linear erythematous gingivitis and necrotizing periodontitis, characterized by intense inflammation and rapid tissue destruction. Immunosuppression caused by HIV favors the proliferation of opportunistic pathogens, such as C. albicans and P. intermedia [230].

HIV- 1 infection acts as a modifying factor in periodontal diseases, and is frequently associated with the occurrence of acute periodontal diseases and exacerbation of preexisting chronic periodontitis. The great bacterial diversity and complexity in the oral microbiota of HIV- 1-infected individuals seems to be related to the chronic periodontitis progression and severity [231].

Antiretroviral therapy can modify the oral microbiota and affect the progression of periodontal disease.

Pathogenic fungi

Although fungi are not as prominent as bacteria, they do play a role in the development of oral diseases, especially in immunocompromised patients.

Candida albicans

The most common fungus found in the oral cavity, especially in individuals with compromised immune systems. C. albicans can cause oral candidiasis, an infection that can result in painful lesions in the oral mucosa and contribute to an imbalanced microbiota, favoring pathogenic bacterial colonization [239, 240].

The impact of oral tissues and fluids in oral-systemic diseases

Oral mucosal tissues and biofluids impact on oral-systemic diseases influencing negatively when in dysbiosis. It explores the relationship between oral health and systemic conditions such as diabetes https://pubmed.ncbi.nlm.nih.gov/33426512/, cardiovascular disease https://www.amjmed.com/article/S0002-9343(23)00755-6/fulltext, oral cancer https://pubmed.ncbi.nlm.nih.gov/33155101/, and lupus https://pubmed.ncbi.nlm.nih.gov/31781106/.

Diabetes

Diabetes is a major risk factor for periodontitis [241]. In diabetes, the increased expression of TLR2 or TLR4 or both results in the release in proinflammatory cytokines, such as TNF-α and IL- 6 by adipocytes [242], which could in severe inflammatory tissue destruction as seen in subjects with periodontitis and uncontrolled diabetes [243]. In gingival tissue samples collected from subjects with periodontitis or diabetes or periodontitis and diabetes or healthy, the expression of TLR2 and TLR4 were quantified. The results showed that chronic periodontitis or diabetes had no significant effect on TLR2 expression in the oral epithelium, but an increased expression was seen at SE and a changed pattern of expression was noticed in GE. Chronic periodontitis decreased the expression of TLR in GE. Higher percentages of TLR2 and TLR4 were found in the connective tissue under SE. Chronic periodontitis and diabetes subjects showed higher percentage of TLR2 and TL4 compared with healthy and diabetics patients [244].

Cardiovascular diseases

The growing interest in salivary biomarkers for diagnosing cardiovascular diseases (CVDs) has been driven by the potential for non-invasive, convenient, and rapid point-of-care testing. Several biomarkers found in saliva have shown promise in identifying acute myocardial infarction and other cardiovascular conditions. Key salivary biomarkers for CVD include myoglobin, cardiac troponin I, creatine phosphokinase MB, MPO, brain natriuretic peptide, exosomal microRNAs (miRNAs), C-reactive protein (CRP), matrix metalloproteinases (MMP- 8, MMP- 9), and tissue inhibitor of metalloproteinase- 1 [245].

Some studies have shown that salivary C-reactive Protein (CRP) levels correlate with serum CRP levels, suggesting that it could be a useful non-invasive marker for systemic inflammation in CVD. Increased salivary CRP may reflect systemic inflammation, a key player in the pathogenesis of cardiovascular diseases, including atherosclerosis and plaque rupture. CRP is a well-established marker of inflammation and is used to assess cardiovascular risk. It is produced by the liver in response to inflammatory cytokines, and elevated levels are associated with increased risk of atherosclerosis, myocardial infarction, and other cardiovascular events [246,247,248].

Oral cancer

Oral cancer, a significant global health concern, has prompted extensive research into utilizing salivary proteomics for early detection and improved diagnostics. Saliva, in its intimate contact with the oral cavity, harbors a plethora of proteins and peptides that can reflect the physiological state of the oral tissues, including the presence of malignant or premalignant lesions.

Advances in mass spectrometry-based proteomics have enabled the identification and quantification of thousands of salivary proteins, opening up new avenues for biomarker discovery. Several studies have reported panels of salivary proteins that show differential expression in oral cancer patients compared to healthy controls [249,250,251,252,253]. These potential biomarkers include proteins involved in inflammation, immune response, cell signaling, and tissue remodeling. Notably, some of these proteins have been detected in saliva even before the clinical manifestation of oral cancer, highlighting the potential of salivary proteomics for early diagnosis. In addition to proteomics, other detection methods and types of markers have been proposed as tools for cancer diagnosis such as salivary miRNAs [254,255,256].

However, the path to clinical implementation of salivary biomarkers for oral cancer is not without challenges. The heterogeneous nature of saliva, inter-individual variability, and the influence of confounding factors such as oral hygiene and diet can complicate the identification of robust and reliable biomarkers [257,258,259]. Ongoing research is focused on addressing these challenges through the development of standardized saliva collection and processing protocols, advanced proteomic technologies, and sophisticated data analysis algorithms [260, 261].

Despite these challenges, the potential of salivary proteomics for oral cancer detection and monitoring remains promising. With continued research and technological advancements, salivary biomarkers could become a valuable tool for early diagnosis, risk assessment, and personalized treatment of oral cancer, ultimately improving patient outcomes and survival rates.

Lupus

Oral bacteria play a crucial role in modulating immune responses and contributing to dysbiosis, which can promote systemic inflammation. The oral cavity, particularly niches such as the gingival sulcus and fluid, is a site where pathogens may induce local and systemic inflammation, contributing to various chronic diseases like type 2 diabetes, rheumatoid arthritis, cardiovascular diseases, systemic lupus erythematosus (SLE) and others [262, 263].

Recent studies have highlighted the role of oral pathogens in exacerbating chronic inflammatory conditions, including SLE. Elevated levels of inflammatory cytokines, such as IL- 1, IL- 6, IFN-γ, and TNF-α, have been observed in lupus patients compared to healthy controls, with some differences in cytokine profiles between lupus patients and those with periodontal disease [264, 265].

Research by our team has shown that lupus patients, particularly those with active disease, exhibit an increased regulation of pro-inflammatory cytokines. Additionally, the oral microbiota of lupus patients is dysbiotic, with significant elevations in T. denticola and T. forsythia, which are associated with higher systemic inflammatory cytokine levels [214]. Some findings suggest that oral pathogens can influence disease activity, and the immune response in SLE may be exacerbated by oral infections, particularly in those with concurrent periodontal disease [205].

Interestingly, salivary biomarkers are emerging as non-invasive indicators for the early detection and monitoring of SLE. Studies show that salivary IgA levels, as well as specific cytokines, correlate with disease activity and can differentiate SLE patients from healthy individuals. Furthermore, salivary cytokines could be valuable tools for early diagnosis, monitoring disease progression, and assessing treatment efficacy [266]. However, further research is necessary to fully understand the pathogenic mechanisms linking the oral microbiome to SLE and other autoimmune diseases, and to validate salivary biomarkers as reliable diagnostic tools for SLE [267].

Future directions

Salivary biomarkers have a promise on how we envision precision medicine. Deeper exploration of salivary biomarkers holds the potential to revolutionize diagnostics and therapeutics. Identifying specific salivary signatures associated with various diseases could enable early detection, personalized treatment plans, and monitoring of therapeutic responses. Therapeutic modulation of salivary immunity is still underexplored. Investigating interventions that can modulate salivary immune responses opens new avenues for disease management. This could include targeted therapies to enhance beneficial immune components, suppress harmful inflammation, or restore a balanced oral microbiome.

Longitudinal studies for disease progression would provide better validation of salivary markers in health and disease longitudinal studies are crucial to understand the long-term impact of salivary immune responses on disease progression. Tracking changes in salivary biomarkers over time can reveal valuable insights into disease trajectories and inform preventive strategies. By integrating salivary and systemic data we will better understand how and why saliva is representative of general health in addition to the oral health metrics. Integrating salivary data with other systemic health information can provide a more comprehensive understanding of disease mechanisms and individual health profiles. This could lead to the development of integrated diagnostic and treatment approaches.

The salivary microbiome has incredible potential in personalized medicine. Further research into the salivary microbiome and its interactions with the host immune system can unlock opportunities for personalized medicine. Tailoring interventions based on an individual’s salivary microbiome profile could improve treatment efficacy and minimize adverse effects.

Conclusions

This comprehensive review highlights the critical role of saliva in oral and systemic health. Saliva, with its diverse cellular components, proteins, cytokines, and immunoglobulins, acts as a dynamic and multifaceted barrier against pathogens. Understanding the intricate immune mechanisms within saliva is essential for unraveling the complex interplay between oral and systemic health. Future research in this field holds immense promise for developing innovative diagnostic tools, personalized therapeutic interventions, and preventive strategies to improve overall health and well-being.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

AJ:

Adherens junction

APCs:

Antigen-presenting cells

BAFF:

B-cell activating factor

BMI:

Body-mass index

BMP:

Bone morphogenetic protein

citH3:

Citrullinated histone H3

CPM:

Count per million

CVD:

Cardiovascular disease

DAMPs:

Damage-associated molecular patterns

DCs:

Dendritic cells

ELAM:

Epithelial leukocyte adhesion molecule

ELISA:

Enzyme-linked immunosorbent assay

E-selectin:

Endothelial-selectin

FC:

Fold-change

FDR:

False-discovery rate

GCF:

Gingival crevicular fluid

GE:

Gingival epithelium

GO:

Gene Ontology

GWAS:

Genome-wide association studies

HbA1c:

Hemoglobin A1C

hBD:

Beta-defensin

HCMV:

Human Cytomegalovirus

ICAM:

Intercellular adhesion molecule

IFN:

Interferon

Ig:

Immunoglobulin

IL:

Interleukin

IL- 1R:

Interleukin-1 receptor

IP- 10:

Interferon gamma-induced protein 10

JE:

Junctional Epithelium

KEGG:

Kyoto Encyclopedia of Genes and Genomes

LBP:

Lipopolysaccharide-binding protein

LCs:

Langerhans cells

Lf:

Lactoferrin

LLOQ:

Lower limit of quantification

LRR:

Leucine-rich repeat

MHC:

Major histocompatibility complex

MPO:

Myeloperoxidase

MUC:

Mucin

NE:

Neutrophil elastase

NETs:

Neutrophil extracellular traps

NK:

Natural killer

NOD:

Nucleotide-binding oligomerization domain

OECs:

Oral epithelial cells

OPG:

Osteoprotegerin

PAMPs:

Pathogen-associated molecular patterns

PCA:

Principal component analysis

PMA:

Phorbol 12-myristate 13-acetate

PPRs:

Pattern-recognition receptors

P-Selectin:

Platelet-selectin

QC:

Quality control

RANKL:

Receptor Activator of Nuclear Factor-kappa B Ligand

RNA-seq:

RNA sequencing

ROS:

Reactive oxygen species

RvE1:

Resolvin E1

SC:

Secretory component

SE:

Sulcular epithelium

sICAM- 1:

Soluble intercellular adhesion molecule-1

SIgA:

Secretory Immunoglobulin A

SPM:

Specialized pro-resolving lipid mediator

ST2:

Suppression of tumorigenicity 2

T2D:

Type 2 diabetes

TJs:

Tight junctions

TLRs:

Toll-like receptors

TNF-α:

Tumor necrosis factor-alpha

TMM:

Trimmed mean of M-values

TSLP:

Thymic Stromal lymphopoietin

References

  1. Choudhury SN, Novotny M, Aevermann BD, et al. A Protocol for Revealing Oral Neutrophil Heterogeneity by Single-Cell Immune Profiling in Human Saliva. 2020. PROTOCOL (Version 2) available at Protocol Exchange. https://doiorg.publicaciones.saludcastillayleon.es/10.21203/rs.3.pex-953/v2.

  2. Theda C, Hwang SH, Czajko A, et al. Quantitation of the cellular content of saliva and buccal swab samples. Sci Rep. 2018;8:6944. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-018-25311-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wong YT, Tayeb MA, Stone TC, Lovat LB, Teschendorff AE, Iwasiow R, et al. A comparison of epithelial cell content of oral samples estimated using cytology and DNA methylation. Epigenetics. 2022;17:327–34.

    Article  PubMed  Google Scholar 

  4. Dawes C. Estimates, from salivary analyses, of the turnover time of the oral mucosal epithelium in humans and the number of bacteria in an edentulous mouth. Arch Oral Biol. 2003;48:329–36.

    Article  CAS  PubMed  Google Scholar 

  5. Rao A, Jump RLP, Pultz NJ, Pultz MJ, Donskey CJ. In vitro killing of nosocomial pathogens by acid and acidified nitrite. Antimicrob Agents Chemother. 2006;50:3901–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vidović A, Vidović Juras D, Vučićević Boras V, Lukač J, Grubišić-Ilić M, Rak D, et al. Determination of leucocyte subsets in human saliva by flow cytometry. Arch Oral Biol. 2012;57:577–83.

    Article  PubMed  Google Scholar 

  7. Rijkschroeff P, Jansen IDC, van der Weijden FA, Keijser BJF, Loos BG, Nicu EA. Oral polymorphonuclear neutrophil characteristics in relation to oral health: a cross-sectional, observational clinical study. Int J Oral Sci. 2016;8:191–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fresneda Alarcon M, McLaren Z, Wright HL. Neutrophils in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus: same foe different M.o. Front Immunol. 2021;12:649693.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Scieszka D, Lin Y-H, Li W, Choudhury S, Yu Y, Freire M. NETome: A model to Decode the human genome and proteome of neutrophil extracellular traps. Sci Data. 2022;9:702.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Fine N, Hassanpour S, Borenstein A, Sima C, Oveisi M, Scholey J, et al. Distinct oral neutrophil subsets define health and periodontal disease States. J Dent Res. 2016;95:931–8.

    Article  CAS  PubMed  Google Scholar 

  11. Vitkov L, Klappacher M, Hannig M, Krautgartner WD. Extracellular neutrophil traps in periodontitis. J Periodontal Res. 2009;44:664–72.

    Article  CAS  PubMed  Google Scholar 

  12. Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20:1126–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lendeckel U, Venz S, Wolke C. Macrophages: shapes and functions. ChemTexts. 2022;8:12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Strizova Z, Benesova I, Bartolini R, Novysedlak R, Cecrdlova E, Foley LK, et al. M1/M2 macrophages and their overlaps - myth or reality? Clin Sci. 2023;137:1067–93.

    Article  CAS  Google Scholar 

  15. Yin L, Li X, Hou J. Macrophages in periodontitis: A dynamic shift between tissue destruction and repair. Jpn Dent Sci Rev. 2022;58:336–47.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Pourgonabadi S, Müller H-D, Mendes JR, Gruber R. Saliva initiates the formation of pro-inflammatory macrophages in vitro. Arch Oral Biol. 2017;73:295–301.

    Article  CAS  PubMed  Google Scholar 

  17. Almubarak A, Tanagala KKK, Papapanou PN, Lalla E, Momen-Heravi F. Disruption of monocyte and macrophage homeostasis in periodontitis. Front Immunol. 2020;11:330.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Qiao D, Cheng S, Song S, Zhang W, Chen B, Yan F, et al. Polarized M2 macrophages induced by glycosylated nano-hydroxyapatites activate bone regeneration in periodontitis therapy. J Clin Periodontol. 2024;51:1054–65.

    Article  CAS  PubMed  Google Scholar 

  19. Viniegra A, Goldberg H, Çil Ç, Fine N, Sheikh Z, Galli M, et al. Resolving macrophages counter osteolysis by anabolic actions on bone cells. J Dent Res. 2018;97:1160–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Miao Y, He L, Qi X, Lin X. Injecting immunosuppressive M2 macrophages alleviates the symptoms of periodontitis in mice. Front Mol Biosci. 2020;7:603817.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hovav A-H. Dendritic cells of the oral mucosa. Mucosal Immunol. 2014;7:27–37.

    Article  CAS  PubMed  Google Scholar 

  22. Allam J-P, Novak N, Fuchs C, Asen S, Bergé S, Appel T, et al. Characterization of dendritic cells from human oral mucosa: A new Langerhans’ cell type with high constitutive FcϵRI expression. J Allergy Clin Immunol. 2003;112:141–8.

    Article  CAS  PubMed  Google Scholar 

  23. The Adaptive Immune System. In. Molecular biology of the cell. W.W. Norton & Company; 2007. pp. 1539–602.

    Google Scholar 

  24. Angel CE, Chen C-JJ, Horlacher OC, Winkler S, John T, Browning J, et al. Distinctive localization of antigen-presenting cells in human lymph nodes. Blood. 2009;113:1257–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cardoso EM, Arosa FA. CD8 + T cells in chronic periodontitis: roles and rules. Front Immunol. 2017;8:145.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Choi Y, Woo KM, Ko SH, Lee YJ, Park SJ, Kim HM, et al. Osteoclastogenesis is enhanced by activated B cells but suppressed by activated CD8(+) T cells. Eur J Immunol. 2001;31:2179–88.

    Article  CAS  PubMed  Google Scholar 

  27. Taubman MA, Kawai T. Involvement of T-lymphocytes in periodontal disease and in direct and indirect induction of bone resorption. Crit Rev Oral Biol Med. 2001;12:125–35.

    Article  CAS  PubMed  Google Scholar 

  28. Jiang Q, Huang X, Yu W, Huang R, Zhao X, Chen C. MTOR signaling in the regulation of CD4 + T cell subsets in periodontal diseases. Front Immunol. 2022;13:827461.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Matsuzaki K, Sugimoto N, Islam R, Hossain ME, Sumiyoshi E, Katakura M, et al. Salivary Immunoglobulin A secretion and polymeric Ig receptor expression in the submandibular glands are enhanced in heat-acclimated rats. Int J Mol Sci. 2020;21:815.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gümüş P, Nizam N, Lappin DF, Buduneli N. Saliva and serum levels of B-cell activating factors and tumor necrosis factor-α in patients with periodontitis. J Periodontol. 2014;85:270–80.

    Article  PubMed  Google Scholar 

  31. Ibrahem HM. B cell dysregulation in primary Sjögren’s syndrome: A review. Jpn Dent Sci Rev. 2019;55:139–44.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Pelaez-Prestel HF, Sanchez-Trincado JL, Lafuente EM, Reche PA. Immune tolerance in the oral mucosa. Int J Mol Sci. 2021;22:12149.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Molero-Abraham M, Sanchez-Trincado JL, Gomez-Perosanz M, Torres-Gomez A, Subiza JL, Lafuente EM, et al. Human oral epithelial cells impair bacteria-mediated maturation of dendritic cells and render T cells unresponsive to stimulation. Front Immunol. 2019;10:1434.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sanchez-Trincado JL, Pelaez-Prestel HF, Lafuente EM, Reche PA. Human oral epithelial cells suppress T cell function via prostaglandin E2 secretion. Front Immunol. 2021;12:740613.

    Article  CAS  PubMed  Google Scholar 

  35. Allam J-P, Peng W-M, Appel T, Wenghoefer M, Niederhagen B, Bieber T, et al. Toll-like receptor 4 ligation enforces tolerogenic properties of oral mucosal Langerhans cells. J Allergy Clin Immunol. 2008;121:368–e741.

    Article  CAS  PubMed  Google Scholar 

  36. Mascarell L, Lombardi V, Louise A, Saint-Lu N, Chabre H, Moussu H, et al. Oral dendritic cells mediate antigen-specific tolerance by stimulating TH1 and regulatory CD4 + T cells. J Allergy Clin Immunol. 2008;122:603–e95.

    Article  CAS  PubMed  Google Scholar 

  37. Iliev ID, Matteoli G, Rescigno M. The Yin and Yang of intestinal epithelial cells in controlling dendritic cell function. J Exp Med. 2007;204:2253–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, et al. A single-cell survey of the small intestinal epithelium. Nature. 2017;551:333–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Justiz Vaillant AA, Jamal Z, Patel P, Ramphul K. In: StatPearls, editor. Immunoglobulin. Treasure Island (FL): StatPearls Publishing; 2024.

    Google Scholar 

  40. Corthésy B. Multi-faceted functions of secretory IgA at mucosal surfaces. Front Immunol. 2013;4:185.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Woof JM, Russell MW. Structure and function relationships in IgA. Mucosal Immunol. 2011;4:590–7.

    Article  CAS  PubMed  Google Scholar 

  42. Bonner A, Almogren A, Furtado PB, Kerr MA, Perkins SJ. The nonplanar secretory IgA2 and near planar secretory IgA1 solution structures rationalize their different mucosal immune responses. J Biol Chem. 2009;284:5077–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Plomp R, de Haan N, Bondt A, Murli J, Dotz V, Wuhrer M. Comparative glycomics of Immunoglobulin A and G from saliva and plasma reveals biomarker potential. Front Immunol. 2018;9:2436.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Kumar Bharathkar S, Parker BW, Malyutin AG, Haloi N, Huey-Tubman KE, Tajkhorshid E, Stadtmueller BM. The structures of secretory and dimeric immunoglobulin A. Elife. 2020;9:e56098. https://doiorg.publicaciones.saludcastillayleon.es/10.7554/eLife.56098. PMID: 33107820; PMCID: PMC7707832.

  45. Bjerkan L, Schreurs O, Engen SA, Jahnsen FL, Baekkevold ES, Blix IJS, et al. The short form of TSLP is constitutively translated in human keratinocytes and has characteristics of an antimicrobial peptide. Mucosal Immunol. 2015;8:49–56.

    Article  CAS  PubMed  Google Scholar 

  46. Sonesson A, Kasetty G, Olin AI, Malmsten M, Mörgelin M, Sørensen OE, et al. Thymic stromal lymphopoietin exerts antimicrobial activities: TSLP exerts antimicrobial activities. Exp Dermatol. 2011;20:1004–10.

    Article  CAS  PubMed  Google Scholar 

  47. Mantis NJ, Rol N, Corthésy B. Secretory IgA’s complex roles in immunity and mucosal homeostasis in the gut. Mucosal Immunol. 2011;4:603–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Steffen U, Koeleman CA, Sokolova MV, Bang H, Kleyer A, Rech J, et al. IgA subclasses have different effector functions associated with distinct glycosylation profiles. Nat Commun. 2020;11:120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jang H, Choudhury S, Yu Y, Sievers BL, Gelbart T, Singh H, et al. Persistent immune and clotting dysfunction detected in saliva and blood plasma after COVID-19. Heliyon. 2023;9:e17958.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bladh O, Aguilera K, Marking U, Kihlgren M, Greilert Norin N, Smed-Sörensen A, et al. Comparison of SARS-CoV-2 spike-specific IgA and IgG in nasal secretions, saliva and serum. Front Immunol. 2024;15:1346749.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Al-Qahtani AA, Alhamlan FS, Al-Qahtani AA. Pro-inflammatory and anti-inflammatory interleukins in infectious diseases: A comprehensive review. Trop Med Infect Dis. 2024;9:13.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Lan Q, Zheng T, Rothman N, Zhang Y, Wang SS, Shen M, et al. Cytokine polymorphisms in the Th1/Th2 pathway and susceptibility to non-Hodgkin lymphoma. Blood. 2006;107:4101–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Zeng Y, Wang L, Zhou H, Qi Y. A meta-analysis of Th1 and Th2 cytokine profiles differentiating tuberculous from malignant pleural effusion. Sci Rep. 2022;12:2743.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev. 2003;8:223–46.

    PubMed  Google Scholar 

  55. Bartolini L, Piras E, Sullivan K, Gillen S, Bumbut A, Lin C-TM, et al. Detection of HHV-6 and EBV and cytokine levels in saliva from children with seizures: results of a multi-Center Cross-sectional study. Front Neurol. 2018;9:834.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Gornowicz A, Bielawska A, Bielawski K, Grabowska SZ, Wójcicka A, Zalewska M, et al. Pro-inflammatory cytokines in saliva of adolescents with dental caries disease. Ann Agric Environ Med. 2012;19:711–6.

    CAS  PubMed  Google Scholar 

  57. Santos MTBR, Diniz MB, Guaré RO, Ferreira MCD, Gutierrez GM, Gorjão R. Inflammatory markers in saliva as indicators of gingival inflammation in cerebral palsy children with and without cervical motor control. Int J Paediatr Dent. 2017;27:364–71.

    Article  PubMed  Google Scholar 

  58. Ramadan DE, Hariyani N, Indrawati R, Ridwan RD, Diyatri I. Cytokines and chemokines in periodontitis. Eur J Dent. 2020;14:483–95.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Roescher N, Tak PP, Illei GG. Cytokines in Sjögren’s syndrome. Oral Dis. 2009;15:519–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dong C, Flavell RA. Cell fate decision: T-helper 1 and 2 subsets in immune responses. Arthritis Res. 2000;2:179–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Diesch T, Filippi C, Fritschi N, Filippi A, Ritz N. Cytokines in saliva as biomarkers of oral and systemic oncological or infectious diseases: A systematic review. Cytokine. 2021;143:155506.

    Article  CAS  PubMed  Google Scholar 

  62. Santolia DD, Gupta IJAR. Mahajan. DB. Saliva as diagnostic tool - expression of gm-csf in oral potentially malignant disorders and oral squamous cell carcinoma patients. Int J Adv Res (Indore). 2016;4:2146–50.

    Article  CAS  Google Scholar 

  63. Malekzadeh H, Robati M, Yousefimanesh H, Ghafourian Boroujerdnia M, Nadripour R. Salivary interferon gamma and interleukin-4 levels in patients suffering from oral lichen planus. Cell J. 2015;17:554–8.

    PubMed  PubMed Central  Google Scholar 

  64. Idris A, Ghazali NB, Koh D. Interleukin 1β-A potential salivary biomarker for cancer progression? Biomark Cancer. 2015;7:25–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Simcić D, Pezelj-Ribarić S, Grzić R, Horvat J, Brumini G, Muhvić-Urek M. Detection of salivary Interleukin 2 and Interleukin 6 in patients with burning mouth syndrome. Mediators Inflamm. 2006;2006:54632.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Wadhwa S, Yoon AJ, Kister K, Bolin I, Chintalapudi N, Besmer A, et al. Detection of SARS-CoV-2 IgG antibodies and inflammatory cytokines in saliva-a pilot study. J Oral Biol Craniofac Res. 2023;13:267–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Rinderknecht C, Filippi C, Ritz N, Fritschi N, Simmen U, Filippi A, et al. Associations between salivary cytokines and oral health, age, and sex in healthy children. Sci Rep. 2022;12:15991.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Schwartz C. Salimetrics’ Growing List of Cytokines Which Can Be Reliably Tested in Saliva Now Includes IL-17A, IL-5, and IL-7 for Fast, Easy Salivary Biomarker Analysis. Salimetrics. 2020. https://salimetrics.com/cytokines-tested-in-saliva-il-17a-il-5-il-7/#:~:text=More%20specific%20to%20the%20salivary,also%20associated%20with%20lymphoid%20leukemias. Accessed 25 Oct 2024.

  69. Principe S, Zapater-Latorre E, Arribas L, Garcia-Miragall E, Bagan J. Salivary IL-8 as a putative predictive biomarker of radiotherapy response in head and neck cancer patients. Clin Oral Investig. 2022;26:437–48.

    Article  PubMed  Google Scholar 

  70. Gupta G. Gingival crevicular fluid as a periodontal diagnostic indicator- II: inflammatory mediators, host-response modifiers and chair side diagnostic aids. J Med Life. 2013;6:7–13.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. IL-12 P70. Quanterix. 2020. https://www.quanterix.com/simoa-assay-kits/il-12-p70/#:~:text=IL%2D12%20stimulates%20growth%20and,with%20autoimmune%20and%20inflammatory%20conditions. Accessed 25 Oct 2024.

  72. Belev B, Vičić I, Sedlić F, Prtorić M, Soče M, Prejac J, et al. Salivary interleukin-13 and transforming growth factor beta as potential biomarkers of cancer cachexia. Cancers (Basel). 2024;16:3035.

    Article  CAS  PubMed  Google Scholar 

  73. Sakai A, Sugawara Y, Kuroishi T, Sasano T, Sugawara S. Identification of IL-18 and Th17 cells in salivary glands of patients with Sjögren’s syndrome, and amplification of IL-17-mediated secretion of inflammatory cytokines from salivary gland cells by IL-18. J Immunol. 2008;181:2898–906.

    Article  CAS  PubMed  Google Scholar 

  74. Alamelu S, Rathinasamy K, Ulaganathan A, Ramamurthy S, Ganesan R, Saket P. Estimation of TNF-α levels in saliva and serum of patients with periodontal health and chronic periodontitis: A case-control study. J Contemp Dent Pract. 2020;21:148–51.

    Article  PubMed  Google Scholar 

  75. Ye J, Wang Y, Wang Z, Ji Q, Huang Y, Zeng T, et al. Circulating Th1, Th2, Th9, Th17, Th22, and Treg levels in aortic dissection patients. Mediators Inflamm. 2018;2018:5697149.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Goswami R, Kaplan MH. A brief history of IL-9. J Immunol. 2011;186:3283–8.

    Article  CAS  PubMed  Google Scholar 

  77. Parkin GM, Kim S, Mikhail A, Malhas R, McMillan L, Hollearn M, et al. Associations between saliva and plasma cytokines in cognitively normal, older adults. Aging Clin Exp Res. 2023;35:117–26.

    Article  PubMed  Google Scholar 

  78. Feng Y, Chen Z, Tu S-Q, Wei J-M, Hou Y-L, Kuang Z-L, et al. Role of interleukin-17A in the pathomechanisms of periodontitis and related systemic chronic inflammatory diseases. Front Immunol. 2022;13:862415.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Techatanawat S, Surarit R, Chairatvit K, Khovidhunkit W, Roytrakul S, Thanakun S, et al. Salivary and serum interleukin-17A and interleukin-18 levels in patients with type 2 diabetes mellitus with and without periodontitis. PLoS ONE. 2020;15:e0228921.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Selman AE, Görgülü NG, Doğan B. Salivary levels of IL-21 as a potential marker of stage III grade C periodontitis. Clin Exp Health Sci. 2021;11:878–83.

    Article  CAS  Google Scholar 

  81. Barone F, Nayar S, Campos J, Cloake T, Withers DR, Toellner K-M, et al. IL-22 regulates lymphoid chemokine production and assembly of tertiary lymphoid organs. Proc Natl Acad Sci USA. 2015;112:11024–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Kamil TF, Ali OH. Association between the cytokine IL-in saliva with periodontal health and disease. https://ejhm.journals.ekb.eg/article_294152_a94220193fecad96c75fd124eba30590.pdf. Accessed 25 Oct 2024.

  83. Andres-Martin F, James C, Catalfamo M. IL-27 expression regulation and its effects on adaptive immunity against viruses. Front Immunol. 2024;15:1395921.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: at the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta. 2014;1843:2563–82.

    Article  CAS  PubMed  Google Scholar 

  85. Ha Y-J, Choi YS, Kang EH, Chung J-H, Cha S, Song YW, et al. Increased expression of interferon-λ in minor salivary glands of patients with primary Sjögren’s syndrome and its synergic effect with interferon-α on salivary gland epithelial cells. Clin Exp Rheumatol. 2018;36(Suppl 112):31–40.

    PubMed  Google Scholar 

  86. Rodero MP, Decalf J, Bondet V, Hunt D, Rice GI, Werneke S, et al. Detection of interferon alpha protein reveals differential levels and cellular sources in disease. J Exp Med. 2017;214:1547–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Cavalli G, Colafrancesco S, Emmi G, Imazio M, Lopalco G, Maggio MC, et al. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and treatment of autoimmune and inflammatory diseases. Autoimmun Rev. 2021;20:102763.

    Article  CAS  PubMed  Google Scholar 

  88. Dubost JJ, Perrier S, Afane M, Viallard JL, Roux-Lombard P, Baudet-Pommel M, et al. IL-1 receptor antagonist in saliva; characterization in normal saliva and reduced concentration in Sjögren’s syndrome (SS). Clin Exp Immunol. 1996;106:237–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Belstrøm D, Damgaard C, Könönen E, Gürsoy M, Holmstrup P, Gürsoy UK. Salivary cytokine levels in early gingival inflammation. J Oral Microbiol. 2017;9:1364101.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Jin J-O, Shinohara Y, Yu Q. Innate immune signaling induces interleukin-7 production from salivary gland cells and accelerates the development of primary Sjögren’s syndrome in a mouse model. PLoS ONE. 2013;8:e77605.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Fehniger TA, Caligiuri MA. Interleukin 15: biology and relevance to human disease. Blood. 2001;97:14–32.

    Article  CAS  PubMed  Google Scholar 

  92. Shah SA, Ishinaga H, Hou B, Okano M, Takeuchi K. Effects of interleukin-31 on MUC5AC gene expression in nasal allergic inflammation. Pharmacology. 2013;91:158–64.

    Article  CAS  PubMed  Google Scholar 

  93. Borgia F, Custurone P, Li Pomi F, Cordiano R, Alessandrello C, Gangemi S. IL-31: state of the Art for an inflammation-oriented Interleukin. Int J Mol Sci. 2022;23:6507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Polz-Dacewicz M, Strycharz-Dudziak M, Dworzański J, Stec A, Kocot J. Salivary and serum IL-10, TNF-α, TGF-β, VEGF levels in oropharyngeal squamous cell carcinoma and correlation with HPV and EBV infections. Infect Agent Cancer. 2016;11:45.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Ling E, Robinson DS. Transforming growth factor-beta1: its anti-inflammatory and pro-fibrotic effects. Clin Exp Allergy. 2002;32:175–8.

    Article  CAS  PubMed  Google Scholar 

  96. Hughes CE, Nibbs RJB. A guide to chemokines and their receptors. FEBS J. 2018;285:2944–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Teixeira AL, Gama CS, Rocha NP, Teixeira MM. Revisiting the role of eotaxin-1/CCL11 in psychiatric disorders. Front Psychiatry. 2018;9:241.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Li J, Thornhill MH. Growth-regulated peptide-alpha (GRO-alpha) production by oral keratinocytes: a comparison with skin keratinocytes. Cytokine. 2000;12:1409–13.

    Article  CAS  PubMed  Google Scholar 

  99. Madhurantakam S, Lee ZJ, Naqvi A, Prasad S. Importance of IP-10 as a biomarker of host immune response: critical perspective as a target for biosensing. Curr Res Biotechnol. 2023;5:100130.

    Article  CAS  Google Scholar 

  100. Grande MA, Belstrøm D, Damgaard C, Holmstrup P, Könönen E, Gursoy M, et al. Salivary concentrations of macrophage activation-related chemokines are influenced by non-surgical periodontal treatment: a 12-week follow-up study. J Oral Microbiol. 2020;12:1694383.

    Article  CAS  PubMed  Google Scholar 

  101. Bhavsar I, Miller CS, Al-Sabbagh M. Macrophage inflammatory Protein-1 alpha (MIP-1 alpha)/CCL3: as a biomarker. Biomarkers in disease: methods, discoveries and applications. Dordrecht: Springer Netherlands; 2015. pp. 223–49.

    Google Scholar 

  102. Menten P, Wuyts A, Van Damme J. Macrophage inflammatory protein-1. Cytokine Growth Factor Rev. 2002;13:455–81.

    Article  CAS  PubMed  Google Scholar 

  103. Chuang J-Y, Yang W-H, Chen H-T, Huang C-Y, Tan T-W, Lin Y-T, et al. CCL5/CCR5 axis promotes the motility of human oral cancer cells. J Cell Physiol. 2009;220:418–26.

    Article  CAS  PubMed  Google Scholar 

  104. Sun J, Nemoto E, Hong G, Sasaki K. Modulation of stromal cell-derived factor 1 alpha (SDF-1α) and its receptor CXCR4 in Porphyromonas gingivalis-induced periodontal inflammation. BMC Oral Health. 2016;17:26.

    Article  PubMed Central  Google Scholar 

  105. Stone WL, Leavitt L, Varacallo M. In: StatPearls, editor. Physiology, growth factor. Treasure Island (FL): StatPearls Publishing; 2024.

    Google Scholar 

  106. Mandel AL, Ozdener H, Utermohlen V. Brain-derived neurotrophic factor in human saliva: ELISA optimization and biological correlates. J Immunoass Immunochem. 2011;32:18–30.

    Article  CAS  Google Scholar 

  107. Eckley CA, Rios L, da Rizzo S. Salivary Egf concentration in adults with reflux chronic laryngitis before and after treatment: preliminary results. Braz J Otorhinolaryngol. 2007;73:156–60.

    Article  PubMed  Google Scholar 

  108. Bryant EM, Richardson R, Graham BM. The relationship between salivary fibroblast growth Factor-2 and cortisol reactivity and psychological outcomes prior to and during the COVID-19 pandemic. J Affect Disord Rep. 2023;13:100606.

    PubMed  PubMed Central  Google Scholar 

  109. Wilczyńska-Borawska M, Borawski J, Bagińska J, Małyszko J, Myśliwiec M. Hepatocyte growth factor in saliva of patients with renal failure and periodontal disease. Ren Fail. 2012;34:942–51.

    Article  PubMed  Google Scholar 

  110. Schenck K, Schreurs O, Hayashi K, Helgeland K. The role of nerve growth factor (NGF) and its precursor forms in oral wound healing. Int J Mol Sci. 2017;18:386.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Yamamoto S, Fukumoto E, Yoshizaki K, Iwamoto T, Yamada A, Tanaka K, et al. Platelet-derived growth factor receptor regulates salivary gland morphogenesis via fibroblast growth factor expression. J Biol Chem. 2008;283:23139–49.

    Article  CAS  PubMed  Google Scholar 

  112. Alshamsi MHA, Koippallil Gopalakrishnan AR, Rahman B, Acharya AB. Evaluation of salivary placental growth factor in health and periodontitis. BMC Oral Health. 2024;24:493.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Ash A, Wilde PJ, Bradshaw DJ, King SP, Pratten JR. Structural modifications of the salivary conditioning film upon exposure to sodium bicarbonate: implications for oral lubrication and mouthfeel. Soft Matter. 2016;12:2794–801.

    Article  CAS  PubMed  Google Scholar 

  114. UniProt. https://www.uniprot.org/uniprotkb/P15692/entry#:~:text=Growth%20factor%20active%20in%20angiogenesis,induces%20permeabilization%20of%20blood%20vessels. Accessed 25 Oct 2024.

  115. Upile T, Jerjes W, Kafas P, Harini S, Singh SU, Guyer M, et al. Salivary VEGF: a non-invasive angiogenic and lymphangiogenic proxy in head and neck cancer prognostication. Int Arch Med. 2009;2:12.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Fujita G, Sato S, Kishino M, Iwai S-I, Nakazawa M, Toyosawa S, et al. Lymphatic vessels and related factors in adenoid cystic carcinoma of the salivary gland. Mod Pathol. 2011;24:885–91.

    Article  PubMed  Google Scholar 

  117. Pan W, Wang Q, Chen Q. The cytokine network involved in the host immune response to periodontitis. Int J Oral Sci. 2019;11:30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Berlutti F, Pilloni A, Pietropaoli M, Polimeni A, Valenti P. Lactoferrin and oral diseases: current status and perspective in periodontitis. Ann Stomatol (Roma). 2011;2:10–8.

    PubMed  Google Scholar 

  119. Wakabayashi H, Yamauchi K, Kobayashi T, Yaeshima T, Iwatsuki K, Yoshie H. Inhibitory effects of lactoferrin on growth and biofilm formation of Porphyromonas gingivalis and prevotella intermedia. Antimicrob Agents Chemother. 2009;53:3308–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Cao X, Ren Y, Lu Q, Wang K, Wu Y, Wang Y, et al. Lactoferrin: A glycoprotein that plays an active role in human health. Front Nutr. 2022;9:1018336.

    Article  PubMed  Google Scholar 

  121. Jenssen H, Hancock REW. Antimicrobial properties of lactoferrin. Biochimie. 2009;91:19–29.

    Article  CAS  PubMed  Google Scholar 

  122. Oram JD, Reiter B. Inhibition of bacteria by lactoferrin and other iron-chelating agents. Biochim Biophys Acta Gen Subj. 1968;170:351–65.

    Article  CAS  Google Scholar 

  123. Actor JK, Hwang S-A, Kruzel ML. Lactoferrin as a natural immune modulator. Curr Pharm Des. 2009;15:1956–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Bolat E, Eker F, Kaplan M, Duman H, Arslan A, Saritaş S, et al. Lactoferrin for COVID-19 prevention, treatment, and recovery. Front Nutr. 2022;9:992733.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Wakabayashi H, Oda H, Yamauchi K, Abe F. Lactoferrin for prevention of common viral infections. J Infect Chemother. 2014;20:666–71.

    Article  CAS  PubMed  Google Scholar 

  126. Frenkel ES, Ribbeck K. Salivary mucins in host defense and disease prevention. J Oral Microbiol. 2015;7:29759.

    Article  PubMed  Google Scholar 

  127. Murray PA, Prakobphol A, Lee T, Hoover CI, Fisher SJ. Adherence of oral Streptococci to salivary glycoproteins. Infect Immun. 1992;60:31–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Wei G-X, Campagna AN, Bobek LA. Factors affecting antimicrobial activity of MUC7 12-mer, a human salivary mucin-derived peptide. Ann Clin Microbiol Antimicrob. 2007;6:14.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Sheng YH, Hasnain SZ. Mucus and mucins: the underappreciated host defence system. Front Cell Infect Microbiol. 2022;12:856962.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Iontcheva I, Oppenheim FG, Troxler RF. Human salivary mucin MG1 selectively forms heterotypic complexes with amylase, proline-rich proteins, Statherin, and histatins. J Dent Res. 1997;76:734–43.

    Article  CAS  PubMed  Google Scholar 

  131. Deniz Tekin E, Calisir M. Investigation of human β-defensins 1, 2 and 3 in human saliva by molecular dynamics. Eur Phys J E Soft Matter. 2022;45:100.

    Article  CAS  PubMed  Google Scholar 

  132. Chaly YV, Paleolog EM, Kolesnikova TS, Tikhonov II, Petratchenko EV, Voitenok NN. Neutrophil alpha-defensin human neutrophil peptide modulates cytokine production in human monocytes and adhesion molecule expression in endothelial cells. Eur Cytokine Netw. 2000;11:257–66.

    CAS  PubMed  Google Scholar 

  133. Brook M, Tomlinson GH, Miles K, Smith RWP, Rossi AG, Hiemstra PS, et al. Neutrophil-derived alpha defensins control inflammation by inhibiting macrophage mRNA translation. Proc Natl Acad Sci U S A. 2016;113:4350–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Daher KA, Selsted ME, Lehrer RI. Direct inactivation of viruses by human granulocyte defensins. J Virol. 1986;60:1068–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Solanki SS, Singh P, Kashyap P, Sansi MS, Ali SA. Promising role of defensins peptides as therapeutics to combat against viral infection. Microb Pathog. 2021;155:104930.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Dale BA, Krisanaprakornkit S. Defensin antimicrobial peptides in the oral cavity: defensin antimicrobial peptides. J Oral Pathol Med. 2001;30:321–7.

    Article  CAS  PubMed  Google Scholar 

  137. Krishnakumari V, Rangaraj N, Nagaraj R. Antifungal activities of human beta-defensins HBD-1 to HBD-3 and their C-terminal analogs Phd1 to Phd3. Antimicrob Agents Chemother. 2009;53:256–60.

    Article  CAS  PubMed  Google Scholar 

  138. Mathews M, Jia HP, Guthmiller JM, Losh G, Graham S, Johnson GK, et al. Production of β-defensin antimicrobial peptides by the oral mucosa and salivary glands. Infect Immun. 1999;67:2740–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Hof W, van ’t, Oudhoff MJ, Veerman ECI, Histatins. Multifunctional salivary antimicrobial peptides. Antimicrobial peptides and innate immunity. Basel: Springer Basel; 2013. pp. 167–81.

    Chapter  Google Scholar 

  140. Puri S, Edgerton M. How does it kill? Understanding the candidacidal mechanism of salivary Histatin 5. Eukaryot Cell. 2014;13:958–64.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Nikawa H, Jin C, Makihira S, Hamada T, Samaranayake LP. Susceptibility of Candida albicans isolates from the oral cavities of HIV-positive patients to histatin-5. J Prosthet Dent. 2002;88:263–7.

    Article  CAS  PubMed  Google Scholar 

  142. Oudhoff MJ, Kroeze KL, Nazmi K, van den Keijbus PAM, van ’t Hof W, Fernandez-Borja M, et al. Structure-activity analysis of Histatin, a potent wound healing peptide from human saliva: cyclization of Histatin potentiates molar activity 1,000-fold. FASEB J. 2009;23:3928–35.

    Article  CAS  PubMed  Google Scholar 

  143. Shah D, Son K-N, Kalmodia S, Lee B-S, Ali M, Balasubramaniam A, et al. Wound healing properties of histatin-5 and identification of a functional domain required for histatin-5-induced cell migration. Mol Ther Methods Clin Dev. 2020;17:709–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Jurczak A, Kościelniak D, Papież M, Vyhouskaya P, Krzyściak W. A study on β-defensin-2 and histatin-5 as a diagnostic marker of early childhood caries progression. Biol Res. 2015;48:61.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Golshani S, Vatanara A, Balalaie S, Kadkhoda Z, Abdollahi M, Amin M. Development of a novel histatin-5 mucoadhesive gel for the treatment of oral mucositis: in vitro characterization and in vivo evaluation. AAPS PharmSciTech. 2023;24:177.

    Article  CAS  PubMed  Google Scholar 

  146. Yeh CK, Dodds MW, Zuo P, Johnson DA. A population-based study of salivary lysozyme concentrations and candidal counts. Arch Oral Biol. 1997;42:25–31.

    Article  CAS  PubMed  Google Scholar 

  147. Lelouard H, Henri S, De Bovis B, Mugnier B, Chollat-Namy A, Malissen B, et al. Pathogenic bacteria and dead cells are internalized by a unique subset of Peyer’s patch dendritic cells that express lysozyme. Gastroenterology. 2010;138:173–84.

    Article  CAS  PubMed  Google Scholar 

  148. Lollike K, Kjeldsen L, Sengeløv H, Borregaard N. Lysozyme in human neutrophils and plasma. A parameter of myelopoietic activity. Leukemia. 1995;9:159–64.

    CAS  PubMed  Google Scholar 

  149. Tonguc Altin K, Topcuoglu N, Duman G, Unsal M, Celik A, Selvi Kuvvetli S, et al. Antibacterial effects of saliva substitutes containing lysozyme or lactoferrin against Streptococcus mutans. Arch Oral Biol. 2021;129:105183.

    Article  CAS  PubMed  Google Scholar 

  150. Kawai Y, Mickiewicz K, Errington J. Lysozyme counteracts β-lactam antibiotics by promoting the emergence of L-form bacteria. Cell. 2018;172:1038–e4910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Song Y, Zhang H, Zhu Y, Zhao X, Lei Y, Zhou W, et al. Lysozyme protects against severe acute respiratory syndrome coronavirus 2 infection and inflammation in human corneal epithelial cells. Invest Ophthalmol Vis Sci. 2022;63:16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Qvarnstrom M, Janket S, Jones JA, Nuutinen P, Baird AE, Nunn ME, et al. Salivary lysozyme and prevalent hypertension. J Dent Res. 2008;87:480–4.

    Article  CAS  PubMed  Google Scholar 

  153. Moslemi M, Sattari M, Kooshki F, Fotuhi F, Modarresi N, Khalili Sadrabad Z, et al. Relationship of salivary lactoferrin and lysozyme concentrations with early childhood caries. J Dent Res Dent Clin Dent Prospects. 2015;9:109–14.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Sun H, Chen Y, Zou X, Li Q, Li H, Shu Y, et al. Salivary secretory Immunoglobulin (SIgA) and lysozyme in malignant tumor patients. Biomed Res Int. 2016;2016:8701423.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Surna A, Kubilius R, Sakalauskiene J, Vitkauskiene A, Jonaitis J, Saferis V, et al. Lysozyme and microbiota in relation to gingivitis and periodontitis. Med Sci Monit. 2009;15:CR66–73.

    CAS  PubMed  Google Scholar 

  156. Said HS, Suda W, Nakagome S, Chinen H, Oshima K, Kim S, et al. Dysbiosis of salivary microbiota in inflammatory bowel disease and its association with oral immunological biomarkers. DNA Res. 2014;21:15–25.

    Article  CAS  PubMed  Google Scholar 

  157. Barros SP, Williams R, Offenbacher S, Morelli T. Gingival crevicular fluid as a source of biomarkers for periodontitis. Periodontol 2000. 2016;70:53–64.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Stepaniuk K, Hinrichs JE. The structure and function of the periodontium. Veterinary periodontology. Wiley; 2013. p. 1–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/9781118705018.ch1.

  159. Subrahmanyam MV, Sangeetha M. Gingival crevicular fluid a marker of the periodontal disease activity. Indian J Clin Biochem. 2003;18:5–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Gouveia F, Gomes S, Validation of an alternative absorbent paper for. collecting gingival crevicular fluid Validação de um papel absorvente para coleta de Fluido Crevicular Gengival. 2009.

  161. Fatima T, Khurshid Z, Rehman A, Imran E, Srivastava KC, Shrivastava D. Gingival crevicular fluid (GCF): A diagnostic tool for the detection of periodontal health and diseases. Molecules. 2021;26:1208.

    Article  CAS  PubMed  Google Scholar 

  162. Fiorellini JPSPG. Anatomy of the periodontium. In: Newman MG, Takei HH, Klokkevold PR, Carranza FA, editors. Carranza’s clinical periodontology. Philadelphia, PA: Saunders; 2015. pp. 9–39.

    Google Scholar 

  163. Passanezi E, Sant’Ana ACP. The periodontium. In: Sant’Ana ACP, Passanezi E, editors. Periodontics– The essential for the clinical practice. Barueri, Brazil: Manole; 2023. pp. 1–26.

    Google Scholar 

  164. Bosshardt DD, Lindhe J, Lang NP, Araújo M. Anatomy and histology of periodontal tissues. In: Berglundh T, Giannobile WV, Lang NP, Sanz M, editors. Lindhe’s clinical periodontology and implant dentistry. New York, NY: John Wiley and Sons (WIE); 2022. pp. 3–49.

    Google Scholar 

  165. Schroeder HEPRG. The normal periodontium. In: Schluger S, Yuodelis R, Page RC, Johnson RH, editors. Periodontal disease. Basic phenomena, clinical management, and occlusal and restorative interrelationships. Philadelphia, PA: Lea & Febiger; 1990. pp. 3–52.

    Google Scholar 

  166. Groeger S, Meyle J. Oral mucosal epithelial cells. Front Immunol. 2019;10:208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Hajishengallis G. Immunomicrobial pathogenesis of periodontitis: keystones, pathobionts, and host response. Trends Immunol. 2014;35:3–11.

    Article  CAS  PubMed  Google Scholar 

  168. Hintermann E, Haake SK, Christen U, Sharabi A, Quaranta V. Discrete proteolysis of focal contact and adherens junction components in Porphyromonas gingivalis-infected oral keratinocytes: a strategy for cell adhesion and migration disabling. Infect Immun. 2002;70:5846–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Lamont RJ, Yilmaz O. In or out: the invasiveness of oral bacteria: invasiveness of oral bacteria. Periodontol 2000. 2002;30:61–9.

    Article  PubMed  Google Scholar 

  170. Akira S, Sato S. Toll-like receptors and their signaling mechanisms. Scand J Infect Dis. 2003;35:555–62.

    Article  CAS  PubMed  Google Scholar 

  171. Cario E, Brown D, McKee M, Lynch-Devaney K, Gerken G, Podolsky DK. Commensal-associated molecular patterns induce selective toll-like receptor-trafficking from apical membrane to cytoplasmic compartments in polarized intestinal epithelium. Am J Pathol. 2002;160:165–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Chiba N, Tada R, Ohnishi T, Matsuguchi T. TLR4/7-mediated host-defense responses of gingival epithelial cells. J Cell Biochem. 2024;125:e30576.

    Article  CAS  PubMed  Google Scholar 

  173. Sugawara Y, Uehara A, Fujimoto Y, Kusumoto S, Fukase K, Shibata K, et al. Toll-like receptors, NOD1, and NOD2 in oral epithelial cells. J Dent Res. 2006;85:524–9.

    Article  CAS  PubMed  Google Scholar 

  174. Uehara A, Sugawara S, Tamai R, Takada H. Contrasting responses of human gingival and colonic epithelial cells to lipopolysaccharides, Lipoteichoic acids and peptidoglycans in the presence of soluble CD14. Med Microbiol Immunol. 2001;189:185–92.

    Article  CAS  PubMed  Google Scholar 

  175. Uehara A, Fujimoto Y, Fukase K, Takada H. Various human epithelial cells express functional Toll-like receptors, NOD1 and NOD2 to produce anti-microbial peptides, but not Proinflammatory cytokines. Mol Immunol. 2007;44:3100–11.

    Article  CAS  PubMed  Google Scholar 

  176. Ren L, Leung WK, Darveau RP, Jin L. The expression profile of lipopolysaccharide-binding protein, membrane-bound CD14, and toll-like receptors 2 and 4 in chronic periodontitis. J Periodontol. 2005;76:1950–9.

    Article  CAS  PubMed  Google Scholar 

  177. Muthukuru M, Jotwani R, Cutler CW. Oral mucosal endotoxin tolerance induction in chronic periodontitis. Infect Immun. 2005;73:687–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Mori Y, Yoshimura A, Ukai T, Lien E, Espevik T, Hara Y. Immunohistochemical localization of Toll-like receptors 2 and 4 in gingival tissue from patients with periodontitis: Toll-like receptors in human periodontitis. Oral Microbiol Immunol. 2003;18:54–8.

    Article  CAS  PubMed  Google Scholar 

  179. Beklen A, Hukkanen M, Richardson R, Konttinen YT. Immunohistochemical localization of Toll-like receptors 1–10 in periodontitis. Oral Microbiol Immunol. 2008;23:425–31.

    Article  CAS  PubMed  Google Scholar 

  180. Harder J, Bartels J, Christophers E, Schröder JM. A peptide antibiotic from human skin. Nature. 1997;387:861.

    Article  CAS  PubMed  Google Scholar 

  181. Harder J, Bartels J, Christophers E, Schroder JM. Isolation and characterization of human beta -defensin-3, a novel human inducible peptide antibiotic. J Biol Chem. 2001;276:5707–13.

    Article  CAS  PubMed  Google Scholar 

  182. Ganz T. Extracellular release of antimicrobial defensins by human polymorphonuclear leukocytes. Infect Immun. 1987;55:568–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. García JR, Krause A, Schulz S, Rodríguez-Jiménez FJ, Klüver E, Adermann K, et al. Human beta-defensin 4: a novel inducible peptide with a specific salt-sensitive spectrum of antimicrobial activity. FASEB J. 2001;15:1819–21.

    Article  PubMed  Google Scholar 

  184. Lu Q, Jin L, Darveau RP, Samaranayake LP. Expression of human beta-defensins-1 and– 2 peptides in unresolved chronic periodontitis. J Periodontal Res. 2004;39:221–7.

    Article  CAS  PubMed  Google Scholar 

  185. Zhao C, Wang I, Lehrer RI. Widespread expression of beta-defensin hBD-1 in human secretory glands and epithelial cells. FEBS Lett. 1996;396:319–22.

    Article  CAS  PubMed  Google Scholar 

  186. Diamond DL, Kimball JR, Krisanaprakornkit S, Ganz T, Dale BA. Detection of beta-defensins secreted by human oral epithelial cells. J Immunol Methods. 2001;256:65–76.

    Article  CAS  PubMed  Google Scholar 

  187. Lu Q, Samaranayake LP, Darveau RP, Jin L. Expression of human beta-defensin-3 in gingival epithelia. J Periodontal Res. 2005;40:474–81.

    Article  CAS  PubMed  Google Scholar 

  188. Hiyoshi T, Domon H, Maekawa T, Tamura H, Isono T, Hirayama S, et al. Neutrophil elastase aggravates periodontitis by disrupting gingival epithelial barrier via cleaving cell adhesion molecules. Sci Rep. 2022;12:8159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Tonetti MS, Greenwell H, Kornman KS. Staging and grading of periodontitis: framework and proposal of a new classification and case definition. J Periodontol. 2018;89(Suppl 1):S159–72.

    PubMed  Google Scholar 

  190. Darveau RP. Periodontitis: a polymicrobial disruption of host homeostasis. Nat Rev Microbiol. 2010;8:481–90.

    Article  CAS  PubMed  Google Scholar 

  191. Benzian H, Watt R, Makino Y, Stauf N, Varenne B. WHO calls to end the global crisis of oral health. Lancet. 2022;400:1909–10.

    Article  PubMed  Google Scholar 

  192. Beck JD, Papapanou PN, Philips KH, Offenbacher S. Periodontal medicine: 100 years of progress. J Dent Res. 2019;98:1053–62.

    Article  CAS  PubMed  Google Scholar 

  193. Hajishengallis G, Lamont RJ. Beyond the red complex and into more complexity: the polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology. Mol Oral Microbiol. 2012;27:409–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Abusleme L, Dupuy AK, Dutzan N, Silva N, Burleson JA, Strausbaugh LD, et al. The subgingival Microbiome in health and periodontitis and its relationship with community biomass and inflammation. ISME J. 2013;7:1016–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Yue Y, Liu Q, Xu C, Loo WTY, Wang M, Wen G, et al. Comparative evaluation of cytokines in gingival crevicular fluid and saliva of patients with aggressive periodontitis. Int J Biol Markers. 2013;28:108–12.

    Article  CAS  PubMed  Google Scholar 

  196. Damgaard C, Danielsen AK, Enevold C, Massarenti L, Nielsen CH, Holmstrup P, et al. Porphyromonas gingivalis in saliva associates with chronic and aggressive periodontitis. J Oral Microbiol. 2019;11:1653123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Page RC, Schroeder HE. Pathogenesis of inflammatory periodontal disease. A summary of current work. Lab Invest. 1976;34:235–49.

    CAS  PubMed  Google Scholar 

  198. Seymour GJ, Berglundh T, Trombelli L. Pathogenesis of gingivitis and periodontitis. In: Berglundh T, Giannobile WV, Lang NP, Sanz M, editors. Lindhe’s clinical periodontology and implant dentistry. Wiley Blackwell; 2022. pp. 235–62.

    Google Scholar 

  199. Offenbacher S, Katz V, Fertik G, Collins J, Boyd D, Maynor G, et al. Periodontal infection as a possible risk factor for preterm low birth weight. J Periodontol. 1996;67(10 Suppl):1103–13.

    CAS  PubMed  Google Scholar 

  200. Nussbaum G, Shapira L. How has neutrophil research improved our Understanding of periodontal pathogenesis? Neutrophils and periodontal pathogenesis. J Clin Periodontol. 2011;38(Suppl 11):49–59.

    Article  PubMed  Google Scholar 

  201. Chakravarti A, Raquil M-A, Tessier P, Poubelle PE. Surface RANKL of Toll-like receptor 4-stimulated human neutrophils activates osteoclastic bone resorption. Blood. 2009;114:1633–44.

    Article  CAS  PubMed  Google Scholar 

  202. Pelletier M, Maggi L, Micheletti A, Lazzeri E, Tamassia N, Costantini C, et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood. 2010;115:335–43.

    Article  CAS  PubMed  Google Scholar 

  203. Gaffen SL, Hajishengallis G. A new inflammatory cytokine on the block: re-thinking periodontal disease and the Th1/Th2 paradigm in the context of Th17 cells and IL-17. J Dent Res. 2008;87:817–28.

    Article  CAS  PubMed  Google Scholar 

  204. Gemmell E, Yamazaki K, Seymour GJ. The role of T cells in periodontal disease: homeostasis and autoimmunity. Periodontol 2000. 2007;43:14–40.

    Article  PubMed  Google Scholar 

  205. Pires JR, Nogueira MRS, Nunes AJF, Degand DRF, Pessoa LC, Damante CA, et al. Deposition of immune complexes in gingival tissues in the presence of periodontitis and systemic lupus erythematosus. Front Immunol. 2021;12:591236.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Bosshardt DD. The periodontal pocket: pathogenesis, histopathology and consequences. Periodontol. 2000;2018(76):43–50.

    Google Scholar 

  207. Tada H, Sugawara S, Nemoto E, Imamura T, Potempa J, Travis J, et al. Proteolysis of ICAM-1 on human oral epithelial cells by gingipains. J Dent Res. 2003;82:796–801.

    Article  CAS  PubMed  Google Scholar 

  208. Tonetti MS, Imboden MA, Lang NP. Neutrophil migration into the gingival sulcus is associated with transepithelial gradients of interleukin-8 and ICAM-1. J Periodontol. 1998;69:1139–47.

    Article  CAS  PubMed  Google Scholar 

  209. Ren L, Jin L, Leung WK. Local expression of lipopolysaccharide-binding protein in human gingival tissues. J Periodontal Res. 2004;39:242–8.

    Article  CAS  PubMed  Google Scholar 

  210. Li X, Liu Y, Yang X, Li C, Song Z. The oral microbiota: community composition, influencing factors, pathogenesis, and interventions. Front Microbiol. 2022;13. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2022.895537.

  211. Radaic A, Kapila YL. The oralome and its dysbiosis: new insights into oral microbiome-host interactions. Comput Struct Biotechnol J. 2021;19:1335–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Hajishengallis G. Periodontitis: from microbial immune subversion to systemic inflammation. Nat Rev Immunol. 2015;15:30–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Marsh PD, Do T, Beighton D, Devine DA. Influence of saliva on the oral microbiota. Periodontol 2000. 2016;70:80–92.

    Article  PubMed  Google Scholar 

  214. Pessoa L, Aleti G, Choudhury S, Nguyen D, Yaskell T, Zhang Y, et al. Host-microbial interactions in systemic lupus erythematosus and periodontitis. Front Immunol. 2019;10:2602.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Slots J. Herpesviral-bacterial synergy in the pathogenesis of human periodontitis. Curr Opin Infect Dis. 2007;20:278–83.

    Article  PubMed  Google Scholar 

  216. Thakkar P, Banks JM, Rahat R, Brandini DA, Naqvi AR. Viruses of the oral cavity: prevalence, pathobiology and association with oral diseases. Rev Med Virol. 2022;32:e2311.

    Article  CAS  PubMed  Google Scholar 

  217. Chow EW, Pang LM, Wang Y. The impact of the host microbiota on Candida albicans infection. Curr Opin Microbiol. 2024;80:102507.

    Article  CAS  PubMed  Google Scholar 

  218. Conti HR, Gaffen SL. IL-17-mediated immunity to the opportunistic fungal pathogen Candida albicans. J Immunol. 2015;195:780–8.

    Article  CAS  PubMed  Google Scholar 

  219. Conti HR, Shen F, Nayyar N, Stocum E, Sun JN, Lindemann MJ, et al. Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J Exp Med. 2009;206:299–311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Gonzales JR, Groeger S, Johansson A, Meyle J. T helper cells from aggressive periodontitis patients produce higher levels of interleukin-1 beta and interleukin-6 in interaction with Porphyromonas gingivalis. Clin Oral Investig. 2014;18:1835–43.

    Article  PubMed  Google Scholar 

  221. Nørskov-Lauritsen N, Claesson R, Birkeholm Jensen A, Åberg CH, Haubek D. Aggregatibacter actinomycetemcomitans: clinical significance of a pathobiont subjected to ample changes in classification and nomenclature. Pathogens. 2019;8:243.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Han YW. Fusobacterium nucleatum: a commensal-turned pathogen. Curr Opin Microbiol. 2015;23:141–7.

    Article  CAS  PubMed  Google Scholar 

  223. Zhang S, Zhao Y, Lalsiamthara J, Peng Y, Qi L, Deng S, et al. Current research progress on prevotella intermedia and associated diseases. Crit Rev Microbiol. 2024:1–18. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/1040841X.2024.2390594.

  224. Sharma A. Virulence mechanisms of Tannerella forsythia: virulence mechanisms of Tannerella forsythia. Periodontol 2000. 2010;54:106–16.

    Article  PubMed  PubMed Central  Google Scholar 

  225. Goetting-Minesky MP, Godovikova V, Fenno JC. Approaches to Understanding mechanisms of dentilisin protease complex expression in Treponema denticola. Front Cell Infect Microbiol. 2021;11:668287.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Bandara HMHN, Samaranayake LP. Viral, bacterial, and fungal infections of the oral mucosa: types, incidence, predisposing factors, diagnostic algorithms, and management. Periodontol 2000. 2019;80:148–76.

    Article  CAS  PubMed  Google Scholar 

  227. Tonoyan L, Vincent-Bugnas S, Olivieri C-V, Doglio A. New viral facets in oral diseases: the EBV paradox. Int J Mol Sci. 2019;20:5861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Contreras A, Botero JE, Slots J. Biology and pathogenesis of cytomegalovirus in periodontal disease: biology and pathogenesis of cytomegalovirus in periodontal disease. Periodontol 2000. 2014;64:40–56.

    Article  PubMed  Google Scholar 

  229. Griffiths P, Reeves M. Pathogenesis of human cytomegalovirus in the immunocompromised host. Nat Rev Microbiol. 2021;19:759–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Li Y, Saxena D, Chen Z, Liu G, Abrams WR, Phelan JA, et al. HIV infection and microbial diversity in saliva. J Clin Microbiol. 2014;52:1400–11.

    Article  PubMed  PubMed Central  Google Scholar 

  231. Pólvora TLS, Nobre ÁVV, Tirapelli C, Taba M Jr, de Macedo LD, Santana RC, et al. Relationship between human immunodeficiency virus (HIV-1) infection and chronic periodontitis. Expert Rev Clin Immunol. 2018;14:315–27.

    Article  PubMed  Google Scholar 

  232. Williams D, Lewis M. Pathogenesis and treatment of oral candidosis. J Oral Microbiol. 2011;3:5771.

    Article  Google Scholar 

  233. Mysak J, Podzimek S, Sommerova P, Lyuya-Mi Y, Bartova J, Janatova T, et al. Porphyromonas gingivalis: major periodontopathic pathogen overview. J Immunol Res. 2014;2014:476068.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Malinowski B, Węsierska A, Zalewska K, Sokołowska MM, Bursiewicz W, Socha M, et al. The role of Tannerella forsythia and Porphyromonas gingivalis in pathogenesis of esophageal cancer. Infect Agent Cancer. 2019;14:3.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Kim HS, Kim CG, Kim WK, Kim K-A, Yoo J, Min BS, et al. Fusobacterium nucleatum induces a tumor microenvironment with diminished adaptive immunity against colorectal cancers. Front Cell Infect Microbiol. 2023;13:1101291.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Belibasakis GN, Belstrøm D, Eick S, Gursoy UK, Johansson A, Könönen E. Periodontal microbiology and microbial etiology of periodontal diseases: historical concepts and contemporary perspectives. Periodontol 2000. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/prd.12473.

    Article  PubMed  Google Scholar 

  237. Jansen HJ, Grenier D, Van der Hoeven JS. Characterization of Immunoglobulin G-degrading proteases of prevotella intermedia and prevotella nigrescens. Oral Microbiol Immunol. 1995;10:138–45.

    Article  CAS  PubMed  Google Scholar 

  238. Tonoyan L, Mounier C, Fassy J, Leymarie S, Mouraret S, Monneyron P, et al. Unveiling the etiopathogenic role of Epstein-Barr virus in periodontitis. J Dent Res. 2025:220345241303138. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/00220345241303138.

  239. Patel M. Oral cavity and Candida albicans: colonisation to the development of infection. Pathogens. 2022;11:335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Macias-Paz IU, Pérez-Hernández S, Tavera-Tapia A, Luna-Arias JP, Guerra-Cárdenas JE. Reyna-Beltrán E. Candida albicans the main opportunistic pathogenic fungus in humans. Rev Argent Microbiol. 2023;55:189–98.

    PubMed  Google Scholar 

  241. Mealey BL, Oates TW, American Academy of Periodontology. Diabetes mellitus and periodontal diseases. J Periodontol. 2006;77:1289–303.

    Article  CAS  PubMed  Google Scholar 

  242. Creely SJ, McTernan PG, Kusminski CM, Fisher ff M, Da Silva NF, Khanolkar M, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. Am J Physiol Endocrinol Metab. 2007;292:E740–7.

    Article  CAS  PubMed  Google Scholar 

  243. Mohammad MK, Morran M, Slotterbeck B, Leaman DW, Sun Y, von Grafenstein H, et al. Dysregulated Toll-like receptor expression and signaling in bone marrow-derived macrophages at the onset of diabetes in the non-obese diabetic mouse. Int Immunol. 2006;18:1101–13.

    Article  CAS  PubMed  Google Scholar 

  244. Promsudthi A, Poomsawat S, Limsricharoen W. The role of T oll-like receptor 2 and 4 in gingival tissues of chronic periodontitis subjects with type 2 diabetes. J Periodontal Res. 2014;49:346–54.

    Article  CAS  PubMed  Google Scholar 

  245. Abdul Rehman S, Khurshid Z, Hussain Niazi F, Naseem M, Al Waddani H, Sahibzada HA, et al. Role of salivary biomarkers in detection of cardiovascular diseases (CVD). Proteomes. 2017;5:21.

    Article  PubMed  PubMed Central  Google Scholar 

  246. Pay JB, Shaw AM. Towards salivary C-reactive protein as a viable biomarker of systemic inflammation. Clin Biochem. 2019;68:1–8.

    Article  CAS  PubMed  Google Scholar 

  247. Naidoo T, Konkol K, Biccard B, Dudose K, McKune AJ. Elevated salivary C-reactive protein predicted by low cardio-respiratory fitness and being overweight in African children. Cardiovasc J Afr. 2012;23:501–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  248. Dongiovanni P, Meroni M, Casati S, Goldoni R, Thomaz DV, Kehr NS, et al. Salivary biomarkers: novel noninvasive tools to diagnose chronic inflammation. Int J Oral Sci. 2023;15:1–12.

    Article  Google Scholar 

  249. Xiao H, Zhang Y, Kim Y, Kim S, Kim JJ, Kim KM, et al. Differential proteomic analysis of human saliva using tandem mass tags quantification for gastric cancer detection. Sci Rep. 2016;6:22165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. Franzmann EJ, Qi Y, Peifer S, Messer K, Messing B, Blanco RG, et al. Salivary CD44 and total protein levels to detect risk for oral and oropharyngeal cancer recurrence: A nonrandomized clinical trial: A nonrandomized clinical trial. JAMA Otolaryngol Head Neck Surg. 2024;150:843–50.

    Article  PubMed  Google Scholar 

  251. Giri K, Maity S, Ambatipudi K. Targeted proteomics using parallel reaction monitoring confirms salivary proteins indicative of metastatic triple-negative breast cancer. J Proteom. 2022;267:104701.

    Article  CAS  Google Scholar 

  252. Chu H-W, Chang K-P, Hsu C-W, Chang IY-F, Liu H-P, Chen Y-T, et al. Identification of salivary biomarkers for oral cancer detection with untargeted and targeted quantitative proteomics approaches. Mol Cell Proteom. 2019;18:1796–806.

    Article  CAS  Google Scholar 

  253. Lohavanichbutr P, Zhang Y, Wang P, Gu H, Nagana Gowda GA, Djukovic D, et al. Salivary metabolite profiling distinguishes patients with oral cavity squamous cell carcinoma from normal controls. PLoS ONE. 2018;13:e0204249.

    Article  PubMed  PubMed Central  Google Scholar 

  254. Li K, Lin Y, Zhou Y, Xiong X, Wang L, Li J, et al. Salivary extracellular MicroRNAs for early detection and prognostication of esophageal cancer: A clinical study. Gastroenterology. 2023;165:932–e459.

    Article  CAS  PubMed  Google Scholar 

  255. Ding Y, Ma Q, Liu F, Zhao L, Wei W. The potential use of salivary MiRNAs as promising biomarkers for detection of cancer: A meta-analysis. PLoS ONE. 2016;11:e0166303.

    Article  PubMed  PubMed Central  Google Scholar 

  256. Balakittnen J, Ekanayake Weeramange C, Wallace DF, Duijf PHG, Cristino AS, Hartel G, et al. A novel saliva-based MiRNA profile to diagnose and predict oral cancer. Int J Oral Sci. 2024;16:14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  257. Jasim H, Olausson P, Hedenberg-Magnusson B, Ernberg M, Ghafouri B. The proteomic profile of whole and glandular saliva in healthy pain-free subjects. Sci Rep. 2016;6:39073.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. Justino AB, Teixeira RR, Peixoto LG, Jaramillo OLB, Espindola FS. Effect of saliva collection methods and oral hygiene on salivary biomarkers. Scand J Clin Lab Invest. 2017;77:415–22.

    Article  CAS  PubMed  Google Scholar 

  259. Louro T, Simões C, Lima W, Carreira L, Castelo PM, Luis H, et al. Salivary protein profile and food intake: A dietary pattern analysis. J Nutr Metab. 2021;2021:6629951.

    Article  PubMed  PubMed Central  Google Scholar 

  260. Ventura TM, da Ribeiro S, Dionizio NR, Sabino AS, Buzalaf IT. Standardization of a protocol for shotgun proteomic analysis of saliva. J Appl Oral Sci. 2018;26:e20170561.

    Article  PubMed  PubMed Central  Google Scholar 

  261. Mann M, Kumar C, Zeng W-F, Strauss MT. Artificial intelligence for proteomics and biomarker discovery. Cell Syst. 2021;12:759–70.

    Article  CAS  PubMed  Google Scholar 

  262. Van Dyke TE, van Winkelhoff AJ. Infection and inflammatory mechanisms. J Clin Periodontol. 2013;40(Suppl 14):S1–7.

    PubMed  Google Scholar 

  263. Freire M, Nelson KE, Edlund A. The oral host-microbial interactome: an ecological chronometer of health? Trends Microbiol. 2021;29:551–61.

    Article  CAS  PubMed  Google Scholar 

  264. McCarthy EM, Smith S, Lee RZ, Cunnane G, Doran MF, Donnelly S, et al. The association of cytokines with disease activity and damage scores in systemic lupus erythematosus patients. Rheumatology (Oxford). 2014;53:1586–94.

    Article  CAS  PubMed  Google Scholar 

  265. Figueredo CMS, Areas A, Sztajnbok FR, Miceli V, Miranda LA, Fischer RG, et al. Higher elastase activity associated with lower IL-18 in GCF from juvenile systemic lupus patients. Oral Health Prev Dent. 2008;6:75–81.

    PubMed  Google Scholar 

  266. Romero-Ramírez S, Sosa-Hernández VA, Cervantes-Díaz R, Carrillo-Vázquez DA, Meza-Sánchez DE, Núñez-Álvarez C, et al. Salivary IgA subtypes as novel disease biomarkers in systemic lupus erythematosus. Front Immunol. 2023;14:1080154.

    Article  PubMed  PubMed Central  Google Scholar 

  267. Zian Z, Bouhoudan A, Mourabit N, Azizi G, Bennani Mechita M. Salivary cytokines as potential diagnostic biomarkers for systemic lupus erythematosus disease. Mediators Inflamm. 2021;2021:8847557.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Dr. Yanbao Yu for productive discussions on saliva biofluid proteomics. This work was completed at the J. Craig Venter Institute.

Funding

This work was supported by U.S. Public Health Service Grants from the National Institute of Dental and Craniofacial Research grant R21DE029625 (MF), Conrad Prebys, and Polybio Foundation grant #23-039 (MF). The funding bodies had no role in the study design, data interpretation, or manuscript preparation of this review article.

Author information

Authors and Affiliations

Authors

Contributions

Writing of the original draft: MF, MM, SS; Writing and editing: MF, MM, SS, AP, JP. Patient recruitment: not applicable. Prepared Graphs: SS and MM; Assisted with sample processing: not applicable; Assisted with sample analysis: not applicable. Funding Acquisition: MF. All authors reviewed the manuscript.

Corresponding author

Correspondence to Marcelo Freire.

Ethics declarations

Ethics approval and consent to participate

No subject’s evaluation participated in this review manuscript.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Matsuoka, M., Soria, S.A., Pires, J.R. et al. Natural and induced immune responses in oral cavity and saliva. BMC Immunol 26, 34 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12865-025-00713-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12865-025-00713-8

Keywords