Next Article in Journal
Characterization of Terpenoids in Aromatic Plants Using Raman Spectroscopy and Gas Chromatography–Mass Spectrometry (GC–MS)
Previous Article in Journal
Diosmin or Hesperidin? Comparison of Antioxidative Action of Two Venoactive Flavonoids in Type 1 Diabetic Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Crude Plant Extracts and Their Anti-Inflammatory Potential in Oral Inflammatory Cell Models: A Systematic Review of In Vitro Studies

1
Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria
2
Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
3
Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(23), 11253; https://doi.org/10.3390/ijms262311253
Submission received: 20 October 2025 / Revised: 10 November 2025 / Accepted: 19 November 2025 / Published: 21 November 2025
(This article belongs to the Special Issue Natural Products and Drug Delivery Systems in Dental Diseases)

Abstract

Plants are a rich source of bioactive compounds with broad pharmaceutical potential, particularly for their anti-inflammatory properties. Oral inflammation underlies many local and systemic diseases, yet conventional anti-inflammatory drugs have adverse effects. Crude plant extracts offer promising, safer alternatives. This systematic review synthesizes evidence on the anti-inflammatory activity of whole plant extracts in vitro oral models of inflammation. It also highlights methodological considerations for improved reproducibility. PubMed searches following PRISMA guidelines identified studies using oral or periodontal cells stimulated with relevant inflammatory triggers and treated with crude plant extracts. Extracts from Camellia sinensis, Salvia officinalis, Paeonia x suffruticosa, Houttuynia cordata, Theobroma cacao, and others consistently reduced pro-inflammatory cytokines such as IL-6, IL-8, IL-1β, and TNF-α, with no reported cytotoxicity at the effective concentrations. Variations in model design, extract characterization, and stimulation protocols were noted. Overall, this review demonstrates that crude plant extracts effectively modulate oral inflammatory responses in vitro. Findings suggest a possible non-cytotoxic anti-inflammatory activity that requires further investigation and underscore the need for methodological standardization to advance clinical translation.

1. Introduction

Inflammation is a tightly regulated physiological process fundamental to host defense against pathogens, tissue injury, and other harmful stimuli [1]. Beyond its defensive role, inflammation interacts dynamically with host metabolism, the commensal microbiome, and resident immune cells, shaping tissue homeostasis and systemic health [2,3,4]. In the oral cavity, inflammation is central to a wide range of local and systemic disorders. Conditions such as pulpitis, periodontitis, and stomatitis are primarily driven by inflammatory processes, which, if uncontrolled, can lead to tissue destruction and contribute to systemic complications via circulating inflammatory mediators [5,6]. Although dental interventions—including pulpectomy, exodontia, and scaling—effectively manage acute and chronic dental inflammation, non-dental inflammatory conditions, such as mucositis and periodontitis, remain challenging. Pharmacological agents are frequently employed, yet their clinical use is often limited by their systemic side effects, cytotoxicity, or patient intolerance. This underscores the need for safe, effective, and accessible alternatives for controlling oral inflammation.
Plant materials have been used medicinally for centuries across various ethnic groups and geographical locations in the world [7]. Their continued use in under-resourced and geographically isolated populations highlights both their accessibility and cultural relevance, as well as their role where socioeconomic barriers to access to medical care exist [8,9]. In recent decades, plant extracts have gained renewed interest due to their pharmacological potency, relative safety, and cost-effectiveness [10]. Bioactive compounds—including flavonoids, polyphenols, terpenoids, and alkaloids—have been shown to modulate inflammatory pathways by influencing cytokine production, chemokine expression, and transcription factor activity. Fractionation and chromatography have enabled the isolation of specific bioactive compounds with anti-inflammatory activity, which are then evaluated in vitro and in vivo. Despite promising preclinical findings, clinical translation remains limited, and few standardized formulations are currently approved for oral inflammatory conditions.
Topical anti-inflammatory agents remain a cornerstone of oral disease management, such as in the management of recurrent aphthous stomatitis (RAS) and radiation-induced mucositis (RIM). For instance, clinical evidence supported the use of benzydamine mouthwashes for the symptomatic relief of RIM [11]. Nonetheless, Aloe vera demonstrated comparable effects to benzydamine in a clinical setting [12]. A recent systematic review and meta-analysis indicated that herbal-derived compounds from turmeric and sumac may outperform conventional therapies in specific contexts, reinforcing the therapeutic potential of plant-based interventions [13]. Nevertheless, the search for safer, more effective, and accessible alternatives continues [14].
Crude plant extracts are not inherently safe by virtue of being natural. Their toxicological profile varies with species, extraction method, concentration, and bioassay, necessitating careful toxicological evaluation. Global health bodies emphasize the need for systematic and vigilant safety assessment of herbal medicines [15,16]. Evidence from systematic reviews and toxicological evaluations indicates that most plant extracts exhibit favorable safety margins when used with traditional dosages [17,18]. Safety reporting remains key when investigating the biological activities of plant extracts.
Mechanistically, oral inflammation involves complex networks of pro-inflammatory mediators, including cytokines (IL-1β, IL-6, TNF-α), chemokines (CCL2, CCL5, CXCL family members), prostaglandins (PGE2), and enzymes such as COX-2. These mediators orchestrate immune cell recruitment, activation, and tissue remodeling, and their dysregulation contributes to pathological outcomes [19,20]. Plant-derived compounds can modulate these pathways by suppressing cytokine and chemokine expression, inhibiting transcription factors such as NF-κB and AP-1, reducing oxidative stress, and downregulating inflammatory enzymes [21]. In vitro models using fibroblasts, epithelial cells, keratinocytes, osteocytes, and immune cell co-cultures have been instrumental in elucidating these mechanisms, but the evidence remains fragmented and often extrapolated from non-oral systems.
Furthermore, methodological heterogeneity—including variations in extraction methods, concentrations, cell models, inflammatory stimuli, and assay endpoints—limits reproducibility and comparability across studies. A systematic synthesis of the current literature on crude plant extracts in oral in vitro models is therefore essential. Such a review can consolidate evidence on anti-inflammatory efficacy, identify reproducible effects, clarify molecular mechanisms, and highlight methodological gaps, ultimately informing both pre-clinical research and translational applications.
This systematic review aims to provide a comprehensive evaluation of the anti-inflammatory potential of whole plant extracts in oral in vitro models. It aims to summarize and review evidence of crude plant extracts in cell models of oral inflammation, characterizing the ethnomedicinal in these models for further mechanistic evaluation. Thus, acknowledging the practical and traditional use of herbal materials as accessible interventions, while identifying methodological limitations, promoting standardization and reproducibility in future research.

2. Materials and Methods

2.1. Search Strategy

This systematic review was designed according to the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) recommendations. Between April 2025 and August 2025, an electronic search was conducted in the PubMed database without time or language restrictions, and without any filters. The search string was developed using a combination of medical subject heading database (MeSH) terms and keywords connected with Boolean operators, to ensure a comprehensive collection of studies. The search string was as follows: ((“Inflammation”[Mesh]) OR (“Inflammation Mediators”[Mesh]) OR (“Stomatitis”[Mesh]) OR (“Mucositis”[Mesh]) OR (“Gingivitis”[Mesh]) OR (“Periodontitis”[Mesh]) OR (“Mouth Diseases”[Mesh]) OR (“Interleukins”[Mesh]) OR (“Interleukin-1”[Mesh]) OR (“Interleukin-1beta”[Mesh]) OR (“Interleukin-6”[Mesh]) OR (“Tumor Necrosis Factor-alpha”[Mesh]) OR (“Cytokines”[Mesh])) AND ((“Plant Extracts”[Mesh]) OR (“Drugs, Chinese Herbal”[Mesh])) AND ((“In Vitro Techniques”[Mesh]) OR (“Cell Culture Techniques”[Mesh]) OR (“gingival fibroblasts”) OR (“periodontal fibroblasts”) OR (“oral epithelial cells”) OR (“oral keratinocytes”)).

2.2. Study Selection and Eligibility Criteria

Initially, eligible studies were identified for compatibility with the aim of this systematic review. Thus, the screening of titles and abstracts for in vitro studies investigating the effects of plant extracts on inflammation was conducted. Following this, full-text screening was carried out, applying the eligibility criteria. Eligibility criteria were specified using the PICO criteria as follows: population was oral inflammatory cell models or in vitro oral inflammatory models, intervention was crude plant extracts regardless of the part of the plant used to obtain the extract, control was positive (inflammation-triggered cells) and negative (cells without stimulation), and outcome was the anti-inflammatory effects.
Inclusion criteria were studies that were in vitro studies using oral cell models or co-culture models of oral cells and immune cells. Only studies with direct inflammatory assay and the use of relevant inflammatory stimuli were included. Studies that utilized crude plant extracts alone or alongside their fractionated compounds were included if a separate reporting of outcomes of the crude plant extract existed. In addition, the criteria applied were studies with clear reporting of methods and analysis.
Exclusion based on study design was performed for review articles, in vivo studies, clinical human studies, and chemical characterization studies. Studies on isolated or fractionated compounds were excluded. Similarly, studies that employed non-oral cell types or immune cells alone were excluded. Additionally, studies that investigated the inflammatory and allergic effects of plant parts, such as pollen and dust, were excluded. Figure 1 illustrates the flow diagram of the study’s retrieval.

2.3. Data Collection Process

Data extraction was conducted using a customized table in Microsoft Excel. Table contents were authors and year, plant name, extract type, concentration of extract, method of extraction, vehicle type, cell type, stimulation time and order, assay, control, outcome, and viability assessment. Moreover, outcomes of each study were extracted according to the following categories: bioassay, outcome, highest non-cytotoxic, and lowest effective concentrations.

2.4. Quality Assessment

Assessment of evidence quality was conducted using the Toxicological data reliability assessment tool (ToxRtool, version 2009), which consists of fifteen items that assess various aspects of experimental studies [22]. These include chemical characterization of the material used, medium/vehicle, system/cell type, justification of the test system, test system preparation and maintenance, concentration, exposure duration, replication, the use of controls, endpoint, validity of method, data documentation and analysis, results reproducibility, study design clarity, and the plausibility of study design and data. Judgment for each item of the tool was scored as 0 if the study failed to meet the criterion, 1 if the study met the criterion, and N/A if the criterion did not apply to the study. According to the assessment tool, a score equal to or above 12 was considered reliable without restrictions, a score between 8 and 11 was considered reliable with restrictions, and a score below 8 was considered unreliable. Two examiners (IR and RG) conducted the quality assessment independently, and discrepancies were discussed until a consensus was reached.

2.5. Synthesis Method

Studies that met the eligibility criteria and were without full-text restrictions were included for synthesis. Data were extracted and tabulated for narrative synthesis. Comparison of outcomes was narratively reported to highlight potential variables that influenced the anti-inflammatory effects of the plant extracts, such as the type of vehicle and the extraction method. Furthermore, data from the inflammatory model and cell types were used to qualitatively contrast the single-cell inflammatory model against co-culture models.

3. Results

3.1. Study Characteristics and Extract Preparation

Twelve in vitro studies published between 2013 and 2022 met the inclusion criteria. Crude plant extracts were prepared from 13 species, including Camellia sinensis, Salvia officinalis, Paeonia suffruticosa, Houttuynia cordata, Listea japonica, Populus nigra, Populus canadensis, Populus balsamifera, Theobroma cacao, Equisetum arvense, Alpinia katsumadai, Juncus effusus, and Zingiber officinale. Extraction methods were primarily aqueous (n = 7) or ethanolic (n = 4), with one study using methanol. Extraction temperatures varied from room temperature (RT) to 95 °C, with specific temperatures reported for Salvia officinalis (95 °C), Populus spp. (60 °C), and Juncus effusus (60 °C). Purification techniques included microfiltration (0.2–0.45 µm) and centrifugation (1200–3000× g, 10–15 min) to ensure sterility and remove particulate matter. Commercial formulations (e.g., Equisetum arvense) employed H2O with butylene glycol as the vehicle. Stock concentrations (SC) ranged from 1% to 200%, while working concentrations (WC) were selected to maximize anti-inflammatory effects without compromising cell viability, typically in the micromolar or sub-percentage range (0.00004–10%). Several studies used multiple working concentrations to establish dose–response relationships. Table 1 summarizes the characteristics of included studies.

3.2. Cell Models and Inflammatory Stimulation

The studies employed a variety of oral cell models, including oral epithelial cell lines, primary gingival fibroblasts, primary periodontal ligament fibroblasts, keratinocytes, and monocytic cell lines (THP-1, RAW264.7). Inflammatory stimuli were mainly bacterial LPS from P. gingivalis, A. actinomycetemcomitans, F. nucleatum, or multiple oral pathogens. Other stimuli included phorbol 12-myristate 13-acetate (PMA) and silver nanoparticles (AgNPs). Several studies assessed basal inflammation without exogenous stimuli. Treatment protocols varied: extracts were applied prior to, simultaneously with, or following inflammatory stimulation, with durations ranging from 30 min to 24 h.

3.3. Cell Viability and Cytotoxicity

Most studies incorporated cytotoxicity assessments to confirm extract safety at tested concentrations. Assays included MTT (n = 7), WST-1 (n = 1), MTS (n = 1), and CellTiter-Blue (n = 1). Generally, extracts were non-cytotoxic across working concentrations, with viability maintained above 80–90% in most models. Notably, Satthakarn et al. (2015) [27] and Shiba et al. (2021) [31] did not perform viability assays, and one study used relatively high working concentrations that may have affected cell health. These data confirm that effective anti-inflammatory concentrations were largely safe for oral cells in vitro.

3.4. Anti-Inflammatory Effects

Crude plant extracts consistently demonstrated anti-inflammatory activity across all studies. Camellia sinensis (aqueous) reduced IL-6, IL-8, and CCL5 in oral epithelial cells, while Salvia officinalis (aqueous, 95 °C) suppressed IL-6 and IL-8 in gingival fibroblasts. Paeonia suffruticosa (ethanolic) downregulated CXCL9, CXCL10, and CXCL11 in fibroblasts, and Houttuynia cordata (aqueous) attenuated basal CXCL5 and CCL2 in primary gingival epithelial cells. Listea japonica (ethanolic) decreased IL-6 and IL-8 in periodontal ligament fibroblasts exposed to multiple LPS species, effective in both pre-treatment and co-treatment protocols. Populus spp. (methanolic) reduced IL-6 and IL-1β in fibroblasts challenged with silver nanoparticles. Theobroma cacao (ethanolic) lowered IL-6 and IL-8 in epithelial cells stimulated with F. nucleatum, while Equisetum arvense (commercial H2O + butylene glycol formulation) suppressed TNF-α in keratinocyte and monocytic cells. Alpinia katsumadai (ethanolic) attenuated COX-2 and PGE2 in fibroblasts, keratinocytes, and macrophages under LPS stimulation; Juncus effusus (aqueous, 60 °C) decreased IL-8 and CCL20 in keratinocytes, and Zingiber officinale (aqueous, RT) reduced IL-6 and IL-8 in primary gingival fibroblasts when applied both before and after LPS exposure. These findings indicate that crude plant extracts, irrespective of extraction method, effectively modulate key pro-inflammatory cytokines and chemokines across diverse oral cell types at cytocompatible concentrations. Table 2 summarizes the outcomes of the included studies.

3.5. Quality Assessment Results

Quality assessment of the collected studies highlights the reliability of the presented evidence. The mean ToxRTool score across studies of this review was 12.6 ± 1.7 (range 9–15), indicating moderate to high reliability. Eight studies (66.6%) were classified as reliable without restrictions, while four studies (33.3%) were reliable with restrictions. Six criteria were fulfilled by all studies: test substance administration, test system (cell culture identification), test system preparation and maintenance, exposure duration, endpoints measured, and data documentation and statistical analysis. Extract identification and replicates had the lowest fulfillment rates (33.3% and 58.3%, respectively). While these findings show a significant reliability of the evidence supporting the anti-inflammatory potential of crude plant extracts, they underscore persistent methodological shortcomings in reporting plant extract behavior within in vitro oral inflammation studies.
Table 3 presents scores and descriptive statistics of quality assessment for studies in this review. Figure 2 illustrates the compliance rate of studies with the ToxRTool assessment criteria. Full details of quality assessment can be found in Table S1 in the Supplementary Materials.

4. Discussion

To date, no systematic reviews have specifically examined the anti-inflammatory potential of crude plant extracts in oral inflammatory cell models. By consolidating in vitro evidence, this review demonstrates that unrefined plant preparations exhibit measurable anti-inflammatory activity across different oral cell types while maintaining cytocompatibility. These findings support the pharmacological validity of traditional herbal practices and identify key methodological challenges that must be addressed for clinical translation.
While this review underscores the limited evidence supporting the anti-inflammatory role of plant extract in oral inflammation, it also reveals important methodological shortcomings of the existing literature. Among the included studies, four—those by Yun et al. (2013) [25], Satthakarn et al. [27], Shin et al. [32], and Al-Shibani et al. [34]—were deemed reliable with restrictions. Despite most studies demonstrating general reliability, only four—Ehrnhöfer-Ressler et al. [24], Yun et al. (2013) [25], Pobłocka-Olech et al. [29], and Ben Lagha et al. [30]—reported adequate characterization of their plant extracts, making this the most frequently unmet criterion in the ToxRTool assessment. While in vitro investigations of crude plant extracts provide valuable insights into their cytoprotective, antioxidant, and anti-inflammatory properties, chemical characterization is crucial for identifying the active constituents responsible for these effects. As highlighted by Brusotti et al., characterization methods should be tailored to the intended biological activity to enhance pharmacological relevance and reduce unwanted metabolites [35].
The herbal extracts investigated in this review consisted of plant species from ten botanical families. Table 4 summarizes the characteristics of plant extracts investigated across studies of this review. The diverse taxonomical and phytochemical properties of the plants underscore their diverse biological activities. The broad pharmacological spectrum, on the other hand, indicates that the putative anti-inflammatory effects of the reported plant extracts are possibly a result of integrative cellular mechanisms that eventually mask inflammation. Chemical characterization and mechanistic investigation of inflammatory signaling promote a targeted therapeutic potential. For instance, polyphenol-driven IL-1β and TNF-α inhibition from Camellia sinensis and Salvia officinalis offers protective antioxidant effects to lessen the inflammatory sequelae of oxidative stress. Whereas salicylates-mediated reduction of COX1 and prostaglandin from Populus spp. offers analgesic effects.
Another recurrent methodological weakness observed in the included studies was the inconsistent use of experimental replicates. Specifically, five studies—those by Yun et al. (2013) [25], Lombardo Bedran et al. [26], Yun et al. (2018) [28], Shin et al. [32], and Al-Shibani et al. [34]—lacked replicates in their inflammatory bioassays. The absence of replication limits data reliability and increases the risk of misinterpreting cytostatic or anti-proliferative effects as genuine anti-inflammatory responses. In addition, many studies reported the anti-inflammatory effects of plant extracts without a dose–response relationship, which is crucial in the validation of the evidence and balancing the efficacy with cytocompatibility.
Across the included studies, aqueous and ethanolic extracts consistently reduced the expression and secretion of pro-inflammatory mediators such as IL-1β, IL-6, and IL-8, as well as chemokines including CCL2, CCL5, and CXCL family members. These effects occurred at non-cytotoxic concentrations—typically between 10 and 125 µg/mL—indicating genuine biological activity rather than metabolic suppression. Collectively, these observations affirm that crude plant extracts can modulate inflammatory signaling in oral cells, supporting their non-cytotoxic anti-inflammatory potential and possibly their efficacy as adjuncts for managing oral inflammation.
The oral cavity represents a complex microenvironment in which epithelial cells, fibroblasts, keratinocytes, immune cells, and the oral microbiome interact to maintain tissue integrity and immune balance [48]. In periodontitis, this balance is disrupted, leading to an excessive cytokine-driven response and tissue destruction [49,50]. Similarly, in oral mucositis, exposure of the oral mucosal tissues to radiation results in damage signals from keratinocytes, tissue fibroblasts, and endothelial cells, resulting in painful inflammation and ulceration of the mucosal tissue [51,52]. Additionally, both in periodontitis and oral mucositis, resident immune cells participate in perpetuating the inflammatory response [53,54,55,56]. Collectively, these observations demonstrate how the interplay of oral cells maintains homeostasis and supports their employment in in vitro studies of oral inflammatory conditions.
The consistency of the test systems employed across studies in this review reinforces the evidence for the anti-inflammatory potential of crude plant extracts in oral inflammation. In vitro models using oral fibroblasts and epithelial cells have proven valuable for mechanistic investigations; however, they often fail to reproduce the cellular interplay characteristic of in vivo inflammation. Kasurinen et al. and Karri et al. reported that co-culture models elicited stronger inflammatory responses compared to monocultures [57,58]. Therefore, future research should prioritize co-culture or three-dimensional models incorporating immune cells to more accurately simulate the inflammatory cascade, enabling more physiologically relevant testing of plant-derived therapeutics. Notably, none of the studies included in this review employed a co-culture model.
On the other hand, oral keratinocytes are a fundamental cell type within the oral mucosa, contributing to both inflammatory signaling and epithelial repair. Pleguezuelos et al. demonstrated that gingival keratinocytes act as early responders to microbial LPS, mediating inflammation through MAP-dependent IL-1ß release [59]. In contrast, Hujiahemaiti et al. reported that a plant-derived compound enhanced oral keratinocyte proliferation, underscoring their importance in tissue regeneration and wound healing [60]. These findings illustrate the methodological variability in how keratinocytes are employed across in vitro models of oral inflammation.
In the context of gingivitis and periodontitis, keratinocytes play a limited role due to their relative scarcity within the junctional epithelium. Conversely, they are functionally prominent in regenerative processes, particularly in the masticatory mucosa, where keratinization significantly influences healing dynamics [61,62]. Therefore, their employment within in vitro studies of oral mucositis is physiologically justified. Only two studies within this review, those by Wada et al. and Shiba et al., incorporated keratinocytes, highlighting a potential methodological gap and the need for purposeful employment of cell types for the studied model of inflammation.
Despite differences in cell type and inflammatory stimuli, methodological consistency across studies was relatively high. Most investigations used bacterial LPS or oral pathogens to elicit inflammation. Studies by Ehrnhöfer-Ressler et al. and Pobłocka-Olech et al. have utilized PMA and AgNP as inflammatory stimuli, respectively, while Satthakarn et al. did not use a positive control [24,27,29]. While chemical stressors induce cellular damage signals, it has been demonstrated that their elicited effects differ from those produced by cytokines and LPS driven by exposure to oral pathogens [63,64]. On the other hand, methods of quantification of cytokine modulation were consistent across studies in this review. They consisted primarily of ELISA or RT-PCR. These assays remain the benchmark for assessing inflammatory outcomes, while multiplex technologies offer higher throughput, provided they are validated against ELISA [65,66,67,68]. Two studies in this review, by Ehrnhöfer-Ressler et al. and Al-Shibani et al., utilized the multiplex immunoassay in their experiments [24,34]. The adoption of standardized stimulation protocols, treatment sequences, and endpoint markers would substantially enhance reproducibility and permit quantitative comparisons across studies.
Variability in extraction methods represents a key determinant of outcome differences [69,70]. In this review, aqueous extractions were predominant, followed by ethanolic or methanolic techniques. Because solvent polarity directly affects the yield and spectrum of bioactive compounds, solvent selection must align with the physicochemical properties of target phytochemicals [71]. Organic solvents often enhance recovery of phenolics and flavonoids, while water-based extractions remain more clinically relevant for topical and oral applications [72]. Systematic reporting and standardization of extraction parameters are therefore essential to ensure reproducibility and facilitate clinical translation. Nonetheless, Jeyaraj et al. reported that aqueous extraction results in a bioactive yield regardless of the solvent type [73]. Therefore, in the context of practical infusion preparations, the use of water as a solvent may also be effective in obtaining the therapeutic benefits of the plant material.
For centuries, herbal materials have been used in traditional medicine to treat a plethora of human ailments, and they continue to serve as accessible therapeutic options in regions with limited healthcare resources [74,75]. Plant-based medicines offer a safer alternative to synthetic anti-inflammatory drugs, demonstrating comparable efficacy to NSAIDs but with fewer adverse effects [76,77,78]. While the effects of plant-based therapies are not as rapid and potent as synthetic drugs, their efficacy with fewer side effects outweighs this limitation. In the context of oral inflammation, their topical application provides a practical advantage, enabling local relief without systemic absorption. Conventional topical therapies, though effective, may cause adverse outcomes such as fungal infections or systemic side effects with prolonged use [79]. Conversely, plant-derived formulations—such as curcumin, honey, and chamomile—have shown promise in reducing the severity of oral inflammatory conditions such as radiation mucositis [80,81]. These formulations exhibit favorable safety profiles compared to synthetic products such as chlorhexidine [82,83]. Although current clinical evidence remains mixed due to variability in herbal formulations and study design, larger standardized trials are warranted to confirm their therapeutic efficacy and safety in oral inflammation.
Safety, however, remains a prerequisite for translation. While most studies reported high cytocompatibility, metabolic assays such as MTT alone do not fully capture sub-lethal or long-term effects [84]. Cellular effects of plant extracts transcend metabolic alteration to potentially impose alterations in junctional adherence, membrane integrity, gene toxicity, and, in a simpler context, assay interferences [84,85,86]. Therefore, comprehensive toxicity testing under GLP conditions, including membrane integrity and genotoxicity assessments, is necessary to establish safety profiles [87]. Additionally, natural variability arising from plant source, harvest time, and extraction technique must be controlled through standardized sourcing and analytical characterization of bioactive constituents [88]. Jantan et al. suggested that the clinical translation of effective immunomodulatory plant-derived compounds requires improved quality and standardization of extraction methods to improve their bioavailability and minimize cytotoxicity [89].
Early mechanistic evidence suggests that many crude extracts modulate inflammation through inhibition of the NF-κB signaling pathway, a central regulator of inflammatory gene expression [90,91,92]. In this review, Theobroma cacao and Equisetum arvense, investigated by Ben Lagha et al. and Shiba et al., respectively, demonstrated NF-κB–dependent anti-inflammatory effects in oral cells, aligning with broader findings across other plant-derived therapeutics [30,31]. Research interest in exploring this role resurged in the late 2010s, when studies began to employ sophisticated molecular approaches, such as gene expression and pathway-level analysis, to elucidate how plant extracts exert their anti-inflammatory effects via the NF-κB signaling system [93,94]. Future investigations integrating transcriptomic or proteomic approaches could unravel synergistic mechanisms among phytochemicals and clarify their collective impact on inflammatory networks.
Studies within this review demonstrated a notable degree of homogeneity in both endpoints and measurement methods. The consistent quantification of key pro-inflammatory cytokines (IL-1ß and IL-6) and chemokines (CCL5 and IL-8) using ELISA strengthens the reliability of the findings by aligning with standardized inflammatory bioassays [65]. However, the selective measurement of only a few cytokines and chemokines may limit the mechanistic depth of interpretation. Notably, IL-8 is a pleiotropic chemokine that can be upregulated by diverse cellular events such as proliferation, angiogenesis, chemical injury, and oxidative stress [95,96]. Thus, relying solely on quantitative cytokine assessment represents a methodological limitation. Integrating signaling pathway analyses—such as NF-κß, MAPK, or Nrf2 activation—would provide deeper mechanistic insight into the molecular basis of the anti-inflammatory effects exerted by plant extracts.
An exception in the endpoints measured was the study by Shin et al., which incorporated prostaglandin E2 (PG-E2) and COX2 as outcome markers for the anti-inflammatory effects of Alpinia katsumadai. Cytokines and chemokines such as IL-6 and IL-8 are considered more robust markers of inflammatory activation due to their rapid induction and greater fold changes compared to PGE2 and COX2 [97,98]. This is due to PGE2 and COX2 being more constitutive in some tissues, reflecting their physiological role in tissue homeostasis.
While this systematic review provides supportive evidence for the anti-inflammatory potential of plant extracts in oral inflammatory cell models, its conclusions should be interpreted carefully due to several caveats. First, the review exclusively includes in vitro studies investigating crude plant extracts rather than isolated bio-compounds, which may miss the mechanistic clarity and specificity offered by isolated compounds [99]. In addition, collecting studies exclusively from the PubMed database may narrow the evidence of the anti-inflammatory potential. Therefore, comprehensive future work is needed to amalgamate the evidence and to explore in depth the methodological limitations of crude plant extracts in in vitro studies. Using crude plant extracts reflects a broader therapeutic application and aligns with traditional medicinal and ethnobotanical practice; however, the observed effects of crude plant extracts may vary across different cell models due to potential interferences between plant components and the biological assay system [100]. Moreover, the reproducibility of in vitro studies that utilize crude plant extracts can be lower than those employing isolated compounds, owing to variability in plant source, growth conditions, and extraction methods [101]. Finally, the heterogeneity in extraction methods, stimulation conditions, and cell systems prevents the conduction of a meta-analysis, which limits evidence generalizability.

5. Conclusions

In conclusion, crude plant extracts emerge as promising, cytocompatible, and biologically active agents capable of modulating key inflammatory mediators in oral tissues. Future research that employs complex models, such as co-culture or 3D models, and standardized experimental design using well-characterized plant extracts is needed. Bridging ethnobotanical knowledge with standardized experimental design, mechanistic analysis, and rigorous safety validation is crucial for translating these natural materials into evidence-based therapeutics. Such an approach may ultimately yield accessible, sustainable, and safe alternatives to conventional anti-inflammatory drugs for the management of oral inflammatory diseases.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms262311253/s1.

Author Contributions

Conceptualization, I.R. and R.G.; Methodology, I.R.; Investigation, I.R.; Writing—Original Draft, I.R.; Writing—Review and Editing, R.G.; Supervision, R.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors on request.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
MeSHmedical subject headings
NDRno dose–response
PG-E2prostaglandin-E2
COX2cyclooxygenase-2
RASrecurrent apthous stomatitis
PRISMApreferred reporting items for systematic reviews and meta-analysis
PMAphorbol 12-myristate 13-acetate
AgNPsilver nanoparticles
MTT3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide
RTroom temperature
SCstock concentration
WCworking concentration
D.H2Odistilled water
DMSOdimethyl sulfoxide
LPSlipopolysaccharide
ELISAenzyme-linked immunosorbent assay
RT-PCRreal-time polymerase chain reaction
WST-1water-soluble tetrazolium salts
MTS(3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium)

References

  1. Menkin, V. The role of inflammation in immunity. Physiol. Rev. 1938, 18, 366–418. [Google Scholar] [CrossRef]
  2. Gaber, T.; Strehl, C.; Buttgereit, F. Metabolic regulation of inflammation. Nat. Rev. Rheumatol. 2017, 13, 267–279. [Google Scholar] [CrossRef]
  3. Kim, D.; Zeng, M.Y.; Núñez, G. The interplay between host immune cells and gut microbiota in chronic inflammatory diseases. Exp. Mol. Med. 2017, 49, e339. [Google Scholar] [CrossRef]
  4. Blander, J.M.; Longman, R.S.; Iliev, I.D.; Sonnenberg, G.F.; Artis, D. Regulation of inflammation by microbiota interactions with the host. Nat. Immunol. 2017, 18, 851–860. [Google Scholar] [CrossRef] [PubMed]
  5. Silva, T.; Garlet, G.P.; Fukada, S.Y.; Silva, J.S.d.; Cunha, F.d.Q. Chemokines in oral inflammatory diseases: Apical periodontitis and periodontal disease. J. Dent. Res. 2007, 86, 306–319. [Google Scholar] [CrossRef] [PubMed]
  6. Fedele, S.; Sabbah, W.; Donos, N.; Porter, S.; D’Aiuto, F. Common oral mucosal diseases, systemic inflammation, and cardiovascular diseases in a large cross-sectional US survey. Am. Heart J. 2011, 161, 344–350. [Google Scholar] [CrossRef] [PubMed]
  7. Lansky, E.P.; Paavilainen, H.M.; Pawlus, A.D.; Newman, R.A. Ficus spp.(fig): Ethnobotany and potential as anticancer and anti-inflammatory agents. J. Ethnopharmacol. 2008, 119, 195–213. [Google Scholar] [CrossRef]
  8. Napagoda, M.T.; Sundarapperuma, T.; Fonseka, D.; Amarasiri, S.; Gunaratna, P. An ethnobotanical study of the medicinal plants used as anti-inflammatory remedies in Gampaha District, Western Province, Sri Lanka. Scientifica 2018, 2018, 9395052. [Google Scholar] [CrossRef]
  9. Ogbole, O.O.; Gbolade, A.A.; Ajaiyeoba, E.O. Ethnobotanical survey of plants used in treatment of inflammatory diseases in Ogun State of Nigeria. Eur. J. Sci. Res. 2010, 43, 183–191. [Google Scholar]
  10. Erdogrul, Ö.T. Antibacterial activities of some plant extracts used in folk medicine. Pharm. Biol. 2002, 40, 269–273. [Google Scholar] [CrossRef]
  11. Ariyawardana, A.; Cheng, K.K.F.; Kandwal, A.; Tilly, V.; Al-Azri, A.R.; Galiti, D.; Chiang, K.; Vaddi, A.; Ranna, V.; Nicolatou-Galitis, O.; et al. Systematic review of anti-inflammatory agents for the management of oral mucositis in cancer patients and clinical practice guidelines. Support. Care Cancer 2019, 27, 3985–3995. [Google Scholar] [CrossRef]
  12. Sahebjamee, M.; Mansourian, A.; Hajimirzamohammad, M.; Zadeh, M.T.; Bekhradi, R.; Kazemian, A.; Manifar, S.; Ashnagar, S.; Doroudgar, K. Comparative efficacy of aloe vera and benzydamine mouthwashes on radiation-induced oral mucositis: A triple-blind, randomised, controlled clinical trial. Oral Health Prev. Dent. 2015, 13, 309–315. [Google Scholar]
  13. Mohandas, R.; Mohapatra, S. Comparative evaluation of the efficacy of herbal and benzydamine mouthwashes in preventing radiation-induced oral mucositis among head and neck cancer patients: A systematic review and network meta-analysis. Evid.-Based Dent. 2025, 26, 118. [Google Scholar] [CrossRef] [PubMed]
  14. Ferrer-Montiel, A. Benzydamine hydrochloride: An overview on a well-established drug with news in mechanisms of action. F1000Research 2024, 13, 350. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  15. World Health Organization. WHO guidelines on safety monitoring of herbal medicines in pharmacovigilance systems. In Who Guidelines on Safety Monitoring of Herbal Medicines in Pharmacovigilance Systems; WHO: Geneva, Switzerland, 2004. [Google Scholar]
  16. European Medicines Agency (EMA); Committee on Herbal Medicinal Products (HMPC). Guideline on Non-Clinical Documentation in Applications for Marketing Authorisation/Registration of Well-Established and Traditional Herbal Medicinal Products (EMA/HMPC/353983/2016). 2019. Available online: https://www.ema.europa.eu/en/non-clinical-documentation-applications-marketing-authorisation-registration-well-established-traditional-herbal-medicinal-products-scientific-guideline (accessed on 9 November 2025).
  17. Posadzki, P.; Watson, L.K.; Ernst, E. Adverse effects of herbal medicines: An overview of systematic reviews. Clin. Med. 2013, 13, 7–12. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Ekor, M. The growing use of herbal medicines: Issues relating to adverse reactions and challenges in monitoring safety. Front. Pharmacol. 2014, 4, 177. [Google Scholar] [CrossRef]
  19. Didilescu, A.C.; Chinthamani, S.; Scannapieco, F.A.; Sharma, A. NLRP3 inflammasome activity and periodontal disease pathogenesis—A bidirectional relationship. Oral Dis. 2024, 30, 4069–4077. [Google Scholar] [CrossRef]
  20. Aral, K.; Berdeli, E.; Cooper, P.R.; Milward, M.R.; Kapila, Y.; Karadede Ünal, B.; Aral, C.A.; Berdeli, A. Differential expression of inflammasome regulatory transcripts in periodontal disease. J. Periodontol. 2020, 91, 606–616. [Google Scholar] [CrossRef]
  21. Kim, K.O.; Lee, D.; Hiep, N.T.; Song, J.H.; Lee, H.-J.; Lee, D.; Kang, K.S. Protective Effect of Phenolic Compounds Isolated from Mugwort (Artemisia argyi) against Contrast-Induced Apoptosis in Kidney Epithelium Cell Line LLC-PK1. Molecules 2019, 24, 195. [Google Scholar] [CrossRef]
  22. Schneider, K.; Schwarz, M.; Burkholder, I.; Kopp-Schneider, A.; Edler, L.; Kinsner-Ovaskainen, A.; Hartung, T.; Hoffmann, S. “ToxRTool”, a new tool to assess the reliability of toxicological data. Toxicol. Lett. 2009, 189, 138–144. [Google Scholar] [CrossRef]
  23. Zhao, L.; La, V.D.; Grenier, D. Antibacterial, antiadherence, antiprotease, and anti-inflammatory activities of various tea extracts: Potential benefits for periodontal diseases. J. Med. Food 2013, 16, 428–436. [Google Scholar] [CrossRef]
  24. Ehrnhöfer-Ressler, M.M.; Fricke, K.; Pignitter, M.; Walker, J.M.; Walker, J.; Rychlik, M.; Somoza, V. Identification of 1,8-cineole, borneol, camphor, and thujone as anti-inflammatory compounds in a Salvia officinalis L. infusion using human gingival fibroblasts. J. Agric. Food Chem. 2013, 61, 3451–3459. [Google Scholar] [CrossRef] [PubMed]
  25. Yun, C.-S.; Choi, Y.-G.; Jeong, M.-Y.; Lee, J.-H.; Lim, S. Moutan Cortex Radicis inhibits inflammatory changes of gene expression in lipopolysaccharide-stimulated gingival fibroblasts. J. Nat. Med. 2013, 67, 576–589. [Google Scholar] [CrossRef] [PubMed]
  26. Lombardo Bedran, T.B.; Morin, M.-P.; Palomari Spolidorio, D.; Grenier, D. Black tea extract and its theaflavin derivatives inhibit the growth of periodontopathogens and modulate interleukin-8 and β-defensin secretion in oral epithelial cells. PLoS ONE 2015, 10, e0143158. [Google Scholar] [CrossRef] [PubMed]
  27. Satthakarn, S.; Chung, W.; Promsong, A.; Nittayananta, W. H outtuynia cordata modulates oral innate immune mediators: Potential role of herbal plant on oral health. Oral Dis. 2015, 21, 512–518. [Google Scholar] [CrossRef] [PubMed]
  28. Yun, I.-G.; Ahn, S.-H.; Yoon, W.-J.; Kim, C.S.; Lim, Y.K.; Kook, J.-K.; Jung, S.; Choi, C.-H.; Lee, T.-H. Litsea japonica leaf extract suppresses proinflammatory cytokine production in periodontal ligament fibroblasts Stimulated with oral pathogenic bacteria or interleukin-1β. Int. J. Mol. Sci. 2018, 19, 2494. [Google Scholar] [CrossRef]
  29. Pobłocka-Olech, L.; Inkielewicz-Stepniak, I.; Krauze-Baranowska, M. Anti-inflammatory and antioxidative effects of the buds from different species of Populus in human gingival fibroblast cells: Role of bioflavanones. Phytomedicine 2019, 56, 1–9. [Google Scholar] [CrossRef] [PubMed]
  30. Ben Lagha, A.; Maquera Huacho, P.; Grenier, D. A cocoa (Theobroma cacao L.) extract impairs the growth, virulence properties, and inflammatory potential of Fusobacterium nucleatum and improves oral epithelial barrier function. PLoS ONE 2021, 16, e0252029. [Google Scholar] [CrossRef]
  31. Shiba, F.; Miyauchi, M.; Chea, C.; Furusho, H.; Iwasaki, S.; Shimizu, R.; Ohta, K.; Nishihara, T.; Takata, T. Anti-inflammatory effect of glycyrrhizin with Equisetum arvense extract. Odontology 2021, 109, 464–473. [Google Scholar] [CrossRef]
  32. Shin, S.W.; Hwang, Y.S. Anti-periodontitis effect of ethanol extracts of Alpinia katsumadai seeds. Nutrients 2021, 14, 136. [Google Scholar] [CrossRef]
  33. Wada, A.; Murakami, K.; Ishikawa, Y.; Amoh, T.; Hirao, K.; Hosokawa, Y.; Hinode, D.; Miyake, Y.; Yumoto, H. Anti-Inflammatory and Protective Effects of Juncus effusus L. Water Extract on Oral Keratinocytes. BioMed Res. Int. 2022, 2022, 9770899. [Google Scholar] [CrossRef]
  34. Al-Shibani, N.; Al-Kattan, R.; Alssum, L.; Allam, E. Effects of ginger (Zingiber officinale) on gingival fibroblasts: An in vitro study. Clin. Exp. Dent. Res. 2022, 8, 906–911. [Google Scholar] [CrossRef] [PubMed]
  35. Brusotti, G.; Cesari, I.; Dentamaro, A.; Caccialanza, G.; Massolini, G. Isolation and characterization of bioactive compounds from plant resources: The role of analysis in the ethnopharmacological approach. J. Pharm. Biomed. Anal. 2014, 87, 218–228. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, D.; Ding, Y.; Chen, G.; Sun, Y.; Zeng, X.; Ye, H. Components identification and nutritional value exploration of tea (Camellia sinensis L.) flower extract: Evidence for functional food. Food Res. Int. 2020, 132, 109100. [Google Scholar] [CrossRef] [PubMed]
  37. Ghorbani, A.; Esmaeilizadeh, M. Pharmacological properties of Salvia officinalis and its components. J. Tradit. Complement. Med. 2017, 7, 433–440. [Google Scholar] [CrossRef]
  38. He, J.; Dong, Y.; Liu, X.; Wan, Y.; Gu, T.; Zhou, X.; Liu, M. Comparison of chemical compositions, antioxidant, and anti-photoaging activities of Paeonia suffruticosa flowers at different flowering stages. Antioxidants 2019, 8, 345. [Google Scholar] [CrossRef]
  39. Shingnaisui, K.; Dey, T.; Manna, P.; Kalita, J. Therapeutic potentials of Houttuynia cordata Thunb. against inflammation and oxidative stress: A review. J. Ethnopharmacol. 2018, 220, 35–43. [Google Scholar] [CrossRef]
  40. Lee, S.Y.; Min, B.S.; Kim, J.H.; Lee, J.; Kim, T.J.; Kim, C.S.; Kim, Y.H.; Lee, H.K. Flavonoids from the leaves of Litsea japonica and their anti-complement activity. Phytother. Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 2005, 19, 273–276. [Google Scholar]
  41. Won, J.; Kim, J.-E.; Choi, D.H.; Han, M.-W.; Lee, D.-H.; Kang, S.C.; Song, Y.-J. Effects of compounds isolated from a Litsea japonica fruit extract on the TNF-α signaling pathway and cell viability. Mol. Cell. Toxicol. 2016, 12, 37–44. [Google Scholar] [CrossRef]
  42. Okińczyc, P.; Widelski, J.; Nowak, K.; Radwan, S.; Włodarczyk, M.; Kuś, P.M.; Susniak, K.; Korona-Głowniak, I. Phytochemical profiles and antimicrobial activity of selected Populus spp. bud extracts. Molecules 2024, 29, 437. [Google Scholar] [CrossRef]
  43. Martínez, R.; Torres, P.; Meneses, M.; Figueroa, J.; Pérez-Álvarez, J.; Viuda-Martos, M. Chemical, technological and in vitro antioxidant properties of cocoa (Theobroma cacao L.) co-products. Food Res. Int. 2012, 49, 39–45. [Google Scholar] [CrossRef]
  44. Pallag, A.; Jurca, T.; Pasca, B.; Sirbu, V.; Honiges, A.; Costuleanu, M. Analysis of phenolic compounds composition by HPLC and assessment of antioxidant capacity in Equisetum arvense L. extracts. Rev. De Chim. 2016, 67, 1623–1627. [Google Scholar]
  45. Zhao, X.M.; You, H.L.; Yang, L.; Zhou, B.; Su, K.M.; Gajendran, B.; Shen, X.C.; Zhang, N.L. Flavonoids and Kavalactones Isolated from Seeds of Alpinia katsumadai Hayata and Their Cytotoxic Activities. Chem. Biodivers. 2025, 22, e202403429. [Google Scholar] [CrossRef]
  46. Park, N.-Y.; Kim, S.-G.; Park, H.-H.; Jeong, K.-T.; Lee, Y.J.; Lee, E. Anti-inflammatory effects of Juncus effusus extract (JEE) on LPS-stimulated RAW 264.7 cells and edema models. Pharm. Biol. 2016, 54, 243–250. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, M.; Zhao, R.; Wang, D.; Wang, L.; Zhang, Q.; Wei, S.; Lu, F.; Peng, W.; Wu, C. Ginger (Zingiber officinale Rosc.) and its bioactive components are potential resources for health beneficial agents. Phytother. Res. 2021, 35, 711–742. [Google Scholar] [CrossRef]
  48. Hasturk, H.; Kantarci, A.; Van Dyke, T.E. Oral inflammatory diseases and systemic inflammation: Role of the macrophage. Front. Immunol. 2012, 3, 118. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Wielento, A.; Lagosz-Cwik, K.B.; Potempa, J.; Grabiec, A.M. The Role of Gingival Fibroblasts in the Pathogenesis of Periodontitis. J. Dent. Res. 2023, 102, 489–496. [Google Scholar] [CrossRef] [PubMed]
  50. Scheres, N.; Laine, M.L.; Sipos, P.M.; Bosch-Tijhof, C.J.; Crielaard, W.; de Vries, T.J.; Everts, V. Periodontal ligament and gingival fibroblasts from periodontitis patients are more active in interaction with Porphyromonas gingivalis. J. Periodontal Res. 2011, 46, 407–416. [Google Scholar] [CrossRef]
  51. Dai, M.; Dai, X.; Liang, Y.; Li, X.; Huang, H.; Zhao, W. Keratinocyte necroptosis promotes the progression of radiation-induced oral mucositis. BMC Oral Health 2025, 25, 941. [Google Scholar] [CrossRef]
  52. Sonis, S.T. The pathobiology of mucositis. Nat. Rev. Cancer 2004, 4, 277–284. [Google Scholar] [CrossRef]
  53. Almubarak, A.; Tanagala, K.K.K.; Papapanou, P.N.; Lalla, E.; Momen-Heravi, F. Disruption of Monocyte and Macrophage Homeostasis in Periodontitis. Front. Immunol. 2020, 11, 330. [Google Scholar] [CrossRef]
  54. Wang, W.; Zheng, C.; Yang, J.; Li, B. Intersection between macrophages and periodontal pathogens in periodontitis. J. Leukoc. Biol. 2021, 110, 577–583. [Google Scholar] [CrossRef]
  55. Handschel, J.; Sunderkötter, C.; Prott, F.-J.; Meyer, U.; Kruse-Lösler, B.; Joos, U. Increase of RM3/1-positive macrophages in radiation-induced oral mucositis. J. Pathol. 2001, 193, 242–247. [Google Scholar] [CrossRef] [PubMed]
  56. Sprinzl, G.; Galvan, O.; De Vries, A.; Ulmer, H.; Gunkel, A.; Lukas, P.; Thumfart, W. Local application of granulocyte-macrophage colony stimulating factor (GM-CSF) for the treatment of oral mucositis. Eur. J. Cancer 2001, 37, 2003–2009. [Google Scholar] [CrossRef] [PubMed]
  57. Kasurinen, S.; Happo, M.S.; Rönkkö, T.J.; Orasche, J.; Jokiniemi, J.; Kortelainen, M.; Tissari, J.; Zimmermann, R.; Hirvonen, M.R.; Jalava, P.I. Differences between co-cultures and monocultures in testing the toxicity of particulate matter derived from log wood and pellet combustion. PLoS ONE 2018, 13, e0192453. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  58. Karri, V.; Lidén, C.; Fyhrquist, N.; Högberg, J.; Karlsson, H.L. Impact of mono-culture vs. Co-culture of keratinocytes and monocytes on cytokine responses induced by important skin sensitizers. J. Immunotoxicol. 2021, 18, 74–84. [Google Scholar] [CrossRef] [PubMed]
  59. Pleguezuelos, O.; Dainty, S.J.; Kapas, S.; Taylor, J.J. A human oral keratinocyte cell line responds to human heat shock protein 60 through activation of ERK1/2 MAP kinases and up- regulation of IL-1beta. Clin. Exp. Immunol. 2005, 141, 307–314. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Hujiahemaiti, M.; Sun, X.; Zhou, J.; Lv, H.; Li, X.; Qi, M.; Chi, M.; Li, C.; Zhou, Y. Effects of quercetin on human oral keratinocytes during re-epithelialization: An in vitro study. Arch. Oral Biol. 2018, 95, 187–194. [Google Scholar] [CrossRef]
  61. Nakamura, M. Histological and immunological characteristics of the junctional epithelium. Jpn. Dent. Sci. Rev. 2018, 54, 59–65. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Logan, R.M.; Stringer, A.M.; Bowen, J.M.; Gibson, R.J.; Sonis, S.T.; Keefe, D.M. Is the pathobiology of chemotherapy-induced alimentary tract mucositis influenced by the type of mucotoxic drug administered? Cancer Chemother. Pharmacol. 2009, 63, 239–251. [Google Scholar] [CrossRef] [PubMed]
  63. Vaseenon, S.; Srisuwan, T.; Chattipakorn, N.; Chattipakorn, S.C. Lipopolysaccharides and hydrogen peroxide induce contrasting pathological conditions in dental pulpal cells. Int. Endod. J. 2023, 56, 179–192. [Google Scholar] [CrossRef] [PubMed]
  64. Chayawatto, S.; Kornsuthisopon, C.; Linsuwanont, P. Effects of Lps and Hydrogen Peroxide on Cell Viability and Expression of Inflammatory Cytokines in Human Dental Pulp Cells: A Preliminary Study. Procedia Multidiscip. Res. 2025, 3, 44. [Google Scholar]
  65. Leng, S.X.; McElhaney, J.E.; Walston, J.D.; Xie, D.; Fedarko, N.S.; Kuchel, G.A. ELISA and multiplex technologies for cytokine measurement in inflammation and aging research. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2008, 63, 879–884. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  66. Xie, Q.; Liu, B.; Yu, X.; Wei, X.; Xiao, Q. Integration of Immune Cell Signatures and Diagnostic Gene Markers in Pancreatitis: A Comprehensive Study on Therapeutic Targets and Predictive Diagnosis. Hum. Mutat. 2025, 2025, 7694723. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. duPont, N.C.; Wang, K.; Wadhwa, P.D.; Culhane, J.F.; Nelson, E.L. Validation and comparison of luminex multiplex cytokine analysis kits with ELISA: Determinations of a panel of nine cytokines in clinical sample culture supernatants. J. Reprod. Immunol. 2005, 66, 175–191. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Ungaro, C.T.; Brown, S.D.; Wolfe, A.S. Comparative Analysis of Serum Cytokine ELISA and Multiplex Techniques. Biomed. J. Sci. Tech. Res. 2020, 32, 25325–25330. [Google Scholar] [CrossRef]
  69. Gligor, O.; Clichici, S.; Moldovan, R.; Muntean, D.; Vlase, A.M.; Nadăș, G.C.; Matei, I.A.; Filip, G.A.; Vlase, L.; Crișan, G. The Effect of Extraction Methods on Phytochemicals and Biological Activities of Green Coffee Beans Extracts. Plants 2023, 12, 712. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Sultana, B.; Anwar, F.; Ashraf, M. Effect of Extraction Solvent/Technique on the Antioxidant Activity of Selected Medicinal Plant Extracts. Molecules 2009, 14, 2167–2180. [Google Scholar] [CrossRef]
  71. Jacotet-Navarro, M.; Laguerre, M.; Fabiano-Tixier, A.-S.; Tenon, M.; Feuillère, N.; Bily, A.; Chemat, F. What is the best ethanol-water ratio for the extraction of antioxidants from rosemary? Impact of the solvent on yield, composition, and activity of the extracts. Electrophoresis 2018, 39, 1946–1956. [Google Scholar] [CrossRef]
  72. Sun, C.; Wu, Z.; Wang, Z.; Zhang, H. Effect of Ethanol/Water Solvents on Phenolic Profiles and Antioxidant Properties of Beijing Propolis Extracts. Evid.-Based Complement. Altern. Med. 2015, 2015, 595393. [Google Scholar] [CrossRef]
  73. Jeyaraj, E.J.; Lim, Y.Y.; Choo, W.S. Effect of Organic Solvents and Water Extraction on the Phytochemical Profile and Antioxidant Activity of Clitoria ternatea Flowers. ACS Food Sci. Technol. 2021, 1, 1567–1577. [Google Scholar] [CrossRef]
  74. Aremu, A.O.; Pendota, S.C. Medicinal Plants for Mitigating Pain and Inflammatory-Related Conditions: An Appraisal of Ethnobotanical Uses and Patterns in South Africa. Front. Pharmacol. 2021, 12, 758583. [Google Scholar] [CrossRef] [PubMed]
  75. Namsa, N.D.; Tag, H.; Mandal, M.; Kalita, P.; Das, A.K. An ethnobotanical study of traditional anti-inflammatory plants used by the Lohit community of Arunachal Pradesh, India. J. Ethnopharmacol. 2009, 125, 234–245. [Google Scholar] [CrossRef] [PubMed]
  76. Shep, D.; Khanwelkar, C.; Gade, P.; Karad, S. Safety and efficacy of curcumin versus diclofenac in knee osteoarthritis: A randomized open-label parallel-arm study. Trials 2019, 20, 214. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  77. Majeed, M.; Majeed, S.; Narayanan, N.K.; Nagabhushanam, K. A pilot, randomized, double-blind, placebo-controlled trial to assess the safety and efficacy of a novel Boswellia serrata extract in the management of osteoarthritis of the knee. Phytother. Res. 2019, 33, 1457–1468. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  78. Bliddal, H.; Rosetzsky, A.; Schlichting, P.; Weidner, M.S.; Andersen, L.A.; Ibfelt, H.H.; Christensen, K.; Jensen, O.N.; Barslev, J. A randomized, placebo-controlled, cross-over study of ginger extracts and ibuprofen in osteoarthritis. Osteoarthr. Cartil. 2000, 8, 9–12. [Google Scholar] [CrossRef] [PubMed]
  79. Thornton, C.P.; Li, M.; Budhathoki, C.; Yeh, C.H.; Ruble, K. Anti-inflammatory mouthwashes for the prevention of oral mucositis in cancer therapy: An integrative review and meta-analysis. Support. Care Cancer 2022, 30, 7205–7218. [Google Scholar] [CrossRef]
  80. Lima, I.C.G.d.S.; de Fátima Souto Maior, L.; Gueiros, L.A.M.; Leão, J.C.; Higino, J.S.; Carvalho, A.A.T. Clinical applicability of natural products for prevention and treatment of oral mucositis: A systematic review and meta-analysis. Clin. Oral Investig. 2021, 25, 4115–4124. [Google Scholar] [CrossRef]
  81. Lee, C.-C.; Chen, Y.-W.; Kang, Y.-N.; Chen, J.-H.; Chen, C.; Lu, C.-Y.; Huang, T.-W.; Gautama, M.S.N. Efficacy of natural products in preventing oral mucositis resulting from cancer therapies: A network meta-analysis of randomized controlled trials. Crit. Rev. Oncol./Hematol. 2024, 199, 104373. [Google Scholar] [CrossRef]
  82. Balto, H.A.; Al-Manei, K.K.; Bin-Mohareb, T.M.; Shakoor, Z.A.; Al-Hadlaq, S.M. Cytotoxic effect of Salvadora persica extracts on human gingival fibroblast cells. Saudi Med. J. 2014, 35, 810–815. [Google Scholar]
  83. Verma, U.P.; Gupta, A.; Yadav, R.K.; Tiwari, R.; Sharma, R.; Balapure, A.K. Cytotoxicity of chlorhexidine and neem extract on cultured human gingival fibroblasts through fluorescence-activated cell sorting analysis: An in-vitro study. Eur. J. Dent. 2018, 12, 344–349. [Google Scholar] [CrossRef]
  84. Ghasemi, M.; Turnbull, T.; Sebastian, S.; Kempson, I. The MTT Assay: Utility, Limitations, Pitfalls, and Interpretation in Bulk and Single-Cell Analysis. Int. J. Mol. Sci. 2021, 22, 12827. [Google Scholar] [CrossRef]
  85. Madorran, E.; Ambrož, M.; Knez, J.; Sobočan, M. An Overview of the Current State of Cell Viability Assessment Methods Using OECD Classification. Int. J. Mol. Sci. 2025, 26, 220. [Google Scholar] [CrossRef] [PubMed]
  86. Sazonova, E.V.; Chesnokov, M.S.; Zhivotovsky, B.; Kopeina, G.S. Drug toxicity assessment: Cell proliferation versus cell death. Cell Death Discov. 2022, 8, 417. [Google Scholar] [CrossRef] [PubMed]
  87. OECD. Guidance Document on Using Cytotoxicity Tests to Estimate Starting Doses for Acute Oral Systematic Toxicity Tests; OECD Publishing: Paris, France, 2010. [Google Scholar]
  88. Chan, S.M.; Khoo, K.S.; Sit, N.W. Interactions between plant extracts and cell viability indicators during cytotoxicity testing: Implications for ethnopharmacological studies. Trop. J. Pharm. Res. 2015, 14, 1991–1998. [Google Scholar] [CrossRef]
  89. Jantan, I.; Ahmad, W.; Bukhari, S.N.A. Corrigendum: Plant-derived immunomodulators: An insight on their preclinical evaluation and clinical trials. Front. Plant Sci. 2018, 9, 1178. [Google Scholar] [CrossRef]
  90. Stalinska, K.; Guzdek, A.; Rokicki, M.; Koj, A. Transcription factors as targets of the anti-inflammatory treatment. A cell culture study with extracts from some Mediterranean diet plants. J. Physiol. Pharmacol. Suppl. 2005, 56, 157–169. [Google Scholar]
  91. Lampronti, I.; Khan, M.; Bianchi, N.; Borgatti, M.; Gambari, R. Inhibitory effects of medicinal plant extracts on interactions between DNA and transcription factors involved in inflammation. Minerva Biotecnol. 2004, 16, 93. [Google Scholar]
  92. Lampronti, I.; Khan, M.; Bianchi, N.; Ather, A.; Borgatti, M.; Vizziello, L.; Fabbri, E.; Gambari, R. Bangladeshi medicinal plant extracts inhibiting molecular interactions between nuclear factors and target DNA sequences mimicking NF-kB binding sites. Med. Chem. 2005, 1, 327–333. [Google Scholar] [CrossRef]
  93. Eom, T.; Kim, I.-H.; Kim, H.-J.; Choi, Y.; Nam, T.-J. Calystegia soldanella Extract Exerts Anti-Oxidative and Anti-Inflammatory Effects via the Regulation of the NF-κB/Nrf-2 Pathways in Mouse Macrophages. Antioxidants 2021, 10, 1639. [Google Scholar] [CrossRef]
  94. Laurindo, L.F.; Santos, A.R.d.O.d.; Carvalho, A.C.A.d.; Bechara, M.D.; Guiguer, E.L.; Goulart, R.d.A.; Vargas Sinatora, R.; Araújo, A.C.; Barbalho, S.M. Phytochemicals and regulation of NF-kB in inflammatory bowel diseases: An overview of in vitro and in vivo effects. Metabolites 2023, 13, 96. [Google Scholar] [CrossRef]
  95. Jain, S.K.; Micinski, D.; Parsanathan, R. l-Cysteine stimulates the effect of vitamin D on inhibition of oxidative stress, IL-8, and MCP-1 secretion in high glucose treated monocytes. J. Am. Coll. Nutr. 2021, 40, 327–332. [Google Scholar] [CrossRef] [PubMed]
  96. Ghafoor, D.D. Correlation between oxidative stress markers and cytokines in different stages of breast cancer. Cytokine 2023, 161, 156082. [Google Scholar] [CrossRef]
  97. Choi, H.M.; Oh, D.H.; Bang, J.S.; Yang, H.I.; Yoo, M.C.; Kim, K.S. Differential effect of IL-1β and TNFα on the production of IL-6, IL-8 and PGE2 in fibroblast-like synoviocytes and THP-1 macrophages. Rheumatol. Int. 2010, 30, 1025–1033. [Google Scholar] [CrossRef] [PubMed]
  98. Leimert, K.B.; Verstraeten, B.S.E.; Messer, A.; Nemati, R.; Blackadar, K.; Fang, X.; Robertson, S.A.; Chemtob, S.; Olson, D.M. Cooperative effects of sequential PGF2α and IL-1β on IL-6 and COX-2 expression in human myometrial cells. Biol. Reprod. 2019, 100, 1370–1385. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  99. Vaou, N.; Stavropoulou, E.; Voidarou, C.; Tsakris, Z.; Rozos, G.; Tsigalou, C.; Bezirtzoglou, E. Interactions between Medical Plant-Derived Bioactive Compounds: Focus on Antimicrobial Combination Effects. Antibiotics 2022, 11, 1014. [Google Scholar] [CrossRef]
  100. Lankatillake, C.; Luo, S.; Flavel, M.; Lenon, G.B.; Gill, H.; Huynh, T.; Dias, D.A. Screening natural product extracts for potential enzyme inhibitors: Protocols, and the standardisation of the usage of blanks in α-amylase, α-glucosidase and lipase assays. Plant Methods 2021, 17, 3. [Google Scholar] [CrossRef]
  101. Mazumder, K.; Nabila, A.; Aktar, A.; Farahnaky, A. Bioactive Variability and In Vitro and In Vivo Antioxidant Activity of Unprocessed and Processed Flour of Nine Cultivars of Australian lupin Species: A Comprehensive Substantiation. Antioxidants 2020, 9, 282. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of study selection.
Figure 1. Flow diagram of study selection.
Ijms 26 11253 g001
Figure 2. Studies’ compliance rate with the ToxRTool criteria.
Figure 2. Studies’ compliance rate with the ToxRTool criteria.
Ijms 26 11253 g002
Table 1. Characteristics of included studies.
Table 1. Characteristics of included studies.
Author and YearPlant NameExtract TypeExtraction TemperatureConcentrationMedium/VehiclePurityCell TypeInflammatory StimulusStimulation Time/OrderCell Viability Assessment
Zhao et al., 2013 [23]Camellia sinensisaqueousRTSC: 2%
WC:0.01%, 0.005%, 0.0025%, 0.0001%
D.H2O0.45 µm filterOral epithelial cell lineP. gingivalis extractExtract for 2 h, then 24 h after stimulationMTT
Ehrnhöfer-Ressler et al., 2013 [24]Salvia officinalisaqueous95 °CSC: 1%
WC: 1%
H2OnoneGingival fibroblast cell linePMAStimulation for 6 h, then plant extract for 6 hWST-1
Yun et al., 2013 [25]Paeonia suffruticosaethanolicRTSC: 10%
WC: 4%
DMSOnonePrimary gingival fibroblastsLPSExtract for 1 h, then LPS for 24 hMTS
Lombardo Bedran et al., 2015 [26]Camellia sinensisaqueous-SC: 4%
WC: 0.02%
0.01%
D.H2O0.2 µm filterOral epithelial cell lineLPS (A. actinomycetemcomitans)Extract for 2 h, then stimulation for 24 hMTT
Satthakarn et al., 2015 [27]Houttuynia cordataaqueous-SC: not mentioned
WC: 0.02%, 0.01%, 0.005%, 0.0025%
H2OnonePrimary gingival epithelial cellsnoneExtract for 18 hCellTiter-Blue
Yun et al., 2018 [28]Listea japonicaethanolicRTWC: 0.01%ethanolnonePrimary PDL fibroblastLPS (P. gingivalis, T. forythea, T. denticola, F. nucleatum)Dual (extract + LPS) for 0, 1, 3, 6, 12, 24 h
Pre-treatment with the extract for 2 h, then LPS for 24 h
MTT
Pobłocka-Olech et al., 2019 [29]Populus spp.: P. nigra, P. berolinensis, P. lasiocarpamethanolic60 °CSC: 5%
WC:
P.n + P.l: 0.0015%
P.b: 0.00075%
methanolnoneGingival fibroblast cell lineAgNPExtract for 1 h, then stimulation for 18 hMTT
Ben Lagha et al., 2021 [30]Theobroma cacaoethanolic-SC: 2%
WC: 0.025%
0.0125%
0.0063%
DMSO0.22 µm filterOral epithelial cell linesF. nucleatumExtract for 30 min, then stimulation for 24 hMTT
Shiba et al., 2021 [31]Equisetum arvense--WC: 0.00004%Commercially formulated: H2O + butylene glycolnoneOral keratinocyte cell line, monocytic cell lineLPS (P. gingivalis, A. actinomycetemcomitans)Co-stimulation for 6 hNone
Shin et al., 2021 [32]Alpinia katsumadaiethanolic-WC: 0.001%ethanolnoneGingival fibroblast and oral keratinocyte cell lines, murine macrophage cell lineLPS (dental plaque, P. gingivalis)Co-stimulation for 24 hMTT
Wada et al., 2022 [33]Juncus effususaqueous60 °CSC: 200%
WC: 10%
2%
Ultra-pure H2O0.45 µm filter + centrigation 1200× g 15 minOral keratinocyte cell lineLPS (P. gingivalis)Co-stimulation for 24 h, extract for 4 h, then stimulation for 24 hNone
Al-Shibani et al., 2022 [34]Zingiber officinaleaqueousRTSC: 20%
WC: 0.005%
D.H2O0.22 µm filter + centrifugation 3000× g 10 minPrimary gingival fibroblastLPS (P. gingivalis)Extract for 24 h, then LPS for 24 h, and vice versaMTT
Abbreviations: RT—room temperature; SC—stock concentration; WC—working concentration; PMA—phorbol 12-myristate 13-acetate; LPS—lipopolysaccharide; MTT—3-[4;5-dimethylthiazol-2-yl]-2;5 diphenyl tetrazolium bromide; WST-1—water-soluble tetrazolium salts; and MTS—(3-(4;5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium).
Table 2. Summary of bioassay outcomes.
Table 2. Summary of bioassay outcomes.
Author and YearAssayOutcomeExtract Highest and Non-Cytotoxic ConcentrationExtract Lowest Effective Concentration
Zhao et al., 2013 [23]ELISA↓ CCL5, IL-6, IL-8100 µg/mL25 µg/mL
Ehrnhöfer-Ressler et al., 2013 [24]Multiplex immunoassay↓ IL-6, IL-8NDRNDR
Yun et al., 2013 [25]RT-PCR↓ CXCL9, CXCL10, CXCL11NDRNDR
Lombardo Bedran et al., 2015 [26]ELISA↓ IL-8200 µg/mL100 µg/mL
Satthakarn et al., 2015 [27]RT-PCR
ELISA
↓ CXCL5
↓ CCL2
200 µg/mL25 µg/mL
Yun et al., 2018 [28]RT-PCR
ELISA
↓ IL-6, IL-8
↓ IL-6, IL-8
100 µg/mL10 µg/mL
Pobłocka-Olech et al., 2019 [29]RT-PCR
ELISA
↓ IL-6, IL-1ß
↓ IL-6, IL-1ß
NDRNDR
Ben Lagha et al., 2021 [30]ELISA↓ IL-6, IL-8250 µg/mL125 µg/mL
Shiba et al., 2021 [31]RT-PCR
ELISA
↓ TNF-α
↓ secretion of TNF-α
NDRNDR
Shin et al., 2021 [32]ELISA↓ PG-E2, COX-2NDRNDR
Wada et al., 2022 [33]ELISA↓ IL-8, CCL-20NDRNDR
Al-Shibani et al., 2022 [34]xMap Milliplex↓ IL-1ß, IL-8NDRNDR
Abbreviations: ↓ decreased, NDR—no dose–response.
Table 3. Summary and statistics of quality assessment of studies.
Table 3. Summary and statistics of quality assessment of studies.
AuthorScoreComment
Zhao et al., 2013 [23]14reliable without restrictions
Ehrnhöfer-Ressler et al., 2013 [24]14reliable without restrictions
Yun et al., 2013 [25]11reliable with restrictions
Lombardo Bedran et al., 2015 [26]13reliable without restrictions
Satthakarn et al., 2015 [27]9reliable with restrictions
Yun et al., 2018 [28]13reliable without restrictions
Pobłocka-Olech et al., 2019 [29]14reliable without restrictions
Ben Lagha et al., 2021 [30]15reliable without restrictions
Shiba et al., 2021 [31]13reliable without restrictions
Shin et al., 2021 [32]11reliable with restrictions
Wada et al., 2022 [33]13reliable without restrictions
Al-Shibani et al., 2022 [34]11reliable with restrictions
Mean12.6
Median13
Standard Deviation1.7
Range6
Minimum9
Maximum15
Confidence Level (95.0%)1.099
Table 4. Summary of characteristics of plant extracts.
Table 4. Summary of characteristics of plant extracts.
Plant Species (Common Name)FamilyMajor Phytochemical GroupsPrincipal Anti-Inflammatory Mechanisms
Camellia sinensis (Green tea) [36]Theaceae Polyphenols (catechins, EGCG, theaflavins)Antioxidant; inhibits NF-κB, COX-2, and iNOS; activates Nrf2
Salvia officinalis (Sage) [37] LamiaceaeRosmarinic acid, carnosic acid, flavonoids, terpenesNF-κB and MAPK suppression; COX-2 inhibition; antioxidant
Paeonia suffruticosa (Tree peony) [38]PaeoniaceaePaeonol, paeoniflorin, flavonoidsInhibits MAPK and NF-κB; reduces IL-6, TNF-α; antioxidant
Houttuynia cordata (Chameleon plant) [39]SaururaceaeFlavonoids, polysaccharides, and volatile oilsImmunomodulatory; suppresses IL-6, TNF-α, and NO; antiviral
Litsea japonica [40,41]LauraceaeMonoterpenes, flavonoids, lignansInhibits NO and TNF-α production; antioxidant
Populus spp. (poplar) [42]SalicaceaePhenolic glycosides (salicin, populin), flavonoidsCOX inhibition; prostaglandin synthesis suppression
Theobroma cacao (Cocoa) [43]MalvaceaePolyphenols (procyanidins), theobromine, flavanolsAntioxidant; inhibits TNF-α, IL-6; enhances NO for vascular repair
Equisetum arvense (Horsetail) [44]EquisetaceaeSilica, flavonoids, phenolic acidsAntioxidant; mild cytokine suppression; enhances collagen synthesis
Alpinia katsumadai (Katsumada galangal) [45]ZingiberaceaeDiarylheptanoids, flavonoids, terpenesInhibits NO, COX-2, and IL-6; modulates MAPK
Juncus effusus (Soft rush) [46]JuncaceaePhenanthrenes, flavonoids, polysaccharidesSuppresses TNF-α, IL-6; antioxidant
Zingiber officinale (Ginger) [47]ZingiberaceaeGingerols, shogaols, zingeroneCOX and LOX inhibition; NF-κB suppression; antioxidant
Abbreviations: EGCG—epigallocatechin gallate; iNOS—inducible nitric oxide synthase; NO—nitric oxide.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rasheed, I.; Gruber, R. Crude Plant Extracts and Their Anti-Inflammatory Potential in Oral Inflammatory Cell Models: A Systematic Review of In Vitro Studies. Int. J. Mol. Sci. 2025, 26, 11253. https://doi.org/10.3390/ijms262311253

AMA Style

Rasheed I, Gruber R. Crude Plant Extracts and Their Anti-Inflammatory Potential in Oral Inflammatory Cell Models: A Systematic Review of In Vitro Studies. International Journal of Molecular Sciences. 2025; 26(23):11253. https://doi.org/10.3390/ijms262311253

Chicago/Turabian Style

Rasheed, Issam, and Reinhard Gruber. 2025. "Crude Plant Extracts and Their Anti-Inflammatory Potential in Oral Inflammatory Cell Models: A Systematic Review of In Vitro Studies" International Journal of Molecular Sciences 26, no. 23: 11253. https://doi.org/10.3390/ijms262311253

APA Style

Rasheed, I., & Gruber, R. (2025). Crude Plant Extracts and Their Anti-Inflammatory Potential in Oral Inflammatory Cell Models: A Systematic Review of In Vitro Studies. International Journal of Molecular Sciences, 26(23), 11253. https://doi.org/10.3390/ijms262311253

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop