Skip to main content
Advances in Nutrition logoLink to Advances in Nutrition
. 2024 Jul 23;15(9):100277. doi: 10.1016/j.advnut.2024.100277

Nutritional Modulation of Host Defense Peptide Synthesis: A Novel Host-Directed Antimicrobial Therapeutic Strategy?

Melanie Whitmore 1, Isabel Tobin 1, Amanda Burkardt 1,, Guolong Zhang 1,
PMCID: PMC11381887  PMID: 39053604

Abstract

The escalating threat of antimicrobial resistance underscores the imperative for innovative therapeutic strategies. Host defense peptides (HDPs), integral components of innate immunity, exhibit profound antimicrobial and immunomodulatory properties. Various dietary compounds, such as short-chain fatty acids, vitamins, minerals, sugars, amino acids, phytochemicals, bile acids, probiotics, and prebiotics have been identified to enhance the synthesis of endogenous HDPs without provoking inflammatory response or compromising barrier integrity. Additionally, different classes of these compounds synergize in augmenting HDP synthesis and disease resistance. Moreover, dietary supplementation of several HDP-inducing compounds or their combinations have demonstrated robust protection in rodents, rabbits, pigs, cattle, and chickens from experimental infections. However, the efficacy of these compounds in inducing HDP synthesis varies considerably among distinct compounds. Additionally, the regulation of HDP genes occurs in a gene-specific, cell type–specific, and species-specific manner. In this comprehensive review, we systematically summarized the modulation of HDP synthesis and the mechanism of action attributed to each major class of dietary compounds, including their synergistic combinations, across a spectrum of animal species including humans. We argue that the ability to enhance innate immunity and barrier function without triggering inflammation or microbial resistance positions the nutritional modulation of endogenous HDP synthesis as a promising host-directed approach for mitigating infectious diseases and antimicrobial resistance. These HDP-inducing compounds, particularly in combinations, harbor substantial clinical potential for further exploration in antimicrobial therapies for both human and other animals.

Keywords: host defense peptides, antimicrobial peptides, antimicrobial resistance, host-directed antimicrobial therapy, nutritional regulation


Statement of Significance.

This review systematically categorizes various dietary compounds capable of inducing host defense peptides by synthesizing a large body of evidence and further summarizes the mechanistic underpinnings of each compound class. This review provides a foundation for future work aimed at developing these compounds as innovative antibiotic alternatives for disease control and prevention, addressing the escalating challenge posed by antimicrobial resistance.

Introduction

The pervasive and indiscriminate usage of antibiotics in human health care and agriculture has led to the emergence of antibiotic-resistant bacteria that are associated with ∼700,000 annual deaths globally [1,2]. Antibiotic resistance has become a major public health concern, arising from the ability of bacteria to survive and proliferate in the presence of antibiotics. Evolutionarily, bacteria exposed to antibiotics experience selective pressure, allowing resistant strains to thrive and multiply through mutation and horizontal gene transfer [3,4]. Biochemically, bacteria gain resistance through mechanisms such as producing enzymes that degrade antibiotics, altering targets for antibiotics, reducing the influx of antibiotics, and increasing the expression of efflux pumps to expel antibiotics [3,4].

In human medicine, antibiotic resistance complicates the treatment of infections, leading to longer hospital stays, higher medical costs, and increased mortality [5]. In veterinary science, the widespread use of antibiotics in livestock for growth promotion and disease prevention contributes to the emergence of resistant bacteria, which can transfer to humans through direct contact or the food chain [6]. A coordinated One Health approach across human health and veterinary medicine sectors is required to preserve the efficacy of current antimicrobials while novel approaches are developed to control infectious diseases [7]. A pressing need exists for novel approaches in the face of rapid resistance development. Modulation of the synthesis of host defense peptides (HDPs), also known as antimicrobial peptides, has emerged as a promising host-directed antimicrobial strategy [8,9].

HDPs, characterized by their short, positively charged, and amphipathic features, constitute an integral component of innate immunity across a wide spectrum of life forms, ranging from mammals, amphibians, and birds to plants, fungi, and bacteria [10,11]. Two major families of HDPs, namely cathelicidins and defensins (DEFs), are produced in vertebrate animals [[10], [11], [12]]. Cathelicidins are distinguished by the presence of a highly conserved cathelin precursor sequence that undergoes enzymatic cleavage to attain biological activity, while DEFs are defined by the conservation of multiple cysteines in defined positions forming intramolecular disulfide bonds [12]. The synthesis of HDPs is predominantly orchestrated by cell types in direct contact with invading pathogens, such as mucosal epithelial cells, neutrophils, and macrophages [12]. HDPs are either constitutively produced for constant host surveillance and protection or inducibly expressed in response to infection or inflammation [12].

As critical components of innate immunity, HDPs exhibit not only a direct antimicrobial activity against a broad spectrum of pathogens but also multifaceted immunomodulatory properties [[10], [11], [12]]. Their primary antimicrobial mechanism involves membrane disruption through electrostatic interactions with negatively charged microbial membranes, followed by membrane disruption and cell lysis owing to the amphiphilic nature of HDPs [[10], [11], [12]]. Additionally, HDPs play pivotal roles in orchestrating immune responses to pathogen invasion by recruiting different types of immune cells to the sites of infection through direct chemoattraction or stimulation of chemokine secretion [12]. Specific HDPs further contribute to host immune responses to pathogens by promoting phagocytosis or the formation of neutrophil extracellular traps [12]. Moreover, certain HDPs have demonstrated ability to neutralize bacterial endotoxins, modulate host signaling pathways, or promote wound healing to reduce inflammation and facilitate tissue repair [12].

Despite the clinical approval of a few HDPs, such as nisin and daptomycin, challenges persist in the application of the synthetic or recombinant form of HDPs due to high production costs and suboptimal pharmacokinetics [[10], [11], [12]]. Recent years have witnessed a burgeoning interest in identifying nutritional compounds capable of inducing endogenous HDP synthesis in humans and animals. These HDP-inducing nutritional compounds, such as fatty acids, vitamins, animal acids, sugars, minerals, bile acids, phytochemicals, epigenetic modulators, probiotics, and prebiotics, have potential to offer a cost effective alternative approach to disease mitigation [8,9,13] (Figure 1). Importantly, such HDP-inducing compounds pose minimum risk of triggering antimicrobial resistance as they target the host rather than the pathogen. This review aims to systematically summarize current findings regarding the regulation of HDP synthesis by diverse classes of nutritional compounds and the underlying mechanisms. Additionally, the review highlights the potential for synergistic induction of HDPs and improved disease mitigation through a combination of compounds, thereby providing a promising avenue for optimizing host-directed therapeutic strategies.

FIGURE 1.

FIGURE 1

Classification of dietary compounds with the ability to induce host defense peptides. Key examples are listed in each category.

Methods

Articles referenced in this systematic review were obtained by searching PubMed for relevant, peer-reviewed, full-text articles that were published in English since the year 1980. The search terms used included (“host defense peptide∗” OR “host defence peptide∗” OR “antimicrobial peptide∗” OR defensin∗ OR cathelicidin∗ OR LL-37) AND (butyrate OR “fatty acid∗” OR vitamin∗ OR “amino acid∗” OR sugar∗ OR mineral∗ OR “bile acid∗” OR polyphenol∗ OR phytochemical∗ OR epigenetic OR probiotic∗ OR prebiotic∗ OR nutrient∗) AND (regulate OR regulation OR modulate OR modulation). References from selected articles were further scanned for relevance and inclusion.

Results

Enhancing HDP expression by different classes of dietary compounds

Fatty acids

Butyrate, propionate, and acetate are major forms of short-chain fatty acids (SCFAs) that are produced by bacterial fermentation of undigested fiber in the intestine [14]. SCFAs and butyrate in particular are capable of upregulating cathelicidin (CAMP) and β-defensin (DEFB) expressions in various cell types including human intestinal epithelia [[15], [16], [17]], lung epithelial cells [18,19], and monocytes [20,21] (Table 1). Besides humans, butyrate induces HDP expression in rodents [[22], [23], [24]], chickens [25,26], cattle [27,28], and pigs [[29], [30], [31], [32], [33]], suggesting the conservation of such an innate defense mechanism. Several butyrate analogs, such as 4-phenylbutyrate (PBA), 4-hydroxybutyrate, isobutyrate, benzylbutyrate, trans-cinnamyl butyrate, and glyceryl tributyrate, had a similar or even superior potency than butyrate in HDP induction in humans [20], rabbits [37], rats [39], and pigs [29,38]. Because of rapid absorption and metabolism of butyrate in the gastrointestinal tract, some of these butyrate analogs may have better potential for in vivo applications.

TABLE 1.

Induction of HDP synthesis by different classes of dietary compounds

Compound Cells or tissues HDPs References
Fatty acids
 Butyrate Human HT-29, SW620 CAMP [15,16]
Human HT-29, Caco-2 DEFB4 [17]
Human A549 DEFB1 [18]
Human EBC-1, THP-1 CAMP [[19], [20], [21]]
C57BL/6 mouse spleen Camp [22]
Mouse Paneth cells Defa1, Defb1, Reg3g [23,24]
Chicken HD11, HTC, primary monocytes, bone marrow cells, crop, jejunum, and cecum explants AvBD3, AvBD4, AvBD5, AvBD8, AvBD9, AvBD10, AvBD14, CATHB1 [25,26]
Bovine MEC DEFB5, DEFB10, TAP, LAP [27]
Bovine MAC-T DEFB5 [28]
Porcine IPEC-J2, 3D4/31, primary monocytes PBD2, PBD3, PG1-5, PEP2C [29]
Porcine ileum and colon, 3D4/2 PBD2, PBD3, PG1-5, PMAP37, PR-39 [30]
Porcine PK-15 PBD3, PBD115, PBD123, PBD128, PEP2C [31]
Porcine IPEC-J2, jejunum, cecum PBD3, PEP2C [32,33]
 Free fatty acids and their derivatives Human HT-29, U937 CAMP [20]
Human SZ95 sebocytes DEFB4 [34]
Porcine IPEC-J2 PBD2, PBD3, PEP2C [29]
Chicken HD11, primary monocytes AvBD9 [35]
 4-Phenylbutyrate Human VA10, U937, A498, HT-29 CAMP [36]
Human VA10 DEFB1 [36]
Rabbit lung and rectum epithelium CAMP [37]
Porcine IPEC-J2 PBD1, PBD3, PEP2C [38]
 4-Hydroxybutyrate Rat bone marrow–derived macrophages Camp, Defb4 [39]
 Isobutyrate, propionate Human SW620 colonocytes CAMP [15]
 Valproic acid Human A549 DEFB1 [18]
 Caprylic acid, nonanoic acid Porcine IPEC-J2 PBD1, PBD2 [40]
 Oleic acid Mouse hair follicle sebaceous glands Defb1 [34]
 Propionate, hexanoate Bovine MEC TAP [41]
Vitamins
 Vitamin D-3 Human keratinocytes, monocytes, neutrophils, neutrophil progenitors, SCC25, Calu-3, U937, HL60, bone marrow cells, HaCat, HT-29, airway epithelial organoids CAMP [[42], [43], [44], [45], [46], [47]]
Human SCC25, Calu-3, primary keratinocytes DEFB4 [42]
Murine intestinal organoids Defa5, Reg3g [48]
Chicken embryonic intestinal epithelial cells and PBMCs Multiple β-defensins [49]
Bovine MEC DEFB1, LAP, S100A7 [50]
Bovine peripheral blood leukocytes DEFB3, DEFB7, DEFB8, DEFB10, LAP, S100A8 [51]
 Calcipotriol Human skin biopsies CAMP [52]
 Retinoic acid Human Bronchial Epithelial Cells DEFB3, DEFB4, CAMP [53]
Human U937 CAMP [54]
Murine intestinal organoids Defa5, Reg3g [48]
Mouse Preadipocytes 3T3-L1 Camp [55]
 Retinol, retinaldehyde Mouse skin DEFB3 [56]
 Nicotinamide Human neutrophils CAMP [57]
Mouse bone marrow cells Camp [57]
 Niacin Piglet ileum, jejunum, colon PBD2, PG1-5, PR39 [58,59]
 Vitamin C Human keratinocytes DEFB1 [60]
Digestive gland of Haliotis discus hannai DEFB [61]
Amino acids
 Isoleucine Human HCT-116 colon cells DEFB1 [62]
Human A549 lung epithelial cells DEFB4 [63]
BALB/c mouse lung DEFB3, DEFB4 [63]
Bovine MDBK kidney cells Bovine β-defensins [64]
Porcine IPEC-J2 PBD1, PBD2, PBD3 [65]
Porcine IPEC-J2, jejunum and ileum PBD1, PBD2, PBD114, PBD129 [66]
Intestine of P. vachelli × L. longirostris DEFB, HAMP [67]
 Leucine Piglet jejunum and ileum, IPEC-J2 PBD1, PBD2, PBD114, PBD129 [66]
Intestine of P. vachelli × L. longirostris DEFB [68]
 Valine Piglet jejunum and ileum, IPEC-J2 PBD1, PBD2, PBD114, PBD129 [66]
 Tryptophan Mouse ileal mucosa Reg3g, Reg3b [69]
Rat jejunal and ileal mucosa Defb2 [70]
Porcine IPEC-J2 PBD1, PBD2, PBD3, PBD115, PG1-5, PR-39 [70]
Piglet jejunal, ileal, colon mucosa PBD2 [71]
 Threonine Porcine IPEC-J2 PBD1, PBD2, PBD3 [72]
Head kidney of Ctenopharyngodon idella DEFB1, HAMP, LEAP-2B [73]
Spleen of Ctenopharyngodon idella DEFB1, HAMP, LEAP-2A, LEAP-2B [73]
 Arginine Human HCT-116 colon cells DEFB1 [62]
Mouse Paneth cells Defa29 [74]
 Glutamine Mouse Paneth cells Reg3γ [74]
Mouse ileum Defa4 [75]
Sugars
 Lactose Human HT-29, Caco-2, T84, THP-1 CAMP [76]
Chicken HD11 macrophages AvBD9, AvBD14 [77]
Piglet jejunal mucosa PBD1, PBD2, PBD3 [78]
 Glucose Human NHK keratinocytes DEFB1, CAMP [60]
Human HEK-293, HCT-116 DEFB1 [79]
Rat kidney DEFB1 [80]
Chicken HD11 macrophages AvBD9, AvBD14 [77]
 Maltose, trehalose Human HT-29 colon epithelia CAMP [76]
 Galactose Chicken HD11 macrophages AvBD9, CATHB1, AvBD14 [77]
 Trehalose Chicken HD11 macrophages AvBD9, CATHB1 [77]
 Maltose, sucrose, fructose Chicken HD11 macrophages AvBD9 [77]
Minerals
 Zinc Human Caco-2 CAMP [81]
Porcine IPEC-J2 PBD1, PBD2, PBD3 [65]
 Zinc gluconate Human skin explants DEFB4, Psoriasin [82]
 Calcium Primary human keratinocytes DEFB3, DEFB4, DEFB104 [83]
Human HaCaT, PHK16-0b DEFB1, DEFB3 [84]
Bile acids
 LCA Human HT-29, keratinocytes CAMP [16,85]
 CDCA, UDCA Human biliary epithelial cells CAMP [86]
 TMCA, THDCA Mouse 3T3-L1 adipocytes Camp [87]
Phytochemicals
 EGCG Human B11 gingival epithelial DEFB1, DEFB4 [88]
Human BEAS-2B DEFB3 [89]
Porcine IPEC-J2 PBD1, PBD2 [90]
Chicken PBMCs AvBD9 [91]
 Quercetin Human HepG2 hepatocytes, rat liver HAMP [92]
Chicken HTC, PBMCs AvBD4-7, AvBD9, AvBD14 [91]
Chicken ileum Multiple β-defensins [93]
Zebrafish liver Defensin, LEAP2 [94]
 Saponarin Human HaCaT keratinocytes CAMP [95]
 Apigenin Human HaCaT keratinocytes CAMP, DEFB1, DEFB3, DEFB4 [96]
 Genistein Human keratinocytes CAMP [97]
 Xanthohumol Porcine 3D4/31 macrophages PBD3, PEP2C, PG1-5 [98]
Porcine jejunal explants PBD3, PG1-5 [98]
 Calycosin Porcine IPEC-J2 PBD2, PBD3, PEP2C [98]
 Datiscetin Chicken HTC, jejunal explants AvBD9 [99]
 Isoloquiritigenin Human Caco-2 colonic epithelial DEFB3 [100]
Porcine IPEC-J2 PBD3, PEP2C, PG1-5 [98]
 Resveratrol Human HepG2 hepatocytes, rat liver HAMP [92]
Human periodontal ligament DEFB4, DEFB3 [101]
Human HaCaT keratinocytes, U937 CAMP [102,103]
Mouse ileum Defa3, Defa5, Defa20 [104]
Rat liver Hamp [92]
Chicken PBMCs AvBD9 [91]
 Polydatin Human HaCat keratinocytes DEFB4 [105]
 Pterostilbene Human U937 monocytes CAMP [103]
Porcine 3D4/31 macrophages PBD3, PG1-5 [98]
 Isorhapontigenin Porcine IPEC-J2 intestinal epithelial PBD3, PEP2C, PG1-5 [98]
Porcine jejunal explants PBD3, PG1-5 [98]
 Ellagic acid Human gingival epithelial DEFB4 [106]
 Caffeic acid Mouse tongue Defb3 [107]
 Anacardic acid Chicken PBMCs AvBD9 [91]
 Curcumin Human U937 monocytes, HT-29 CAMP [108]
Grass carp liver, PBMCs HAMP, LEAP-2, DEFB [109]
 Forskolin Human HT-29, Caco-2, HaCaT, INT407, A549 CAMP [110]
Chicken HD11, HTC macrophages AvBD9 [26]
 Andrographolide Human Caco-2 colonic epithelial DEFB3 [100]
 Oridonin Human Caco-2 colonic epithelial DEFB3 [100]
 Tetrandrine Chicken HTC cells, jejunal explants AvBD9 [99]
 Sanguinarine Chicken HTC cells, jejunal explants AvBD9 [99]
 Deoxyshikonin Porcine IPEC-J2, 3D4/31 cells PBD3, PEP2C, PG1-5 [98]
Porcine jejunal explants PBD3, PG1-5 [98]
Epigenetic modulators
 Trichostatin A Human SW620 colon epithelial CAMP [15]
Human A549 lung epithelial, human NCI-H727 lung epithelial DEFB1 [18]
Human primary gingival epithelial DEFB4 [111]
Human Caco-2 colonic epithelial, primary colonic CAMP, DEFB3, DEFB4 [112]
Human VK2/E6E7 vaginal keratinocytes DEFB1 [113]
Chicken HTC macrophages AvBD9 [99]
Porcine IPEC-J2 intestinal epithelial PBD3 [114]
Primary bovine mammary epithelial DEFB3, DEFB4, DEFB7, DEFB10 [115]
Rat testis, cauda Defb1, Defb2 [116]
 Vorinostat Human THP-1 monocytes DEFA1, DEFA5, DEFA6, DEFB4 [117]
Human Caco-2/TC7 cells DEFB4 [118]
Human Caco-2 and primary colonic epithelial cells CAMP, DEFB3, DEFB4 [112]
Human Huh7 hepatocytes LEAP-1 [119]
Porcine IPEC-J2 intestinal epithelial, porcine 3D4/31 macrophages PBD2, PBD3 [114]
Chicken HTC macrophages AvBD4, AvBD8, AvBD9, AvBD10, AvBD14 [120]
 Entinostat Human HT-29 colonic epithelial CAMP [121,122]
Rabbit ileal and rectal epithelium CAMP [121,123]
Chicken crop and jejunum AvBD9, AvBD10, AvBD14, CATHB1 [124]
 Aroylated phenylenediamines Human HT-29 colonic epithelial CAMP [121]
 ADP HO53 Human BCi and VA10 bronchial epithelial cells CAMP, DEFB1 [125]
 ADP HO56 Human BCi and VA10 bronchial epithelial cells CAMP, DEFB1 [125]
 Mocetinostat Porcine IPEC-J2 intestinal epithelial PBD3 [114]
Chicken HTC macrophages Multiple β-defensins [126]
Chicken jejunal explants AvBD9 [126]
Chicken HTC and HD11 macrophages AvBD9 [120]
 RGFP966 Human Huh7 hepatocytes LEAP-1 [119]
 Panobinostat Porcine IPEC-J2 intestinal epithelial PBD2, PBD3, PG1-5 [114]
 LAQ824 Porcine IPEC-J2 intestinal epithelial PBD2, PBD3, PG1-5 [114]
 SB939 Porcine IPEC-J2 intestinal epithelial PBD2, PBD3, PG1-5 [114]
 Apicidin Human A549 lung epithelial DEFB1 [18]
Chicken HTC macrophages AvBD9 [99]
Porcine IPEC-J2 intestinal epithelial PBD2, PBD3 [114]
 Depudecin Human A549 lung epithelial DEFB1 [18]
Chicken HTC macrophages AvBD9 [99]
Porcine IPEC-J2 intestinal epithelial PBD3 [114]
 SGI-1027 Chicken HTC macrophages AvBD9 [120]
 BIX01294 Chicken HTC macrophages AvBD9 [120]
 UNC1999 Chicken HTC macrophages AvBD9 [120]
 5-Azacytidine Human gingival epithelial cells DEFB4 [111]
Human OSC-19, BSC-OF, SAS, HSC-2, HSC-4, HSY oral squamous DEFB4 [127]
Human HSC-3 and HSC-4 oral squamous, HaCaT keratinocytes, TR146 buccal epithelial LL-37 [128]
Human VK2/E6E7 keratinocytes DEFB1 [113]
Human HC-OA chondrocytes CAMP [129]
Bovine mammary epithelial DEFB3, DEFB5, DEFB10, EBD, LAP, TAP [115]
A. pernyi larvae Attacin, cercropin, and lebocin [130]
Rat cauda Defb1, Defb2, Defb27 [116]
Rat testis Defb1, Defb2, Defb30 [116]
Rat caput Defb30, Defb36 [116]
Probiotics
 Lactobacillus gasseri Human VK2/E6E7 keratinocytes DEFB1 [113]
 Lactobacillus fermentum K11-Lb3 and K2-Lb6 Human Caco-2 colonic epithelial DEFB4 [131]
 Ligiactobacillus salivarius SMXD51 Human Caco-2/TC7 cells DEFB4 [132]
 Lacticaseibacillus paracasei CBA L74 Human Caco-2 colonic epithelial CAMP, DEFB4 [133]
 Lactobacillus helveticus SBT2171 Human Caco-2 colonic epithelial, HSC-4 tongue epithelial DEFB4 [134]
Human Ca9-22 gingival cells DEFB3, DEFB4 [135]
Mouse gingival tissue Defb4, Defb14 [135]
 Lactobacillus rhamnosus GG Human SW480 colonic epithelial DEFB4 [136]
 Bifidobacterium longum Human SW480 colonic epithelial DEFB4 [136]
 L. paracasei SD1, L. rhamnosus SD4, L. fermentum SD7, L. rhamnosus SD11, L. rhamnosus GG Primary human gingival epithelial cells DEFB4, DEFB104 [137]
 L. rhamnosus Lcr35 Human VK2/E6E7 vaginal epithelia DEFB4 [138]
 Lactobacillus crispatus Human HeLa cells DEFB3, DEFB4 [139]
 Lactobacillus johnsonii NCC 533 Human epidermis CAMP, DEFB1, DEFB4, DEFB103B [140]
 Lactobacillus delbrueckii subsp. bulgaricus 8481 Human serum DEFB4 [141]
 BB12 Human infant stool samples CAMP, DEFB4 [142]
 Escherichia coli Nissle 1917, Pediococcus pentosaceus, Lactobacillus acidophilus, L. fermentum Human Caco-2 colonic epithelial DEFB4 [143]
 Lactobacillus reuteri 5454, B. animalis ssp. Mouse ileum Reg3b, Reg3g [144]
 L. paracasei subsp. paracasei CNCM I-1518 Rat ileum Defb1 [145]
 Lactobacillus casei CRF28, UW1 C57BL/6 mouse ileum Defa-rs1 [146]
 L. casei 32G, CRF28, BL23, M36 C57BL/6 mouse ileum Reg3β [146]
 Lactobacillus plantarum ZLP001 Piglet jejunum, ileum, IPEC-J2, 3D4/31 PBD2, PBD3, PBD114, PBD129, PG 1–5, PEP2C [147,148]
 Lactobacillus amylovorus SLZX20-1 Porcine IPEC-J2 PBD1, PEP2C, PG1-5 [149]
 Bacillus subtilis CP9 Porcine IPEC-J2 PG1 [150]
 B. subtilis and L. salivarius Piglet duodenum PBD2 [151]
 L. rhamnosus GG Piglet jejunum PBD1, PMAP-37 [152]
 L. reuteri
Clostridium butyricum
Chicken cecum AvBD4
AvBD1, CATH3
[153]
 L. casei BL23 Bovine MEC TAP [154]
 Saccharomyces cerevisiae CNCM I-1077 Bovine rumen and colon epithelium DEFB1 [155]
Prebiotics
 Inulin Mouse Paneth cells Defb1, Defa1, Defa4, Defa29, Defa5, Reg3g [24]
 Long-chain inulin-type fructans Mouse colon Defb1, Camp [156]
 Dendrobium huoshanense polysaccharides Mouse jejunum and ileum Total β-defensin protein [157]

Abbreviations: AvBD, avian β-defensin; CAMP, cathelicidin; CDCA, chenodeoxycholic acid; DEFA, α-defensin; DEFB,β-defensin; EGCG, epigallocatechin gallate; HAMP, hepcidin antimicrobial peptide; HDP, host defense peptide; LAP, lingual antimicrobial peptide; LCA, lithocholic acid; LEAP, liver-enriched antimicrobial peptide; PBD, porcine β-defensin; Reg3, regenerating islet-derived protein 3; TAP, tracheal antimicrobial peptide; TDCA, taurodeoxycholic acid; THDCA, taurohyodeoxycholic acid; TMCA, α-tauromuricholic acid; UDCA, ursodeoxycholic acid.

Consistent with their HDP-inducing activity in cells, oral supplementation of sodium butyrate or PBA improved the clinical outcomes of shigellosis in both humans and rabbits by counteracting Shigella-induced downregulation of CAMP expression in the intestinal tract [37,158]. In chickens, dietary supplementation of butyrate significantly reduced Salmonella sp. colonization in the cecum of experimentally infected chickens [25]. Additionally, oral supplementation with sodium butyrate reduced the Corynebacterium pseudotuberculosis load, enhanced Camp expression, and alleviated lesions in the spleens of infected mice [22]. Similarly, butyrate, caprylic acid, and nonanoic acid reduced the bacterial load, mitigated intestinal inflammation, and upregulated HDP expressions in piglets challenged with Escherichia coli 0157:H7 [30,40]. A recent study also showed the ability of butyrate to counteract deoxynivalenol-mediated suppression of intestinal HDPs in weaned piglets [33].

Comparison of the HDP-inducing activity among free saturated fatty acids of 1–18 hydrocarbons in humans, pigs, and chickens revealed that SCFAs are the most potent, whereas fatty acids with longer aliphatic chains quickly lose HDP-inducing potency [20,29,39]. However, relative HDP-inducing potency of individual fatty acids varies among animal species. Butyrate appears to be the most efficacious HDP inducer in chicken and porcine epithelial and monocytic cells [29,35], whereas valerate, hexanoate, and heptanoate are capable of triggering stronger HDP expression than butyrate when used at higher concentrations in humans [20]. It will be interesting to evaluate the in vivo efficacy of valerate, hexanoate, and heptanoate and their analogs in HDP induction and bacterial clearance in humans.

It is abundantly clear that different HDPs are regulated differently by fatty acids. For example, PBA stimulated human CAMP and DEFB1 expression but not DEFB3, DEFB4, and DEFB104 in human lung epithelial cells [36]. Lauric acid, palmitic acid, and oleic acid upregulated DEFB4 but not CAMP or DEFB1 in human sebocytes [34]. In mice, oleic acid preferentially promoted the induction of mouse Defb2 in the hair follicle sebaceous glands of mouse ear skin [34]. Propionate and hexanoate potentiated bovine tracheal antimicrobial peptide expression but not DEFB5 [41].

A growing body of evidence has suggested that SCFAs and butyrate in particular induce HDP expression primarily by acting as histone deacetylase (HDAC) inhibitors [159]. HDAC1, HDAC2, and HDAC3 were downregulated by butyrate in human THP-1 monocytes [21], associated with an increase in histone acetylation along the promoters of human, bovine, and porcine HDPs, which is associated with active gene transcription [19,28,30]. Free fatty acid receptors, such as GPR41/FFAR3, GPR43/FFAR2, and GPR109a/HCAR2, are also involved in SCFA-mediated HDP induction, leading to downstream activation of the mammalian target of rapamycin and signal transducers and activator of transcription (STAT) 3 [23,160] (Figure 2).

FIGURE 2.

FIGURE 2

Molecular mechanisms of HDP induction by butyrate, vitamin D-3, bile acids, and lactose. An arrowhead indicates stimulation of a pathway, while a T-bar indicates inhibition of a pathway. See text for details. AC, acetyl group; AP1, activator protein-1; BRG1, Brahma-related gene 1; CDCA, chenodeoxycholic acid; C/EBPα, CCAAT enhancer binding protein α; CRE, cAMP-response element; CREB, cAMP-response element-binding protein; FXR, farnesoid X receptor; GPR, G protein-coupled receptors; HDAC, histone deacetylase; HDP, host defense peptide; LCA, lithocholic acid; MAPK, mitogen-activated protein kinases; mTOR, mammalian target of rapamycin; p50/p65, NF-κB proteins p50 and p65 heterodimer; PU.1, an ETS-family transcription factor; RXR, retinoid X receptor; SRC, steroid receptor coactivator; STAT, signal transducer and activator of transcription; TLR, toll-like receptor; UDCA, ursodeoxycholic acid; VDR; vitamin D receptor.

Additionally, Toll-like receptor (TLR) 2–mediated NF-κB activation is required for butyrate-induced expression of porcine β-defensins (PBDs) in porcine kidney and intestinal cells, while HDAC inhibition is insufficient [31,32,38]. NF-κB signaling is also required for butyrate-induced DEFB2 production by human colonic epithelial cells [118]. In fact, TLR2 expression is enhanced in bovine epithelial cells treated with butyrate, while concurrent p38 phosphorylation implicates the involvement of the mitogen-activated protein kinase (MAPK) pathway [27,28]. Canonical MAPK family members including MEK, extracellular signal-regulated kinase (ERK), and JNK regulate butyrate-induced CAMP expression in human colon, gastric, and bronchial cell lines but not in heptocarcinoma cell lines [15,36]. Furthermore, butyrate-enhanced CAMP expression in human colonic epithelial cells is mediated by PU.1, a transcription factor activated by MAPK [16]. Additional transcription factors, such as activator protein (AP) 1 and cAMP (cyclic adenosine monophosphate)-response element-binding protein (CREB), are key regulators in butyrate-induced human CAMP and DEFB4 expression [19,110,161].

Vitamins

Vitamin D-3 is a potent HDP inducer in several human cell types including epithelial cells, keratinocytes, monocytes, and neutrophils, leading to an increase in the antimicrobial activity of the host cells [[42], [43], [44], [45]]. For example, the growth of Mycobacterium tuberculosis was significantly suppressed in macrophages exposed to 1,25-dihydroxyvitamin D-3, also known as calcitriol [46,162] (Table 1 [[15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154], [155], [156], [157]]). The positive influence of treatment with vitamin D-3 or its analogs on HDP production was clearly demonstrated in vivo as well. Topical treatment with vitamin D-3 resulted in the upregulation of human CAMP in acute skin injuries [52]. Similarly, psoriatic skin biopsies treated with vitamin D-3 had elevated concentrations of LL-37 and reduced proinflammatory cytokines IL-12/23 p40, IL-1α, IL-1β, and TNF-α [163].

Conversely, a deficiency in vitamin D-3 is often associated with an increased susceptibility to infections, likely due to decreased synthesis of HDPs [164]. For instance, patients who experience severe congenital neutropenia, because of a lack of vitamin D-3, experience recurrent infections, whereas patients given vitamin D-3 show restored LL-37 concentrations and reduced infections [47]. In patients with atopic dermatitis, downregulation of serum 25-hydroxyvitamin D-3 and LL-37 concentrations was observed, with a reciprocal increase in the expression of proinflammatory cytokines [165]. In patients with cystic fibrosis, serum vitamin D concentrations are often deficient, perhaps contributing to the frequency of lung infections [166]. Although serum vitamin D concentrations are positively correlated with tuberculosis disease risk [167], a recent clinical trial with 8851 children with latent tuberculosis infection found no significant improvement in disease risk when a weekly dose of 14,000 IU of vitamin D-3 was given for 3 y [168].

In addition to humans, vitamin D-3 enhances the expression of HDPs in pigs [169], chickens [49], and cattle [50,51]. Administration of vitamin D-3 to cattle reduced internalization of Staphylococcus aureus in bovine mammary epithelial cells by inducing the expression of lingual antimicrobial peptide, DEFB10, and psoriasin [50]. Similarly, an increase in expression of HDPs after vitamin D-3 supplementation was accompanied by enhanced clearance of Mycobacterium bovis Bacillus Calmette-Guérin (BCG) vaccine in bovine peripheral blood leukocytes [51]. It is noted that there is clearly a species-specific difference in the HDP-inducing ability of vitamin D-3. Although it is a strong inducer of cathelicidins and several DEFBs in humans and cattle, vitamin D-3 fails to do so in mice, dogs, and sheep, which is likely due to an absence of the vitamin D response (VDR) elements in the promoter of HDP genes in those species [43,170]. However, intestinal α-defensins (DEFAs) appear to be regulated by vitamin D-3 in mice [48].

Vitamin D-3 induces CAMP and DEFB4 gene expression by binding to VDR to activate respective gene promoters containing VDR elements in different human cell types [42,43] (Figure 2). More recently, transcription factors PU.1 and C/EBPα were found to participate in vitamin D-3 regulation of CAMP and DEFB4 gene expression [171]. Binding of PU.1 and C/EBPα recruits Brahma-related gene 1, a component of the SWI/SNF chromatin remodeling complex, to the CAMP gene promoter, leading to an increase in H4 acetylation [171]. Additionally, steroid receptor activator 3, which has intrinsic histone acetyltransferase activity, is critical for vitamin D-3–mediated induction of CAMP, as steroid receptor activator 3 knockdown prevented the upregulation of the CAMP gene [45].

Retinoic acid, a metabolite of vitamin A, has conflicting effects on HDP induction. Retinoic acid induces CAMP, DEFB3, and DEFB4 gene expression in human bronchial epithelial cells [53] but inhibits DEFB3, DEFB4, and DEFB104 in primary keratinocytes in response to proinflammatory cytokines, bacteria, phorbol myristate acetate, or calcium [83]. Although retinoic acid induces CAMP in human U937 monocytes, no change is seen in K562 lymphoblasts [54]. In mice, retinoic acid upregulated Defa1, Defa5, Defa21, and regenerating islet-derived protein 3-γ (Reg3g) in intestinal organoids [48], Camp in primary preadipocytes [55], along with Reg3b and Reg3g in lymphocytes [172]. Retinol and retinaldehyde significantly increased Defb3 expression in mouse skin, suggesting that topical retinols may be a potential treatment of cutaneous infections via increased expression of HDPs [56]. The expression of porcine cathelicidin PR-39 was enhanced in bone marrow cells by retinoic acid [173]. However, retinoic acid failed to induce HDP expression in ovine respiratory epithelial cells [174].

Niacin, also known as vitamin B-3, was found to elevate HDP gene expression in porcine IPEC-J2 cells as well as throughout the intestinal tract of piglets [58,59]. Notably, supplementation of niacin alleviated the disease severity of porcine deltacoronavirus [58] as well as enterotoxigenic E. coli K88 in weaned piglets [59]. Niacin appeared to induce porcine HDP gene expression through epigenetic modifications. Improved HDP mRNA concentrations coincided with increased expression concentrations of sirtuin (SIRT) 1 and reduced HDAC7 in the intestine of piglets in response to niacin supplementation [59]. Niacin enhanced phosphorylation of histone H3 at Ser10 (H3S10p) in the ileum as well as acetylation of lysine 9 on histone 3 (H3K9ac) and H3K27ac in the colon of piglets challenged with ETEC K88 [59].

Nicotinamide, an amide form of niacin, improved the expression of CAMP in human neutrophils but not in monocytes [57]. Increased Camp protein concentrations were also detected in bone marrow mononuclear cells of mice supplemented with nicotinamide, while the treatment enhanced S. aureus killing by ≤1000-fold in a systemic murine infection model [57]. Vitamin C, also known as ascorbic acid, increased DEFB1 but not CAMP gene expression in human keratinocytes, [60]. Vitamin C was similarly found to increase DEFB mRNA concentrations in the digestive gland of abalone [61].

Amino acids

Essential, branched-chain amino acids, including isoleucine, leucine, and valine, are each capable of upregulating HDP expression. Isoleucine increased DEFBs across multiple animal species, including humans [62,63], mice [63], cattle [64], pigs [65,175], and catfish [67] (Table 1 [[15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154], [155], [156], [157]]). For example, isoleucine supplementation increased Defb3 and Defb4 expression, which led to a decrease in intestinal bacillary loads and tissue damage after mice were inoculated with a multidrug resistant M. tuberculosis H37Rv [63]. Leucine increased Defa1 in mouse Paneth cells [23], 4 DEFBs in the jejunum and ileum of piglets [66], as well as DEFB transcription in the intestine of hybrid catfish [68]. Valine supplementation improved intestinal DEFB mRNA concentrations in piglets [66] and upregulated DEFB1 and CATH7 in cultured mammary epithelial cells of goats [176].

Tryptophan and threonine are essential amino acids to mammals that have also been shown to upregulate HDP production. Tryptophan improved both mRNA and protein concentrations of Defb2 in the jejunal and ileal mucosa of rats [177], as well as Reg3g and Reg3b mRNA concentrations in mouse ileal mucosa [69]. Tryptophan also increased the expression of multiple HDP genes in porcine IPEC-J2 cells [70]. Furthermore, diets with adequate tryptophan (0.21%, 0.28%, or 0.35%) elevated PBD2 protein concentration across the small intestine of piglets compared with a 0.14% tryptophan diet [71]. L-threonine similarly induced the transcription of DEFBs in porcine IPEC-J2 cells [72]. Supplementation with threonine compared with a threonine-deficient diet enhanced mRNA concentrations of hepcidin antimicrobial peptide (HAMP), liver-enriched antimicrobial peptide (LEAP) 2, and DEFB1 in the head kidney and spleen of grass carp [73].

Furthermore, arginine upregulated DEFB1 in human HCT-116 colonic epithelial cells [62,74]. Dietary arginine enhanced Defa29 and Reg3g expression in mouse Paneth cells and decreased E. coli colonization in the jejunum of challenged mice [74]. In porcine IPEC-J2 cells, L-arginine triggered DEFB transcription and could ameliorate LPS-induced inflammation [178]. Glutamine is a non-essential amino acid capable of HDP induction in mice. Defa4 mRNA was increased in the small intestine of mice supplemented with glutamine [75]. Similarly, glutamine augmented Defa29 and Reg3g mRNA expression and decreased E. coli colonization in mice [74].

Isoleucine, tryptophan, and threonine involve SIRT1 to initiate a signaling cascade that enhances HDP gene expression; however, whether SIRT1 is activated or inhibited varies among individual amino acids. In porcine intestinal epithelial cells and the intestine of hybrid catfish, isoleucine activated a SIRT1/ERK/90RSK signaling pathway to upregulate several HDP genes [66,67]. However, threonine suppressed SIRT1 expression in porcine intestinal epithelial cells, which enhanced acetylation of p65, promoted translocation of p65 into the nucleus, and activated NF-κB [72]. Tryptophan, on the contrary, had no direct effect on SIRT1 expression but suppressed LPS-induced SIRT1 expression in mouse ileal mucosa and porcine intestinal epithelial cells [69]. The mammalian target of rapamycin pathway is involved in induction of DEFBs by tryptophan [177] and arginine [178].

Sugars

Lactose, a disaccharide sugar, was shown to induce CAMP transcription in a dose-dependent and time-dependent manner in human HT-29 and T84 colonic epithelial cells and THP-1 monocytes/macrophages [76] (Table 1 [[15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154], [155], [156], [157]]). In fact, several other monosaccharide and disaccharide sugars such as glucose, galactose, trehalose, and maltose also showed varied potency in inducing CAMP gene expression [76]. Glucose supplementation upregulated DEFB1 and CAMP gene expression in human keratinocytes, leading to greater antimicrobial activity against both Listeria monocytogenes and S. aureus [60]. Interestingly, high glucose induced DEFB1 expression in human kidney and colon cells [79]. Similarly, Defb1 was also upregulated in the kidneys of hyperglycemic rats [80]. However, the expression of human CAMP, DEFB3, and DEFB4 were downregulated by glucose [179,180]. Multiple sugars, such as lactose, glucose, galactose, trehalose, maltose, sucrose, and fructose, increased HDP gene expression in chicken cells [77]. In pigs, lactose induced several DEFB genes in the jejunal mucosa and mitigated the negative effect of Rotavirus on animal growth [78]. Lactose-mediated HDP induction involves the MAPK pathway and histone acetylation. In HT-29 colonic epithelial cells, an inhibition of p38 MAPK and JNK reduced CAMP induction by lactose [76], and histone H4 acetylation was increased in chicken HD11 macrophage cells in response to lactose [77]. Additional studies are needed to clarify the mechanisms by which sugars upregulate HDP.

Minerals

Zinc exerts a plethora of benefits for mounting an effective immune response to infectious agents [181]. Among them is zinc’s ability to induce HDP synthesis. Zinc was found to induce CAMP in human Caco-2 colonic epithelial cells [81] and DEFB4 in LPS-induced inflammatory skin explants [82] (Table 1 [[15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154], [155], [156], [157]]). Zinc also upregulated DEFB concentrations in porcine intestinal epithelial cells [65]. On the contrary, zinc deficiency was accompanied by a decrease in Paneth cell DEFA synthesis in both humans [182] and mice [183]. Calcium is capable of inducing multiple DEFB gene transcription in human keratinocytes [83,84], while DEFB4 expression was reduced when calcium was chelated [184]. Chelation of calcium prevented cathelicidin-mediated killing of human Jurkat T leukemia cells [185] and inhibited DEFB-mediated fungicidal activity against Candida albicans [186].

Bile acids

Several bile acids have been reported to upregulate HDP gene expression. A primary bile acid, chenodeoxycholic acid, and a secondary bile acid, ursodeoxycholic acid, induced CAMP transcription in human biliary epithelial cells [86] (Table 1 [[15], [16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85], [86], [87], [88], [89], [90], [91], [92], [93], [94], [95], [96], [97], [98], [99], [100], [101], [102], [103], [104], [105], [106], [107], [108], [109], [110], [111], [112], [113], [114], [115], [116], [117], [118], [119], [120], [121], [122], [123], [124], [125], [126], [127], [128], [129], [130], [131], [132], [133], [134], [135], [136], [137], [138], [139], [140], [141], [142], [143], [144], [145], [146], [147], [148], [149], [150], [151], [152], [153], [154], [155], [156], [157]]). Another secondary bile acid, lithocholic acid (LCA), upregulated CAMP in human HT-29 colonic epithelial cells [16] and primary keratinocytes [85] but not in human Caco-2 colonic epithelial cells [85]. In mouse 3T3-L1 adipocytes, α-tauromuricholic acid and taurohyodeoxycholic acid, increased Camp mRNA expression, but cholic acid, deoxycholic acid, and taurodeoxycholic acid had no effect [87]. Two nuclear receptors, farnesoid X receptor and VDR, are involved in the upregulation of CAMP by bile acids (Figure 2). Although chenodeoxycholic acid induced CAMP through farnesoid X receptor, ursodeoxycholic acid activated VDR [187]. LCA likewise signaled through VDR to improve CAMP gene expression [16,85]. Moreover, MEK-ERK signaling was involved in CAMP gene regulation by LCA in primary human keratinocytes [85], while VDR recruited PU.1 to the CAMP gene promoter in HT-29 colon epithelial cells [16].

Phytochemicals

Polyphenols are a diverse group of naturally occurring compounds found in plants with anti-inflammatory, antioxidant, and anticancer properties [188]. Several classes of polyphenols, such as flavonoids, stilbenes, and phenolic acids, are capable of inducing HDP synthesis [189] (Table 1). For example, epigallocatechin gallate (EGCG), a flavonoid found in green tea, enhanced DEFB synthesis in humans [88,89], pigs [90], and chickens [91]. Quercetin is another flavonoid with an ability to improve HDP expression in rats [92], chickens [91,93], and zebrafish [94]. Saponarin (a flavone), genistein (an isoflavone), and apigenin (a trihydroxyflavone) enhanced CAMP transcription in human keratinocytes [[95], [96], [97]]. Additionally, xanthohumol (a prenylated flavonoid), calycosin (an isoflavone), datiscetin (a tetrahydroxyflavone), and isoliquiritigenin (a chalcone flavonoid) have been found to induce multiple DEFB genes in pigs [98], chickens [99], and humans [95,100], respectively. Resveratrol, a stilbene in grapes, stimulated HDP gene expression in humans, mice, rats, and chickens [91,92,[101], [102], [103], [104]]. Similarly, polydatin and pterostilbene, 2 resveratrol derivatives, were shown to increase HDP production in human keratinocytes [105] and monocytes [103] as well as in porcine macrophages [98]. Isorhapontigenin, a stilbenoid, augmented the expression concentrations of several DEFBs in porcine intestinal epithelial cells [98]. Phenolic acids, such as ellagic acid, caffeic acid, and anacardic acid, increased the DEFB expression in humans [106], mice [107], and chickens [91]. Curcumin, belonging to the curcuminoid group of polyphenols, stimulated CAMP expression in human monocytes, colonic epithelial cells, and keratinocytes [108] and enhanced HAMP, LEAP2, and DEFB mRNA concentrations in grass carp [109].

A few nonpolyphenol phytochemicals have also been found to upregulate HDP synthesis. For instance, forskolin (FSK), a diterpenoid produced by the Indian Coleus plant, was shown to enhance the expression of CAMP in human HT-29 epithelial cells [110], but suppress CAMP and DEFB1 concentrations in butyrate-differentiated HT-29 cells [161]. Chicken macrophage cells treated with FSK increased avian β-defensin 9 (AvBD9) transcription in chicken macrophage cells as well as in the crop of orally supplemented chickens [26]. Two diterpenoids, andrographolide and oridonin, were found to induce DEFB3 expression in human colonic epithelial cells [100]. The supernatant of cells treated with andrographolide and isoliquiritigenin limited the growth of 4 pathogenic bacteria [100]. A number of natural products and phytochemicals in particular were recently identified with the HDP-inducing activity in chicken and porcine cells through high throughput screening [98,99]. For instance, 1 plant alkaloids, tetrandrine and sanguinarine, were each capable of inducing chicken HDP gene expression [99], while deoxyshikonin, a plant naphthoquinone, increased PBD expression [98].

Many phytochemicals upregulate HDP synthesis through MAPK signaling pathways. For example, EGCG induced DEFB1, DEFB3, and DEFB4 via MAPK p38/ERK/JNK pathways in human bronchial or gingival epithelial cells [88,89] and required p38 activation for PBD2 induction in porcine intestinal epithelium [90]. Andrographolide, oridonin, and isoliquiritigenin activated MAPK pathways downstream of the epidermal growth factor (EGF) receptor and recruited c-Fos, c-Jun, and Elk1 or cMyc to the DEFB3 promoter in human colon epithelial cells [100]. Although andrographolide upregulated DEFB3 by EGFR/ERK/JNK, oridonin and isoliquiritigenin instead worked through an EGFR/ERK/p38 signaling pathway [100]. Sulforaphane required VDR and ERK1/2 but not p38 to increase DEFB4 transcription in human intestinal epithelium [17]. In chicken macrophage cells, inhibitors of p38 or JNK nearly abolished FSK-induced AvBD9 [26]. MAP kinases are required for the upregulation of HDP by many phytochemicals, albeit the specific signaling cascade varies by treatment.

A NF-κB pathway was activated by resveratrol, genistein, or sulforaphane for HDP upregulation. Resveratrol-induced CAMP in human keratinocytes required the spingosine-1-phosphate (S1P) signaling, followed by transactivation of NF-κB and transcription factor C/EPBα [102]. A similar ER-β→S1P→NF-κB →C/EPBα mechanism was found for CAMP induction in keratinocytes treated with genistein [97]. Sulforaphane incubated with a NF-κB inhibitor reduced DEFB4 transcription in human intestinal epithelium [17]. NF-κB was also required for quercetin-induced AvBD9 induction in chicken monocytes [91]. However, dietary quercetin supplementation downregulated NF-κB mRNA expression in the chicken ileum although multiple AvBD genes were enhanced [93]. The involvement of NF-κB in HDP regulation by phytochemicals should be confirmed in vivo for additional species.

Interestingly, resveratrol, EGCG, quercetin, anacardic acid, and garcinol are natural cyclooxygenase (COX)-2 inhibitors that are able to upregulate HDPs in chicken monocytes [91]. Soponarin and apigenin were also found to inhibit COX-2 in human keratinocytes stimulated with inflammatory cytokines [95], mouse macrophages, and rat basophils [96]. Although inhibition of COX-2 signaling has previously been linked to HDP expression [190], its involvement in HDP regulation after treatment with these polyphenols remains to be studied.

Nuclear factor erythroid 2-related factor 2 (Nrf2) facilitates HDP expression in the liver in response to phytochemical treatment. Quercetin treatment increased binding of Nrf2 to an antioxidant response element at the HAMP promoter in parallel to enhanced HAMP transcription in human hepatocytes [92]. Curcumin likewise upregulated Nrf2 mRNA concentrations while increasing HAMP, LEAP2, and DEFB in grass carp liver [109].

FSK elevates intracellular cAMP concentrations, and binding of CREB to the human cathelicidin promoter promoted active gene transcription [110]. However, cAMP inhibited by 2',3'-dideoxyadenosine improved quercetin-induced AvBD9 in chicken monocytes [91]. The role of cAMP in HDP regulation needs to be clarified for different species and HDP genes.

Epigenetic modulators

Epigenetic modifications, such as acetylation of histones and methylation of histones and DNA, play an essential role in regulating chromatin accessibility and gene expression [191]. Histone acetylation is facilitated by histone acetyltransferases to favor a more relaxed chromatin structure, whereas the removal of acetyl groups by HDACs favors a more condensed chromatin configuration [191]. Histones and DNA can also be methylated by DNA methyltransferases (DNMT) and histone methyltransferases (HMT), respectively, to affect gene transcription [191]. HDAC inhibitors have been demonstrated to be among the most potent inducers of HDPs in several high throughput screenings [99,114,121,124,126,192]. Members of all 4 major classes of HDAC inhibitors, including benzamides, hydroxamates, cyclic peptides, and SCFAs, have been shown to be HDP inducers. For example, trichostatin A, a hydroxamate HDCA inhibitor, induced mRNA expression of HDPs in humans [15,18], pigs [114], chickens [99], and cattle [115] (Table 1). Other hydroxamte HDAC inhibitors, such as vorinostat also known as suberoylanilide hydroxamic acid, induced the mRNA expression of DEFA1, DEFA5, DEFA6, and DEFB4 in human monocytic cells infected with Leishmania donovani, contributing to increased antiparasitic activity [117]. Vorinostat also enhanced the expression of several HDP genes in human intestinal epithelium [112,118] and hepatocytes [119], chicken macrophages, [120,126] and porcine intestinal epithelial cells and lung alveolar macrophages [114].

However, among all HDAC inhibitors, benzamides appear to be the most potent HDP inducers in humans, pigs, and chickens as revealed in several recent high throughput assays [114,121,124,126,192]. For example, entinostat, a benzamide HDAC inhibitor, potently enhanced DEFB1 and CAMP mRNA expression in intestinal epithelial cells [121,122] and protected rabbits from experimental cholera [123]. Its analogous compounds, known as aroylated phenylenediamines, had similar effects in human bronchial epithelial cells, associated with a significant reduction in the intracellular invasion of Pseudomonas aeruginosa [125]. Entinostat also similarly induced HDP gene expression in chicken jejunal explants, and an oral inoculation of entinostat led to an increased expression of multiple HDP genes in the crop and jejunum of broiler chicks [124]. Mocetinostat, an entinostat analog, potently increased AvBD9 mRNA concentrations in chicken jejunal explants [126]. Polyphenols, such as sulforaphane, curcumin, and EGCG, are known natural HDAC inhibitors [193] with an HDP-inducing activity [17,89,108].

In addition to HDAC inhibitors, inhibitors of DNMT and HMT are HDP inducers. For instance, 5-azacytidine, a well-known DNMT inhibitor, increased CAMP and DEFB concentrations in gingival epithelial cells [111], oral carcinoma cells [127,128], keratinocytes [113], and chondrocytes [129]. Additionally, 5-azacytidine stimulated HDP expression in bovine mammary epithelial cells [115], chicken macrophage cells [120], the larvae of silkworm [130], as well as in the caput, cauda, and testis of the male rats [116]. Polyphenols such as EGCG, quercetin, and genistein, known for their DNMT inhibitory properties [194], also possess the ability to induce HDP synthesis as discussed in greater detail in an earlier section. Similarly, inhibitors of HMT, such as BIX01294 and UNC1999, have been shown to promote the expression of multiple HDPs in chicken macrophages [120].

Inhibition of HDACs, DNMTs, and HMTs may facilitate chromatin relaxation and promote gene transcription. Several HDAC inhibitors have demonstrated the capacity to promote histone acetylation at HDP promoter regions, accompanied by increased HDP transcription [18,112,117,195]. HDAC inhibition may also activate transcription factors such as NF-κB to turn on the transcription of HDPs such as DEFB4 owing to the presence of multiple NF-κB binding sites in the promoter region [118] (Figure 3). Consistently, trichostatin A increased activation and translocation of NF-κB through acetylation of p65 and phosphorylation of the IKK complex, ultimately leading to an increased binding of NF-κB to the DEFB4 promoter in human colonic epithelial cells [112]. Entinostat and its structural analog, HO53, promoted STAT3 activation leading to increased HIF1α expression and binding of HIF-1α to the CAMP promoter [122,125]. Similarly, RGFP966, an HDAC3 inhibitor, increased acetylation of STAT3, C/EBPα, and HIF-1α in human liver cells, enhancing the affinity of these transcription factors to the LEAP1 promoter [119].

FIGURE 3.

FIGURE 3

Epigenetic mechanisms of HDP gene induction. See text for details. Ac, acetyl group; C/EBPα, CCAAT enhancer binding protein α; DNMTi, DNA methyltransferase inhibitor; HDACi, histone deacetylase inhibitor; HIF1α, hypoxia-inducible factor-1α; HMTi, histone methyltransferase inhibitor; IKK, Iκ B kinase; P, phosphate group; Me, methyl group; TSA, trichostatin A; p50/p65, NF-κB proteins p50 and p65 heterodimer; STAT, signal transducer and activator of transcription.

DNMT inhibitors, such as AZA, reduced DNA methylation at DEFB1 [113] and CAMP [128,129] promoter regions, resulting in an increase in their transcription. Although the mechanism to support HMT inhibitors, BIX01294 and UNC1999, has not been well studied, a strong correlation has been found between histone demethylase, JMJD3, and expression of DEFB3, S100A7, S100A8, and CAMP in human keratinocytes [196]. Moreover, JMJD3 knockdown led to significantly increased histone methylation concentrations and reduced mRNA expression of these HDPs, highlighting the importance of histone methylation in shaping HDP gene expression patterns [196].

Probiotics

Probiotics are beneficial microbes that promote intestinal health through a myriad of mechanisms [197]. A variety of lactic acid bacteria have been found to induce HDP expression. For example, several Lactobacillus species induced HDP in human intestinal epithelia [[131], [132], [133], [134],136], gingival epithelia [135,137], vaginal epithelia [113,138], cervical epithelia [139], and epidermal cells [140] (Table 1). Oral supplementation of Lactobacillus delbruckii subsp. bulgaricus 8481 decreased IL-8 and increased DEFB4 in elderly patients [141]. In infants, Bifidobacterium animalis subsp. lactis BB-12 elevated both CAMP and DEFB4 protein concentrations [142]. E. coli Nissle 1917 were found to enhance DEFB4 expression in human intestinal epithelial cells [143,198].

Probiotics also induced HDP expression in rodents, pigs, chickens, cattle, and sheep. For instance, lactic acid bacteria were found to enhance HDP production and alleviate colitis [144], vancomycin-resistant enterococcus infection [199], and cirrhosis in mice [145]. In pigs, Lactobacillus reuteri I5007, Lactobacillus amylovorus SLZX20, and Lactobacillus plantarum ZLP001 increased the mRNA expression of multiple DEFBs in porcine intestinal epithelial cell lines [147,149,200], while L. plantarum counteracted intestinal barrier dysfunction induced by enterotoxigenic E. coli [148]. Treatment of intestinal cells with Bacillus subtilis CP9 enhanced PG1 transcription, but not PBD3, while having anti-ETEC properties [150]. Co-administration of Lactobacillus salivarius B1 and B. subtilis RJGP16 significantly induced the expression of PBD2 in the duodenum of neonatal piglets [151]. Likewise, both L. reuteri D8 and Lactobacillus rhamnosus Gorbach-Goldin (GG) increased body weight gain and decreased the incidence of diarrhea by upregulating HDPs in the jejunum of piglets [152,201].

In chickens, modulation of intestinal HDPs by L. reuteri is gene specific: AvBD1 and CATH3 mRNA expressions were increased, while CATH2 and AvBD10 were decreased, with no change observed with AvBD2 or AvBD12 [153,202]. In bovine mammary epithelial cells, Lactobacillus casei BL23 sustained the expression of DEFs during infection with S. aureus [154]. Furthermore, DEFB1 was increased in colon mucosa from dairy cows supplemented with live Saccharomyces cerevisiae CNCM I-1077 [155]. S. cerevisiae similarly upregulated SBD1 in ovine ruminal epithelial cells [203]. However, not all probiotics have the capacity to induce HDP synthesis. For instance, Bifidobacterium breve M16V did not affect the expression of DEFB1, DEFB4, or CAMP in preterm human infants [204], and 3 of 7 L. casei strains tested failed to upregulated Reg3b or a DEFA in the ileum of mice [146].

Probiotics such as Lactobacillus [133,147] or S. cerevisiae [203] triggered TLR2 along with MAPK and/or NF-κB signaling pathways to induce HDP gene expression in human, porcine, and ovine cells (Figure 4). L. rhamnosus GG and L. plantarum increased VDR protein expression in both mouse and human intestinal epithelial cells and induce CAMP gene expression through VDR [205]. On the contrary, L. gasseri, but not L. reuteri, induced DEFB1 gene expression associated with increased concentrations of histone H3 acetylation, H3K4me3, and H2A.Z in the proximal promoter region of the DEFB1 gene [113]. E. coli Nissle 1917, Pediococcus pentosaceus, Lactobacillus fermentum, and Lactobacillus acidophilus required NF-κB signaling in addition to AP-1 activation to induce DEFB4 [143,198]. An increase in HDP was paralleled by expression of nucleotide-binding oligomerization domain (NOD1/2) in cells treated with L. rhamnosus GG., B. longum spp. Infantis S12, or BI 5764 [136,144]. However, L. reuteri I5007 enhanced HDPs though upregulation of peroxisome proliferator-activated receptor-γ and GPR41 in porcine epithelial cells [200], while L. casei BL23 had no effect on NOD2 nor NF-κB [154]. These discrepancies may be due to differences in probiotic-host interaction or variation in signaling mechanisms between host species. Regardless, the escalating interest in the use of probiotics to promote host health warrants continued research into the mechanisms behind probiotic-induced HDP expression.

FIGURE 4.

FIGURE 4

Molecular mechanisms of HDP gene expression by probiotics. See text for details. Ac, acetyl group; AP1, activator protein-1; HDP, host defense peptide; MAPK, mitogen-activated protein kinases; NF-κB, nuclear factor-kappa B; TLR, toll-like receptor, VDR, vitamin D receptor.

Prebiotics

Supplementation of inulin to a Western-style diet increased the expression of multiple Paneth cell DEFAs and DEFB1 as well as tight junction proteins in the intestinal tissues of mice, resulting in improved intestinal barrier integrity and reduced endotoxemia [24] (Table 1). Similarly, long-chain, but not short-chain, inulin-type fructans delayed the onset of type 1 diabetes by promoting gut health, including a significant increase in Defb1 and Camp in the colon of mice [156]. Additionally, oral administration of polysaccharides extracted from Dendrobium huoshanense, a medicinal plant, enhanced DEFB production and barrier function of the intestinal tract of mice [157]. The HDP-inducing activity of other prebiotics remains to be studied.

Synergistic induction of HDP synthesis by different classes of dietary compounds

Several different classes of dietary compounds have been found to synergize with each other to potentiate HDP synthesis (Table 2) [16,26,35,36,76,77,91,99,100,103,105,[120], [121], [122],206,207]. For example, vitamin D-3 is synergistic with butyrate or PBA in CAMP synthesis in bronchial epithelial cells and macrophages, showing a bacteriostatic effect against M. tuberculosis [36,206]. The combination of vitamin D-3 and PBA was also shown to induce HDPs in human dendritic cells, leading to effective killing of S. aureus [207]. Vitamin D-3 further synergized with entinostat, resveratrol, or pterostilbene to induce CAMP synthesis in human colon cells, monocytes, or keratinocytes [103,121,122]. Synergy in augmenting CAMP expression was also observed when vitamin D-3 was combined with calcium in normal and cystic fibrosis bronchial epithelial cells [208].

TABLE 2.

Synergistic induction of HDP synthesis by combinations of different classes of dietary compounds

Compounds Cells or tissues HDPs References
Butyrate + lithocholic acid Human HT-29 CAMP [16]
Butyrate + forskolin Chicken HD11, HTC, jejunal explants AvBD9 [26]
Butyrate + propionate + acetate Chicken HD11 macrophages AvBD9 [35]
Butyrate/PBA + lactose Human HT-29 CAMP [76]
Butyrate + galactose/trehalose/lactose Chicken HD11 macrophages AvBD9, CATHB1 [77]
Butyrate + COX-2 inhibitors Chicken HTC, PBMCs AvBD9 [91]
Butyrate + wortmannin/tetrandrine/datiscetin Chicken HTC, chicken monocytes AvBD9 [99]
Vitamin D-3 + PBA Human VA10, MDM, PBMCs CAMP [36,206,207]
Vitamin D-3 + resveratrol/pterostilbene Human U937, HaCaT CAMP [103]
Vitamin D-3 + entinostat Human HT-29 CAMP [121,122]
Vitamin D-3 + calcium Human NHBE CAMP [208]
Andrographolide + isoliquiritigenin Human Caco-2 DEFB3 [100]
Polydatin + resveratrol Human HaCaT DEFB4 [105]
HDACi + HMTi/DNMTi Chicken HTC, HD11 macrophages AvBD9 [120]

Abbreviations: AvBD, avian β-defensin; CAMP, cathelicidin; CATH, chicken cathelicidin; COX, cyclooxygenase; DEF, defensin; DNMTi, DNA methyltransferase inhibitor; HDACi, histone deacetylase inhibitor; HDP, host defense peptide; HMTi, histone methyltransferase inhibitor; PBA, phenylbutyrate.

In addition to vitamin D-3, butyrate synergizes with several other classes of dietary compounds to enhance HDP synthesis. When combined with lactose, butyrate or 4-PBA showed a synergistic effect on human CAMP expression [76]. Similarly, butyrate synergized with other sugars, such as galactose or trehalose, to upregulate HDP expression in chicken macrophages [77]. Administration of butyrate and FSK showed a synergistic increase in AvBD9 in the crop and jejunum of chickens [26]. An even higher magnitude of HDP gene induction and protection against necrotic enteritis was observed with the combination of butyrate, lactose, and FSK over any of the 2-compound combinations in chickens [209]. Butyrate in combination with wortmannin, a naturally occurring fungal metabolite and a specific phosphoinositide 3-kinase inhibitor, synergistically increased HDP expression in chickens [99]. Additionally, butyrate synergized with tetrandrine or datiscetin in chicken HDP gene induction [99]. Several polyphenols and COX-2 inhibitors, namely quercetin, resveratrol, anacardic acid, EGCG, and garcinol had a strong synergy with butyrate in HDP transcription in chicken cells [91]. Additionally, butyrate synergized with other SCFAs such as acetate and propionate in enhancing chicken HDP expression both in vitro and in vivo [35].

Furthermore, polydatin, a natural precursor to resveratrol, synergistically increased DEFB4 production in human keratinocytes when combined with resveratrol [105]. Andrographolide and isoliquiritigenin cooperated to enhance DEFB3 expression and antibacterial activity of human colonic epithelial cells [100]. Different classes of epigenetic compounds also appear to act in a corporative manner in increasing HDP gene expression. For instance, an HDAC inhibitor paired with an inhibitor of either an HMT or a DNMT led to a drastic synergy in the transcription of multiple HDP genes in chicken macrophages [120]. Additionally, probiotics such as E. coli Nissle 1917 synergizes with HDCA inhibitors to enhance DEFB4 expression in human intestinal cell lines, but not in human colonic biopsies [118].

The mechanism for synergy between HDP inducers remains elusive. HDAC inhibitors including butyrate increase histone acetylation in favor of active gene transcription [30], so a proposed mechanism for synergy with butyrate involved additional enrichment of histone acetylation. Butyrate and lactose treatment led to the hyperacetylation of H4 preceding a synergistic response in chicken HDP gene induction [77]. However, no additional histone acetylation was observed for butyrate and quercetin in relation to their synergy in chicken HDP transcription [91]. Changes in histone acetylation are insufficient to describe the synergy observed between butyrate and a secondary HDP inducer.

Other studies looked at the signaling pathways activated by individual compounds compared with compound combinations. A similar involvement of p38 MAPK, JNK, NF-κB, and cAMP signaling pathways was observed in response to butyrate and lactose as for butyrate alone [77]. Likewise, the signaling pathways activated by lactose and PBA for synergistic induction of CAMP coincided with those found for the individual treatments [210].

The mechanism appears to be due to the cooperation between epigenetic modulation and regulation of signaling pathways and/or specific transcription factors as opposed to synergy in boosting any individual pathway. For example, induction of CAMP transcription involved cooperation among VDR, C/EBPα, PU.1, and chromatin remodeling [171]. On the contrary, inhibition of ERK1/2 and MAPK-p38 by resveratrol was found to contribute to the synergy with vitamin D-3 in augmenting CAMP gene expression [103]. The synergistic effect of andrographolide and isoliquiritigenin on DEFB3 expression was supported by higher phosphorylation of H3S10 and recruitment of transcription factors Fos and ELK1 than use of either molecule alone [100]. A plausible mechanism for synergy between HDP inducers could be due to increased transcription factor binding when 1 or both modulators can also induce epigenetic changes in favor of an accessible HDP gene promoter.

Conclusions and Future Prospects

Dietary modulation of endogenous HDP synthesis has potential to be developed as an alternative approach to antimicrobial therapy. Several HDP inducers, such as butyrate, 4-PBA, and vitamin D-3, have demonstrated protective efficacy against infectious diseases across multiple animal species. It is noted that nutritional modulation of endogenous HDP synthesis is unlikely to drive antimicrobial resistance, as these HDP-inducing dietary factors act on the host without exerting direct antimicrobial activity. Additionally, unlike other immune boosters that often nonspecifically trigger inflammation, many of the HDP-inducing compounds stimulate the synthesis of HDPs without eliciting a proinflammatory response while some even have anti-inflammatory effects [31,69,100]. This is beneficial, as inflammation can lead to detrimental effects on the host, such as tissue damage. However, additional human and animal trials are warranted to realize the clinical potential of HDP inducers as novel host-directed antimicrobials.

It is noted that different classes of dietary compounds have a strong capacity to induce HDPs in multiple cell types and animal species, although cell-specific and species-specific HDP induction is evident. Importantly, many compounds show a synergy in promoting HDP synthesis when combined. Mechanisms of dietary compound-mediated HDP induction are being investigated and a detailed understanding of the molecular mechanisms may allow further HDP-inducing efficiency. Current evidence indicates the involvement of histone acetylation as well as MAPK, NF-κB, VDR, cAMP, and COX-2 signaling pathways. Transcription factors such as AP-1, CREB, STAT3, and sphingosine-1-phosphate have also been implicated in nutritional regulation of HDPs. Further investigation may yield dietary compounds or their combinations as promising candidates as effective alternatives to antibiotics for both human and animal applications.

Author contributions

The authors’ responsibilities were as follows – GZ: conceived the study; MW, IT, AB, GZ: drafted the manuscript; IT, MW: contributed to visualization; GZ: edited the manuscript and has primary responsibility for final content; and all authors: read and approved the final manuscript.

Conflict of interest

The authors report no conflicts of interest.

Funding

This research was supported by the USDA National Institute of Food and Agriculture grants (2020-67016-31619 and 2023-67015-39095), the Ralph F. and Leila W. Boulware Endowment Fund, and Oklahoma Agricultural Experiment Station Project H-3112. MW was supported by a USDA National Institute of Food and Agriculture Predoctoral Fellowship grant (2021-67034-35184). However, the funders had no role in the design of the study, the collection or interpretation of data, the writing of the manuscript, or the decision to publish the results.

Acknowledgments

We apologize for not being able to include all references owing to space constraints. All figures were created with BioRender.

References

  • 1.Schrader S.M., Vaubourgeix J., Nathan C. Biology of antimicrobial resistance and approaches to combat it. Sci. Transl. Med. 2020;12(549) doi: 10.1126/scitranslmed.aaz6992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ghosh C., Sarkar P., Issa R., Haldar J. Alternatives to conventional antibiotics in the era of antimicrobial resistance. Trends Microbiol. 2019;27(4):323–338. doi: 10.1016/j.tim.2018.12.010. [DOI] [PubMed] [Google Scholar]
  • 3.Christaki E., Marcou M., Tofarides A. Antimicrobial resistance in bacteria: mechanisms, evolution, and persistence. J. Mol. Evol. 2020;88(1):26–40. doi: 10.1007/s00239-019-09914-3. [DOI] [PubMed] [Google Scholar]
  • 4.Darby E.M., Trampari E., Siasat P., Gaya M.S., Alav I., Webber M.A., et al. Molecular mechanisms of antibiotic resistance revisited. Nat. Rev. Microbiol. 2023;21(5):280–295. doi: 10.1038/s41579-022-00820-y. [DOI] [PubMed] [Google Scholar]
  • 5.Watkins R.R., Bonomo R.A. Overview: the ongoing threat of antimicrobial resistance. Infect. Dis. Clin. North. Am. 2020;34(4):649–658. doi: 10.1016/j.idc.2020.04.002. [DOI] [PubMed] [Google Scholar]
  • 6.Vidovic N., Vidovic S. Antimicrobial resistance and food animals: influence of livestock environment on the emergence and dissemination of antimicrobial resistance. Antibiotics (Basel) 2020;9(2):52. doi: 10.3390/antibiotics9020052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.McEwen S.A., Collignon P.J. Antimicrobial resistance: a one health perspective. Microbiol. Spectr. 2018;6(2) doi: 10.1128/microbiolspec.ARBA-0009-2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.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. doi: 10.3389/fimmu.2020.01209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wu J., Ma N., Johnston L.J., Ma X. Dietary nutrients mediate intestinal host defense peptide expression. Adv. Nutr. 2020;11(1):92–102. doi: 10.1093/advances/nmz057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Magana M., Pushpanathan M., Santos A.L., Leanse L., Fernandez M., Ioannidis A., et al. The value of antimicrobial peptides in the age of resistance. Lancet Infect. Dis. 2020;20(9):e216–e230. doi: 10.1016/S1473-3099(20)30327-3. [DOI] [PubMed] [Google Scholar]
  • 11.Lazzaro B.P., Zasloff M., Rolff J. Antimicrobial peptides: application informed by evolution. Science. 2020;368(6490) doi: 10.1126/science.aau5480. eaau5480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mookherjee N., Anderson M.A., Haagsman H.P., Davidson D.J. Antimicrobial host defence peptides: functions and clinical potential. Nat. Rev. Drug Discov. 2020;19(5):311–332. doi: 10.1038/s41573-019-0058-8. [DOI] [PubMed] [Google Scholar]
  • 13.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(2):160–169. doi: 10.1016/j.aninu.2018.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Koh A., De Vadder F., Kovatcheva-Datchary P., Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–1345. doi: 10.1016/j.cell.2016.05.041. [DOI] [PubMed] [Google Scholar]
  • 15.Schauber J., Svanholm C., Termen S., Iffland K., Menzel T., Scheppach W., et al. Expression of the cathelicidin LL-37 is modulated by short chain fatty acids in colonocytes: relevance of signalling pathways. Gut. 2003;52(5):735–741. doi: 10.1136/gut.52.5.735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Termén S., Tollin M., Rodriguez E., Sveinsdóttir S.H., Jóhannesson B., Cederlund A., et al. PU.1 and bacterial metabolites regulate the human gene CAMP encoding antimicrobial peptide LL-37 in colon epithelial cells. Mol. Immunol. 2008;45(15):3947–3955. doi: 10.1016/j.molimm.2008.06.020. [DOI] [PubMed] [Google Scholar]
  • 17.Schwab M., Reynders V., Loitsch S., Steinhilber D., Schröder O., Stein J. The dietary histone deacetylase inhibitor sulforaphane induces human beta-defensin-2 in intestinal epithelial cells. Immunology. 2008;125(2):241–251. doi: 10.1111/j.1365-2567.2008.02834.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kallsen K., Andresen E., Heine H. Histone deacetylase (HDAC) 1 controls the expression of beta defensin 1 in human lung epithelial cells. PLoS One. 2012;7(11) doi: 10.1371/journal.pone.0050000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.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(12):1972–1981. doi: 10.1016/j.molimm.2005.11.014. [DOI] [PubMed] [Google Scholar]
  • 20.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. doi: 10.1016/j.peptides.2013.10.008. [DOI] [PubMed] [Google Scholar]
  • 21.Zhang K., Hussain T., Wang J., Li M., Wang W., Ma X., et al. Sodium butyrate abrogates the growth and pathogenesis of Mycobacterium bovis via regulation of cathelicidin (LL37) expression and NF-κB signaling. Front. Microbiol. 2020;11:433. doi: 10.3389/fmicb.2020.00433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhou Z., Yang H., Li H., Li X., Li X., Wu B., et al. Sodium butyrate ameliorates Corynebacterium pseudotuberculosis infection in RAW264.7 macrophages and C57BL/6 mice. Microb. Pathog. 2019;131:144–149. doi: 10.1016/j.micpath.2019.04.008. [DOI] [PubMed] [Google Scholar]
  • 23.Takakuwa A., Nakamura K., Kikuchi M., Sugimoto R., Ohira S., Yokoi Y., et al. Butyric acid and leucine induce α-defensin secretion from small intestinal Paneth cells. Nutrients. 2019;11(11):2817. doi: 10.3390/nu11112817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Beisner J., Filipe Rosa L., Kaden-Volynets V., Stolzer I., Günther C., Bischoff S.C. Prebiotic inulin and sodium butyrate attenuate obesity-induced intestinal barrier dysfunction by induction of antimicrobial peptides. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.678360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sunkara L.T., Achanta M., Schreiber N.B., Bommineni Y.R., Dai G., Jiang W., et al. Butyrate enhances disease resistance of chickens by inducing antimicrobial host defense peptide gene expression. PLoS One. 2011;6(11) doi: 10.1371/journal.pone.0027225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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(2):171–180. doi: 10.1016/j.molimm.2013.09.003. [DOI] [PubMed] [Google Scholar]
  • 27.Alva-Murillo N., Medina-Estrada I., Báez-Magaña M., Ochoa-Zarzosa A., López-Meza J.E. The activation of the TLR2/p38 pathway by sodium butyrate in bovine mammary epithelial cells is involved in the reduction of Staphylococcus aureus internalization. Mol. Immunol. 2015;68(2 Pt B):445–455. doi: 10.1016/j.molimm.2015.09.025. [DOI] [PubMed] [Google Scholar]
  • 28.Dai H., Wei G., Wang Y., Ma N., Chang G., Shen X. Sodium butyrate promotes lipopolysaccharide-induced innate immune responses by enhancing mitogen-activated protein kinase activation and histone acetylation in bovine mammary epithelial cells. J. Dairy Sci. 2020;103(12):11636–11652. doi: 10.3168/jds.2020-18198. [DOI] [PubMed] [Google Scholar]
  • 29.Zeng X., Sunkara L.T., Jiang W., Bible M., Carter S., Ma X., et al. Induction of porcine host defense peptide gene expression by short-chain fatty acids and their analogs. PLoS One. 2013;8(8) doi: 10.1371/journal.pone.0072922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Xiong H., Guo B., Gan Z., Song D., Lu Z., Yi H., et al. Butyrate upregulates endogenous host defense peptides to enhance disease resistance in piglets via histone deacetylase inhibition. Sci. Rep. 2016;6 doi: 10.1038/srep27070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dou X., Han J., Song W., Dong N., Xu X., Zhang W., et al. Sodium butyrate improves porcine host defense peptide expression and relieves the inflammatory response upon Toll-like receptor 2 activation and histone deacetylase inhibition in porcine kidney cells. Oncotarget. 2017;8(16):26532–26551. doi: 10.18632/oncotarget.15714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dou X., Gao N., Lan J., Han J., Yang Y., Shan A. TLR2/EGFR are two sensors for pBD3 and pEP2C induction by sodium butyrate independent of HDAC inhibition. J. Agric. Food Chem. 2020;68(2):512–522. doi: 10.1021/acs.jafc.9b06569. [DOI] [PubMed] [Google Scholar]
  • 33.Wang S., Zhang C., Yang J., Wang X., Wu K., Zhang B., et al. Sodium butyrate protects the intestinal barrier by modulating intestinal host defense peptide expression and gut microbiota after a challenge with deoxynivalenol in weaned piglets. J. Agric. Food Chem. 2020;68(15):4515–4527. doi: 10.1021/acs.jafc.0c00791. [DOI] [PubMed] [Google Scholar]
  • 34.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. Invest. Dermatol. 2010;130(4):985–994. doi: 10.1038/jid.2009.384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sunkara L.T., Jiang W., Zhang G. Modulation of antimicrobial host defense peptide gene expression by free fatty acids. PLoS One. 2012;7(11) doi: 10.1371/journal.pone.0049558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Steinmann J., Halldorsson S., Agerberth B., Gudmundsson G.H. Phenylbutyrate induces antimicrobial peptide expression, Antimicrob. Agents Chemother. 2009;53(12):5127–5133. doi: 10.1128/AAC.00818-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sarker P., Ahmed S., Tiash S., Rekha R.S., Stromberg R., Andersson J., et al. Phenylbutyrate counteracts Shigella mediated downregulation of cathelicidin in rabbit lung and intestinal epithelia: a potential therapeutic strategy. PLoS One. 2011;6(6) doi: 10.1371/journal.pone.0020637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dou X., Han J., Ma Q., Cheng B., Shan A., Gao N., et al. TLR2/4-mediated NF-κB pathway combined with the histone modification regulates β-defensins and interleukins expression by sodium phenyl butyrate in porcine intestinal epithelial cells, Food Nutr. Res. 2018;62 doi: 10.29219/fnr.v62.1493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pineda Molina C., Hussey G.S., Eriksson J., Shulock M.A., Cárdenas Bonilla L.L., Giglio R.M., et al. 4-Hydroxybutyrate promotes endogenous antimicrobial peptide expression in macrophages. Tissue Eng. Part A. 2019;25(9–10):693–706. doi: 10.1089/ten.TEA.2018.0377. [DOI] [PubMed] [Google Scholar]
  • 40.Wang J., Huang N., Xiong J., Wei H., Jiang S., Peng J. Caprylic acid and nonanoic acid upregulate endogenous host defense peptides to enhance intestinal epithelial immunological barrier function via histone deacetylase inhibition. Int. Immunopharmacol. 2018;65:303–311. doi: 10.1016/j.intimp.2018.10.022. [DOI] [PubMed] [Google Scholar]
  • 41.Alva-Murillo N., Ochoa-Zarzosa A., López-Meza J.E. Short chain fatty acids (propionic and hexanoic) decrease Staphylococcus aureus internalization into bovine mammary epithelial cells and modulate antimicrobial peptide expression. Vet. Microbiol. 2012;155(2–4):324–331. doi: 10.1016/j.vetmic.2011.08.025. [DOI] [PubMed] [Google Scholar]
  • 42.Wang T.T., Nestel F.P., Bourdeau V., Nagai Y., Wang Q., Liao J., et al. Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J. Immunol. 2004;173(5):2909–2912. doi: 10.4049/jimmunol.173.5.2909. [DOI] [PubMed] [Google Scholar]
  • 43.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(9):1067–1077. doi: 10.1096/fj.04-3284com. [DOI] [PubMed] [Google Scholar]
  • 44.Liao S., Huang Y., Zhang J., Xiong Q., Chi M., Yang L., et al. Vitamin D promotes epithelial tissue repair and host defense responses against influenza H1N1 virus and Staphylococcus aureus infections. Respir. Res. 2023;24(1):175. doi: 10.1186/s12931-023-02477-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Schauber J., Oda Y., Büchau A.S., Yun Q.C., Steinmeyer A., Zügel U., et al. Histone acetylation in keratinocytes enables control of the expression of cathelicidin and CD14 by 1,25-dihydroxyvitamin D3. J. Invest. Dermatol. 2008;128(4):816–824. doi: 10.1038/sj.jid.5701102. [DOI] [PubMed] [Google Scholar]
  • 46.Martineau A.R., Wilkinson K.A., Newton S.M., Floto R.A., Norman A.W., Skolimowska K., et al. IFN-gamma- and TNF-independent vitamin D-inducible human suppression of mycobacteria: the role of cathelicidin LL-37. J. Immunol. 2007;178(11):7190–7198. doi: 10.4049/jimmunol.178.11.7190. [DOI] [PubMed] [Google Scholar]
  • 47.Karlsson J., Carlsson G., Larne O., Andersson M., Pütsep K. Vitamin D3 induces pro-LL-37 expression in myeloid precursors from patients with severe congenital neutropenia. J. Leukoc. Biol. 2008;84(5):1279–1286. doi: 10.1189/jlb.0607437. [DOI] [PubMed] [Google Scholar]
  • 48.Filipe Rosa L., Petersen P.P., Görtz L.F., Stolzer I., Kaden-Volynets V., Günther C., et al. Vitamin A- and D-deficient diets disrupt intestinal antimicrobial peptide defense involving Wnt and STAT5 signaling pathways in mice. Nutrients. 2023;15(2):376. doi: 10.3390/nu15020376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhang L., Lu L., Li S., Zhang G., Ouyang L., Robinson K., et al. 1,25-Dihydroxyvitamin-D3 induces avian beta-defensin gene expression in chickens. PLoS One. 2016;11(5) doi: 10.1371/journal.pone.0154546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Téllez-Pérez A.D., Alva-Murillo N., Ochoa-Zarzosa A., López-Meza J.E. Cholecalciferol (vitamin D) differentially regulates antimicrobial peptide expression in bovine mammary epithelial cells: implications during Staphylococcus aureus internalization. Vet. Microbiol. 2012;160(1–2):91–98. doi: 10.1016/j.vetmic.2012.05.007. [DOI] [PubMed] [Google Scholar]
  • 51.Flores-Villalva S., Remot A., Carreras F., Winter N., Gordon S.V., Meade K.G. Vitamin D induced microbicidal activity against Mycobacterium bovis BCG is dependent on the synergistic activity of bovine peripheral blood cell populations. Vet. Immunol. Immunopathol. 2023;256 doi: 10.1016/j.vetimm.2022.110536. [DOI] [PubMed] [Google Scholar]
  • 52.Heilborn J.D., Weber G., Grönberg A., Dieterich C., Ståhle M. Topical treatment with the vitamin D analogue calcipotriol enhances the upregulation of the antimicrobial protein hCAP18/LL-37 during wounding in human skin in vivo. Exp. Dermatol. 2010;19(4):332–338. doi: 10.1111/j.1600-0625.2009.00997.x. [DOI] [PubMed] [Google Scholar]
  • 53.Jacobo-Delgado Y.M., Torres-Juarez F., Rodríguez-Carlos A., Santos-Mena A., Enciso-Moreno J.E., Rivas-Santiago C., et al. Retinoic acid induces antimicrobial peptides and cytokines leading to Mycobacterium tuberculosis elimination in airway epithelial cells. Peptides. 2021;142 doi: 10.1016/j.peptides.2021.170580. [DOI] [PubMed] [Google Scholar]
  • 54.Elloumi H.Z., Holland S.M. Complex regulation of human cathelicidin gene expression: novel splice variants and 5'UTR negative regulatory element. Mol. Immunol. 2008;45(1):204–217. doi: 10.1016/j.molimm.2007.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Liggins M.C., Li F., Zhang L.J., Dokoshi T., Gallo R.L. Retinoids enhance the expression of cathelicidin antimicrobial peptide during reactive dermal adipogenesis. J. Immunol. 2019;203(6):1589–1597. doi: 10.4049/jimmunol.1900520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.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(9):1082–1084. doi: 10.1111/j.1365-4632.2009.04260.x. [DOI] [PubMed] [Google Scholar]
  • 57.Kyme P., Thoennissen N.H., Tseng C.W., Thoennissen G.B., Wolf A.J., Shimada K., et al. C/EBPepsilon mediates nicotinamide-enhanced clearance of Staphylococcus aureus in mice. J. Clin. Invest. 2012;122(9):3316–3329. doi: 10.1172/JCI62070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chen Y., Li P., Zhen R., Wang L., Feng J., Xie Y., et al. Effects of niacin on intestinal epithelial barrier, intestinal immunity, and microbial community in weaned piglets challenged by PDCoV. Int. Immunopharmacol. 2022;111 doi: 10.1016/j.intimp.2022.109054. [DOI] [PubMed] [Google Scholar]
  • 59.Zhen R., Feng J., He D., Chen Y., Chen T., Cai W., et al. Effects of niacin on resistance to enterotoxigenic Escherichia coli infection in weaned piglets. Front. Nutr. 2022;9 doi: 10.3389/fnut.2022.865311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Cruz Díaz L.A., Flores Miramontes M.G., Chávez Hurtado P., Allen K., Gonzalez Ávila 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 doi: 10.1155/2015/714580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Luo K., Li X., Wang L., Rao W., Wu Y., Liu Y., et al. Ascorbic acid regulates the immunity, anti-oxidation and apoptosis in abalone Haliotis discus hannai Ino. Antioxidants (Basel) 2021;10(9):1449. doi: 10.3390/antiox10091449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sherman H., Chapnik N., Froy O. Albumin and amino acids upregulate the expression of human beta-defensin 1. Mol. Immunol. 2006;43(10):1617–1623. doi: 10.1016/j.molimm.2005.09.013. [DOI] [PubMed] [Google Scholar]
  • 63.Rivas-Santiago C.E., Rivas-Santiago B., León D.A., Castañeda-Delgado J., Hernández Pando R. Induction of beta-defensins by l-isoleucine as novel immunotherapy in experimental murine tuberculosis. Clin. Exp. Immunol. 2011;164(1):80–89. doi: 10.1111/j.1365-2249.2010.04313.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Fehlbaum P., Rao M., Zasloff M., Anderson G.M. An essential amino acid induces epithelial beta-defensin expression. Proc. Natl. Acad. Sci. U.S.A. 2000;97(23):12723–12728. doi: 10.1073/pnas.220424597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Mao X., Qi S., Yu B., He J., Yu J., Chen D. Zn(2+) and L-isoleucine induce the expressions of porcine beta-defensins in IPEC-J2 cells. Mol. Biol. Rep. 2013;40(2):1547–1552. doi: 10.1007/s11033-012-2200-0. [DOI] [PubMed] [Google Scholar]
  • 66.Ren M., Zhang S., Liu X., Li S., Mao X., Zeng X., et al. Different lipopolysaccharide branched-chain amino acids modulate porcine intestinal endogenous β-defensin expression through the Sirt1/ERK/90RSK pathway. J. Agric. Food Chem. 2016;64(17):3371–3379. doi: 10.1021/acs.jafc.6b00968. [DOI] [PubMed] [Google Scholar]
  • 67.Zhao Y., Yan M.Y., Jiang Q., Yin L., Zhou X.Q., Feng L., et al. Isoleucine improved growth performance, and intestinal immunological and physical barrier function of hybrid catfish Pelteobagrus vachelli × Leiocassis longirostris. Fish Shellfish Immunol. 2021;109:20–33. doi: 10.1016/j.fsi.2020.09.035. [DOI] [PubMed] [Google Scholar]
  • 68.Zhao J., Zhao Y., Liu H., Cao Q., Feng L., Zhang Z., et al. Dietary leucine improves fish intestinal barrier function by increasing humoral immunity, antioxidant capacity, and tight junction. Int. J. Mol. Sci. 2023;24(5):4716. doi: 10.3390/ijms24054716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gao N., Yang Y., Liu S., Fang C., Dou X., Zhang L., et al. Gut-derived metabolites from dietary tryptophan supplementation quench intestinal inflammation through the AMPK-SIRT1-autophagy pathway. J. Agric. Food Chem. 2022;70(51):16080–16095. doi: 10.1021/acs.jafc.2c05381. [DOI] [PubMed] [Google Scholar]
  • 70.Gao N., Dou X., Yin T., Yang Y., Yan D., Ma Z., et al. Tryptophan promotes intestinal immune defense through calcium-sensing receptor (CaSR)-dependent metabolic pathways. J. Agric. Food Chem. 2021;69(45):13460–13473. doi: 10.1021/acs.jafc.1c05820. [DOI] [PubMed] [Google Scholar]
  • 71.Rao Z., Li J., Shi B., Zeng Y., Liu Y., Sun Z., et al. Dietary tryptophan levels impact growth performance and intestinal microbial ecology in weaned piglets via tryptophan metabolites and intestinal antimicrobial peptides. Animals. (Basel) 2021;11(3):817. doi: 10.3390/ani11030817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wang C., Yang Y., Gao N., Lan J., Dou X., Li J., et al. L-Threonine upregulates the expression of β-defensins by activating the NF-κB signaling pathway and suppressing SIRT1 expression in porcine intestinal epithelial cells. Food Funct. 2021;12(13):5821–5836. doi: 10.1039/D1FO00269D. [DOI] [PubMed] [Google Scholar]
  • 73.Dong Y.W., Jiang W.D., Wu P., Liu Y., Kuang S.Y., Tang L., et al. Novel insight into nutritional regulation in enhancement of immune status and mediation of inflammation dynamics integrated study in vivo and in vitro of teleost grass carp (Ctenopharyngodon idella): administration of threonine. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.770969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu G., Ren W., Fang J., Hu C.A., Guan G., Al-Dhabi N.A., et al. L-glutamine and L-arginine protect against enterotoxigenic Escherichia coli infection via intestinal innate immunity in mice. Amino Acids. 2017;49(12):1945–1954. doi: 10.1007/s00726-017-2410-9. [DOI] [PubMed] [Google Scholar]
  • 75.Wang X., Pierre J.F., Heneghan A.F., Busch R.A., Kudsk K.A. Glutamine improves innate immunity and prevents bacterial enteroinvasion during parenteral nutrition. JPEN J. Parenter. Enteral. Nutr. 2015;39(6):688–697. doi: 10.1177/0148607114535265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Cederlund A., Kai-Larsen Y., Printz G., Yoshio H., Alvelius G., Lagercrantz H., et al. Lactose in human breast milk an inducer of innate immunity with implications for a role in intestinal homeostasis. PLoS One. 2013;8(1) doi: 10.1371/journal.pone.0053876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yang Q., Fong L.A., Lyu W., Sunkara L.T., Xiao K., Zhang G. Synergistic induction of chicken antimicrobial host defense peptide gene expression by butyrate and sugars. Front. Microbiol. 2021;12 doi: 10.3389/fmicb.2021.781649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yu W., Xiao X., Chen D., Yu B., He J., Zheng P., et al. Effect of dietary lactose supplementation on growth performance and intestinal epithelium functions in weaned pigs challenged by Rotavirus. Animals. 2022;12(18):2336. doi: 10.3390/ani12182336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.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(2):452–456. doi: 10.1016/j.bbrc.2007.12.158. [DOI] [PubMed] [Google Scholar]
  • 80.Page R.A., Malik A.N. Elevated levels of beta defensin-1 mRNA in diabetic kidneys of GK rats. Biochem. Biophys. Res. Commun. 2003;310(2):513–521. doi: 10.1016/j.bbrc.2003.09.034. [DOI] [PubMed] [Google Scholar]
  • 81.Talukder P., Satho T., Irie K., Sharmin T., Hamady D., Nakashima Y., et al. Trace metal zinc stimulates secretion of antimicrobial peptide LL-37 from Caco-2 cells through ERK and p38 MAP kinase. Int. Immunopharmacol. 2011;11(1):141–144. doi: 10.1016/j.intimp.2010.10.010. [DOI] [PubMed] [Google Scholar]
  • 82.Poiraud C., Quereux G., Knol A.C., Zuliani T., Allix R., Khammari A., et al. Human beta-defensin-2 and psoriasin, two new innate immunity targets of zinc gluconate. Eur. J. Dermatol. 2012;22(5):634–639. doi: 10.1684/ejd.2012.1800. [DOI] [PubMed] [Google Scholar]
  • 83.Harder J., Meyer-Hoffert U., Wehkamp K., Schwichtenberg L., Schröder J.M. Differential gene induction of human beta-defensins (hBD-1, -2, -3, and -4) in keratinocytes is inhibited by retinoic acid. J. Invest. Dermatol. 2004;123(3):522–529. doi: 10.1111/j.0022-202X.2004.23234.x. [DOI] [PubMed] [Google Scholar]
  • 84.Abiko Y., Nishimura M., Kusano K., Yamazaki M., Arakawa T., Takuma T., et al. Upregulated expression of human beta defensin-1 and -3 mRNA during differentiation of keratinocyte immortalized cell lines, HaCaT and PHK16-0b. J. Dermatol. Sci. 2003;31(3):225–228. doi: 10.1016/s0923-1811(03)00007-0. [DOI] [PubMed] [Google Scholar]
  • 85.Peric M., Koglin S., Dombrowski Y., Gross K., Bradac E., Ruzicka T., et al. VDR and MEK-ERK dependent induction of the antimicrobial peptide cathelicidin in keratinocytes by lithocholic acid. Mol. Immunol. 2009;46(16):3183–3187. doi: 10.1016/j.molimm.2009.08.010. [DOI] [PubMed] [Google Scholar]
  • 86.D’Aldebert E., Biyeyeme Bi Mve M.J., Mergey M., Wendum D., Firrincieli D., Coilly A., et al. Bile salts control the antimicrobial peptide cathelicidin through nuclear receptors in the human biliary epithelium. Gastroenterology. 2009;136(4):1435–1443. doi: 10.1053/j.gastro.2008.12.040. [DOI] [PubMed] [Google Scholar]
  • 87.Hochberg A., Patz M., Karrasch T., Schäffler A., Schmid A. Serum levels and adipose tissue gene expression of cathelicidin antimicrobial peptide (CAMP) in obesity and during weight loss, Horm. Metab. Res. 2021;53(3):169–177. doi: 10.1055/a-1323-3050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lombardo 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(5):615–623. doi: 10.1111/jre.12142. [DOI] [PubMed] [Google Scholar]
  • 89.Mou Q., Jiang Y., Zhu L., Zhu Z., Ren T. EGCG induces β-defensin 3 against influenza A virus H1N1 by the MAPK signaling pathway. Exp. Ther. Med. 2020;20(4):3017–3024. doi: 10.3892/etm.2020.9047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.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(5):1048–1058. doi: 10.1002/mnfr.201500992. [DOI] [PubMed] [Google Scholar]
  • 91.Yang Q., Burkardt A.C., Sunkara L.T., Xiao K., Zhang G. Natural cyclooxygenase-2 inhibitors synergize with butyrate to augment chicken host defense peptide gene expression. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.819222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Bayele H.K., Balesaria S., Srai S.K. Phytoestrogens modulate hepcidin expression by Nrf2: implications for dietary control of iron absorption. Free Radic. Biol. Med. 2015;89:1192–1202. doi: 10.1016/j.freeradbiomed.2015.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ying L., Wu H., Zhou S., Lu H., Ding M., Wang B., et al. Toll-like receptors signaling pathway of quercetin regulating avian beta-defensin in the ileum of broilers. Front. Cell Dev. Biol. 2022;10 doi: 10.3389/fcell.2022.816771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Wang J., Zhang C., Zhang J., Xie J., Yang L., Xing Y., et al. The effects of quercetin on immunity, antioxidant indices, and disease resistance in zebrafish (Danio rerio) Fish Physiol. Biochem. 2020;46(2):759–770. doi: 10.1007/s10695-019-00750-2. [DOI] [PubMed] [Google Scholar]
  • 95.Min S.Y., Park C.H., Yu H.W., Park Y.J. Anti-inflammatory and anti-allergic effects of saponarin and its impact on signaling pathways of RAW 264.7, RBL-2H3, and HaCaT cells. Int. J. Mol. Sci. 2021;22(16):8432. doi: 10.3390/ijms22168431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Park C.H., Min S.Y., Yu H.W., Kim K., Kim S., Lee H.J., et al. Effects of apigenin on RBL-2H3, RAW264.7, and HaCaT cells: anti-allergic, anti-inflammatory, and skin-protective activities. Int. J. Mol. Sci. 2020;21(13):4620. doi: 10.3390/ijms21134620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Park K., Kim Y.I., Shin K.O., Seo H.S., Kim J.Y., Mann T., et al. The dietary ingredient, genistein, stimulates cathelicidin antimicrobial peptide expression through a novel S1P-dependent mechanism. J. Nutr. Biochem. 2014;25(7):734–740. doi: 10.1016/j.jnutbio.2014.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wang J., Lyu W., Zhang W., Chen Y., Luo F., Wang Y., et al. Discovery of natural products capable of inducing porcine host defense peptide gene expression using cell-based high throughput screening. J. Anim. Sci. Biotechnol. 2021;12(1):14. doi: 10.1186/s40104-020-00536-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lyu W., Deng Z., Sunkara L.T., Becker S., Robinson K., Matts R., et al. High throughput screening for natural host defense peptide-inducing compounds as novel alternatives to antibiotics. Front. Cell Infect. Microbiol. 2018;8:191. doi: 10.3389/fcimb.2018.00191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Sechet E., Telford E., Bonamy C., Sansonetti P.J., Sperandio B. Natural molecules induce and synergize to boost expression of the human antimicrobial peptide beta-defensin-3. Proc. Natl. Acad. Sci. U.S.A. 2018;115(42):E9869–E9878. doi: 10.1073/pnas.1805298115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lee S.I., Park K.H., Kim S.J., Kang Y.G., Lee Y.M., et al. Mechanical stress-activated immune response genes via sirtuin 1 expression in human periodontal ligament cells. Clin. Exp. Immunol. 2012;168(1):113–124. doi: 10.1111/j.1365-2249.2011.04549.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Park K., Elias P.M., Hupe M., Borkowski A.W., Gallo R.L., Shin K.O., et al. Resveratrol stimulates sphingosine-1-phosphate signaling of cathelicidin production. J Invest Dermatol. 2013;133(8):1942–1949. doi: 10.1038/jid.2013.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Guo C., Sinnott B., Niu B., Lowry M.B., Fantacone M.L., Gombart A.F. Synergistic induction of human cathelicidin antimicrobial peptide gene expression by vitamin D and stilbenoids. Mol. Nutr. Food Res. 2014;58(3):528–536. doi: 10.1002/mnfr.201300266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhuang Y., Huang H., Liu S., Liu F., Tu Q., Yin Y., He S. Resveratrol improves growth performance, intestinal morphology, and microbiota composition and metabolism in mice. Front. Microbiol. 2021;12 doi: 10.3389/fmicb.2021.726878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ravagnan G., De Filippis A., Cartenì M., De Maria S., Cozza V., Petrazzuolo M., et al. Polydatin, a natural precursor of resveratrol, induces beta-defensin production and reduces inflammatory response. Inflammation. 2013;36(1):26–34. doi: 10.1007/s10753-012-9516-8. [DOI] [PubMed] [Google Scholar]
  • 106.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(3):214–221. doi: 10.1111/jop.12223. [DOI] [PubMed] [Google Scholar]
  • 107.de Barros P.P., Rossoni R.D., Garcia M.T., Kaminski V.L., Loures F.V., Fuchs B.B., et al. The anti-biofilm efficacy of caffeic acid phenethyl ester (CAPE) in vitro and a murine model of oral candidiasis. Front. Cell. Infect. Microbiol. 2021;11 doi: 10.3389/fcimb.2021.700305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Guo C., Rosoha E., Lowry M.B., Borregaard N., Gombart A.F. Curcumin induces human cathelicidin antimicrobial peptide gene expression through a vitamin D receptor-independent pathway. J. Nutr. Biochem. 2013;24(5):754–759. doi: 10.1016/j.jnutbio.2012.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ming J., Ye J., Zhang Y., Xu Q., Yang X., Shao X., et al. Optimal dietary curcumin improved growth performance, and modulated innate immunity, antioxidant capacity and related genes expression of NF-κB and Nrf2 signaling pathways in grass carp (Ctenopharyngodon idella) after infection with Aeromonas hydrophila. Fish Shellfish Immunol. 2020;97:540–553. doi: 10.1016/j.fsi.2019.12.074. [DOI] [PubMed] [Google Scholar]
  • 110.Chakraborty K., Maity P.C., Sil A.K., Takeda Y., Das S. cAMP stringently regulates human cathelicidin antimicrobial peptide expression in the mucosal epithelial cells by activating cAMP-response element-binding protein, AP-1, and inducible cAMP early repressor. J. Biol. Chem. 2009;284(33):21810–21827. doi: 10.1074/jbc.M109.001180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Yin L., Chung W.O. Epigenetic regulation of human beta-defensin 2 and CC chemokine ligand 20 expression in gingival epithelial cells in response to oral bacteria. Mucosal Immunol. 2011;4(4):409–419. doi: 10.1038/mi.2010.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fischer N., Sechet E., Friedman R., Amiot A., Sobhani I., Nigro G., et al. Histone deacetylase inhibition enhances antimicrobial peptide but not inflammatory cytokine expression upon bacterial challenge. Proc. Natl. Acad. Sci. U.S.A. 2016;113(21):E2993–E3001. doi: 10.1073/pnas.1605997113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lee J., Jang A., Kim J.W., Han J.H., Chun B.H., Jung H.S., et al. Distinct histone modifications modulate DEFB1 expression in human vaginal keratinocytes in response to Lactobacillus spp. Probiotics Antimicrob. Proteins. 2017;9(4):406–414. doi: 10.1007/s12602-017-9286-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Deng Z., Wang J., Lyu W., Wieneke X., Matts R., Ma X., et al. Development of a cell-based high-throughput screening assay to identify porcine host defense peptide-inducing compounds. J. Immunol. Res. 2018;2018 doi: 10.1155/2018/5492941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kweh M.F., Merriman K.E., Nelson C.D. Short communication: inhibition of DNA methyltransferase and histone deacetylase increases beta-defensin expression but not the effects of lipopolysaccharide or 1,25-dihydroxyvitamin D3 in bovine mammary epithelial cells. J. Dairy Sci. 2019;102(6):5706–5712. doi: 10.3168/jds.2018-16141. [DOI] [PubMed] [Google Scholar]
  • 116.Sangeeta K., Yenugu S. Male reproductive tract antimicrobial expression in the extremes of ages of rats. Gene. 2019;710:218–232. doi: 10.1016/j.gene.2019.05.053. [DOI] [PubMed] [Google Scholar]
  • 117.Roy G., Brar H.K., Muthuswami R., Madhubala R. Epigenetic regulation of defense genes by histone deacetylase1 in human cell line-derived macrophages promotes intracellular survival of Leishmania donovani. PLoS Negl. Trop. Dis. 2020;14(4) doi: 10.1371/journal.pntd.0008167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Stebe-Frick S., Ostaff M.J., Stange E.F., Malek N.P., Wehkamp J. Histone deacetylase-mediated regulation of the antimicrobial peptide hBD2 differs in intestinal cell lines and cultured tissue. Sci. Rep. 2018;8(1) doi: 10.1038/s41598-018-31125-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zhou Y., Wang Q., Yang Q., Tang J., Xu C., Gai D., et al. Histone deacetylase 3 inhibitor suppresses hepatitis C virus replication by regulating Apo-A1 and LEAP-1 expression. Virol. Sin. 2018;33(5):418–428. doi: 10.1007/s12250-018-0057-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Whitmore M.A., Li H., Lyu W., Khanam S., Zhang G. Epigenetic regulation of host defense peptide synthesis: synergy between histone deacetylase inhibitors and DNA/histone methyltransferase inhibitors. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.874706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ottosson H., Nylén F., Sarker P., Miraglia E., Bergman P., Gudmundsson G.H., et al. Potent inducers of endogenous antimicrobial peptides for host directed therapy of infections. Sci. Rep. 2016;6 doi: 10.1038/srep36692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Miraglia E., Nylén F., Johansson K., Arnér E., Cebula M., Farmand S., et al. Entinostat up-regulates the CAMP gene encoding LL-37 via activation of STAT3 and HIF-1alpha transcription factors. Sci. Rep. 2016;6 doi: 10.1038/srep33274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Sarker P., Banik A., Stromberg R., Gudmundsson G.H., Raqib R., Agerberth B. Treatment with entinostat heals experimental cholera by affecting physical and chemical barrier functions of intestinal epithelia. Antimicrob. Agents Chemother. 2017;61(7) doi: 10.1128/AAC.02570-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Deng Z., Lyu W., Zhang G. High-throughput identification of epigenetic compounds to enhance chicken host defense peptide gene expression. Antibiotics. (Basel) 2022;11(7):933. doi: 10.3390/antibiotics11070933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Myszor I.T., Parveen Z., Ottosson H., Bergman P., Agerberth B., Strömberg R., et al. Novel aroylated phenylenediamine compounds enhance antimicrobial defense and maintain airway epithelial barrier integrity. Sci. Rep. 2019;9(1):7114. doi: 10.1038/s41598-019-43350-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lyu W., Mi D., Vinson P.N., Xiao Y., Zhang G. Large-scale identification of multiple classes of host defense peptide-inducing compounds for antimicrobial therapy. Int. J. Mol. Sci. 2022;23(15):8400. doi: 10.3390/ijms23158400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Kamino Y., Kurashige Y., Uehara O., Sato J., Nishimura M., Yoshida K., et al. HBD-2 is downregulated in oral carcinoma cells by DNA hypermethylation, and increased expression of hBD-2 by DNA demethylation and gene transfection inhibits cell proliferation and invasion. Oncol. Rep. 2014;32(2):462–468. doi: 10.3892/or.2014.3260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Chen X., Qi G., Qin M., Zou Y., Zhong K., Tang Y., et al. DNA methylation directly downregulates human cathelicidin antimicrobial peptide gene (CAMP) promoter activity. Oncotarget. 2017;8(17):27943–27952. doi: 10.18632/oncotarget.15847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Wang G., Li Y., Yang G., Yang T., He L., Wang Y. Cathelicidin antimicrobial peptide (CAMP) gene promoter methylation induces chondrocyte apoptosis. Hum. Genomics. 2021;15(1):24. doi: 10.1186/s40246-021-00321-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kausar S., Abbas M.N., Gul I., Liu R., Li Q., Zhao E., et al. Molecular identification of two DNA methyltransferase genes and their functional characterization in the anti-bacterial immunity of Antheraea pernyi. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.855888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ghadimi D., Hassan M., Njeru P.N., de Vrese M., Geis A., Shalabi S.I., et al. Suppression subtractive hybridization identifies bacterial genomic regions that are possibly involved in hBD-2 regulation by enterocytes. Mol. Nutr. Food Res. 2011;55(10):1533–1542. doi: 10.1002/mnfr.201100052. [DOI] [PubMed] [Google Scholar]
  • 132.Messaoudi S., Madi A., Prévost H., Feuilloley M., Manai M., Dousset X., et al. In vitro evaluation of the probiotic potential of Lactobacillus salivarius SMXD51. Anaerobe. 2012;18(6):584–589. doi: 10.1016/j.anaerobe.2012.10.004. [DOI] [PubMed] [Google Scholar]
  • 133.Paparo L., Aitoro R., Nocerino R., Fierro C., Bruno C., Canani R.B. Direct effects of fermented cow’s milk product with Lactobacillus paracasei CBA L74 on human enterocytes. Benef. Microbes. 2018;9(1):165–172. doi: 10.3920/BM2017.0038. [DOI] [PubMed] [Google Scholar]
  • 134.Kobatake E., Kabuki T. S-layer protein of Lactobacillus helveticus SBT2171 promotes human β-defensin 2 expression via TLR2–JNK signaling. Front. Microbiol. 2019;10:2414. doi: 10.3389/fmicb.2019.02414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kobatake E., Kobayashi R., Kabuki T., Kurita-Ochiai T. Lactobacillus helveticus SBT2171 upregulates the expression of β-defensin and ameliorates periodontal disease caused by Porphyromonas gingivalis. Microbiol. Immunol. 2019;63(8):293–302. doi: 10.1111/1348-0421.12719. [DOI] [PubMed] [Google Scholar]
  • 136.Huang F.C., Lu Y.T., Liao Y.H. Beneficial effect of probiotics on Pseudomonas aeruginosa-infected intestinal epithelial cells through inflammatory IL-8 and antimicrobial peptide human beta-defensin-2 modulation. Innate Immun. 2020;26(7):592–600. doi: 10.1177/1753425920959410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Pahumunto N., Duangnumsawang Y., Teanpaisan R. Effects of potential probiotics on the expression of cytokines and human beta-defensins in human gingival epithelial cells and in vivo efficacy in a dog model. Arch. Oral. Biol. 2022;142 doi: 10.1016/j.archoralbio.2022.105513. [DOI] [PubMed] [Google Scholar]
  • 138.Miquel S., Verlaguet J., Garcin S., Bertran T., Evrard B., Forestier C., Vareille-Delarbre M. Lacticaseibacillus rhamnosus Lcr35 stimulates epithelial vaginal defenses upon Gardnerella vaginalis infection. Infect. Immun. 2022;90(9) doi: 10.1128/iai.00309-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Rizzo A., Losacco A., Carratelli C.R. Lactobacillus crispatus modulates epithelial cell defense against Candida albicans through toll-like receptors 2 and 4, interleukin 8 and human beta-defensins 2 and 3. Immunol. Lett. 2013;156(1–2):102–109. doi: 10.1016/j.imlet.2013.08.013. [DOI] [PubMed] [Google Scholar]
  • 140.Rosignoli C., Thibaut de Ménonville S., Orfila D., Béal M., Bertino B., Aubert J., et al. A topical treatment containing heat-treated Lactobacillus johnsonii NCC 533 reduces Staphylococcus aureus adhesion and induces antimicrobial peptide expression in an in vitro reconstructed human epidermis model. Exp. Dermatol. 2018;27(4):358–365. doi: 10.1111/exd.13504. [DOI] [PubMed] [Google Scholar]
  • 141.Moro-García M.A., Alonso-Arias R., Baltadjieva M., Fernández Benítez C., Fernández Barrial M.A., Díaz Ruisánchez E., et al. Oral supplementation with Lactobacillus delbrueckii subsp. bulgaricus 8481 enhances systemic immunity in elderly subjects. Age. 2013;35(4):1311–1326. doi: 10.1007/s11357-012-9434-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Nocerino R., De Filippis F., Cecere G., Marino A., Micillo M., Di Scala C., et al. The therapeutic efficacy of Bifidobacterium animalis subsp. lactis BB-12® in infant colic: a randomised, double blind, placebo-controlled trial, Aliment. Pharmacol. Ther. 2020;51(1):110–120. doi: 10.1111/apt.15561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Schlee M., Harder J., Köten B., Stange E.F., Wehkamp J., Fellermann K. Probiotic Lactobacilli and VSL#3 induce enterocyte beta-defensin 2. Clin. Exp. Immunol. 2008;151(3):528–535. doi: 10.1111/j.1365-2249.2007.03587.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hrdý J., Alard J., Couturier-Maillard A., Boulard O., Boutillier D., Delacre M., et al. Lactobacillus reuteri 5454 and Bifidobacterium animalis ssp. lactis 5764 improve colitis while differentially impacting dendritic cells maturation and antimicrobial responses. Sci. Rep. 2020;10(1):5345. doi: 10.1038/s41598-020-62161-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Sánchez E., Nieto J.C., Vidal S., Santiago A., Martinez X., Sancho F.J., et al. Fermented milk containing Lactobacillus paracasei subsp. paracasei CNCM I-1518 reduces bacterial translocation in rats treated with carbon tetrachloride. Sci. Rep. 2017;7 doi: 10.1038/srep45712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Aktas B., De Wolfe T.J., Safdar N., Darien B.J., Steele J.L. The Impact of Lactobacillus casei on the composition of the cecal microbiota and innate immune system is strain specific. PLoS One. 2016;11(5) doi: 10.1371/journal.pone.0156374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wang J., Zhang W., Wang S., Liu H., Zhang D., Wang Y., et al. Swine-derived probiotic Lactobacillus plantarum modulates porcine intestinal endogenous host defense peptide synthesis through TLR2/MAPK/AP-1 signaling pathway. Front. Immunol. 2019;10:2691. doi: 10.3389/fimmu.2019.02691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wang J., Ji H., Wang S., Liu H., Zhang W., Zhang D., et al. Probiotic Lactobacillus plantarum promotes intestinal barrier function by strengthening the epithelium and modulating gut microbiota. Front. Microbiol. 2018;9:1953. doi: 10.3389/fmicb.2018.01953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Shen J., Zhang J., Zhao Y., Lin Z., Ji L., Ma X. Tibetan pig-derived probiotic Lactobacillus amylovorus SLZX20-1 improved intestinal function via producing enzymes and regulating intestinal microflora. Front. Nutr. 2022;9 doi: 10.3389/fnut.2022.846991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Sudan S., Zhan X., Li J. A novel probiotic Bacillus subtilis strain confers cytoprotection to host pig intestinal epithelial cells during enterotoxic Escherichia coli infection. Microbiol. Spectr. 2022;10(4) doi: 10.1128/spectrum.01257-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Deng J., Li Y., Zhang J., Yang Q. Co-administration of Bacillus subtilis RJGP16 and Lactobacillus salivarius B1 strongly enhances the intestinal mucosal immunity of piglets. Res. Vet. Sci. 2013;94(1):62–68. doi: 10.1016/j.rvsc.2012.07.025. [DOI] [PubMed] [Google Scholar]
  • 152.Wang Y., Gong L., Wu Y.P., Cui Z.W., Wang Y.Q., Huang Y., et al. Oral administration of Lactobacillus rhamnosus GG to newborn piglets augments gut barrier function in pre-weaning piglets. J. Zhejiang Univ. Sci. B. 2019;20(2):180–192. doi: 10.1631/jzus.B1800022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Terada T., Nii T., Isobe N., Yoshimura Y. Effects of probiotics Lactobacillus reuteri and Clostridium butyricum on the expression of toll-like receptors, pro- and anti-inflammatory cytokines, and antimicrobial peptides in broiler chick intestine. J. Poult. Sci. 2020;57(4):310–318. doi: 10.2141/jpsa.0190098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Souza R.F.S., Rault L., Seyffert N., Azevedo V., Le Loir Y., Even S. Lactobacillus casei BL23 modulates the innate immune response in Staphylococcus aureus-stimulated bovine mammary epithelial cells. Benef. Microbes. 2018;9(6):985–995. doi: 10.3920/BM2018.0010. [DOI] [PubMed] [Google Scholar]
  • 155.Bach A., Guasch I., Elcoso G., Chaucheyras-Durand F., Castex M., Fàbregas F., et al. Changes in gene expression in the rumen and colon epithelia during the dry period through lactation of dairy cows and effects of live yeast supplementation. J. Dairy Sci. 2018;101(3):2631–2640. doi: 10.3168/jds.2017-13212. [DOI] [PubMed] [Google Scholar]
  • 156.Chen K., Chen H., Faas M.M., de Haan B.J., Li J., Xiao P., et al. Specific inulin-type fructan fibers protect against autoimmune diabetes by modulating gut immunity, barrier function, and microbiota homeostasis. Mol. Nutr. Food Res. 2017;61(8) doi: 10.1002/mnfr.201601006. [DOI] [PubMed] [Google Scholar]
  • 157.Xie S.Z., Liu B., Ye H.Y., Li Q.M., Pan L.H., Zha X.Q., et al. Dendrobium huoshanense polysaccharide regionally regulates intestinal mucosal barrier function and intestinal microbiota in mice. Carbohydr. Polym. 2019;206:149–162. doi: 10.1016/j.carbpol.2018.11.002. [DOI] [PubMed] [Google Scholar]
  • 158.Raqib R., Sarker P., Mily A., Alam N.H., Arifuzzaman A.S., Rekha R.S., et al. Efficacy of sodium butyrate adjunct therapy in shigellosis: a randomized, double-blind, placebo-controlled clinical trial. BMC Infect. Dis. 2012;12:111. doi: 10.1186/1471-2334-12-111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Hinnebusch B.F., Meng S., Wu J.T., Archer S.Y., Hodin R.A. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J. Nutr. 2002;132(5):1012–1017. doi: 10.1093/jn/132.5.1012. [DOI] [PubMed] [Google Scholar]
  • 160.Zhao Y., Chen F., Wu W., Sun M., Bilotta A.J., Yao S., 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(3):752–762. doi: 10.1038/mi.2017.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Chakraborty K., Ghosh S., Koley H., Mukhopadhyay A.K., Ramamurthy T., Saha D.R., et al. Bacterial exotoxins downregulate cathelicidin (hCAP-18/LL-37) and human beta-defensin 1 (HBD-1) expression in the intestinal epithelial cells. Cell Microbiol. 2008;10(12):2520–2537. doi: 10.1111/j.1462-5822.2008.01227.x. [DOI] [PubMed] [Google Scholar]
  • 162.Selvaraj P., Prabhu Anand S., Harishankar M., Alagarasu K. Plasma 1,25 dihydroxy vitamin D3 level and expression of vitamin D receptor and cathelicidin in pulmonary tuberculosis. J. Clin. Immunol. 2009;29(4):470–478. doi: 10.1007/s10875-009-9277-9. [DOI] [PubMed] [Google Scholar]
  • 163.Sato-Deguchi E., Imafuku S., Chou B., Ishii K., Hiromatsu K., Nakayama J. Topical vitamin D₃ analogues induce thymic stromal lymphopoietin and cathelicidin in psoriatic skin lesions. Br. J. Dermatol. 2012;167(1):77–84. doi: 10.1111/j.1365-2133.2012.10917.x. [DOI] [PubMed] [Google Scholar]
  • 164.Muehleisen B., Bikle D.D., Aguilera C., Burton D.W., Sen G.L., Deftos L.J., Gallo R.L. PTH/PTHrP and vitamin D control antimicrobial peptide expression and susceptibility to bacterial skin infection. Sci. Transl. Med. 2012;4(135):135ra66. doi: 10.1126/scitranslmed.3003759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Kanda N., Hau C.S., Tada Y., Sato S., Watanabe S. Decreased serum LL-37 and vitamin D3 levels in atopic dermatitis: relationship between IL-31 and oncostatin M. Allergy. 2012;67(6):804–812. doi: 10.1111/j.1398-9995.2012.02824.x. [DOI] [PubMed] [Google Scholar]
  • 166.Finklea J.D., Grossmann R.E., Tangpricha V. Vitamin D and chronic lung disease: a review of molecular mechanisms and clinical studies. Adv. Nutr. 2011;2(3):244–253. doi: 10.3945/an.111.000398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Aibana O., Huang C.C., Aboud S., Arnedo-Pena A., Becerra M.C., Bellido-Blasco J.B., et al. Vitamin D status and risk of incident tuberculosis disease: a nested case-control study, systematic review, and individual-participant data meta-analysis. PLoS Med. 2019;16(9) doi: 10.1371/journal.pmed.1002907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Ganmaa D., Uyanga B., Zhou X., Gantsetseg G., Delgerekh B., Enkhmaa D., et al. Vitamin D supplements for prevention of tuberculosis infection and disease. N. Engl. J. Med. 2020;383(4):359–368. doi: 10.1056/NEJMoa1915176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Tian G., Liang X., Chen D., Mao X., Yu J., Zheng P., et al. Vitamin D3 supplementation alleviates Rotavirus infection in pigs and IPEC-J2 cells via regulating the autophagy signaling pathway. J. Steroid Biochem. Mol. Biol. 2016;163:157–163. doi: 10.1016/j.jsbmb.2016.05.004. [DOI] [PubMed] [Google Scholar]
  • 170.Coyle C., Wheelhouse N., Jacques M., Longbottom D., Svoboda P., Pohl J., et al. Ovine trophoblasts express cathelicidin host defence peptide in response to infection. J. Reprod. Immunol. 2016;117:10–16. doi: 10.1016/j.jri.2016.06.006. [DOI] [PubMed] [Google Scholar]
  • 171.Wei R., Dhawan P., Baiocchi R.A., Kim K.Y., Christakos S. PU.1 and epigenetic signals modulate 1,25-dihydroxyvitamin D3 and C/EBPalpha regulation of the human cathelicidin antimicrobial peptide gene in lung epithelial cells. J. Cell Physiol. 2019;234(7):10345–10359. doi: 10.1002/jcp.27702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Mielke L.A., Jones S.A., Raverdeau M., Higgs R., Stefanska A., Groom J.R., et al. Retinoic acid expression associates with enhanced IL-22 production by γδ T cells and innate lymphoid cells and attenuation of intestinal inflammation. J. Exp. Med. 2013;210(6):1117–1124. doi: 10.1084/jem.20121588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Wu H., Zhang G., Minton J.E., Ross C.R., Blecha F. Regulation of cathelicidin gene expression: induction by lipopolysaccharide, interleukin-6, retinoic acid, and Salmonella enterica serovar typhimurium infection. Infect. Immun. 2000;68(10):5552–5558. doi: 10.1128/IAI.68.10.5552-5558.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Grubor B., Meyerholz D.K., Lazic T., DeMacedo M.M., Derscheid R.J., Hostetter J.M., et al. Regulation of surfactant protein and defensin mRNA expression in cultured ovine type II pneumocytes by all-trans retinoic acid and VEGF. Int. J. Exp. Pathol. 2006;87(5):393–403. doi: 10.1111/j.1365-2613.2006.00494.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ren M., Cai S., Zhou T., Zhang S., Li S., Jin E., et al. Isoleucine attenuates infection induced by E. coli challenge through the modulation of intestinal endogenous antimicrobial peptide expression and the inhibition of the increase in plasma endotoxin and IL-6 in weaned pigs. Food Funct. 2019;10(6):3535–3542. doi: 10.1039/c9fo00218a. [DOI] [PubMed] [Google Scholar]
  • 176.Tsugami Y., Nii T., Isobe N. Valine treatment enhances antimicrobial component production in mammary epithelial cells and the milk of lactating goats without influencing the tight junction barrier. J. Mammary Gland Biol. Neoplasia. 2023;28(1):3. doi: 10.1007/s10911-023-09529-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Tang Z., Shi B., Sun W., Yin Y., Chen Q., Mohamed T., et al. Tryptophan promoted β-defensin-2 expression via the mTOR pathway and its metabolites: kynurenine banding to aryl hydrocarbon receptor in rat intestine. RSC Adv. 2020;10(6):3371–3379. doi: 10.1039/c9ra10477a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lan J., Dou X., Li J., Yang Y., Xue C., Wang C., et al. L-Arginine ameliorates lipopolysaccharide-induced intestinal inflammation through inhibiting the TLR4/NF-κB and MAPK pathways and stimulating β-defensin expression in vivo and in vitro. J. Agric. Food Chem. 2020;68(9):2648–2663. doi: 10.1021/acs.jafc.9b07611. [DOI] [PubMed] [Google Scholar]
  • 179.Yin J., Yu F.S. LL-37 via EGFR transactivation to promote high glucose-attenuated epithelial wound healing in organ-cultured corneas. Invest. Ophthalmol. Vis. Sci. 2010;51(4):1891–1897. doi: 10.1167/iovs.09-3904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Lan C.C., Wu C.S., Huang S.M., Kuo H.Y., Wu I.H., Liang C.W., et al. High-glucose environment reduces human beta-defensin-2 expression in human keratinocytes: implications for poor diabetic wound healing. Br. J. Dermatol. 2012;166(6):1221–1229. doi: 10.1111/j.1365-2133.2012.10847.x. [DOI] [PubMed] [Google Scholar]
  • 181.Wessels I., Fischer H.J., Rink L. Dietary and physiological effects of zinc on the immune system. Annu. Rev. Nutr. 2021;41:133–175. doi: 10.1146/annurev-nutr-122019-120635. [DOI] [PubMed] [Google Scholar]
  • 182.Kelly P., Feakins R., Domizio P., Murphy J., Bevins C., Wilson J., et al. Paneth cell granule depletion in the human small intestine under infective and nutritional stress. Clin. Exp. Immunol. 2004;135(2):303–309. doi: 10.1111/j.1365-2249.2004.02374.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zhong W., Wei X., Hao L., Lin T.D., Yue R., Sun X., et al. Paneth cell dysfunction mediates alcohol-related steatohepatitis through promoting bacterial translocation in mice: role of zinc deficiency. Hepatology. 2020;71(5):1575–1591. doi: 10.1002/hep.30945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Krisanaprakornkit S., Jotikasthira D., Dale B.A. Intracellular calcium in signaling human beta-defensin-2 expression in oral epithelial cells. J. Dent. Res. 2003;82(11):877–882. doi: 10.1177/154405910308201106. [DOI] [PubMed] [Google Scholar]
  • 185.Mader J.S., Mookherjee N., Hancock R.E., Bleackley R.C. The human host defense peptide LL-37 induces apoptosis in a calpain- and apoptosis-inducing factor-dependent manner involving Bax activity. Mol. Cancer Res. 2009;7(5):689–702. doi: 10.1158/1541-7786.MCR-08-0274. [DOI] [PubMed] [Google Scholar]
  • 186.Vylkova S., Nayyar N., Li W., Edgerton M. Human beta-defensins kill Candida albicans in an energy-dependent and salt-sensitive manner without causing membrane disruption. Antimicrob. Agents Chemother. 2007;51(1):154–161. doi: 10.1128/AAC.00478-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Fuchs C.D., Trauner M. Role of bile acids and their receptors in gastrointestinal and hepatic pathophysiology. Nat. Rev. Gastroenterol. Hepatol. 2022;19(7):432–450. doi: 10.1038/s41575-021-00566-7. [DOI] [PubMed] [Google Scholar]
  • 188.Wang S., Du Q., Meng X., Zhang Y. Natural polyphenols: a potential prevention and treatment strategy for metabolic syndrome. Food Funct. 2022;13(19):9734–9753. doi: 10.1039/d2fo01552h. [DOI] [PubMed] [Google Scholar]
  • 189.Tobin I., Zhang G. Regulation of host defense peptide synthesis by polyphenols. Antibiotics (Basel) 2023;12(4):660. doi: 10.3390/antibiotics12040660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Noguchi T., Shiba H., Komatsuzawa H., Mizuno N., Uchida Y., Ouhara K., et al. Syntheses of prostaglandin E2 and E-cadherin and gene expression of beta-defensin-2 by human gingival epithelial cells in response to Actinobacillus actinomycetemcomitans. Inflammation. 2003;27(6):341–349. doi: 10.1023/b:ifla.0000006702.27906.e9. [DOI] [PubMed] [Google Scholar]
  • 191.Perri F., Longo F., Giuliano M., Sabbatino F., Favia G., Ionna F., et al. Epigenetic control of gene expression: potential implications for cancer treatment. Crit. Rev. Oncol. Hematol. 2017;111:166–172. doi: 10.1016/j.critrevonc.2017.01.020. [DOI] [PubMed] [Google Scholar]
  • 192.Lyu W., Deng Z., Zhang G. High-throughput screening for epigenetic compounds that induce human beta-defensin 1 synthesis. Antibiotics. (Basel) 2023;12(2):186. doi: 10.3390/antibiotics12020186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Bouyahya A., El Hachlafi N., Aanniz T., Bourais I., Mechchate H., Benali T., et al. Natural bioactive compounds targeting histone deacetylases in human cancers: recent updates. Molecules. 2022;27(8):2568. doi: 10.3390/molecules27082568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Akone S.H., Ntie-Kang F., Stuhldreier F., Ewonkem M.B., Noah A.M., Mouelle S.E.M., et al. Natural products impacting DNA methyltransferases and histone deacetylases. Front. Pharmacol. 2020;11:992. doi: 10.3389/fphar.2020.00992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Barrier M.L., Myszor I.T., Sahariah P., Sigurdsson S., Carmena-Bargueño M., Pérez-Sánchez H., et al. Aroylated phenylenediamine HO53 modulates innate immunity, histone acetylation and metabolism. Mol. Immunol. 2023;155:153–164. doi: 10.1016/j.molimm.2023.02.003. [DOI] [PubMed] [Google Scholar]
  • 196.Gschwandtner M., Zhong S., Tschachler A., Mlitz V., Karner S., Elbe-Burger A., et al. Fetal human keratinocytes produce large amounts of antimicrobial peptides: involvement of histone-methylation processes. J. Invest. Dermatol. 2014;134(8):2192–2201. doi: 10.1038/jid.2014.165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Latif A., Shehzad A., Niazi S., Zahid A., Ashraf W., Iqbal M.W., et al. Probiotics: mechanism of action, health benefits and their application in food industries. Front. Microbiol. 2023;14 doi: 10.3389/fmicb.2023.1216674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Wehkamp J., Harder J., Wehkamp K., Wehkamp-von Meissner B., Schlee M., Enders C., et al. NF-kappaB- and AP-1-mediated induction of human beta defensin-2 in intestinal epithelial cells by Escherichia coli Nissle 1917: a novel effect of a probiotic bacterium. Infect. Immun. 2004;72(10):5750–5758. doi: 10.1128/IAI.72.10.5750-5758.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Li X., Song L., Zhu S., Xiao Y., Huang Y., Hua Y., et al. Two strains of Lactobacilli effectively decrease the colonization of VRE in a mouse model. Front. Cell Infect. Microbiol. 2019;9:6. doi: 10.3389/fcimb.2019.00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Liu H., Hou C., Wang G., Jia H., Yu H., Zeng X., et al. Lactobacillus reuteri I5007 modulates intestinal host defense peptide expression in the model of IPEC-J2 cells and neonatal piglets. Nutrients. 2017;9(6):559. doi: 10.3390/nu9060559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Wang M., Wu H., Lu L., Jiang L., Yu Q. Lactobacillus reuteri promotes intestinal development and regulates mucosal immune function in newborn piglets. Front. Vet. Sci. 2020;7:42. doi: 10.3389/fvets.2020.00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Nii T., Jirapat J., Isobe N., Yoshimura Y. Effects of oral administration of Lactobacillus reuteri on mucosal barrier function in the digestive tract of broiler chicks. J. Poult. Sci. 2020;57(1):67–76. doi: 10.2141/jpsa.0190035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Jin X., Zhang M., Yang Y.F. Saccharomyces cerevisiae β-glucan-induced SBD-1 expression in ovine ruminal epithelial cells is mediated through the TLR-2-MyD88-NF-κB/MAPK pathway. Vet. Res. Commun. 2019;43(2):77–89. doi: 10.1007/s11259-019-09747-x. [DOI] [PubMed] [Google Scholar]
  • 204.Strunk T., Hibbert J., Doherty D., Granland C., Trend S., Simmer K., et al. Probiotics and antimicrobial protein and peptide levels in preterm infants, Acta. Paediatr. 2017;106(11):1747–1753. doi: 10.1111/apa.13826. [DOI] [PubMed] [Google Scholar]
  • 205.Wu S., Yoon S., Zhang Y.G., Lu R., Xia Y., Wan J., et al. Vitamin D receptor pathway is required for probiotic protection in colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2015;309(5):G341–G349. doi: 10.1152/ajpgi.00105.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Coussens A.K., Wilkinson R.J., Martineau A.R. Phenylbutyrate is bacteriostatic against Mycobacterium tuberculosis and regulates the macrophage response to infection, synergistically with 25-hydroxy-vitamin D3. PLoS. Pathog. 2015;11(7) doi: 10.1371/journal.ppat.1005007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.van der Does A.M., Kenne E., Koppelaar E., Agerberth B., Lindbom L. Vitamin D3 and phenylbutyrate promote development of a human dendritic cell subset displaying enhanced antimicrobial properties. J. Leukoc. Biol. 2014;95(6):883–891. doi: 10.1189/jlb.1013549. [DOI] [PubMed] [Google Scholar]
  • 208.Yim S., Dhawan P., Ragunath C., Christakos S., Diamond G. Induction of cathelicidin in normal and CF bronchial epithelial cells by 1,25-dihydroxyvitamin D(3) J. Cyst. Fibros. 2007;6(6):403–410. doi: 10.1016/j.jcf.2007.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Yang Q., Whitmore M.A., Robinson K., Lyu W., Zhang G. Butyrate, forskolin, and lactose synergistically enhance disease resistance by inducing the expression of the genes involved in innate host defense and barrier function. Antibiotics. (Basel). 2021;10(10):1175. doi: 10.3390/antibiotics10101175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Cederlund A., Nylén 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(3):365–376. doi: 10.1159/000355931. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Advances in Nutrition are provided here courtesy of American Society for Nutrition

RESOURCES