Next Article in Journal
Evaluation of Piperacillin/Sulbactam, Piperacillin/Tazobactam and Cefoperazone/Sulbactam Dosages in Gram-Negative Bacterial Bloodstream Infections by Monte Carlo Simulation
Next Article in Special Issue
The Antibacterial and Antifungal Capacity of Eight Commercially Available Types of Mouthwash against Oral Microorganisms: An In Vitro Study
Previous Article in Journal
Hydrocinnamic Acid and Perillyl Alcohol Potentiate the Action of Antibiotics against Escherichia coli
Previous Article in Special Issue
Antibiotics as Adjunctive Therapy in the Non-Surgical Treatment of Peri-Implantitis: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Induction of Endogenous Antimicrobial Peptides to Prevent or Treat Oral Infection and Inflammation

by
Kimberly A. Morio
1,
Robert H. Sternowski
2 and
Kim A. Brogden
3,*
1
Apex Endodontics, Hiawatha, IA 52233, USA
2
Softronics, Co., Ltd., Marion, IA 52302, USA
3
College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(2), 361; https://doi.org/10.3390/antibiotics12020361
Submission received: 16 January 2023 / Revised: 3 February 2023 / Accepted: 5 February 2023 / Published: 9 February 2023

Abstract

:
Antibiotics are often used to treat oral infections. Unfortunately, excessive antibiotic use can adversely alter oral microbiomes and promote the development of antibiotic-resistant microorganisms, which can be difficult to treat. An alternate approach could be to induce the local transcription and expression of endogenous oral antimicrobial peptides (AMPs). To assess the feasibility and benefits of this approach, we conducted literature searches to identify (i) the AMPs expressed in the oral cavity; (ii) the methods used to induce endogenous AMP expression; and (iii) the roles that expressed AMPs may have in regulating oral inflammation, immunity, healing, and pain. Search results identified human neutrophil peptides (HNP), human beta defensins (HBD), and cathelicidin AMP (CAMP) gene product LL-37 as prominent AMPs expressed by oral cells and tissues. HNP, HBD, and LL-37 expression can be induced by micronutrients (trace elements, elements, and vitamins), nutrients, macronutrients (mono-, di-, and polysaccharides, amino acids, pyropeptides, proteins, and fatty acids), proinflammatory agonists, thyroid hormones, and exposure to ultraviolet (UV) irradiation, red light, or near infrared radiation (NIR). Localized AMP expression can help reduce infection, inflammation, and pain and help oral tissues heal. The use of a specific inducer depends upon the overall objective. Inducing the expression of AMPs through beneficial foods would be suitable for long-term health protection. Additionally, the specialized metabolites or concentrated extracts that are utilized as dosage forms would maintain the oral and intestinal microbiome composition and control oral and intestinal infections. Inducing AMP expression using irradiation methodologies would be applicable to a specific oral treatment area in addition to controlling local infections while regulating inflammatory and healing processes.

Graphical Abstract

1. Introduction

Antibiotics are used throughout all disciplines of dentistry for the treatment of both soft and hard tissue infections [1]. These treatments include infections associated with the pulp and periapical tissues, oral mucosal and oropharyngeal tissues, and oral lesions in immunocompromised patients, especially during radiotherapy and chemotherapy. Antibiotics are also used for prophylaxis against infections in individuals with joint replacements and heart defects prior to teeth cleaning or surgical procedures [1]. While the use of antibiotics has clear benefits, their overuse can alter the composition of the oral microbiomes, create niches on mucosal surfaces for opportunistic microorganisms such as Candida species to colonize, promote the development of antibiotic-resistant microorganisms, and result in complications associated with hypersensitivity reactions and allergic disorders [1,2,3].
Antimicrobial peptides (AMPs) have been recently proposed as alternatives to antibiotics for treating oral infections [4]. However, there have been concerns related to producing and using AMPs as small-peptide therapeutics. AMP molecule stability, toxicity, elimination half-life, and clearance are obstacles that have hindered the development of AMPs [5]. There can also be difficulties in correctly folding the complex three-dimensional structures seen in larger AMPs [6,7]. Finally, the antimicrobial activities of AMPs are modest in host environments. AMPs do not withstand the physiological conditions in the host environment, including the activity of host proteinases, differences in site pH, differences in ionic conditions, and the presence of polyanionic glycosaminoglycans [8,9].
Host-directed therapy (HDT) is a concept proposed to boost innate immune mechanisms for controlling antibiotic-resistant microorganisms [10,11]. Included in HDT is the use of inducers or “elicitors” to express endogenous AMPs for treating infection and inflammation [6,10,12,13,14,15,16,17,18].
In this study, we hypothesized that this approach could be used to induce AMP transcription and expression in the oral cavity. We started by conducting an initial literature search to identify AMPs expressed in the oral cavity. Human neutrophil peptide (HNP)-1, -2, and -3 defensins [19], the human beta defensins (HBD) 1, 2, and 3 [16], and the cathelicidin AMP (CAMP) gene product LL-37 [9] were found to be among the most prominent AMPs expressed by oral cells and tissues [20]. Next, we conducted a second literature search to identify the approaches that could be applied to the transcription and expression of HNPs, HBDs, and LL-37 in oral cells and tissues. Finally, we conducted a third literature search to identify the additional functions that induced AMPs could have in regulating oral inflammation, immunity, tissue healing, and pain. The results of these three searches suggest that approaches to induce AMP transcription and expression are feasible and that induced AMP expression in the oral cavity would create an environment rich in molecules with antimicrobial activity able to regulate a variety of host defense activities and orchestrate tissue recovery events.

2. AMPs in the Oral Cavity

To date, between 3000 [5] and 5000 [21] natural and synthetic AMPs have been identified throughout the Animalia, Plantae, Fungi, Protista, and Monera kingdoms. These AMPs are tracked in 11 general and 9 specific databases [7]. Ramazi et al. noted that AMPs are a diverse family of peptides and have historically been grouped together based on their species of origin, similarities in their specific activities, similarities in their three-dimensional structures, similarities in their amino acid sequences, and the functions of their activities [7]. The availability of sequenced data across species can now be mined using computational methods and machine learning algorithms to identify potentially new AMPs. These algorithms can also be used to design new AMPs with novel antimicrobial activities [7].

2.1. AMPs Expressed in Oral Tissues

AMPs are expressed in cells and tissues of the oral cavity [22]. We searched the PubMed literature database using oral tissues, tongue, tonsils, salivary glands, gingival tissues, oral epithelium, sulcular epithelium, junctional epithelium, saliva, gingival crevicular fluid, antimicrobial peptides, defensins, cathelicidins, HBD, HNP, and LL-37 as search terms. These terms were linked in various combinations, using Boolean operators to identify AMPs reported to be expressed in oral tissues and present in oral secretions. The literature was screened for potential articles that were downloaded and read by the investigators, who selected articles that were within the search objectives for this section.
In oral tissues, HNP-1-3, HBD1-2, and S100A7 are produced by the tongue [23,24,25], HNP-4, HBD1-3, LL-37, and LEAP-1-2 are produced in the palatine tonsils [26], and HBD1-2 and LL-37 are produced by the salivary glands [23,27,28,29]. In gingival tissues, HBD1-2 are among the peptides produced by the oral epithelium, HNP-1-3 and HBD1-2 are among those produced by the sulcular epithelium, and HNP-1-3 are among those produced from the junctional epithelium [30]. LL-37, which occurs in the sulcular epithelium and junctional epithelium, is thought to be a product from neutrophils that might be present [30].
AMPs are also transcribed and expressed in deeper sites and dental pulp, as these tissues have the capacity to produce AMPs [31,32,33,34]. DEFB1 and DEFB4B transcription and HBD1 and HBD2 expression were detected by RT-PCR in pulp tissue and detected by immunohistochemical staining in the cytoplasm of odontoblasts [31].

2.2. AMPs in Oral Secretions

Of the more than 2290 proteins detected in saliva [35,36], approximately 46 or more have reported antimicrobial activities [22,37]. Similarly, of the 100–200 proteins detected in the gingival crevicular fluid [38], seven or more are proteins have reported antimicrobial activities [39]. The physiological ranges of AMP concentrations in the saliva are high enough to support their claims as antimicrobial agents and regulators of inflammatory, immune, and healing processes (Supplemental Table S1). Of the major AMPs present, saliva contains 0.26–11.37 μM HNP-1, 0.03–5.39 μM HNP-2, 0.00–0.77 μM HNP-3, 0.00–0.04 μM HBD1, 0.00–0.03 μM HBD2, 0.00–1.36 μM HBD3, 0.56–1.11 μM LL-37 in adults and 4.45 μM LL-37 in infants [8,22,23,40,41].

3. Induction of Endogenous AMPs

AMPs in cells, tissues, and salivary tissues are induced by a variety of nutritional, hormonal, or physical stimuli (Scheme 1, Table 1). We searched the PubMed literature database using micronutrients, trace elements, elements, vitamins, nutrients, macronutrients, monosaccharides, disaccharides, polysaccharides, amino acids, pyropeptides, proteins, fatty acids, proinflammatory agonists, hormones, ultraviolet (UV) C, UVB, UVA, light, near infrared radiation (NIR), antimicrobial peptides, defensins, cathelicidins, HBD, HNP, and LL-37 as search terms. These terms were also linked in various combinations using Boolean operators to identify molecules that induce AMP transcription and expression in cells, keratinocytes, oral cells, and oral tissues. Results from this search were downloaded and read by the authors. These results formed the basis for Table 1, Figure 1, Supplemental Tables S2 and S3, and Supplemental Figures S1–S4.

3.1. Micronutrients

3.1.1. Trace Elements

Selenium [42,43,44], zinc [42,43], copper [42,43], and iron [42,43] are among the trace elements that contribute to immune competence, including maintenance of the skin barrier function, antibody production, and cellular response. Zinc is also commonly involved in signal transduction [45]. These elements are abundant in whole grains, dairy products, and seafood, and can also be consumed as supplements to boost immune function.
Experimentally, trace elements induce AMP transcription and expression (Table 1). Chickens given supplemental selenium in their diet had increased transcripts of avian β-defensins (AvBD) 6, 8, and 13 in their gastrointestinal tract and spleen [46]. Caco-2 cells treated with 20 and 50 μM zinc had increased expression of LL-37 [47]. With 20 μM zinc, LL-37 concentrations increased from 0.29 to 0.58 ng/mL in 48 h [47]. Zinc is thought to induce the phosphorylation of the extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 MAP kinases [47], which regulate LL-37 expression (Supplemental Figure S1, Figure 1). Copper binding motifs built into the amino terminus of native AMPs, CM15, and citropin 1.1 presented increased antimicrobial activity against antibiotic-resistant microorganisms [48,49]. Finally, hepcidin is a cysteine-rich antimicrobial peptide that is expressed in the liver in response to iron loading and inflammation [50].
Table 1. Inducers of endogenous antimicrobial peptides (AMPs) to treat oral infection and inflammation.
Table 1. Inducers of endogenous antimicrobial peptides (AMPs) to treat oral infection and inflammation.
Family of InducersInducer of AMP Transcription or ExpressionReferences
Micronutrients
Trace elementsSelenium[46]
Zinc[47]
Copper[48,49]
Iron[50]
ElementsCalcium[51]
VitaminsA (retinoic acid)[52,53]
B3 (niacin)[54,55]
C (ascorbic acid)[56]
D, D3, and calcitriol (1,25(OH)2D3)[57,58,59,60,61,62,63,64,65,66,67,68]
E (alpha-tocopherol)[69]
Nutrients and macronutrients
Mono-, di-, and polysaccharidesGlucose[56,70,71]
Lactose[72]
β-glucans[73]
Amino acids, pyroglutamyl peptides, and proteinsArginine[74]
Isoleucine[74,75,76,77]
Pyroglutamyl peptides, pyroglutamyl dipeptides, and pyroglutamyl polypeptides[78,79]
Bovine serum albumin (BSA)[74]
Free fatty acids (FFA) and histone deacetylase (HDAC) inhibitorsShort FFA (≤5 carbons) including butyrate and phenylbutyrate[15,57,72,80,81,82,83,84,85,86,87,88,89,90,91,92]
Medium FFA (6–11 carbons) including hexanoate and heptanoate[15]
Long FFA (≥12 carbons) including laurate, palmitate, and oleate[15,93]
Proinflammatory agonists
Pam3csk4 peptide (Toll-like receptor (TLR) 2)[61]
Lipopolysaccharide (LPS) (TLR4)[23,26,94,95,96]
CpG (TLR9)[97]
IL-1β[23,98]
TNF-α[95,96,98]
IFN-γ[98]
Hormones
Triiodothyronine (T3)[86]
Thyroxine (T4)[86]
Irradiation
UltravioletUltraviolet C, 100–280 nm[56]
Ultraviolet B, 280–315 nm[94,95,96,99,100,101,102]
Ultraviolet A, 340–400 nm[56]
Red lightLaser, 625 nm[103]
Near infraredLaser, 810 nm[104]

3.1.2. Elements

Calcium is an abundant mineral found in many food groups, including vegetables and dairy products. Calcium can be consumed as a supplement to improve skeletal, cardiovascular, and neuromuscular health and is commonly involved in cellular signaling as an enzyme cofactor or signal transducer [105].
Experimentally, calcium is known to have direct effects on innate and adaptive immune functions (Table 1). For example, when added to keratinocytes, 1.7 mM calcium can induce increased levels of DEFB4B and DEFB103A transcription and HBD2 and HBD3 expression [51]. One modeled pathway [106] featured calcium signaling through EGFR and the MAPK signaling module to ERK1/2 (Figure 1).

3.1.3. Vitamins

Vitamins are micronutrients essential for normal growth and development. They are not synthesized but are required in small quantities in the diet. They form a diverse family containing vitamins A (retinoic acid), B1–B12, C (ascorbic acid), D, and E (alpha-tocopherol).
Vitamins are involved in AMP transcription and peptide expression (Table 1). Vitamin A (retinoic acid) promotes the expression of HBD2, HBD3, LL-37, and RNase7 in alveolar macrophages and respiratory epithelial cells [52],the expression of BD3 in mouse skin [53], and the expression of resistin, a small cysteine-rich AMP, in epidermal keratinocytes [107].
Little information is available on the roles of vitamins B1 (thiamin), B2 (riboflavin), B5 (pantothenic acid), B6 (pyridoxine), B7 (biotin), B9 (folic acid), or B12 (cyanocobalamin) in AMP transcription and peptide expression. However, vitamin B3 (niacin) promoted the expression of porcine β defensin (pBD) 2, protegrin 1–5, and PR39 in the gastrointestinal tract of weaned pigs, which contributed to their elimination of enterotoxigenic Escherichia coli K88 infections [54,55].
Vitamin C (ascorbic acid) induced DEFB1 transcription in normal keratinocytes [56], and vitamin E (alpha-tocopherol) induced HBD1-2 expression in human gingival fibroblasts stimulated with Porphyromonas gingivalis lipopolysaccharide (LPS) [69].
Vitamin D has pleotropic activities, and it is important in the cellular uptake and metabolism of calcium, phosphate, and other minerals necessary for mineral metabolism and bone development [108,109]. Vitamin D regulates inflammatory events [110], prevents cancer [110], regulates endocrine system events, regulates central nervous system events, and initiates and regulates innate and adaptive immune responses [65,67]. The two main isoforms are ergocalciferol (vitamin D2) and cholecalciferol (vitamin D3) [109].
Some vitamin D3 comes from the dietary intake of fish, eggs, and vegetables high in vitamin D or dairy products and fruit juices fortified with vitamin D [109]. However, most vitamin D comes from the conversion of 7-dehydrocholesterol to pre-vitamin D3 by ultraviolet B (UVB) as a result of normal exposure to sunlight [108,109,111]. Pre-vitamin D3 can also come from the photochemical conversion of lumisterol and tachysterol [108,109,111]. Pre-vitamin D3 then becomes vitamin D3 and is hydroxylated in the liver to form 25(OH)D and hydroxylated in the kidneys to form 1,25(OH)2D3 (e.g., 1,25-dihydroxvitamin D3 or calcitriol) [111], while 1,25(OH)2D3 (calcitriol) is the physiologically active form [109].
Important to this review is the ability of vitamin D to regulate antimicrobial innate immune responses (Figure 1). This regulation occurs through the vitamin D receptor (VDR) and induces hCAP-18 gene expression [59], CAMP gene expression [58,66], and HBD2 gene expression [65]. The promoters of these genes contain consensus vitamin D response elements (VDRE) that mediate 1,25(OH)2D3-dependent gene expression [65]: 1,25(OH)2D3 induces antimicrobial peptide gene expression in isolated human keratinocytes [65], monocytes [65], neutrophils [65] and neutrophil progenitors [62], myeloid cells [66], EBV-transformed B-lymphocytes [62], and human cell lines [65].
Signaling (Supplemental Figure S2) occurs through the induction of pathogen recognition receptors (PRRs) (e.g., Toll-like receptor or TLR) resulting in the downstream expression of CYP27B1 (1α hydroxylase), an enzyme that converts calcifediol to 1,25(OH)2D3 (calcitriol) [67,112,113]. 1,25(OH)2D3 (calcitriol) then combines with VDR and VDRE [109]. This conversion initiates CAMP and DEFB4B transcription, resulting in LL-37 and HBD2 expression [67].
The use of micronutrients (trace elements, elements, and vitamins) to induce endogenous AMP transcription and expression is unlikely to have side effects if such micronutrients are obtained as consumable nutrients in foods. However, if consumed in excess or misused as supplements, micronutrients can induce a variety of off-target side effects. An estimated 10,176 of 32,000 (31.8%) emergency department visits per year (2004–2013) were estimated to result from adverse events related to the consumption of micronutrients [114]. Within this group, 1504 (4.7%) visits were associated with iron-induced nausea, vomiting, and abdominal pain; 1088 (3.4%) visits were associated with calcium-induced swallowing problems; and 5376 (16.8%) visits were associated with vitamin consumption. Alterations in calcium metabolism affect cellular signaling leading to changes in cell proliferation, apoptosis, autophagy, and cancer [115]. Supplements of vitamin B6 (>500 mg/d) can result in photosensitivity and neurotoxicity; supplements of vitamin E (800–1200 mg/d) can result in diarrhea, weakness, and blurred vision; and excess supplements of vitamin A can result in a loss of bone mineral density with an increase in fracture risk [116].

3.2. Nutrients and Macronutrients

Many nutrients and macronutrients can induce AMP transcription and expression [11,17]. These nutrient types include the broad categories of saccharides and polysaccharides, amino acids and proteins, and short-, medium-, and long-chain fatty acids (Table 1, Figure 1).

3.2.1. Mono-, Di-, and Polysaccharides

Glucose, lactose, and complex polysaccharides such as β-glucans all induce the expression of AMPs (Table 1). Glucose induces DEFB1 transcription in normal keratinocytes [56]. Lactose in human breast milk is thought to be involved in immune protection of the gastrointestinal tract of nursing infants by upregulating CAMP transcription and LL-37 expression [72]. Lactose isolated from human milk was found to induce CAMP transcription and LL-37 expression in the colonic epithelial cell line T84, THP-1 monocytes, and macrophages [72]. LL-37 transcription was suppressed by two different p38 antagonists, suggesting signaling via the p38 signaling pathway (Supplemental Figure S1).
β-glucans have potent immune regulatory functions [73]: they regulate the inflammatory and antimicrobial activities of neutrophils and macrophages. β-glucans are polymers of D-glucose that are linked by β-glycosidic bonds forming structural chains. These chains are found throughout the Plantae, Fungi, and Monera kingdoms as energy stores in plants and algae and as structural components in the cell walls of fungi, yeasts, and bacteria [117]; β-1,3-1,6-glucans bind a number of cellular receptors, including dectin-1, CR3, lactosylceramide, TLR2, 4, and 6, and CD36. After exposure to yeast β-D-glucans, AvBD, cathelicidin, and LEAP-2 gene expression was found to be altered in chickens [73]. Macrophages, splenocytes, and heterophils presented increased phagocytosis and bacterial killing activities. These glucans were observed to amplify humoral and cell-mediated immune responses, effectively clearing enteric infections in treated birds [73].

3.2.2. Amino Acids, Pyroglutamyl Peptides, and Proteins

Amino acids regulate gene expression via the mTORC1, AMPK, and MAPK signaling pathways [11,118]. Amino acids involved in the cell signaling pathways include arginine, glutamine, glutamate, glycine, leucine, isoleucine, proline, and tryptophan. Of these, arginine, isoleucine, and several of its analogues can specifically induce epithelial HBD expression [74,75,76]. For example, when added to HCT-116 human colon cells, arginine and isoleucine can induce increased levels of DEFB1 transcription and HBD1 expression. The administration of l-isoleucine induced a significant increase in HBD3 and HBD4, which was associated with decreased bacillary loads and tissue damage in animals infected with the Mycobacterium tuberculosis antibiotic-sensitive strain H37Rv and the M. tuberculosis strains of MDR clinical isolates, suggesting that the induction of HBDs might aid in controlling this infection [76]. Induction was transcriptional in nature (Supplemental Figure S3, Figure 1) and involved activation of the NFκB/rel family [75].
Pyroglutamyl peptides form a family of bioactive molecules [78,79] containing a free amino terminal glutaminyl group that cyclizes to form a lactam (e.g., a pyroglutaminyl group abbreviated as pyroGlu) [78]. PyroGlu peptides are common in fermented foods fish, sake, soybean paste (miso), and soy sauce (shoyu), as well as the protein lysates of corn or wheat gluten [78,79]. These peptides often remain in protein hydrolysates due to their resistance to proteinase and peptidase digestion [78]. Members include pyroGlu-Leu (pyroglutamyl leucine), pyroGlu-Tyr (pyroglutamyl-tyrosine), pyroGlu-Asn-Ile (pyroglutamyl-asparaginyl-isoleucine), and pyroGlu-Asn-Ile-Asp-Asn-Pro (pyroglutamyl-asparaginyl-isoleucyl-asparagyl-asparaginyl-proline).
PyroGlu peptides are thought to enhance the production of AMPs in the gastrointestinal tract to help maintain normal intestinal microbiota (Table 1). They regulate the colonic microbiota composition, reverse colonic dysbiosis in animals on high-fat diets, and restore colonic dysbiosis in dextran sulfate-induced colitis in mice. For example, in rats fed a high-fat diet, pyroGlu given orally increased the induction of defensin alpha 9 and rattusin, which was associated with the suppressed proliferation of Gram-positive bacteria (Firmicutes) [78].
Bovine serum albumin (BSA) has also been found to regulate DEFB1 transcription and HBD1 expression [74]. BSA is a globular non-glycosylated protein isolated from bovine serum [119]. It is composed of 583 amino acids and has a mass of 66,400 Da [119]. When added to HCT-116 human colon cells, BSA induced increased levels of DEFB1 transcription and HBD1 expression. Induced DEFB1 transcription was related to c-myc over-expression, which suggested that DEFB1 transcription may be regulated via c-myc signaling [74].

3.2.3. Free Fatty Acids

Free fatty acids (FFAs) form a large family of carboxylic acids of saturated or unsaturated aliphatic hydrocarbon chains. FFAs are formed as breakdown products from the hydrolysis of fats and oils and readily occur in the oral cavity, in the gastrointestinal tract, and on the skin. FFAs are grouped by size into those containing five or fewer carbons (short-chain fatty acids or SCFAs), six to eleven carbons (medium chain fatty acids or MCFAs), or twelve or more carbons (long-chain fatty acids or LCFAs).
FFAs have very potent innate immune properties, including direct antimicrobial activities against viruses, bacteria, and fungi [120] and the ability to induce AMP transcription and expression (Table 1). Generally, FFAs shorter than four carbons or longer than seven carbons have only a marginal ability to induce CAMP transcription and LL-37 expression [15]. In laboratory studies, SCFAs including butyrate, phenylbutyrate, glyceryl tributyrate, benzyl butyrate, and valerate were found to be potent inducers of LL-37 [15,57,72,80,82,83,84,85,86,87]; MCFAs including hexanoate (six carbons) and heptanoate (seven carbons) are moderate inducers of LL-37 [15]; and LCFAs including laurate, palmitate, and oleate are marginal inducers of LL-37 [15] but moderate inducers of HBD2 expression [93]. Sodium butyrate upregulates LL-37 gene expression in PNEC cells and NCI-H292 cells [80] and induces pBD2, pBD3, epididymis protein 2 splicing variant C (pEP2C), and protegrin expression in porcine IPEC-J2 intestinal epithelial cells, 3D4/31 macrophages, and primary monocytes [83]. Phenylbutyrate increases CAMP transcription in VA10, HT-29, A498, and U937 cell lines [87]. DEFB1 transcription was increased in the lung epithelial cell line VA10, but decreased in the monocytic cell line U937 [87]. Glyceryl tributyrate, benzyl butyrate, and 4-phenylbutyrate were comparable to butyrate in activity [83]. Valerate induced CAMP gene expression in HT-29 and U937 cell lines [15]. Hexanoate and heptanoate induced LL-37 gene expression in HT-29 and U937 cell lines [15] and laurate, palmitate, and oleate induced expression of HBD2 in human sebocytes [93]. In clinical trials, sodium butyrate provided protection for individuals infected with shigellosis [85].
The induction of AMP transcription and expression by FFAs occurs through multiple pathways (Supplemental Figures S1–S3 and Figure 1). Phenylbutyrate and its analogue alpha-methylhydrocinnamate induce CAMP transcription through the ERK1/2 and c-Jun N-terminal kinase (JNK) signaling pathways [87]. Butyrate and valproate also act as histone deacetylase (HDAC) inhibitors (Supplemental Figure S4, Figure 1). HDAC is an enzyme that removes the acetyl group from DNA histones, making it less accessible to transcription factors. HDAC regulates a number of important cellular proteins, including histones h3 and h4, MMP9, and others, including AMPs. The inhibition of this enzymatic process by butyrate and valproate likely makes DNA more accessible to transcription factors, thus increasing DEFB4B transcription (Supplemental Figure S4).

3.2.4. Foodstuffs

Foodstuffs contain complex macronutrients that can induce AMP transcription in oral cells, suggesting that their consumption contributes to the presence of AMPs in the oral cavity (see Section 2.1 and Section 2.2 above). For example, human milk oligosaccharide 3-fucosyllactose with and without 2’-fucosyllactose induced DEFB4B transcription in human gingival cells, suggesting that these oligosaccharides may stimulate the oral mucosal expression of HBD2 without inducing a proinflammatory response [121]. Avocado extracts contain natural sugars that induce DEFB4B and DEFB103B transcription in keratinocytes [122,123]. Fruit extracts containing gallic acid induce the expression of HBD2 in primary human gingival epithelial cells [124].
Green tea extracts containing polyphenols induce the expression of pBD1 and pBD2 in porcine jejunal epithelial cells IPEC-J2 [125] and DEFB1, as well as DEFB4B expression in gingival epithelial cells [126]. Black tea extracts containing theaflavins induce HBD1, 2, and 4 expression in oral epithelial cells [127]. Peonies are a component of herbal medicine, and their extracts containing paeoniflorin upregulate HBD2 expression in human bronchial epithelial cells through the p38 MAPK, ERK1/2, and NF-κB signaling pathways [128].
Finally, foodstuffs containing complex macronutrients that can induce AMP transcription have been proposed to protect the overall general health of the oral cavity and gastrointestinal tract. In a unique synergy, flours from amaranth, millet, soybean, and sesame grains increased DEFB4B transcription in human HFK cells co-stimulated with E. coli [12].
Overall, the consumption of nutrients, macronutrients, and foodstuffs is thought to be generally safe [116]. The use of nutrients, macronutrients (mono-, di-, and polysaccharides, amino acids, pyropeptides, proteins, and fatty acids), and foodstuffs to induce endogenous AMP transcription and expression would be beneficial and suitable for long-term health protection. The specialized metabolites or concentrated extracts that are formulated as dosage forms would provide therapeutic consideration. However, a few can induce a variety of off-target side effects if consumed in excess. Arginine (>6 g/day), for example, can induce nausea and diarrhea [129], and excess consumption of protein powders can result in ketosis [116].

3.3. Proinflammatory Agonists

These agonists are well-known and potent inducers of proinflammatory and AMP responses in a variety of cells and tissues (Supplemental Figure S3, Figure 1). They involve a vast number of microbial antigens and products with unique pathogen-associated molecular patterns (PAMPs), as well as host-derived proinflammatory cytokines released from damaged, stressed, or abnormal cells and tissues. To recognize and respond to the presence of these molecules, host cells contain both surface PRRs and cytokine receptors. This process also occurs in oral tissues [130]. These PRRs encompass the TLR family, Nod-like receptor/nucleotide-binding oligomerization domain-like receptor (NLR), retinoic acid-inducible gene I-like receptor (RLR), C-type lectin receptor (CLR), and AIM2-like receptor (ALR) groups. PRRs that initiate AMP transcription include TLR2-6 and 9 [131].
Lipoproteins, lipoteichoic acid, triacyl bacterial lipopeptides, and fungal zymosans are TLR2 and TLR6 ligands [61]; double stranded viral RNA is a TLR3 ligand; and bacterial LPS [23,26,95] and P. gingivalis recombinant hemagglutinin B (rHagB) [132] are TLR4 ligands. LPS induces DEFB1, DEFB4B, and CAMP transcription and HBD1, HBD2, and LL-37 expression in a variety of cell types [95,96]. Unmethylated DNA CpG is a TLR9 ligand.
Proinflammatory cytokines including IL-1β and TNF-α bind and signal via IL1R [133], TNFR-1, and TNFR-2 [134]. ILR1 belongs to the Toll-IL1-receptor (TIR) superfamily, and TNFRs belong to the TNF receptor superfamily. IL-1β and TNF-α induce DEFB1, DEFB4B, and CAMP transcription and HBD1, HBD2, and LL-37 expression in a variety of cell types [95,96]. Agonist–receptor engagement then results in the initiation of specific cascades through the MAPK [131] and NF-κB [131] signaling modules (Supplemental Figure S3, Figure 1).
The use of proinflammatory agonists such as LPS to induce endogenous AMP transcription and expression is unlikely to be a popular or feasible approach. Systematic LPS administration can systemically induce inflammation, proinflammatory cytokine production, fever, and septic shock [135]. LPS present in food and food supplements, however, seems to be tolerated [135].

3.4. Thyroid Hormones

Thyroid hormones regulate metabolism and immune functions [136], and an excess or deficiency of these hormones can have profound effects in the oral cavity [137]. It is possible that these hormones may also induce the transcription and expression of AMPs in the oral cavity (Table 1). This activity is supported by a report showing that colonic epithelial cells treated with thyroid hormones triiodothyronine (T3) and thyroxine (T4) induced CAMP transcription [86]. LL-37 expression was induced by both T3 (2.5 nM–1.0 μM for 3–30 h) and T4 (2.5–10 nM for 24 h) [86].
There are a number of possible mechanisms for this phenomenon, one of which is receptor mediated. T3 and T4 bind to cellular thyroid hormone receptors (TRs), which are ligand-activated transcription factors (TFs). TRs then bind to thyroid hormone response elements (TREs) together with retinoid X receptors (RXR) in the promotor regions of their target genes to regulate gene expression. A good example is the presence of TR/TRE binding sites in the promoter region of the CAMP gene [86]. Another mechanism may repress HDACs similar to butyrate and phenylbutyrate (Supplemental Figure S4). Finally, RXR may also be involved with VDR and VDRE in vitamin D-induced CAMP and DEFB4B transcription and LL-37 and HBD2 expression [138].
While thyroid hormones are associated with changes in the oral cavity, altering thyroid hormone concentrations to induce endogenous AMP transcription and expression would not likely be a valid approach. Any advantages would be offset by numerous and more severe consequences that could be associated with metabolic complications [137].

3.5. Irradiation

AMP transcription and expression is induced in cells and tissues after exposure to radiation within select ranges in the electromagnetic spectrum [139,140,141]. Irradiation induces AMP expression in normal human skin and regulates AMP expression in the skin of individuals with dermal diseases (Table 1 and Table 2). It also has the potential to induce AMP expression in oral tissues for treatment of oral infections and inflamed areas to enhance healing processes [18,142].

3.5.1. UVC

The use of UVC irradiation is relatively new compared to that of other areas in the electromagnetic spectrum. UVC irradiation (100–280 nm) alone is directly antimicrobial and rapidly kills microorganisms but is not toxic to oral cells [143,144]. Irradiation also induces the transcription and expression of chemokines, cytokines, and growth factors that are beneficial to endodontic tissue regeneration [143]. In exposed cells, UVC induces AMP expression. For example, Antonio Cruz Diaz et al. found that UVC exposure of human keratinocytes for 5 and 10 min induced CAMP and DEFB1 transcription [56].
UVC is thought to signal through the p38 MAPK, ERK1/2, and JNK pathways [18]. Abbreviated pathways are shown in Supplemental Figure S1 and Figure 1. UVC stimulates EGFR signaling to p38 MAPK. UVC also activates EGFR and signals through SRC > RAS > raf > MAP2K1 to ERK1/2. UVC can also activate PKC and MAP2K1 to ERK1/2. ERK1/2 then activates transcription factor AP-1. Alternately, UVC can activate sphingomyelinase, which activates JNK. Activated (phosphorylated) JNK in turn signals to AP-1.

3.5.2. UVB

UVB irradiation (280-315 nm) alone is directly antimicrobial but also induces cells and tissues to express AMPs (Supplemental Table S2). In cultured cells, irradiation induces DEFB4B, DEFB103A, RNASE7, S100A7, and CAMP transcription and HBD2, HBD3, RNase7, S100A7, and hCAP18/LL-37 expression [100,102,104]. For example, keratinocytes from neonatal foreskin and human keratinocyte HaCat cells treated with UVB irradiation (30 or 100 mJ/cm2) induced DEFB1 and DEFB4B transcription, whereas transformed human keratinocyte A431 cells did not [96]. In tissue biopsies, UVB irradiation increased the expression of HBD and LL-37 [95]; upregulated skin chemerin [99], a potent chemoattractant with antimicrobial activity [145]; and induced levels of CAMP transcription [102].
In clinical studies, UBV irradiation induces AMP expression in normal human skin and regulates AMP expression in the skin of individuals with atopic eczema, psoriasis, dermatitis, and vitiligo [141]. UVB increased hCAP18 and vitamin D receptor expression in healthy skin biopsies at 24 h posttreatment [102]. In patients with atopic eczema, HBD1 expression was decreased, and HBD2 expression was increased, in the epidermis, which is thought to lead to the recurrent skin infections seen in this condition [146]. After treatment of atopic eczema skin with NB-UVB phototherapy, values returned to normal. HBD1 expression was increased, and HBD2 expression was decreased, compared to responses in healthy controls [146].
UVB is thought to signal through the p38 MAPK, ERK1/2, and JNK pathways [18]. Abbreviated pathways are shown in Supplemental Figure S1 and Figure 1. UVB can stimulate the phosphorylation of p38 MAPK or stimulate EGFR signaling to p38 MAPK. UVB activates ERK1/2 in multiple pathway combinations: PKC > ERK1/2; PCK > JNK > ERK1/2; PI3K > PKC > ERK1/2; and/or PI3K > PKC > JNK > ERK1/2. Activated ERK1/2 then signals to AP-1. UVB activates JNK in multiple pathway combinations: UVB activates PKC > JNK and/or PI3K > PKC > JNK. Activated (phosphorylated) JNK, in turn, signals to AP-1.

3.5.3. UVA

UVA (340–400 nm) is a longer wavelength that also regulates AMP transcription and expression in keratinocytes. In human keratinocytes, Cruz Diaz et al. found that 5 min of UVA irradiation induced DEFB1 transcription, whereas 10 and 20 min of UVA irradiation reduced DEFB1 transcription [56].
Clinically, UVA can regulate AMP transcription, which is correlated with clinical improvement of localized scleroderma [147]. Prior to treatment, DEFB1, DEFB4B, and DEFB103A transcription levels were higher in localized scleroderma compared to normal skin. After treatment with UVA, clinical scores and areas of treatment improved. DEFB1 transcription decreased in localized scleroderma compared to normal skin. DEFB103A transcription decreased in localized scleroderma but increased in normal skin [147].
UVA signals through the p38 MAPK, ERK1/2, and JNK pathways [18]. Abbreviated pathways are shown in Supplemental Figure S1 and Figure 1. UVA can phosphorylate p38 MAPK. Activated p38 MAPK then signals to STAT1. UVA can activate EGFR and signal to ERK1/2. UVA can also signal to PLC with Ca2+ to PRKCA through RAS to ERK1/2. ERK1/2 then signals through RPS6KA5 to STAT1. Finally, UVA can activate JNK directly. Activated (phosphorylated) JNK then signals to AP-1 and STAT1.

3.5.4. Red Light

The red-light region occurs within the visible spectrum at 625–740 nm. Lasers emitting light at 625 nm can induce CAMP and DEFB1 transcription and LL-37 expression in immortalized gingival fibroblasts infected with P. gingivalis [103].

3.5.5. Near-Infrared Irradiation (NIR)

NIR occurs just outside the visible spectrum at 800 to 2500 nm. NIR at 810 nm induces higher DEFB4B transcription in oral fibroblast cells than in oral keratinocytes [104]. Transcription was higher at lower fluences, and the induction of HBD2 occurred via activation of the TGF-B1 pathway and signaling through the Smad and non-Smad pathways [104].
Using irradiation to induce AMP transcription and expression would be an attractive approach. Irradiation can be strictly controlled in the clinic and used locally in the oral cavity. This measure can avoid systemic or long-term over exposure. The dental community has been using UV irradiation in the oral cavity for over 50 years to whiten teeth and polymerize compounds used in restorations and fillings [142]. Since the 1980s, narrow-band UVB (311–312 nm) has been used successfully to reduce the erythema response in psoriasis patients, and UVA (340–400 nm) has been used to treat atopic dermatitis [141]. However, safety can be compromised if exposure doses exceed therapeutic levels.

3.6. Synergy among Inducers

Increases in AMP transcription and expression are induced in cells after exposure to multiple AMP inducers (Supplemental Table S3). Co-exposure to lactose and butyrate or phenylbutyrate [86], butyrate and cAMP [82], phenylbutyrate and vitamin D3 [87], and vitamin D3 and LPS [65] were all found to induce higher AMP expression levels. In a well-studied example of synergy among lactose and phenylbutyrate, the regulatory pathways were assessed in their induction of the CAMP gene [86]. The colonic epithelial cell line HT-29 was treated with lactose, phenylbutyrate, and lactose + phenylbutyrate. Induced proteins assessed via mass spectroscopy and pathway analysis showed no additional metabolic pathways detectable in colonic cells treated with lactose + phenylbutyrate compared to proteins/pathways activated in cells treated with lactose or phenyl butyrate separately. Metabolic pathways activated with lactose included glycolysis and the pentose phosphate pathway, and the metabolic pathways activated with phenylbutyrate included those associated with the biosynthesis of steroids and butyrate/propionate metabolism [86].
Synergy also occurs among proinflammatory cytokines [98]. Various combinations of IL-1β, TNF-α, and IFN-γ induced DEFB4B and DEFB103A transcription. Depending on the combination of proinflammatory cytokines and HBD examined, DEFB4B and DEFB103A transcription can vary from a 4- to 150-fold increase.

4. Roles of AMPs in Inflammation, Immunity, Healing, and Pain

Induced HNPs, HBDs, and LL-37 have additional roles beyond their antimicrobial activities. Their local presence can regulate the intensity of inflammation; regulate innate and adaptive immune functions, and accelerate angiogenesis, vasculogenesis, and wound healing activities. We searched the PubMed literature database using innate immunity, chemotaxis, cell migration, proliferation, cytokine response, gene expression, microbial antigens, pathway signaling, adaptive immunity, antibody response, Th1, Th2, wound healing, angiogenesis, cytotoxicity, apoptosis, and pain together with antimicrobial peptides, defensins, cathelicidins, HBD, HNP, and LL-37 as search terms. These terms were linked in various combinations using Boolean operators to identify AMPs reported to also have roles in inflammation, immunity, healing, and pain. Appropriate articles were downloaded and read by the authors. The information from this search was used to construct Supplemental Table S1 and Table 2.
Although there are wide ranges in the concentrations of AMPs used in many studies (Supplemental Table S1, Table 2), generally AMP-induced activities are concentration-dependent [148]. At lower concentrations (e.g., 0.01 to 0.22 μM), AMPs have chemotactic activities, bind microbial antigens, enhance cytokine responses, promote cell migration, initiate angiogenesis, enhance antibody responses, attenuate cytokine responses, induce cell proliferation, suppress apoptosis, promote cell migration, activate pathway signaling, and enhance wound closure (Table 2). At intermediate concentrations (e.g., 1.00 to 2.54 μM), AMPs attenuate signaling pathways, begin to induce cell cytotoxicity, enhance gene expression, and decrease cell numbers in experimental assays (Table 2). At higher concentrations (e.g., 4.45 to 58.00 μM), AMPs attenuate the expression of genes, increase the production of cell markers, induce Th1 cytokine profiles, delay wound closure, and enhance wound healing (Table 2). For example, at lower concentrations, defensins do not induce TNF-α or IL-1β expression in monocytes or macrophages [149,150], but at higher concentrations, defensins induce chemokine and cytokine production in epithelial cells, keratinocytes, monocytes, and macrophages [151,152,153]. LL-37 induces CXCL8 in epithelial cells and macrophages [154]. At their highest concentrations, AMPs are cytotoxic for a variety of cells.

4.1. Roles of AMPs in Inflammation

AMPs regulate inflammatory processes in part by (i) altering proinflammatory agonist binding to cells and (ii) altering intracellular cytokine signaling pathways. Regulation occurs in both directions. In some circumstances, AMPs increase the expression of proinflammatory cytokines in stimulated cells [152,155,156,157], but in other circumstances, AMPs decrease the expression of proinflammatory cytokines [158,159].
AMPs can directly bind to microbial agonists [160,161,162,163,164,165], which likely prevents their binding to cells. AMPs can also alter the binding of microbial agonists to cells [166,167,168]. HNP-1,2 and HBD3 bind to P. gingivalis recombinant fimbrillin A (rFimA) [160]; HNP-1,2 and HBD3 bind to P. gingivalis rHagB [160,163]; and LL-37 binds to LPS [169]. The binding of AMPs to microbial agonists attenuates proinflammatory cytokine responses. HNP-1-3 attenuates the expression of proinflammatory cytokines from macrophages [170]. HNP-1 attenuates the expression of IL-1β, but not TNF-α, from LPS-treated monocytes [171]. Histatin 5, HBD1, and HBD3 attenuate the expression of IL6, IL10, GM-CSF, and TNF-α from rHagB-treated dendritic cells [163,164], and LL-37 attenuates the expression of proinflammatory cytokines to TLR2, 4, and 9 agonists [172,173].
AMPs alter the intracellular signaling pathways. As an example, HBD3 rapidly enters cells and once in the cytoplasm, prevents the expression of genes related to the activation of proinflammatory responses to LPS/KDO2-Lipid A [158]. The inhibitory effects likely occur downstream of TLR4 activation by LPS. Furthermore, HBD3 dramatically reduced the number of genes expressed in KDO2-lipid A-treated macrophages [158]. LL-37-treated PBMC, CD14+ monocytes, dendritic cells, B-lymphocytes, and T-lymphocytes all produced different profiles of intracellular cytokines [174]. Scott et al. showed that LL-37 increased the expression of 29 genes and decreased the expression of 20 genes encoding chemokines and chemokine receptors [154]. Additionally, Mookherjee et al. showed that LL-37 increased the expression of 475 genes in stimulated CD14+ monocytes [174]. LL-37 activated the p38, ERK1/2, and JNK; IκBα/NFκB; and PI3K signaling pathways and the AP-1, AP-2, E2F1, EGR, NFκB, and SP-1 transcription factors [174].

4.2. Roles of AMPs in Immunity

Once expressed, AMPs regulate important innate and adaptive immune-related activities [9,175,176,177,178]. In innate immunity, AMPs induce chemotactic activities, induce cell migration, increase the production of cellular markers, induce cell proliferation, suppress apoptosis, and increase cellular cytotoxicity (Table 2).
In adaptive immunity, AMPs induce adaptive immune responses, enhance antibody responses, and increase cell survival (Table 2). LL-37 (11.12 μM) was found to induce dendritic cell differentiation, increase FITC-labeled dextran antigen uptake, increase co-stimulatory molecule CD11b, CD86, and CD83 expression, and enhance the Th1 cytokine response [179]. Dendritic cells exposed to LPS produced a T helper type 1 (TH1) cell inducing cytokine profile [179].

4.3. Roles of AMPs in Angiogenesis, Vasculogenesis, and Wound Healing

AMPs clearly accelerate wound healing events [178]. AMPs chemoattract a variety of cells important for wound healing. They increase cell migration, increase angiogenesis, increase vasculogenesis, and facilitate wound closure (Table 2). LL-37 attracts fibroblasts, microvascular endothelial cells, and human umbilical vein endothelial cells [180]. AMPs can also increase cell proliferation [152,181], which facilitates the closure of cell monolayers in scratch models. LL-37 increases fibroblast proliferation, induces human microvascular endothelial cell and human umbilical vein endothelial cell proliferation, and stimulates reepithelialization [180,182,183].
At concentrations of 0.3–5.9 μM, AMPs participate in cell proliferation and wound repair activities. These activities include a large variety of cells involved in the repair of injured tissues (epithelial cells, PLE epithelial cells, corneal epithelial cells, and fibroblasts), cells involved in allergic inflammation (mast cells and eosinophils), cells involved in angiogenesis (endothelial cells), and some cancer cell lines (A549 carcinoma cells, NCI tumor cells, and human epithelial carcinoma KB cell line).

4.4. Roles of AMPs in Pain Nociception

A new and exciting area of AMP functions is the potential of AMPs to be antinociceptive. While inducers of endogenous AMP expression are not yet known to be directly antinociceptive, it is tempting to speculate that they may have stimulatory activities on the MAPK (p38, JNK, and ERK1/2) and NF-κB signaling pathways that produce AMPs and cytokines involved in pain reduction [18]. Blockage of the peripheral activation of ERK1/2 was thought to be antinociceptive on melittin-induced persistent spontaneous nociception and hyperalgesia [184].
Recent work in a variety of fields has demonstrated that small peptides with antimicrobial activities are also involved in nociceptive and antinociceptive responses. These peptides come from insects [184,185,186,187], marine organisms and fish [188,189], cationic arginine-rich peptides [190], lanthipeptides [191], and endocrine cells [192]. Many peptides have been reported to have stimulatory or inhibitory activities on the NF-κB [185] and MAPK (ERK1/2, JNK and p38) [184] signaling pathways. These findings could form the basis to assess the roles of HNPs, HBDs, and LL-37 in pain reduction.

5. Potential Applications of Inducing Endogenous AMPs

The use of a specific inducer would depend upon the overall objective [6,10,12,13,14,15,16,17,18]. AMPs could be induced as prophylactic or preventative measures to bolster innate resistance for improved health or preventing infection [193]. An enhanced antimicrobial barrier would have all the advantages of an alert innate immune system on standby in individuals prone to oral infections including stomatitis and denture stomatitis or about to undergo a surgical procedure. Alternately, AMPs could be induced to treat an emerging or established oral infection. Expressed AMPs would also be available to control acute inflammation and pain antinociception. The presence of AMPs would have the added advantage of inducing additional innate and adaptive immune responses and then facilitating or expediting local wound healing, tissue regeneration, and regrowth.
Inducing the expression of AMPs through micronutrients, nutrients, and macronutrients (Table 1) would be most applicable to improving general health within the oral cavity and gastrointestinal tract. This nutrient-based approach could be used to deliver dietary supplements to maintain normal animal and human growth and health. The actions of the supplements would occur in the epithelium lining, oral cavity, and gastrointestinal tract. Doses would not be readily known, and treatments would generally not be toxic.
Inducing the expression of AMPs by irradiation would be primarily applicable to clearing focal infections. Treatments would likely be highly focused and controlled in a clinical setting. Expressed AMPs would have direct antimicrobial activities and regulate local defenses to attenuate inflammation, ameliorate pain, and help oral tissues heal. Applications include controlling infections in the oral cavity and include root canals. The wavelength and dose of UV irradiation can be easily controlled to facilitate healing and tissue regeneration, and treatments would not be toxic at therapeutic levels.
Dental procedures often have accompanying pain. Treating dental infections, as well as reducing the associated pain, would have popular benefits. However, the ability of AMPs to be antinociceptive is not yet fully known and remains under investigation.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/antibiotics12020361/s1. Supplemental Figure S1. MAPK-dependent induction of antimicrobial peptides (AMPs) [194,195,196]; Supplemental Figure S2. Vitamin D induction of antimicrobial peptides (AMPs); Supplemental Figure S3. NF-κB-dependent induction of antimicrobial peptides (AMPs) [195,196,197,198]; Supplemental Figure S4. Butyrate (BA) and phenylbutyrate (PBA) inhibition of histone deacetylase (HDAC) [199,200,201]; Supplemental Table S1. Many of the roles antimicrobial peptides (AMPs) play in inflammation: innate and adaptive immunity, angiogenesis, vasculogenesis, wound healing, and pain antinociception [202,203,204,205,206,207,208,209,210,211,212,213,214,215,216,217,218,219,220]; Supplemental Table S2. Irradiation induction of antimicrobial peptides (AMPs); and Supplemental Table S3. Synergistic activity of molecules on induction of antimicrobial peptides (AMPs) [221].

Author Contributions

The authors contributed to the study in the following aspects: conceptualization, K.A.M. and K.A.B.; writing—original draft preparation, K.A.M. and K.A.B.; writing—review and editing, K.A.M., R.H.S. and K.A.B. 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

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. We have no financial affiliation (e.g., employment, direct payment, stock holdings, retainers, consultantships, patent licensing arrangements, or honoraria) or involvement with any commercial organization with direct financial interest in the subject or materials discussed in this manuscript, nor have any such arrangements existed in the past three years. Kimberly A. Morio is an endodontist at Apex Endodontics and an adjunct instructor at the University of Iowa; Robert H. Sternowski is the president of Softronics, Ltd.; and Kim A. Brogden is an Emeritus Professor at the University of Iowa.

References

  1. Ahmadi, H.; Ebrahimi, A.; Ahmadi, F. Antibiotic therapy in dentistry. Int. J. Dent. 2021, 2021, 6667624. [Google Scholar] [CrossRef] [PubMed]
  2. Salgado-Peralvo, A.O.; Pena-Cardelles, J.F.; Kewalramani, N.; Garcia-Sanchez, A.; Mateos-Moreno, M.V.; Velasco-Ortega, E.; Ortiz-Garcia, I.; Jimenez-Guerra, A.; Vegh, D.; Pedrinaci, I.; et al. Is antibiotic prophylaxis necessary before dental implant procedures in patients with orthopaedic prostheses? a systematic review. Antibiotics 2022, 11, 93. [Google Scholar] [CrossRef] [PubMed]
  3. Siddique, S.; Chhabra, K.G.; Reche, A.; Madhu, P.P.; Kunghadkar, A.; Kalmegh, S. Antibiotic stewardship program in dentistry: Challenges and opportunities. J. Family Med. Prim. Care 2021, 10, 3951–3955. [Google Scholar] [CrossRef] [PubMed]
  4. Griffith, A.; Mateen, A.; Markowitz, K.; Singer, S.R.; Cugini, C.; Shimizu, E.; Wiedman, G.R.; Kumar, V. Alternative antibiotics in dentistry: Antimicrobial peptides. Pharmaceutics 2022, 14, 1679. [Google Scholar] [CrossRef]
  5. Chen, C.H.; Lu, T.K. Development and challenges of antimicrobial peptides for therapeutic applications. Antibiotics 2020, 9, 24. [Google Scholar] [CrossRef]
  6. Prado Montes de Oca, E. Antimicrobial peptide elicitors: New hope for the post-antibiotic era. Innate Immun. 2013, 19, 227–241. [Google Scholar] [CrossRef]
  7. Ramazi, S.; Mohammadi, N.; Allahverdi, A.; Khalili, E.; Abdolmaleki, P. A review on antimicrobial peptides databases and the computational tools. Database 2022, 2022. [Google Scholar] [CrossRef]
  8. Bowdish, D.M.; Davidson, D.J.; Hancock, R.E. A re-evaluation of the role of host defence peptides in mammalian immunity. Curr. Protein Pept. Sci. 2005, 6, 35–51. [Google Scholar] [CrossRef]
  9. Alford, M.A.; Baquir, B.; Santana, F.L.; Haney, E.F.; Hancock, R.E.W. Cathelicidin host defense peptides and inflammatory signaling: Striking a balance. Front. Microbiol. 2020, 11, 1902. [Google Scholar] [CrossRef]
  10. Bergman, P.; Raqib, R.; Rekha, R.S.; Agerberth, B.; Gudmundsson, G.H. Host directed therapy against infection by boosting innate immunity. Front. Immunol. 2020, 11, 1209. [Google Scholar] [CrossRef]
  11. Wu, J.; Ma, N.; Johnston, L.J.; Ma, X. Dietary nutrients mediate intestinal host defense peptide expression. Adv. Nutr. 2020, 11, 92–102. [Google Scholar] [CrossRef]
  12. Nishimura, E.; Kato, M.; Hashizume, S. Human beta-defensin-2 induction in human foreskin keratinocyte by synergetic stimulation with foods and Escherichia coli. Cytotechnology 2003, 43, 135–144. [Google Scholar] [CrossRef]
  13. Campbell, Y.; Fantacone, M.L.; Gombart, A.F. Regulation of antimicrobial peptide gene expression by nutrients and by-products of microbial metabolism. Eur. J. Nutr. 2012, 51, 899–907. [Google Scholar] [CrossRef]
  14. van der Does, A.M.; Bergman, P.; Agerberth, B.; Lindbom, L. Induction of the human cathelicidin LL-37 as a novel treatment against bacterial infections. J. Leukoc. Biol. 2012, 92, 735–742. [Google Scholar] [CrossRef]
  15. Jiang, W.; Sunkara, L.T.; Zeng, X.; Deng, Z.; Myers, S.M.; Zhang, G. Differential regulation of human cathelicidin LL-37 by free fatty acids and their analogs. Peptides 2013, 50, 129–138. [Google Scholar] [CrossRef]
  16. Wang, G. Human antimicrobial peptides and proteins. Pharmaceuticals 2014, 7, 545–594. [Google Scholar] [CrossRef]
  17. Chen, J.; Zhai, Z.; Long, H.; Yang, G.; Deng, B.; Deng, J. Inducible expression of defensins and cathelicidins by nutrients and associated regulatory mechanisms. Peptides 2020, 123, 170177. [Google Scholar] [CrossRef]
  18. Morio, K.A.; Sternowski, R.H.; Zeng, E.; Brogden, K.A. Antimicrobial peptides and biomarkers induced by ultraviolet irradiation have the potential to reduce endodontic inflammation and facilitate tissue healing. Pharmaceutics 2022, 14, 1979. [Google Scholar] [CrossRef]
  19. Lehrer, R.I.; Lu, W. a-Defensins in human innate immunity. Immunol. Rev. 2012, 245, 84–112. [Google Scholar] [CrossRef]
  20. Niu, J.Y.; Yin, I.X.; Mei, M.L.; Wu, W.K.K.; Li, Q.L.; Chu, C.H. The multifaceted roles of antimicrobial peptides in oral diseases. Mol. Oral Microbiol. 2021, 36, 159–171. [Google Scholar] [CrossRef]
  21. Bahar, A.A.; Ren, D. Antimicrobial peptides. Pharmaceuticals 2013, 6, 1543–1575. [Google Scholar] [CrossRef] [PubMed]
  22. Gorr, S.U. Antimicrobial peptides of the oral cavity. Periodontol. 2000 2009, 51, 152–180. [Google Scholar] [CrossRef] [PubMed]
  23. Mathews, M.; Jia, H.P.; Guthmiller, J.M.; Losh, G.; Graham, S.; Johnson, G.K.; Tack, B.F.; McCray, P.B., Jr. Production of beta-defensin antimicrobial peptides by the oral mucosa and salivary glands. Infect. Immun. 1999, 67, 2740–2745. [Google Scholar] [CrossRef] [PubMed]
  24. Meyer, J.E.; Harder, J.; Sipos, B.; Maune, S.; Kloppel, G.; Bartels, J.; Schroder, J.M.; Glaser, R. Psoriasin (S100A7) is a principal antimicrobial peptide of the human tongue. Mucosal Immunol. 2008, 1, 239–243. [Google Scholar] [CrossRef]
  25. Lundy, F.T.; Orr, D.F.; Gallagher, J.R.; Maxwell, P.; Shaw, C.; Napier, S.S.; Gerald Cowan, C.; Lamey, P.J.; Marley, J.J. Identification and overexpression of human neutrophil alpha-defensins (human neutrophil peptides 1, 2 and 3) in squamous cell carcinomas of the human tongue. Oral Oncol. 2004, 40, 139–144. [Google Scholar] [CrossRef]
  26. Kamekura, R.; Imai, R.; Takano, K.; Yamashita, K.; Jitsukawa, S.; Nagaya, T.; Ito, F.; Hirao, M.; Tsubota, H.; Himi, T. Expression and localization of human efensins in palatine tonsils. Adv. Otorhinolaryngol. 2016, 77, 112–118. [Google Scholar] [CrossRef]
  27. Sahasrabudhe, K.S.; Kimball, J.R.; Morton, T.H.; Weinberg, A.; Dale, B.A. Expression of the antimicrobial peptide, human beta-defensin 1, in duct cells of minor salivary glands and detection in saliva. J. Dent. Res. 2000, 79, 1669–1674. [Google Scholar] [CrossRef]
  28. Dunsche, A.; Acil, Y.; Dommisch, H.; Siebert, R.; Schroder, J.M.; Jepsen, S. The novel human beta-defensin-3 is widely expressed in oral tissues. Eur. J. Oral Sci. 2002, 110, 121–124. [Google Scholar] [CrossRef]
  29. Woo, J.S.; Jeong, J.Y.; Hwang, Y.J.; Chae, S.W.; Hwang, S.J.; Lee, H.M. Expression of cathelicidin in human salivary glands. Arch. Otolaryngol. Head Neck Surg. 2003, 129, 211–214. [Google Scholar] [CrossRef]
  30. Dale, B.A.; Kimball, J.R.; Krisanaprakornkit, S.; Roberts, F.; Robinovitch, M.; O’Neal, R.; Valore, E.V.; Ganz, T.; Anderson, G.M.; Weinberg, A. Localized antimicrobial peptide expression in human gingiva. J. Periodontal Res. 2001, 36, 285–294. [Google Scholar] [CrossRef]
  31. Dommisch, H.; Winter, J.; Acil, Y.; Dunsche, A.; Tiemann, M.; Jepsen, S. Human beta-defensin (hBD-1, -2) expression in dental pulp. Oral Microbiol. Immunol. 2005, 20, 163–166. [Google Scholar] [CrossRef]
  32. Ghattas Ayoub, C.; Aminoshariae, A.; Bakkar, M.; Ghosh, S.; Bonfield, T.; Demko, C.; Montagnese, T.A.; Mickel, A.K. Comparison of IL-1beta, TNF-alpha, hBD-2, and hBD-3 expression in the dental pulp of smokers versus nonsmokers. J. Endod. 2017, 43, 2009–2013. [Google Scholar] [CrossRef]
  33. Zhai, Y.; Wang, Y.; Rao, N.; Li, J.; Li, X.; Fang, T.; Zhao, Y.; Ge, L. Activation and biological properties of human beta defensin 4 in stem cells derived From human exfoliated deciduous teeth. Front. Physiol. 2019, 10, 1304. [Google Scholar] [CrossRef]
  34. Loureiro, C.; Buzalaf, M.A.R.; Pessan, J.P.; Ventura, T.M.O.; Pela, V.T.; Ribeiro, A.P.F.; Jacinto, R.C. Proteomic analysis of infected root canals with apical periodontitis in patients with type 2 diabetes mellitus: A cross-sectional study. Int. Endod. J. 2022, 55, 910–922. [Google Scholar] [CrossRef]
  35. Loo, J.A.; Yan, W.; Ramachandran, P.; Wong, D.T. Comparative human salivary and plasma proteomes. J. Dent. Res. 2010, 89, 1016–1023. [Google Scholar] [CrossRef]
  36. Lau, W.W.; Hardt, M.; Zhang, Y.H.; Freire, M.; Ruhl, S. The human salivary proteome wiki: A community-driven research platform. J. Dent. Res. 2021, 100, 1510–1519. [Google Scholar] [CrossRef]
  37. Waniczek, D.; Swietochowska, E.; Snietura, M.; Kiczmer, P.; Lorenc, Z.; Muc-Wierzgon, M. Salivary concentrations of chemerin, alpha-defensin 1, and TNF-alpha as potential biomarkers in the early diagnosis of colorectal cancer. Metabolites 2022, 12, 704. [Google Scholar] [CrossRef]
  38. Pei, F.; Wang, M.; Wang, Y.; Pan, X.; Cen, X.; Huang, X.; Jin, Y.; Zhao, Z. Quantitative proteomic analysis of gingival crevicular fluids to identify novel biomarkers of gingival recession in orthodontic patients. J. Proteom. 2022, 266, 104647. [Google Scholar] [CrossRef]
  39. Kido, J.; Bando, M.; Hiroshima, Y.; Iwasaka, H.; Yamada, K.; Ohgami, N.; Nambu, T.; Kataoka, M.; Yamamoto, T.; Shinohara, Y.; et al. Analysis of proteins in human gingival crevicular fluid by mass spectrometry. J. Periodontal Res. 2012, 47, 488–499. [Google Scholar] [CrossRef]
  40. Tao, R.; Jurevic, R.J.; Coulton, K.K.; Tsutsui, M.T.; Roberts, M.C.; Kimball, J.R.; Wells, N.; Berndt, J.; Dale, B.A. Salivary antimicrobial peptide expression and dental caries experience in children. Antimicrob. Agents Chemother. 2005, 49, 3883–3888. [Google Scholar] [CrossRef] [Green Version]
  41. Dale, B.A.; Tao, R.; Kimball, J.R.; Jurevic, R.J. Oral antimicrobial peptides and biological control of caries. BMC Oral Health 2006, 6, S13. [Google Scholar] [CrossRef] [PubMed]
  42. Maggini, S.; Wintergerst, E.S.; Beveridge, S.; Hornig, D.H. Selected vitamins and trace elements support immune function by strengthening epithelial barriers and cellular and humoral immune responses. Br. J. Nutr. 2007, 98 (Suppl. S1), S29–S35. [Google Scholar] [CrossRef] [PubMed]
  43. Wintergerst, E.S.; Maggini, S.; Hornig, D.H. Contribution of selected vitamins and trace elements to immune function. Ann. Nutr. Metab. 2007, 51, 301–323. [Google Scholar] [CrossRef] [PubMed]
  44. Bae, M.; Kim, H. Mini-review on the roles of vitamin C, vitamin D, and selenium in the immune system against COVID-19. Molecules 2020, 25, 5346. [Google Scholar] [CrossRef]
  45. Haase, H.; Rink, L. Signal transduction in monocytes: The role of zinc ions. Biometals 2007, 20, 579–585. [Google Scholar] [CrossRef]
  46. Xu, S.Z.; Lee, S.H.; Lillehoj, H.S.; Bravo, D. Dietary sodium selenite affects host intestinal and systemic immune response and disease susceptibility to necrotic enteritis in commercial broilers. Br. Poult. Sci. 2015, 56, 103–112. [Google Scholar] [CrossRef]
  47. Talukder, P.; Satho, T.; Irie, K.; Sharmin, T.; Hamady, D.; Nakashima, Y.; Kashige, N.; Miake, F. Trace metal zinc stimulates secretion of antimicrobial peptide LL-37 from Caco-2 cells through ERK and p38 MAP kinase. Int. Immunopharmacol. 2011, 11, 141–144. [Google Scholar] [CrossRef]
  48. Portelinha, J.; Duay, S.S.; Yu, S.I.; Heilemann, K.; Libardo, M.D.J.; Juliano, S.A.; Klassen, J.L.; Angeles-Boza, A.M. Antimicrobial peptides and copper(II) ions: Novel therapeutic opportunities. Chem. Rev. 2021, 121, 2648–2712. [Google Scholar] [CrossRef]
  49. Agbale, C.M.; Sarfo, J.K.; Galyuon, I.K.; Juliano, S.A.; Silva, G.G.O.; Buccini, D.F.; Cardoso, M.H.; Torres, M.D.T.; Angeles-Boza, A.M.; de la Fuente-Nunez, C.; et al. Antimicrobial and antibiofilm activities of helical antimicrobial peptide sequences incorporating metal-binding motifs. Biochemistry 2019, 58, 3802–3812. [Google Scholar] [CrossRef]
  50. Ganz, T. Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation. Blood 2003, 102, 783–788. [Google Scholar] [CrossRef] [Green Version]
  51. Pernet, I.; Reymermier, C.; Guezennec, A.; Branka, J.E.; Guesnet, J.; Perrier, E.; Dezutter-Dambuyant, C.; Schmitt, D.; Viac, J. Calcium triggers beta-defensin (hBD-2 and hBD-3) and chemokine macrophage inflammatory protein-3 alpha (MIP-3alpha/CCL20) expression in monolayers of activated human keratinocytes. Exp. Dermatol. 2003, 12, 755–760. [Google Scholar] [CrossRef]
  52. Jacobo-Delgado, Y.M.; Torres-Juarez, F.; Rodriguez-Carlos, A.; Santos-Mena, A.; Enciso-Moreno, J.E.; Rivas-Santiago, C.; Diamond, G.; Rivas-Santiago, B. Retinoic acid induces antimicrobial peptides and cytokines leading to Mycobacterium tuberculosis elimination in airway epithelial cells. Peptides 2021, 142, 170580. [Google Scholar] [CrossRef]
  53. Lee, S.E.; Lee, J.S.; Kim, M.R.; Kim, M.Y.; Kim, S.C. Topical retinoids induce beta-defensin 3 expression in mouse skin. Int. J. Dermatol. 2010, 49, 1082–1084. [Google Scholar] [CrossRef]
  54. Chen, Y.; Li, P.; Zhen, R.; Wang, L.; Feng, J.; Xie, Y.; Yang, B.; Xiong, Y.; Niu, J.; Wu, Q.; et al. Effects of niacin on intestinal epithelial barrier, intestinal immunity, and microbial community in weaned piglets challenged by PDCoV. Int. Immunopharmacol. 2022, 111, 109054. [Google Scholar] [CrossRef]
  55. Zhen, R.; Feng, J.; He, D.; Chen, Y.; Chen, T.; Cai, W.; Xiong, Y.; Qiu, Y.; Jiang, Z.; Wang, L.; et al. Effects of niacin on resistance to enterotoxigenic Escherichia coli infection in weaned piglets. Front. Nutr. 2022, 9, 865311. [Google Scholar] [CrossRef]
  56. Cruz Diaz, L.A.; Flores Miramontes, M.G.; Chavez Hurtado, P.; Allen, K.; Gonzalez Avila, M.; Prado Montes de Oca, E. Ascorbic acid, ultraviolet C rays, and glucose but not hyperthermia are elicitors of human beta-defensin 1 mRNA in normal keratinocytes. Biomed. Res. Int. 2015, 2015, 714580. [Google Scholar] [CrossRef]
  57. Robinson, K.; Ma, X.; Liu, Y.; Qiao, S.; Hou, Y.; Zhang, G. Dietary modulation of endogenous host defense peptide synthesis as an alternative approach to in-feed antibiotics. Anim. Nutr. 2018, 4, 160–169. [Google Scholar] [CrossRef]
  58. Strandberg, K.L.; Richards, S.M.; Gunn, J.S. Cathelicidin antimicrobial peptide expression is not induced or required for bacterial clearance during Salmonella enterica infection of human monocyte-derived macrophages. Infect. Immun. 2012, 80, 3930–3938. [Google Scholar] [CrossRef]
  59. Martineau, A.R.; Wilkinson, K.A.; Newton, S.M.; Floto, R.A.; Norman, A.W.; Skolimowska, K.; Davidson, R.N.; Sorensen, O.E.; Kampmann, B.; Griffiths, C.J.; et al. IFN-gamma- and TNF-independent vitamin D-inducible human suppression of mycobacteria: The role of cathelicidin LL-37. J. Immunol. 2007, 178, 7190–7198. [Google Scholar] [CrossRef]
  60. Misawa, Y.; Baba, A.; Ito, S.; Tanaka, M.; Shiohara, M. Vitamin D(3) induces expression of human cathelicidin antimicrobial peptide 18 in newborns. Int. J. Hematol. 2009, 90, 561–570. [Google Scholar] [CrossRef] [Green Version]
  61. Chen, S.; Ge, L.; Gombart, A.F.; Shuler, F.D.; Carlson, M.A.; Reilly, D.A.; Xie, J. Nanofiber-based sutures induce endogenous antimicrobial peptide. Nanomedicine 2017, 12, 2597–2609. [Google Scholar] [CrossRef] [PubMed]
  62. Karlsson, J.; Carlsson, G.; Larne, O.; Andersson, M.; Putsep, K. Vitamin D3 induces pro-LL-37 expression in myeloid precursors from patients with severe congenital neutropenia. J. Leukoc. Biol. 2008, 84, 1279–1286. [Google Scholar] [CrossRef] [PubMed]
  63. Jiang, J.; Zhang, Y.; Indra, A.K.; Ganguli-Indra, G.; Le, M.N.; Wang, H.; Hollins, R.R.; Reilly, D.A.; Carlson, M.A.; Gallo, R.L.; et al. 1alpha,25-dihydroxyvitamin D3-eluting nanofibrous dressings induce endogenous antimicrobial peptide expression. Nanomedicine 2018, 13, 1417–1432. [Google Scholar] [CrossRef] [PubMed]
  64. Su, Y.; Ganguli-Indra, G.; Bhattacharya, N.; Logan, I.E.; Indra, A.K.; Gombart, A.F.; Wong, S.L.; Xie, J. Codelivery of 1alpha,25-dihydroxyvitamin D3 and CYP24A1 inhibitor VID400 by nanofiber dressings promotes endogenous antimicrobial peptide LL-37 induction. Mol. Pharm. 2022, 19, 974–984. [Google Scholar] [CrossRef]
  65. Wang, T.T.; Nestel, F.P.; Bourdeau, V.; Nagai, Y.; Wang, Q.; Liao, J.; Tavera-Mendoza, L.; Lin, R.; Hanrahan, J.W.; Mader, S.; et al. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J. Immunol. 2004, 173, 2909–2912. [Google Scholar] [CrossRef]
  66. Gombart, A.F.; Borregaard, N.; Koeffler, H.P. Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3. FASEB J. 2005, 19, 1067–1077. [Google Scholar] [CrossRef]
  67. White, J.H. Emerging roles of vitamin D-induced antimicrobial peptides in antiviral innate immunity. Nutrients 2022, 14, 284. [Google Scholar] [CrossRef]
  68. Subramanian, K.; Bergman, P.; Henriques-Normark, B. Vitamin D promotes pneumococcal killing and modulates inflammatory responses in primary human neutrophils. J. Innate Immun. 2017, 9, 375–386. [Google Scholar] [CrossRef]
  69. Derradjia, A.; Alanazi, H.; Park, H.J.; Djeribi, R.; Semlali, A.; Rouabhia, M. Alpha-tocopherol decreases interleukin-1beta and -6 and increases human beta-defensin-1 and -2 secretion in human gingival fibroblasts stimulated with Porphyromonas gingivalis lipopolysaccharide. J. Periodontal Res. 2016, 51, 295–303. [Google Scholar] [CrossRef]
  70. Malik, A.N.; Al-Kafaji, G. Glucose regulation of beta-defensin-1 mRNA in human renal cells. Biochem. Biophys. Res. Commun. 2007, 353, 318–323. [Google Scholar] [CrossRef]
  71. Barnea, M.; Madar, Z.; Froy, O. Glucose and insulin are needed for optimal defensin expression in human cell lines. Biochem. Biophys. Res. Commun. 2008, 367, 452–456. [Google Scholar] [CrossRef]
  72. Cederlund, A.; Kai-Larsen, Y.; Printz, G.; Yoshio, H.; Alvelius, G.; Lagercrantz, H.; Stromberg, R.; Jornvall, H.; Gudmundsson, G.H.; Agerberth, B. Lactose in human breast milk an inducer of innate immunity with implications for a role in intestinal homeostasis. PLoS ONE 2013, 8, e53876. [Google Scholar] [CrossRef]
  73. Shao, Y.; Wang, Z.; Tian, X.; Guo, Y.; Zhang, H. Yeast β-D-glucans induced antimicrobial peptide expressions against Salmonella infection in broiler chickens. Int. J. Biol. Macromol. 2016, 85, 573–584. [Google Scholar] [CrossRef]
  74. Sherman, H.; Chapnik, N.; Froy, O. Albumin and amino acids upregulate the expression of human beta-defensin 1. Mol. Immunol. 2006, 43, 1617–1623. [Google Scholar] [CrossRef]
  75. Fehlbaum, P.; Rao, M.; Zasloff, M.; Anderson, G.M. An essential amino acid induces epithelial beta-defensin expression. Proc. Natl. Acad. Sci. USA 2000, 97, 12723–12728. [Google Scholar] [CrossRef]
  76. Rivas-Santiago, C.E.; Rivas-Santiago, B.; Leon, D.A.; Castaneda-Delgado, J.; Hernandez Pando, R. Induction of beta-defensins by l-isoleucine as novel immunotherapy in experimental murine tuberculosis. Clin. Exp. Immunol. 2011, 164, 80–89. [Google Scholar] [CrossRef]
  77. Konno, Y.; Ashida, T.; Inaba, Y.; Ito, T.; Tanabe, H.; Maemoto, A.; Ayabe, T.; Mizukami, Y.; Fujiya, M.; Kohgo, Y. Isoleucine, an essential amino acid, induces the expression of human β defensin 2 through the activation of the G-protein coupled receptor-ERK pathway in the intestinal epithelia. Food Nutr. Sci. 2012, 3, 548. [Google Scholar] [CrossRef]
  78. Shirako, S.; Kojima, Y.; Tomari, N.; Nakamura, Y.; Matsumura, Y.; Ikeda, K.; Inagaki, N.; Sato, K. Pyroglutamyl leucine, a peptide in fermented foods, attenuates dysbiosis by increasing host antimicrobial peptide. NPJ Sci. Food 2019, 3, 18. [Google Scholar] [CrossRef]
  79. Kiyono, T.; Wada, S.; Ohta, R.; Wada, E.; Takagi, T.; Naito, Y.; Yoshikawa, T.; Sato, K. Identification of pyroglutamyl peptides with anti-colitic activity in Japanese rice wine, sake, by oral administration in a mouse model. J. Funct. Foods 2016, 27, 612–621. [Google Scholar] [CrossRef]
  80. Liu, Q.; Liu, J.; Roschmann, K.I.L.; van Egmond, D.; Golebski, K.; Fokkens, W.J.; Wang, D.; van Drunen, C.M. Histone deacetylase inhibitors up-regulate LL-37 expression independent of toll-like receptor mediated signalling in airway epithelial cells. J. Inflamm. 2013, 10, 15. [Google Scholar] [CrossRef] [Green Version]
  81. Sunkara, L.T.; Jiang, W.; Zhang, G. Modulation of antimicrobial host defense peptide gene expression by free fatty acids. PLoS ONE 2012, 7, e49558. [Google Scholar] [CrossRef] [PubMed]
  82. Sunkara, L.T.; Zeng, X.; Curtis, A.R.; Zhang, G. Cyclic AMP synergizes with butyrate in promoting beta-defensin 9 expression in chickens. Mol. Immunol. 2014, 57, 171–180. [Google Scholar] [CrossRef] [PubMed]
  83. Zeng, X.; Sunkara, L.T.; Jiang, W.; Bible, M.; Carter, S.; Ma, X.; Qiao, S.; Zhang, G. Induction of porcine host defense peptide gene expression by short-chain fatty acids and their analogs. PLoS ONE 2013, 8, e72922. [Google Scholar] [CrossRef] [PubMed]
  84. Schauber, J.; Iffland, K.; Frisch, S.; Kudlich, T.; Schmausser, B.; Eck, M.; Menzel, T.; Gostner, A.; Luhrs, H.; Scheppach, W. Histone-deacetylase inhibitors induce the cathelicidin LL-37 in gastrointestinal cells. Mol. Immunol. 2004, 41, 847–854. [Google Scholar] [CrossRef]
  85. Raqib, R.; Sarker, P.; Bergman, P.; Ara, G.; Lindh, M.; Sack, D.A.; Nasirul Islam, K.M.; Gudmundsson, G.H.; Andersson, J.; Agerberth, B. Improved outcome in shigellosis associated with butyrate induction of an endogenous peptide antibiotic. Proc. Natl. Acad. Sci. USA 2006, 103, 9178–9183. [Google Scholar] [CrossRef]
  86. Cederlund, A.; Nylen, F.; Miraglia, E.; Bergman, P.; Gudmundsson, G.H.; Agerberth, B. Label-free quantitative mass spectrometry reveals novel pathways involved in LL-37 expression. J. Innate Immun. 2014, 6, 365–376. [Google Scholar] [CrossRef]
  87. Steinmann, J.; Halldorsson, S.; Agerberth, B.; Gudmundsson, G.H. Phenylbutyrate induces antimicrobial peptide expression. Antimicrob. Agents Chemother. 2009, 53, 5127–5133. [Google Scholar] [CrossRef]
  88. Xiong, H.; Guo, B.; Gan, Z.; Song, D.; Lu, Z.; Yi, H.; Wu, Y.; Wang, Y.; Du, H. Butyrate upregulates endogenous host defense peptides to enhance disease resistance in piglets via histone deacetylase inhibition. Sci. Rep. 2016, 6, 27070. [Google Scholar] [CrossRef]
  89. Schauber, J.; Svanholm, C.; Termen, S.; Iffland, K.; Menzel, T.; Scheppach, W.; Melcher, R.; Agerberth, B.; Luhrs, H.; Gudmundsson, G.H. Expression of the cathelicidin LL-37 is modulated by short chain fatty acids in colonocytes: Relevance of signalling pathways. Gut 2003, 52, 735–741. [Google Scholar] [CrossRef]
  90. Schwab, M.; Reynders, V.; Shastri, Y.; Loitsch, S.; Stein, J.; Schroder, O. Role of nuclear hormone receptors in butyrate-mediated up-regulation of the antimicrobial peptide cathelicidin in epithelial colorectal cells. Mol. Immunol. 2007, 44, 2107–2114. [Google Scholar] [CrossRef]
  91. Zhao, Y.; Chen, F.; Wu, W.; Sun, M.; Bilotta, A.J.; Yao, S.; Xiao, Y.; Huang, X.; Eaves-Pyles, T.D.; Golovko, G.; et al. GPR43 mediates microbiota metabolite SCFA regulation of antimicrobial peptide expression in intestinal epithelial cells via activation of mTOR and STAT3. Mucosal. Immunol. 2018, 11, 752–762. [Google Scholar] [CrossRef]
  92. Kida, Y.; Shimizu, T.; Kuwano, K. Sodium butyrate up-regulates cathelicidin gene expression via activator protein-1 and histone acetylation at the promoter region in a human lung epithelial cell line, EBC-1. Mol. Immunol. 2006, 43, 1972–1981. [Google Scholar] [CrossRef]
  93. Nakatsuji, T.; Kao, M.C.; Zhang, L.; Zouboulis, C.C.; Gallo, R.L.; Huang, C.M. Sebum free fatty acids enhance the innate immune defense of human sebocytes by upregulating beta-defensin-2 expression. J. Investig. Dermatol. 2010, 130, 985–994. [Google Scholar] [CrossRef]
  94. Kim, J.E.; Kim, B.J.; Jeong, M.S.; Seo, S.J.; Kim, M.N.; Hong, C.K.; Ro, B.I. Expression and modulation of LL-37 in normal human keratinocytes, HaCaT cells, and inflammatory skin diseases. J. Korean Med. Sci. 2005, 20, 649–654. [Google Scholar] [CrossRef]
  95. Kim, B.J.; Rho, Y.K.; Lee, H.I.; Jeong, M.S.; Li, K.; Seo, S.J.; Kim, M.N.; Hong, C.K. The effect of calcipotriol on the expression of human beta defensin-2 and LL-37 in cultured human keratinocytes. Clin. Dev. Immunol. 2009, 2009, 645898. [Google Scholar] [CrossRef]
  96. Seo, S.J.; Ahn, S.W.; Hong, C.K.; Ro, B.I. Expressions of beta-defensins in human keratinocyte cell lines. J. Dermatol. Sci. 2001, 27, 183–191. [Google Scholar] [CrossRef]
  97. Santamaria, M.H.; Perez Caballero, E.; Corral, R.S. Unmethylated CpG motifs in Toxoplasma gondii DNA induce TLR9- and IFN-beta-dependent expression of alpha-defensin-5 in intestinal epithelial cells. Parasitology 2016, 143, 60–68. [Google Scholar] [CrossRef]
  98. Joly, S.; Organ, C.C.; Johnson, G.K.; McCray, P.B., Jr.; Guthmiller, J.M. Correlation between beta-defensin expression and induction profiles in gingival keratinocytes. Mol. Immunol. 2005, 42, 1073–1084. [Google Scholar] [CrossRef]
  99. Yin, Q.; Xu, X.; Lin, Y.; Lv, J.; Zhao, L.; He, R. Ultraviolet B irradiation induces skin accumulation of plasmacytoid dendritic cells: A possible role for chemerin. Autoimmunity 2014, 47, 185–192. [Google Scholar] [CrossRef]
  100. Glaser, R.; Navid, F.; Schuller, W.; Jantschitsch, C.; Harder, J.; Schroder, J.M.; Schwarz, A.; Schwarz, T. UV-B radiation induces the expression of antimicrobial peptides in human keratinocytes in vitro and in vivo. J. Allergy Clin. Immunol. 2009, 123, 1117–1123. [Google Scholar] [CrossRef]
  101. Di Nuzzo, S.; Sylva-Steenland, R.M.; Koomen, C.W.; de Rie, M.A.; Das, P.K.; Bos, J.D.; Teunissen, M.B. Exposure to UVB induces accumulation of LFA-1+ T cells and enhanced expression of the chemokine psoriasin in normal human skin. Photochem. Photobiol. 2000, 72, 374–382. [Google Scholar] [CrossRef] [PubMed]
  102. Mallbris, L.; Edstrom, D.W.; Sundblad, L.; Granath, F.; Stahle, M. UVB upregulates the antimicrobial protein hCAP18 mRNA in human skin. J. Investig. Dermatol. 2005, 125, 1072–1074. [Google Scholar] [CrossRef] [PubMed]
  103. Kim, J.; Kim, S.; Lim, W.; Choi, H.; Kim, O. Effects of the antimicrobial peptide cathelicidin (LL-37) on immortalized gingival fibroblasts infected with Porphyromonas gingivalis and irradiated with 625-nm LED light. Lasers Med. Sci. 2015, 30, 2049–2057. [Google Scholar] [CrossRef] [PubMed]
  104. Tang, E.; Khan, I.; Andreana, S.; Arany, P.R. Laser-activated transforming growth factor-beta1 induces human beta-defensin 2: Implications for laser therapies for periodontitis and peri-implantitis. J. Periodontal Res. 2017, 52, 360–367. [Google Scholar] [CrossRef]
  105. Fedrizzi, L.; Lim, D.; Carafoli, E. Calcium and signal transduction. Biochem. Mol. Biol. Educ. 2008, 36, 175–180. [Google Scholar] [CrossRef]
  106. KEGG. Calcium Signaling Pathway—Homo Sapiens (Human). Available online: https://www.genome.jp/pathway/hsa04020 (accessed on 2 January 2023).
  107. Harris, T.A.; Gattu, S.; Propheter, D.C.; Kuang, Z.; Bel, S.; Ruhn, K.A.; Chara, A.L.; Edwards, M.; Zhang, C.; Jo, J.H.; et al. Resistin-like molecule alpha provides vitamin-A-dependent antimicrobial protection in the skin. Cell Host Microbe 2019, 25, 777–788.e778. [Google Scholar] [CrossRef]
  108. Christakos, S.; Dhawan, P.; Verstuyf, A.; Verlinden, L.; Carmeliet, G. Vitamin D: Metabolism, molecular mechanism of action, and pleiotropic effects. Physiol. Rev. 2016, 96, 365–408. [Google Scholar] [CrossRef]
  109. Pilz, S.; Zittermann, A.; Trummer, C.; Theiler-Schwetz, V.; Lerchbaum, E.; Keppel, M.H.; Grubler, M.R.; Marz, W.; Pandis, M. Vitamin D testing and treatment: A narrative review of current evidence. Endocr. Connect. 2019, 8, R27–R43. [Google Scholar] [CrossRef]
  110. Chen, J.; Tang, Z.; Slominski, A.T.; Li, W.; Zmijewski, M.A.; Liu, Y.; Chen, J. Vitamin D and its analogs as anticancer and anti-inflammatory agents. Eur. J. Med. Chem. 2020, 207, 112738. [Google Scholar] [CrossRef]
  111. Keane, J.T.; Elangovan, H.; Stokes, R.A.; Gunton, J.E. Vitamin D and the liver-correlation or cause? Nutrients 2018, 10, 496. [Google Scholar] [CrossRef] [Green Version]
  112. Liu, P.T.; Stenger, S.; Li, H.; Wenzel, L.; Tan, B.H.; Krutzik, S.R.; Ochoa, M.T.; Schauber, J.; Wu, K.; Meinken, C.; et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science 2006, 311, 1770–1773. [Google Scholar] [CrossRef]
  113. Edfeldt, K.; Liu, P.T.; Chun, R.; Fabri, M.; Schenk, M.; Wheelwright, M.; Keegan, C.; Krutzik, S.R.; Adams, J.S.; Hewison, M.; et al. T-cell cytokines differentially control human monocyte antimicrobial responses by regulating vitamin D metabolism. Proc. Natl. Acad. Sci. USA 2010, 107, 22593–22598. [Google Scholar] [CrossRef]
  114. Geller, A.I.; Shehab, N.; Weidle, N.J.; Lovegrove, M.C.; Wolpert, B.J.; Timbo, B.B.; Mozersky, R.P.; Budnitz, D.S. Emergency Department Visits for Adverse Events Related to Dietary Supplements. N. Engl. J. Med. 2015, 373, 1531–1540. [Google Scholar] [CrossRef]
  115. Patergnani, S.; Danese, A.; Bouhamida, E.; Aguiari, G.; Previati, M.; Pinton, P.; Giorgi, C. Various aspects of calcium signaling in the regulation of apoptosis, autophagy, cell proliferation, and cancer. Int. J. Mol. Sci. 2020, 21, 8323. [Google Scholar] [CrossRef]
  116. Ronis, M.J.J.; Pedersen, K.B.; Watt, J. Adverse Effects of Nutraceuticals and Dietary Supplements. Annu. Rev. Pharmacol. Toxicol. 2018, 58, 583–601. [Google Scholar] [CrossRef]
  117. De Marco Castro, E.; Calder, P.C.; Roche, H.M. Beta-1,3/1,6-glucans and immunity: State of the art and future directions. Mol. Nutr. Food Res. 2021, 65, e1901071. [Google Scholar] [CrossRef]
  118. Paudel, S.; Wu, G.; Wang, X. Amino acids in cell signaling: Regulation and function. Adv. Exp. Med. Biol. 2021, 1332, 17–33. [Google Scholar] [CrossRef]
  119. Topala, T.; Bodoki, A.; Oprean, L.; Oprean, R. Bovine serum albumin interactions with metal complexes. Clujul. Med. 2014, 87, 215–219. [Google Scholar] [CrossRef]
  120. Bergsson, G.; Hilmarsson, H.; Thormar, H. Antibacterial, antiviral, and antifungal activities of lipids. In Lipids and Essential Oils as Antimicrobial Agents; Thormar, H., Thormar, H., Eds.; John Wiley & Sons: Chichester, UK, 2011; p. 34. [Google Scholar]
  121. Gursoy, U.K.; Salli, K.; Soderling, E.; Gursoy, M.; Hirvonen, J.; Ouwehand, A.C. Regulation of hBD-2, hBD-3, hCAP18/LL37, and proinflammatory cytokine secretion by human milk oligosaccharides in an organotypic oral mucosal model. Pathogens 2021, 10, 739. [Google Scholar] [CrossRef]
  122. Paoletti, I.; Buommino, E.; Tudisco, L.; Baudouin, C.; Msika, P.; Tufano, M.A.; Baroni, A.; Donnarumma, G. Patented natural avocado sugars modulate the HBD-2 expression in human keratinocytes through the involvement of protein kinase C and protein tyrosine kinases. Arch. Dermatol. Res. 2010, 302, 201–209. [Google Scholar] [CrossRef] [Green Version]
  123. Paoletti, I.; Buommino, E.; Fusco, A.; Baudouin, C.; Msika, P.; Tufano, M.A.; Baroni, A.; Donnarumma, G. Patented natural avocado sugar modulates the HBD-2 and HBD-3 expression in human keratinocytes through toll-like receptor-2 and ERK/MAPK activation. Arch. Dermatol. Res. 2012, 304, 619–625. [Google Scholar] [CrossRef] [PubMed]
  124. Promsong, A.; Chung, W.O.; Satthakarn, S.; Nittayananta, W. Ellagic acid modulates the expression of oral innate immune mediators: Potential role in mucosal protection. J. Oral Pathol. Med. 2015, 44, 214–221. [Google Scholar] [CrossRef] [PubMed]
  125. Wan, M.L.; Ling, K.H.; Wang, M.F.; El-Nezami, H. Green tea polyphenol epigallocatechin-3-gallate improves epithelial barrier function by inducing the production of antimicrobial peptide pBD-1 and pBD-2 in monolayers of porcine intestinal epithelial IPEC-J2 cells. Mol. Nutr. Food Res. 2016, 60, 1048–1058. [Google Scholar] [CrossRef] [PubMed]
  126. Lombardo Bedran, T.B.; Feghali, K.; Zhao, L.; Palomari Spolidorio, D.M.; Grenier, D. Green tea extract and its major constituent, epigallocatechin-3-gallate, induce epithelial beta-defensin secretion and prevent beta-defensin degradation by Porphyromonas gingivalis. J. Periodontal Res. 2014, 49, 615–623. [Google Scholar] [CrossRef]
  127. 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 beta-defensin secretion in oral epithelial cells. PLoS ONE 2015, 10, e0143158. [Google Scholar] [CrossRef]
  128. Gan, Y.; Cui, X.; Ma, T.; Liu, Y.; Li, A.; Huang, M. Paeoniflorin upregulates beta-defensin-2 expression in human bronchial epithelial cell through the p38 MAPK, ERK, and NF-kappaB signaling pathways. Inflammation 2014, 37, 1468–1475. [Google Scholar] [CrossRef]
  129. Holecek, M. Side effects of amino acid supplements. Physiol Res 2022, 71, 29–45. [Google Scholar] [CrossRef]
  130. Jang, J.H.; Shin, H.W.; Lee, J.M.; Lee, H.W.; Kim, E.C.; Park, S.H. An overview of pathogen recognition receptors for innate immunity in dental pulp. Mediators Inflamm. 2015, 2015, 794143. [Google Scholar] [CrossRef]
  131. Froy, O. Regulation of mammalian defensin expression by Toll-like receptor-dependent and independent signalling pathways. Cell Microbiol 2005, 7, 1387–1397. [Google Scholar] [CrossRef]
  132. Gaddis, D.E.; Michalek, S.M.; Katz, J. TLR4 signaling via MyD88 and TRIF differentially shape the CD4+ T cell response to Porphyromonas gingivalis hemagglutinin B. J. Immunol. 2011, 186, 5772–5783. [Google Scholar] [CrossRef] [Green Version]
  133. Fields, J.K.; Gunther, S.; Sundberg, E.J. Structural basis of IL-1 family cytokine signaling. Front. Immunol. 2019, 10, 1412. [Google Scholar] [CrossRef]
  134. Idriss, H.T.; Naismith, J.H. TNF alpha and the TNF receptor superfamily: Structure-function relationship(s). Microsc. Res. Tech. 2000, 50, 184–195. [Google Scholar] [CrossRef]
  135. Wassenaar, T.M.; Zimmermann, K. Lipopolysaccharides in Food, Food Supplements, and Probiotics: Should We be Worried? Eur. J. Microbiol. Immunol. 2018, 8, 63–69. [Google Scholar] [CrossRef]
  136. De Vito, P.; Incerpi, S.; Pedersen, J.Z.; Luly, P.; Davis, F.B.; Davis, P.J. Thyroid hormones as modulators of immune activities at the cellular level. Thyroid 2011, 21, 879–890. [Google Scholar] [CrossRef]
  137. Chandna, S.; Bathla, M. Oral manifestations of thyroid disorders and its management. Indian J. Endocrinol. Metab. 2011, 15, S113–S116. [Google Scholar] [CrossRef]
  138. Tangestani, H.; Boroujeni, H.K.; Djafarian, K.; Emamat, H.; Shab-Bidar, S. Vitamin D and the gut microbiota: A narrative literature review. Clin. Nutr. Res. 2021, 10, 181–191. [Google Scholar] [CrossRef]
  139. Kennedy Crispin, M.; Fuentes-Duculan, J.; Gulati, N.; Johnson-Huang, L.M.; Lentini, T.; Sullivan-Whalen, M.; Gilleaudeau, P.; Cueto, I.; Suarez-Farinas, M.; Lowes, M.A.; et al. Gene profiling of narrowband UVB-induced skin injury defines cellular and molecular innate immune responses. J. Investig. Dermatol. 2013, 133, 692–701. [Google Scholar] [CrossRef]
  140. Ou, Y.; Petersen, P.M. Application of ultraviolet light sources for in vivo disinfection. Jpn. J. Appl. Phys. 2021, 60, 100501. [Google Scholar] [CrossRef]
  141. Vieyra-Garcia, P.A.; Wolf, P. A deep dive into UV-based phototherapy: Mechanisms of action and emerging molecular targets in inflammation and cancer. Pharmacol. Ther. 2021, 222, 107784. [Google Scholar] [CrossRef]
  142. Morio, K.A.; Sternowski, R.H.; Brogden, K.A. Using ultraviolet (UV) light emitting diodes (LED) to create sterile root canals and to treat endodontic infections. Curr. Opin. Biomed. Eng. 2022, 23, 100397. [Google Scholar] [CrossRef]
  143. Morio, K.A.; Thayer, E.L.; Bates, A.M.; Brogden, K.A. 255-nm light-emitting diode kills Enterococcus faecalis and induces the production of cellular biomarkers in human embryonic palatal mesenchyme cells and gingival fibroblasts. J. Endod. 2019, 45, 774–783.e776. [Google Scholar] [CrossRef] [PubMed]
  144. Morio, K.A.; Sternowski, R.H.; Brogden, K.A. Dataset of endodontic microorganisms killed at 265 nm wavelength by an ultraviolet C light emitting diode in root canals of extracted, instrumented teeth. Data Brief 2022, 40, 107750. [Google Scholar] [CrossRef] [PubMed]
  145. Karampela, I.; Christodoulatos, G.S.; Vallianou, N.; Tsilingiris, D.; Chrysanthopoulou, E.; Skyllas, G.; Antonakos, G.; Marinou, I.; Vogiatzakis, E.; Armaganidis, A.; et al. Circulating chemerin and Its kinetics may be a useful diagnostic and prognostic biomarker in critically ill patients with sepsis: A prospective study. Biomolecules 2022, 12, 301. [Google Scholar] [CrossRef] [PubMed]
  146. Gambichler, T.; Skrygan, M.; Tomi, N.S.; Altmeyer, P.; Kreuter, A. Changes of antimicrobial peptide mRNA expression in atopic eczema following phototherapy. Br. J. Dermatol. 2006, 155, 1275–1278. [Google Scholar] [CrossRef]
  147. Kreuter, A.; Hyun, J.; Skrygan, M.; Sommer, A.; Bastian, A.; Altmeyer, P.; Gambichler, T. Ultraviolet A1-induced downregulation of human beta-defensins and interleukin-6 and interleukin-8 correlates with clinical improvement in localized scleroderma. Br. J. Dermatol. 2006, 155, 600–607. [Google Scholar] [CrossRef]
  148. Brogden, N.K.; Mehalick, L.; Fischer, C.L.; Wertz, P.W.; Brogden, K.A. The emerging role of peptides and lipids as antimicrobial epidermal barriers and modulators of local inflammation. Skin Pharmacol. Physiol. 2012, 25, 167–181. [Google Scholar] [CrossRef]
  149. Chaly, Y.V.; Paleolog, E.M.; Kolesnikova, T.S.; Tikhonov, I.I.; Petratchenko, E.V.; Voitenok, N.N. Neutrophil alpha-defensin human neutrophil peptide modulates cytokine production in human monocytes and adhesion molecule expression in endothelial cells. Eur. Cytokine Netw. 2000, 11, 257–266. [Google Scholar]
  150. Barabas, N.; Rohrl, J.; Holler, E.; Hehlgans, T. Beta-defensins activate macrophages and synergize in pro-inflammatory cytokine expression induced by TLR ligands. Immunobiology 2013, 218, 1005–1011. [Google Scholar] [CrossRef]
  151. Van Wetering, S.; MannesseLazeroms, S.P.G.; Dijkman, J.H.; Hiemstra, P.S. Effect of neutrophil serine proteinases and defensins on lung epithelial cells: Modulation of cytotoxicity and IL-8 production. J. Leukoc. Biol. 1997, 62, 217–226. [Google Scholar] [CrossRef]
  152. Niyonsaba, F.; Ushio, H.; Nakano, N.; Ng, W.; Sayama, K.; Hashimoto, K.; Nagaoka, I.; Okumura, K.; Ogawa, H. Antimicrobial peptides human beta-defensins stimulate epidermal keratinocyte migration, proliferation and production of proinflammatory cytokines and chemokines. J. Investig. Dermatol. 2007, 127, 594–604. [Google Scholar] [CrossRef]
  153. Petrov, V.; Funderburg, N.; Weinberg, A.; Sieg, S. Human beta defensin-3 induces chemokines from monocytes and macrophages: Diminished activity in cells from HIV-infected persons. Immunology 2013, 140, 413–420. [Google Scholar] [CrossRef]
  154. Scott, M.G.; Davidson, D.J.; Gold, M.R.; Bowdish, D.; Hancock, R.E. The human antimicrobial peptide LL-37 is a multifunctional modulator of innate immune responses. J. Immunol. 2002, 169, 3883–3891. [Google Scholar] [CrossRef]
  155. Boniotto, M.; Jordan, W.J.; Eskdale, J.; Tossi, A.; Antcheva, N.; Crovella, S.; Connell, N.D.; Gallagher, G. Human beta-defensin 2 induces a vigorous cytokine response in peripheral blood mononuclear cells. Antimicrob. Agents Chemother. 2006, 50, 1433–1441. [Google Scholar] [CrossRef]
  156. Presicce, P.; Giannelli, S.; Taddeo, A.; Villa, M.L.; Della Bella, S. Human defensins activate monocyte-derived dendritic cells, promote the production of proinflammatory cytokines, and up-regulate the surface expression of CD91. J. Leukoc. Biol. 2009, 86, 941–948. [Google Scholar] [CrossRef]
  157. Jin, G.; Kawsar, H.I.; Hirsch, S.A.; Zeng, C.; Jia, X.; Feng, Z.; Ghosh, S.K.; Zheng, Q.Y.; Zhou, A.; McIntyre, T.M.; et al. An antimicrobial peptide regulates tumor-associated macrophage trafficking via the chemokine receptor CCR2, a model for tumorigenesis. PLoS ONE 2010, 5, e10993. [Google Scholar] [CrossRef]
  158. Semple, F.; MacPherson, H.; Webb, S.; Cox, S.L.; Mallin, L.J.; Tyrrell, C.; Grimes, G.R.; Semple, C.A.; Nix, M.A.; Millhauser, G.L.; et al. Human beta-defensin 3 affects the activity of pro-inflammatory pathways associated with MyD88 and TRIF. Eur. J. Immunol. 2011, 41, 3291–3300. [Google Scholar] [CrossRef]
  159. Semple, F.; Webb, S.; Li, H.N.; Patel, H.B.; Perretti, M.; Jackson, I.J.; Gray, M.; Davidson, D.J.; Dorin, J.R. Human beta-defensin 3 has immunosuppressive activity in vitro and in vivo. Eur. J. Immunol. 2010, 40, 1073–1078. [Google Scholar] [CrossRef]
  160. Dietrich, D.E.; Xiao, X.; Dawson, D.V.; Belanger, M.; Xie, H.; Progulske-Fox, A.; Brogden, K.A. Human alpha- and beta-defensins bind to immobilized adhesins from Porphyromonas gingivalis. Infect. Immun. 2008, 76, 5714–5720. [Google Scholar] [CrossRef]
  161. Gallo, S.A.; Wang, W.; Rawat, S.S.; Jung, G.; Waring, A.J.; Cole, A.M.; Lu, H.; Yan, X.; Daly, N.L.; Craik, D.J.; et al. Theta-defensins prevent HIV-1 Env-mediated fusion by binding gp41 and blocking 6-helix bundle formation. J. Biol. Chem. 2006, 281, 18787–18792. [Google Scholar] [CrossRef]
  162. Wei, G.; de Leeuw, E.; Pazgier, M.; Yuan, W.; Zou, G.; Wang, J.; Ericksen, B.; Lu, W.Y.; Lehrer, R.I.; Lu, W. Through the looking glass, mechanistic insights from enantiomeric human defensins. J. Biol. Chem. 2009, 284, 29180–29192. [Google Scholar] [CrossRef]
  163. Pingel, L.C.; Kohlgraf, K.G.; Hansen, C.J.; Eastman, C.G.; Dietrich, D.E.; Burnell, K.K.; Srikantha, R.N.; Xiao, X.; Belanger, M.; Progulske-Fox, A.; et al. Human beta-defensin 3 binds to hemagglutinin B (rHagB), a non-fimbrial adhesin from Porphyromonas gingivalis, and attenuates a pro-inflammatory cytokine response. Immunol. Cell Biol. 2008, 86, 643–649. [Google Scholar] [CrossRef] [PubMed]
  164. Borgwardt, D.S.; Martin, A.D.; Van Hemert, J.R.; Yang, J.; Fischer, C.L.; Recker, E.N.; Nair, P.R.; Vidva, R.; Chandrashekaraiah, S.; Progulske-Fox, A.; et al. Histatin 5 binds to Porphyromonas gingivalis hemagglutinin B (HagB) and alters HagB-induced chemokine responses. Sci. Rep. 2014, 4, 3904. [Google Scholar] [CrossRef] [PubMed]
  165. Scott, A.; Weldon, S.; Buchanan, P.J.; Schock, B.; Ernst, R.K.; McAuley, D.F.; Tunney, M.M.; Irwin, C.R.; Elborn, J.S.; Taggart, C.C. Evaluation of the ability of LL-37 to neutralise LPS in vitro and ex vivo. PLoS ONE 2011, 6, e26525. [Google Scholar] [CrossRef] [PubMed]
  166. Wright, S.D.; Ramos, R.A.; Tobias, P.S.; Ulevitch, R.J.; Mathison, J.C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249, 1431–1433. [Google Scholar] [CrossRef]
  167. Gallo, R.L.; Hooper, L.V. Epithelial antimicrobial defence of the skin and intestine. Nat. Rev. Immunol. 2012, 12, 503–516. [Google Scholar] [CrossRef]
  168. Van Hemert, J.R.; Recker, E.N.; Dietrich, D.; Progulske-Fox, A.; Kurago, Z.B.; Walters, K.S.; Cavanaugh, J.E.; Brogden, K.A. Human beta-defensin-3 alters, but does not inhibit, the binding of Porphyromonas gingivalis haemagglutinin B to the surface of human dendritic cells. Int. J. Antimicrob. Agents 2012, 40, 75–79. [Google Scholar] [CrossRef]
  169. Nagaoka, I.; Hirota, S.; Niyonsaba, F.; Hirata, M.; Adachi, Y.; Tamura, H.; Heumann, D. Cathelicidin family of antibacterial peptides CAP18 and CAP11 inhibit the expression of TNF-alpha by blocking the binding of LPS to CD14(+) cells. J. Immunol. 2001, 167, 3329–3338. [Google Scholar] [CrossRef]
  170. Miles, K.; Clarke, D.J.; Lu, W.; Sibinska, Z.; Beaumont, P.E.; Davidson, D.J.; Barr, T.A.; Campopiano, D.J.; Gray, M. Dying and necrotic neutrophils are anti-inflammatory secondary to the release of alpha-defensins. J. Immunol. 2009, 183, 2122–2132. [Google Scholar] [CrossRef]
  171. Shi, J.; Aono, S.; Lu, W.; Ouellette, A.J.; Hu, X.; Ji, Y.; Wang, L.; Lenz, S.; van Ginkel, F.W.; Liles, M.; et al. A novel role for defensins in intestinal homeostasis: Regulation of IL-1beta secretion. J. Immunol. 2007, 179, 1245–1253. [Google Scholar] [CrossRef]
  172. Mookherjee, N.; Brown, K.L.; Bowdish, D.M.; Doria, S.; Falsafi, R.; Hokamp, K.; Roche, F.M.; Mu, R.; Doho, G.H.; Pistolic, J.; et al. Modulation of the TLR-mediated inflammatory response by the endogenous human host defense peptide LL-37. J. Immunol. 2006, 176, 2455–2464. [Google Scholar] [CrossRef]
  173. Hilchie, A.L.; Wuerth, K.; Hancock, R.E. Immune modulation by multifaceted cationic host defense (antimicrobial) peptides. Nat. Chem. Biol. 2013, 9, 761–768. [Google Scholar] [CrossRef]
  174. Mookherjee, N.; Hamill, P.; Gardy, J.; Blimkie, D.; Falsafi, R.; Chikatamarla, A.; Arenillas, D.J.; Doria, S.; Kollmann, T.R.; Hancock, R.E. Systems biology evaluation of immune responses induced by human host defence peptide LL-37 in mononuclear cells. Mol. Biosyst. 2009, 5, 483–496. [Google Scholar] [CrossRef]
  175. Lai, Y.; Gallo, R.L. AMPed up immunity: How antimicrobial peptides have multiple roles in immune defense. Trends Immunol. 2009, 30, 131–141. [Google Scholar] [CrossRef]
  176. Brogden, K.A.; Bates, A.M.; Fischer, C.L. Antimicrobial peptides in host defense: Functions beyond antimicrobial activity. In Antimicrobial Peptides—Role in Human Health and Disease; Harder, J., Schroeder, J.M., Kaufmann, S.H., Mercer, A.A., Weber, B., Eds.; Birkhauser Advances in Infectious Diseases; Springer International Publishing: Cham, Switzerland, 2016; pp. 129–146. [Google Scholar] [CrossRef]
  177. Drayton, M.; Deisinger, J.P.; Ludwig, K.C.; Raheem, N.; Muller, A.; Schneider, T.; Straus, S.K. Host defense peptides: Dual antimicrobial and immunomodulatory action. Int. J. Mol. Sci. 2021, 22, 11172. [Google Scholar] [CrossRef]
  178. Takahashi, M.; Umehara, Y.; Yue, H.; Trujillo-Paez, J.V.; Peng, G.; Nguyen, H.L.T.; Ikutama, R.; Okumura, K.; Ogawa, H.; Ikeda, S.; et al. The antimicrobial peptide human beta-defensin-3 accelerates wound healing by promoting angiogenesis, cell migration, and proliferation through the FGFR/JAK2/STAT3 signaling pathway. Front. Immunol. 2021, 12, 712781. [Google Scholar] [CrossRef]
  179. Davidson, D.J.; Currie, A.J.; Reid, G.S.; Bowdish, D.M.; MacDonald, K.L.; Ma, R.C.; Hancock, R.E.; Speert, D.P. The cationic antimicrobial peptide LL-37 modulates dendritic cell differentiation and dendritic cell-induced T cell polarization. J. Immunol. 2004, 172, 1146–1156. [Google Scholar] [CrossRef]
  180. Ramos, R.; Silva, J.P.; Rodrigues, A.C.; Costa, R.; Guardao, L.; Schmitt, F.; Soares, R.; Vilanova, M.; Domingues, L.; Gama, M. Wound healing activity of the human antimicrobial peptide LL37. Peptides 2011, 32, 1469–1476. [Google Scholar] [CrossRef]
  181. Warnke, P.H.; Voss, E.; Russo, P.A.; Stephens, S.; Kleine, M.; Terheyden, H.; Liu, Q. Antimicrobial peptide coating of dental implants: Biocompatibility assessment of recombinant human beta defensin-2 for human cells. Int. J. Oral Maxillofac. Implants 2013, 28, 982–988. [Google Scholar] [CrossRef]
  182. Heilborn, J.D.; Nilsson, M.F.; Kratz, G.; Weber, G.; Sorensen, O.; Borregaard, N.; Stahle-Backdahl, M. The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of human skin wounds and is lacking in chronic ulcer epithelium. J. Investig. Dermatol. 2003, 120, 379–389. [Google Scholar] [CrossRef]
  183. Nakatsuji, T.; Gallo, R.L. Antimicrobial peptides: Old molecules with new ideas. J. Investig. Dermatol. 2012, 132, 887–895. [Google Scholar] [CrossRef] [Green Version]
  184. Hao, J.; Liu, M.G.; Yu, Y.Q.; Cao, F.L.; Li, Z.; Lu, Z.M.; Chen, J. Roles of peripheral mitogen-activated protein kinases in melittin-induced nociception and hyperalgesia. Neuroscience 2008, 152, 1067–1075. [Google Scholar] [CrossRef] [PubMed]
  185. Son, D.J.; Lee, J.W.; Lee, Y.H.; Song, H.S.; Lee, C.K.; Hong, J.T. Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol. Ther. 2007, 115, 246–270. [Google Scholar] [CrossRef] [PubMed]
  186. Rykaczewska-Czerwinska, M.; Oles, P.; Oles, M.; Kuczer, M.; Konopinska, D.; Plech, A. Effect of alloferon 1 on central nervous system in rats. Acta Pol. Pharm. 2015, 72, 205–211. [Google Scholar] [CrossRef] [PubMed]
  187. Merlo, L.A.; Bastos, L.F.; Godin, A.M.; Rocha, L.T.; Nascimento, E.B., Jr.; Paiva, A.L.; Moraes-Santos, T.; Zumpano, A.A.; Bastos, E.M.; Heneine, L.G.; et al. Effects induced by Apis mellifera venom and its components in experimental models of nociceptive and inflammatory pain. Toxicon 2011, 57, 764–771. [Google Scholar] [CrossRef]
  188. Logashina, Y.A.; Solstad, R.G.; Mineev, K.S.; Korolkova, Y.V.; Mosharova, I.V.; Dyachenko, I.A.; Palikov, V.A.; Palikova, Y.A.; Murashev, A.N.; Arseniev, A.S.; et al. New disulfide-stabilized fold provides sea anemone peptide to exhibit both antimicrobial and TRPA1 potentiating properties. Toxins 2017, 9, 154. [Google Scholar] [CrossRef]
  189. Chen, W.F.; Huang, S.Y.; Liao, C.Y.; Sung, C.S.; Chen, J.Y.; Wen, Z.H. The use of the antimicrobial peptide piscidin (PCD)-1 as a novel anti-nociceptive agent. Biomaterials 2015, 53, 1–11. [Google Scholar] [CrossRef]
  190. Edwards, A.B.; Mastaglia, F.L.; Knuckey, N.W.; Meloni, B.P. Neuroprotective cationic arginine-rich peptides (CARPs): An assessment of their clinical safety. Drug Saf. 2020, 43, 957–969. [Google Scholar] [CrossRef]
  191. Walker, M.C.; Eslami, S.M.; Hetrick, K.J.; Ackenhusen, S.E.; Mitchell, D.A.; van der Donk, W.A. Precursor peptide-targeted mining of more than one hundred thousand genomes expands the lanthipeptide natural product family. BMC Genomics 2020, 21, 387. [Google Scholar] [CrossRef]
  192. Ghia, J.E.; Crenner, F.; Metz-Boutigue, M.H.; Aunis, D.; Angel, F. Effects of a chromogranin-derived peptide (CgA 47-66) in the writhing nociceptive response induced by acetic acid in rats. Regul. Pept. 2004, 119, 199–207. [Google Scholar] [CrossRef]
  193. Wood, S.M.; Kennedy, J.S.; Arsenault, J.E.; Thomas, D.L.; Buck, R.H.; Shippee, R.L.; DeMichele, S.J.; Winship, T.R.; Schaller, J.P.; Montain, S.; et al. Novel nutritional immune formula maintains host defense mechanisms. Mil. Med. 2005, 170, 975–985. [Google Scholar] [CrossRef] [Green Version]
  194. Krisanaprakornkit, S.; Kimball, J.R.; Dale, B.A. Regulation of human beta-defensin-2 in gingival epithelial cells: The involvement of mitogen-activated protein kinase pathways, but not the NF-kappaB transcription factor family. J. Immunol. 2002, 168, 316–324. [Google Scholar] [CrossRef]
  195. Li, G.; Domenico, J.; Jia, Y.; Lucas, J.J.; Gelfand, E.W. NF-kappaB-dependent induction of cathelicidin-related antimicrobial peptide in murine mast cells by lipopolysaccharide. Int. Arch. Allergy Immunol. 2009, 150, 122–132. [Google Scholar] [CrossRef]
  196. Steubesand, N.; Kiehne, K.; Brunke, G.; Pahl, R.; Reiss, K.; Herzig, K.H.; Schubert, S.; Schreiber, S.; Folsch, U.R.; Rosenstiel, P.; et al. The expression of the beta-defensins hBD-2 and hBD-3 is differentially regulated by NF-kappaB and MAPK/AP-1 pathways in an in vitro model of Candida esophagitis. BMC Immunol. 2009, 10, 36. [Google Scholar] [CrossRef]
  197. Wehkamp, K.; Schwichtenberg, L.; Schroder, J.M.; Harder, J. Pseudomonas aeruginosa- and IL-1beta-mediated induction of human beta-defensin-2 in keratinocytes is controlled by NF-kappaB and AP-1. J. Investig. Dermatol. 2006, 126, 121–127. [Google Scholar] [CrossRef]
  198. Drover, V.A.; Nguyen, D.V.; Bastie, C.C.; Darlington, Y.F.; Abumrad, N.A.; Pessin, J.E.; London, E.; Sahoo, D.; Phillips, M.C. CD36 mediates both cellular uptake of very long chain fatty acids and their intestinal absorption in mice. J. Biol. Chem. 2008, 283, 13108–13115. [Google Scholar] [CrossRef]
  199. Ren, Y.; Li, S.; Zhu, R.; Wan, C.; Song, D.; Zhu, J.; Cai, G.; Long, S.; Kong, L.; Yu, W. Discovery of STAT3 and Histone Deacetylase (HDAC) Dual-Pathway Inhibitors for the Treatment of Solid Cancer. J. Med. Chem. 2021, 64, 7468–7482. [Google Scholar] [CrossRef]
  200. Li, A.; Gan, Y.; Wang, R.; Liu, Y.; Ma, T.; Huang, M.; Cui, X. IL-22 up-regulates beta-defensin-2 expression in human alveolar epithelium via STAT3 but not NF-kappaB signaling pathway. Inflammation 2015, 38, 1191–1200. [Google Scholar] [CrossRef]
  201. Sinha, R.; Yen, P.M. Cellular action of thyroid hormone. In Endotext [Internet]; Feingold, K.R., Anawalt, B., Boyce, A., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2018. [Google Scholar]
  202. Kohlgraf, K.G.; Ackermann, A.; Lu, X.; Burnell, K.; Belanger, M.; Cavanaugh, J.E.; Xie, H.; Progulske-Fox, A.; Brogden, K.A. Defensins attenuate cytokine responses yet enhance antibody responses to Porphyromonas gingivalis adhesins in mice. Future Microbiol. 2010, 5, 115–125. [Google Scholar] [CrossRef]
  203. Harvey, L.E.; Kohlgraf, K.G.; Mehalick, L.A.; Raina, M.; Recker, E.N.; Radhakrishnan, S.; Prasad, S.A.; Vidva, R.; Progulske-Fox, A.; Cavanaugh, J.E.; et al. Defensin DEFB103 bidirectionally regulates chemokine and cytokine responses to a pro-inflammatory stimulus. Sci. Rep. 2013, 3, 1232. [Google Scholar] [CrossRef]
  204. Ruan, Y.; Shen, T.; Wang, Y.; Hou, M.; Li, J.; Sun, T. Antimicrobial peptide LL-37 attenuates LTA induced inflammatory effect in macrophages. Int. Immunopharmacol. 2013, 15, 575–580. [Google Scholar] [CrossRef]
  205. Oudhoff, M.J.; Blaauboer, M.E.; Nazmi, K.; Scheres, N.; Bolscher, J.G.; Veerman, E.C. The role of salivary histatin and the human cathelicidin LL-37 in wound healing and innate immunity. Biol. Chem. 2010, 391, 541–548. [Google Scholar] [CrossRef] [PubMed]
  206. Yin, J.; Yu, F.S. LL-37 via EGFR transactivation to promote high glucose-attenuated epithelial wound healing in organ-cultured corneas. Investig. Ophthalmol. Vis. Sci. 2010, 51, 1891–1897. [Google Scholar] [CrossRef] [PubMed]
  207. Aarbiou, J.; Verhoosel, R.M.; Van Wetering, S.; De Boer, W.I.; Van Krieken, J.H.; Litvinov, S.V.; Rabe, K.F.; Hiemstra, P.S. Neutrophil defensins enhance lung epithelial wound closure and mucin gene expression in vitro. Am. J. Respir. Cell Mol. Biol. 2004, 30, 193–201. [Google Scholar] [CrossRef] [PubMed]
  208. Joly, S.; Librant, J.; Brogden, K.A.; Burnell, K.K.; Johnson, G.; Guthmiller, J.M. Beta-defensins differentially induce cytokines in gingival keratinocyte cultures. In Proceedings of the 83rd General Session IADR meeting, Baltimore, MD, USA, 9–12 March 2005. [Google Scholar]
  209. Niyonsaba, F.; Ushio, H.; Nagaoka, I.; Okumura, K.; Ogawa, H. The human beta-defensins (-1, -2, -3, -4) and cathelicidin LL-37 induce IL-18 secretion through p38 and ERK MAPK activation in primary human keratinocytes. J. Immunol. 2005, 175, 1776–1784. [Google Scholar] [CrossRef] [PubMed]
  210. Khine, A.A.; Del Sorbo, L.; Vaschetto, R.; Voglis, S.; Tullis, E.; Slutsky, A.S.; Downey, G.P.; Zhang, H. Human neutrophil peptides induce interleukin-8 production through the P2Y6 signaling pathway. Blood 2006, 107, 2936–2942. [Google Scholar] [CrossRef]
  211. van Wetering, S.; Mannesse-Lazeroms, S.P.; van Sterkenburg, M.A.; Hiemstra, P.S. Neutrophil defensins stimulate the release of cytokines by airway epithelial cells: Modulation by dexamethasone. Inflamm. Res. 2002, 51, 8–15. [Google Scholar] [CrossRef]
  212. Territo, M.C.; Ganz, T.; Selsted, M.E.; Lehrer, R. Monocyte-chemotactic activity of defensins from human neutrophils. J. Clin. Investig. 1989, 84, 2017–2020. [Google Scholar] [CrossRef]
  213. Shaykhiev, R.; Beisswenger, C.; Kandler, K.; Senske, J.; Puchner, A.; Damm, T.; Behr, J.; Bals, R. Human endogenous antibiotic LL-37 stimulates airway epithelial cell proliferation and wound closure. Am. J. Physiol. Lung Cell Mol. Physiol. 2005, 289, L842–L848. [Google Scholar] [CrossRef]
  214. Niyonsaba, F.; Iwabuchi, K.; Someya, A.; Hirata, M.; Matsuda, H.; Ogawa, H.; Nagaoka, I. A cathelicidin family of human antibacterial peptide LL-37 induces mast cell chemotaxis. Immunology 2002, 106, 20–26. [Google Scholar] [CrossRef]
  215. Nishimura, M.; Abiko, Y.; Kurashige, Y.; Takeshima, M.; Yamazaki, M.; Kusano, K.; Saitoh, M.; Nakashima, K.; Inoue, T.; Kaku, T. Effect of defensin peptides on eukaryotic cells: Primary epithelial cells, fibroblasts and squamous cell carcinoma cell lines. J. Dermatol. Sci. 2004, 36, 87–95. [Google Scholar] [CrossRef]
  216. Baroni, A.; Donnarumma, G.; Paoletti, I.; Longanesi-Cattani, I.; Bifulco, K.; Tufano, M.A.; Carriero, M.V. Antimicrobial human beta-defensin-2 stimulates migration, proliferation and tube formation of human umbilical vein endothelial cells. Peptides 2009, 30, 267–272. [Google Scholar] [CrossRef]
  217. Lillard, J.W., Jr.; Boyaka, P.N.; Chertov, O.; Oppenheim, J.J.; McGhee, J.R. Mechanisms for induction of acquired host immunity by neutrophil peptide defensins. Proc. Natl. Acad. Sci. USA 1999, 96, 651–656. [Google Scholar] [CrossRef]
  218. Aarbiou, J.; Ertmann, M.; van Wetering, S.; van Noort, P.; Rook, D.; Rabe, K.F.; Litvinov, S.V.; van Krieken, J.H.; de Boer, W.I.; Hiemstra, P.S. Human neutrophil defensins induce lung epithelial cell proliferation in vitro. J. Leukoc. Biol. 2002, 72, 167–174. [Google Scholar] [CrossRef]
  219. Chamorro, C.I.; Weber, G.; Gronberg, A.; Pivarcsi, A.; Stahle, M. The human antimicrobial peptide LL-37 suppresses apoptosis in keratinocytes. J. Investig. Dermatol. 2009, 129, 937–944. [Google Scholar] [CrossRef]
  220. Brogden, K.A.; Heidari, M.; Sacco, R.E.; Palmquist, D.; Guthmiller, J.M.; Johnson, G.K.; Jia, H.P.; Tack, B.F.; McCray, P.B. Defensin-induced adaptive immunity in mice and its potential in preventing periodontal disease. Oral Microbiol. Immunol. 2003, 18, 95–99. [Google Scholar] [CrossRef]
  221. Schrumpf, J.A.; van Sterkenburg, M.A.; Verhoosel, R.M.; Zuyderduyn, S.; Hiemstra, P.S. Interleukin 13 exposure enhances vitamin D-mediated expression of the human cathelicidin antimicrobial peptide 18/LL-37 in bronchial epithelial cells. Infect. Immun. 2012, 80, 4485–4494. [Google Scholar] [CrossRef] [Green Version]
Scheme 1. The benefits of inducing endogenous antimicrobial peptide (AMP) transcription and expression.
Scheme 1. The benefits of inducing endogenous antimicrobial peptide (AMP) transcription and expression.
Antibiotics 12 00361 sch001
Figure 1. A generalized scheme showing the pathways for the induction of AMP transcription and expression via nutritional, hormonal, and physical stimuli through the MAPK signaling, vitamin D signaling, HDAC inhibition signaling, NF-kB signaling, and TR/RXT signaling modules. All signaling cascades converge onto a few transcription factors (TF) that bind to TF-specific motifs and response elements in the promoter regions of the respective AMP genes. Detailed information on each of these signaling pathways is included in Supplemental Figures S1–S4.
Figure 1. A generalized scheme showing the pathways for the induction of AMP transcription and expression via nutritional, hormonal, and physical stimuli through the MAPK signaling, vitamin D signaling, HDAC inhibition signaling, NF-kB signaling, and TR/RXT signaling modules. All signaling cascades converge onto a few transcription factors (TF) that bind to TF-specific motifs and response elements in the promoter regions of the respective AMP genes. Detailed information on each of these signaling pathways is included in Supplemental Figures S1–S4.
Antibiotics 12 00361 g001
Table 2. Roles of AMPs in inflammation, immunity, healing, and pain. The activities below were compiled from results reported in Supplemental Table S1.
Table 2. Roles of AMPs in inflammation, immunity, healing, and pain. The activities below were compiled from results reported in Supplemental Table S1.
AMP Concentration (μM)ActivityAMPs Involved (Reported μM Concentrations)
Roles of AMPs in inflammation
0.02–0.30Binds microbial antigensLL-37 (0.02 μM), HBD3 (0.19 μM), HBD1 (0.25 μM), and Histatin 5 (0.30 μM)
1.94Attenuated pathway signalingHBD3 (1.94 μM)
4.45Attenuated gene expressionLL-37 (4.45 μM)
0.19–2.50Attenuated cytokine responseHBD3 (0.19 μM), LL-37 (0.22–2.50 μM), HBD1 (0.25 μM), and HNP-1,2 (0.29 μM)
1.11Activated pathway signalingLL-37 (1.11 μM)
2.32Enhanced gene expressionHNP-1-3 (2.32 μM)
0.03–29.00Enhanced cytokine responseHBD1 (0.03–5.08 μM), LL-37 (0.11–11.12 μM), HBD2 (0.46–4.61 μM), HNP-1-3 (0.87–29.0 μM), and HBD3 (0.97–3.87 μM)
Roles of AMPs in immunity
0.01–29.00Chemotactic activityHNP-1-3 (0.01–29.00 μM) and LL-37 (0.22-4.45 μM)
1.00–2.32Promoted cell migrationLL-37 (1.00 μM) and HNP-1-3 (2.32 μM)
11.12Increased cell markersLL-37 (11.12 μM)
0.22-11.12Induced proliferationLL-37 (0.22–2.22 μM), HNP-1-3 (0.29–2.90 μM), HBD3 (0.97–1.55 μM), HBD2 (1.15–2.31 μM), and HBD1 (1.27 μM)
Induced Th1 cytokine profileLL-37 (11.12 μM)
0.19–1.45Enhanced antibody responseHBD3 (0.19 μM), HBD2 (0.23 μM), HBD1 (0.25 μM), and HNP-1-3 (0.29–1.45 μM)
1.00Suppressed apoptosisLL-37 (1.0 μM)
2.54–12.70Decreased cell numbersHBD1 (2.54–12.70 μM)
2.22–14.50Cell cytotoxicityLL-37 (2.2–11.12 μM), HBD2 (6.92 μM), HNP-1-3 (14.50 μM)
Roles of AMPs in angiogenesis, vasculogenesis, and wound healing
0.12–58.00AngiogenesisHBD2 (0.12 μM) and HBD3 (58.00 μM)
0.11–2.31Promoted cell migrationHBD3 (0.11–0.97 μM), HBD2 (0.12–2.31 μM), and HBD4 (2.22 μM)
1.16–58.00Enhanced wound closureLL-37 (0.11–2.50 μM), HBD2 (0.12 μM), HNP-1-3 (1.16–2.32 μM), and HBD3 (58.00 μM)
14.50Delayed wound closureHNP-1-3 (14.50 μM)
58.00Enhanced wound healingHBD3 (58.00 μM)
Roles of AMPs in pain nociception
0.2–6.0 mg/kgPain antinociceptionAlloferon (0.10 μM), PCD-1 (3.69 μM), Ueq 12-1 (0.2 mg/kg), CgA (0.5 mg/kg), and AMV (6 mg/kg)
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

Morio, K.A.; Sternowski, R.H.; Brogden, K.A. Induction of Endogenous Antimicrobial Peptides to Prevent or Treat Oral Infection and Inflammation. Antibiotics 2023, 12, 361. https://doi.org/10.3390/antibiotics12020361

AMA Style

Morio KA, Sternowski RH, Brogden KA. Induction of Endogenous Antimicrobial Peptides to Prevent or Treat Oral Infection and Inflammation. Antibiotics. 2023; 12(2):361. https://doi.org/10.3390/antibiotics12020361

Chicago/Turabian Style

Morio, Kimberly A., Robert H. Sternowski, and Kim A. Brogden. 2023. "Induction of Endogenous Antimicrobial Peptides to Prevent or Treat Oral Infection and Inflammation" Antibiotics 12, no. 2: 361. https://doi.org/10.3390/antibiotics12020361

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

Article Metrics

Back to TopTop