Download as pdf or txt
Download as pdf or txt
You are on page 1of 29

Periodontology 2000, Vol. 51, 2009, 152180 Printed in Singapore.

All rights reserved

2009 John Wiley & Sons A/S

PERIODONTOLOGY 2000

Antimicrobial peptides of the oral cavity


S V E N -U L R I K G O R R

Antimicrobial proteins and peptides constitute a diverse class of host-defense molecules that act early to combat invasion and infection with bacteria and other microorganisms. These peptides have engendered considerable interest in the past decade as a biological paradigm in innate immunity and as a potential source of novel antibiotics. Many recent reviews have explored the biological diversity and function of this class of peptides [e.g. (36, 50, 73, 80, 103, 126, 127, 191, 209, 221, 226, 255)]. This review focuses especially on antimicrobial proteins and peptides of the oral cavity and on their role in periodontal disease. The potential use of antimicrobial peptides as templates for novel antibiotics is also discussed.

Oral bacteria and infection


Oral infections and attendant inammatory diseases are among the most common human infections. The oral cavity is a major entry point for bacteria and other microorganisms and is typically considered host to over 700 species of bacteria, including known pathogens and commensals, and over 400 of these bacterial species are found in the periodontal pocket. Not all bacteria are found in every individual. For example, only 69 of the 400 species of periodontal bacteria were detected in multiple subjects in one study (188). A recent study, using pyrosequencing of short sequence tags for the 16S ribosomal DNA V6 region, led to much higher estimates of microbial diversity in saliva and plaque (124). A preliminary estimate identied 5669 and 10,052 phylotypes (species) in saliva and plaque, respectively, using operational taxonomic units at 3% difference. This may represent about 50% of the total species present (124). However, 95% of the sequences were represented by the 1000 most

abundant operational taxonomic units, which is similar to previous estimates. Although there are a large number of oral microbes, only a relatively small number of species have been consistently associated with periodontal disease (190, 218, 230) (Table 1). It should be noted that most of the bacteria associated with disease are found in the gingival crevices and periodontal pockets of both healthy and diseased sites (45, 230). However, an age difference has been noted in the types of bacteria associated with periodontal disease, whereby Aggregatibacter actinomycetemcomitans is more prevalent in the young while Porphyromonas gingivalis is the dominant bacterial agent later in life (217). It is probable that differences in environmental factors and in the hosts acquired and innate immune responses, including antimicrobial peptides, pattern recognition receptors, chemokines and cytokines, play a role in determining the microbial balance of oral tissues and the subsequent outcome of bacterial colonization. Consistent with this view, population studies have identied population subgroups with high, moderate or low susceptibility to inammatory diseases, including periodontitis (151). In general, the unique proles of antimicrobial proteins found in different mucosal secretions provide an appropriate response to the microorganisms invading each epithelium. As an example, the oral cavity and airways share exposure to similar environments, and many antimicrobial proteins are common to both locations. However, differences in gene expression and regulation suggest that the innate immune responses differ in these locations (57). Table 1 lists the expression of antimicrobial peptides produced by host cells in response to periodontal pathogens. Because of the lack of available data, the absence of an antimicrobial peptide from the host response may simply be a result of limited data

152

Antimicrobial peptides of the oral cavity

Table 1. Effect of periodontal pathogens on antimicrobial peptide expression in human gingival epithelial cells
Periodontal pathogens Aggregatibacter (Actinobacillus) actinomycetemcomitans1 Antimicrobial peptide hBD-2 hBD-3 hCAP18 Fibronectin Porphyromonas gingivalis
1,3

Expression Up
4

References 140, 186 186 186 246 210 43 240 240 122 114

Up Up Down5 Up Up Up
6

PSP SPLUNC2 hBD-2 hBD-1 hBD-3 Adrenomedullin LL-37 Calgranulin Cystatin C

Up Up = Up
7

170 64 114 114 114 114 114 114 35 114 114 136 114 114 170

Down Down = = = Down6 Down = = = Up Up Up


7

Tannerella forsythia (T. forsythensis)1,3

hBD-3 hBD-1 hBD-2 LL-37

Treponema denticola2,3

hBD-1 hBD-3 hBD-2 LL-37

Fusobacterium nucleatum

hBD-1 hBD-2 hBD-3 LL-37 Calgranulin

Eubacterium nodatum2 Prevotella intermedia2 hBD-1 hBD-2 hBD-3 LL-37 Prevotella nigrescens2
PubMed was searched for the listed bacterial species AND each antimicrobial peptide listed in Table 2 AND the keyword epithelial to identify relevant references. Gene expression in response to each pathogen is shown as up-regulated (Up), down-regulated (Down) or unchanged (=). 1 Consensus pathogens (190). 2 Suspected periodontal pathogens with strong supporting data (230). 3 Bacteria in the red complex described by Socransky et al. (218). 4 A. actinomycetemcomitans has no effect on hBD-2 expression in cells from a patient with localized aggressive periodontitis (140). 5 Cells treated with A. actinomycetemcomitans protease. 6 Some studies found that hBD-1 is expressed constitutively in HGEC (56, 114, 137). 7 Expression was tested in the oral squamous cell carcinoma cell line H400 (194). hBD, human b-defensin; PSP, parotid secretory protein.

Up6 Up Up Up

114 114 114 114

availability, rather than a lack of response. Several of the antimicrobial proteins and peptides are cationic and it has been found that depletion of cationic

proteins in human airway uid also removes the antibacterial activity (44). A similar experiment has not been reported for saliva. However, we recently

153

Gorr

found that ion-exchange fractionation of whole saliva revealed anti-pseudomonal activity in some fractions that was not apparent in the starting material (S. U. Gorr, unpublished data).

Functional families of antimicrobial proteins in the oral cavity


Despite the rich microbial environment of the oral cavity, cuts, abrasions and invasive procedures, such as tooth extraction, rarely lead to infection, indicating the effectiveness of host-defense mechanisms (266). Indeed, oral epithelial cells, neutrophils and salivary glands secrete at least 45 known antimicrobial gene products that are found in saliva (Table 2). A subset of these antimicrobial peptides is also found in gingival crevicular uid (Table 3). Indeed, all of the antimicrobial peptides found in gingival crevicular uid are also found in saliva. Several antimicrobial peptides are more highly concentrated in gingival crevicular uid than in saliva. The concentrations of calgranulins, bronectin, substance P and calcitonin gene related peptide are 10010,000-fold higher in gingival crevicular uid than in whole saliva. Adrenomedulin and beta-2-microglobulin are enriched about 30-fold in gingival crevicular uid. Of the antimicrobial peptides for which gingival crevicular uid concentrations are shown in Table 3, only the alpha-defensins are found at a concentration 1000-fold lower in gingival crevicular uid than in saliva (Tables 2 and 3). The antimicrobial peptides that are highly expressed in gingival crevicular uid are unlikely to be caused by contamination of the gingival crevicular uid samples with saliva (83). Several of these antimicrobial peptides are post-translationally processed (167, 199) or exhibit polymorphisms (182, 257) that further diversify the antimicrobial response of oral tissues and saliva. Antimicrobial peptides are early responders of the innate immune system that search and destroy invading pathogens. The large variety of antimicrobial peptides presumably allows for an effective response to the large variety of microorganisms that invade the mouth and airways. In addition, a host response that involves multiple antimicrobial peptides to a single pathogen is less likely to be met with antimicrobial resistance. Thus, multiple antimicrobial peptides with different minimal inhibitory concentrations act on oral microbes (49). Table 4A,B shows representative antimicrobial activities for antimicrobial peptides found in the oral cavity.

Comparison with the data shown in Table 3 suggests that the concentration of these antimicrobial peptides in gingival crevicular uid is well below the minimal inhibitory concentration for most microbes. However, we do not have sufcient data to determine if multiple antimicrobial peptides are involved in the killing or elimination of individual microorganisms in vivo.

Cationic peptides
Cationic peptides are typically bactericidal and or bacteriostatic. Their activities against typical oral pathogens are listed in Table 4A. Adrenomedullin This 185 amino acid protein is proteolytically processed and C-terminally amidated to produce the 52 amino acid mature adrenomedullin, a cationic amphipathic peptide with one disulde bond. It is found in gingival crevicular uid and glandular and whole saliva. Whole saliva contains higher concentrations of adrenomedullin than glandular saliva, suggesting that oral epithelial cells contribute to the salivary expression (123). In gingival crevicular uid, the amount of adrenomedullin is about twice as high in periodontal disease sites than in healthy sites (158). Alpha-defensins Human neutrophil peptides 14 are 94-amino acid cationic pre-pro-peptides. Human neutrophil peptide-1 (amino acids 6595 of the DEFA1 product) and human neutrophil peptide-3 (amino acids 6594 of the DEFA3 product) differ only at the N-terminal residue. Human neutrophil peptide-2 does not contain this residue and can be derived from both precursor proteins. Human neutrophil peptide-4 is a product of the DEFA4 gene, which contains an 83-base segment not found in the other genes. The mature peptides contain three disulde bridges and are mainly expressed in neutrophils and saliva (79). Alpha-defensins are expressed in neutrophils and have been identied in the gingival crevicular uid of both healthy and diseased sites, although different experimental methodologies lead to somewhat different conclusions on the relative expression in healthy and diseased individuals (56, 159, 193, 195). Defensins exhibit broad antibacterial activity to both gram-positive and gram-negative bacteria (143). The peptides bind to or are inserted into the bacterial cell membrane, causing membrane permeabilization and cell lysis. Defensins are most active against negatively

154

Antimicrobial peptides of the oral cavity

Table 2. Antimicrobial proteins and peptides found in saliva


Protein Adrenomedullin HNP-1 HNP-2 HNP-3 hBD-1 hBD-2 hBD-3 Cathelicidin LL-37 Histatin 1 Histatin 3 Statherin CCL28 Azurocidin CAP37 heparin-binding protein Substance P Calcitonin gene related peptide Neuropeptide Y Vasoactive intestinal peptide Mucin 7 Gene ADM DEFA1 DEFA1 DEFA3 DEFA3 DEFB1 DEFB4 DEFB103A CAMP HTN1 HTN3 STATH CCL28 AZU1 26.5 0.9 MS Whole saliva (lg ml) 0.06 8.6 5.6 02.7 0.15 0.150 0.31 1.6 MS 10.1 7.3 69 2.7 MS 34.7 10.2 75 MS MS Parotid saliva (lg ml) 0.02 MS SM SL saliva (lg ml) 0.035 MS References 123 79 79 74 164 164 231 229, 16 116 116 46 99 102

TAC1 CALCA NPY VIP

7.5 10)6 23.5 10


)6

6.2 10)6 1.5 10


)6

52 52 52 52

41.4 10)6 39.9 10


)6

44.4 10)6 29.9 10


)6

MUC7

40

MS

116157

189 21

GP340 salivary agglutinin DMBT1 Surfactant protein-A Beta-2-microglobulin Proline-rich proteins

DMBT1

MS

6.3

67

SFTPA1 B2M PRH1 PRH2 PRB1 PRB3 PRB2 PRB4

0.9 0.38 MS MS MS MS 512 81


2

215 MS 652 103


2

169 111 111

MS MS MS

MS MS MS MS 0.5 MS 150, 236 129

MS 1.20.13 1.93

MS MS MS

Fibronectin Calgranulin A

FN1 S100A8

155

Gorr

Table 2. Continued
Protein Calgranulin B Psoriasin Lactoferrin Cystatin A Cystatin B Cystatin C Cystatin D Cystatin S Cystatin SA Cystatin SN Secretory leukocyte protease inhibitor SKALP Elan Gene S100A9 S100A7 LTF CSTA CSTB CST3 CST5 CST4 CST2 CST1 SLPI Whole saliva (lg ml) 1.93 MS 20 MS MS 0.9 3.8 53116 78 39 2.9 4.7 MS MS 0.4 MS 1.0 MS MS 0.55 MS MS MS MS MS MS MS 1.1 101, 238 69 18, 101 18 18 147, 212 207, 212, 228 Parotid saliva (lg ml) MS SM SL saliva (lg ml) MS References 129

PI3

0.020

PS4

SMG4

235 142

Lactoperoxidase (salivary peroxidase) Myeloperoxidase

LPO

1.9

6.2

MS

WS = Unstim (233) PS = Stim (207)

MPO

MS

MS

233 185

Lysozyme C Peptidoglycan recognition protein 1

LYZ PGLYRP1

40

4.4

MS MS

7, 130, 207, 212

Common name, gene symbol, concentrations in whole saliva; parotid saliva and submandibular sublingual (SM SL) saliva (lg ml). Values in bold italic are from stimulated saliva. MS = mass spectrometry detection of proteins in unstimulated whole saliva (260, 264) or in stimulated glandular saliva (55). Saliva was collected from healthy subjects, unless stated otherwise. It should be noted that the concentrations shown are representative average values only. Individual variation and variation between studies can be signicant; see references for details. 1 Salivary agglutinin (DMBT1) puried from parotid saliva represented 0.4% of total protein (67). 2 Calculation based on an average parotid salivary ow rate of 0.7 ml min gland (100) and SM SL ow rate of 0.4 ml min gland (261). 3 Measured as calprotectin (calgranulin A and B complex) (129). 4 Found in parotid saliva and submandibular glands (SMG) (142). HNP, human neutrophil peptide, a-defensin.

charged phospholipids and their activity is inhibited by increased salt concentrations, suggesting that ionic interactions between membrane lipids and the cationic peptide are involved in their activity (71, 72). Beta-defensins hBD1, hBD2 and hBD3 are cationic peptides with three disulde bonds. Alpha- and beta-defensin differ in the spacing and the pairing of the cysteine residues (72). They are expressed in epithelial cells of the oral cavity, lung and nasal epithelia, kidney, pancreas, uterus and eye (1), and are found in the gingival crevicular uid (56) and in the saliva. In gingival

epithelial cells, hBD2 expression is correlated with cellular differentiation and is regulated by calcium and phospholipase D (135). Cathelicidin Cathelicidin is the 18 kDa precursor of the antimicrobial peptides FALL-39 and LL-37. LL-37 is a cationic alpha-helical peptide, expressed in neutrophils, epithelial cells, saliva and gingival crevicular uid (176, 195). In addition to antibacterial activity (Table 4A), LL-37 also binds to and neutralizes lipopolysaccharide from gram-negative bacteria. In Candida albicans, the peptide causes

156

Antimicrobial peptides of the oral cavity

Table 3. Antimicrobial proteins and peptides found in gingival crevicular uid


Protein Gingival crevicular uid: healthy (lg ml) 1.8 (158) MS MS MS 0.0012 (195) MS (56) (56) Up-regulated 15-fold in aggressive periodontitis and 60-fold in chronic periodontitis, respectively (195) Gingival crevicular uid: periodontitis

Adrenomedullin HNP-1 HNP-2 HNP-3 HNP(13)

Twofold increase in periodontitis (158)

HNP-4 hBD-1 hBD-2 hBD-3 Cathelicidin LL-37 Statherin Beta-2-microglobulin

(195) MS 9.4 (174) MS MS 106 (152)

Up-regulated in aggressive periodontitis and chronic periodontitis (195)

Increased threefold in mild periodontitis and 10-fold in severe periodontitis, respectively (174)

Proline rich protein Proline rich peptide P-B Fibronectin

Decreased twofold in periodontitis and 30-fold in gingivitis (152). Less intact FN is found in periodontitis than in healthy control or treated sites (227) Increased 23-fold in periodontitis, decreased 23-fold after periodontal therapy (132, 154, 155) Increased in periodontitis, decreased 23-fold after periodontal therapy (132) Calprotectin concentration increased with the gingival index (180); 35-fold increase in periodontitis (>4 mm pocket depth) (125); 70 ng ll detected in children (172) Concentration highly variable. No consistent change with disease (2, 3, 70, 112) Main cystatin activity in gingival crevicular uid of periodontal patients (30) No change in children with gingivitis (237) 79.7 pg ll in periodontitis, increased 34-fold at 2 and 4 weeks post treatment (179)

Calgranulin A

240 (154) (132) 570 (125)

Calgranulin B Calprotectin

Lactoferrin

600

Cystatin A

MS

Cystatin C Secretory Leukocyte

1.15 (children) (237) No data

157

Gorr

Table 3. Continued
Protein Gingival crevicular uid: healthy (lg ml) Gingival crevicular uid: periodontitis

Protease Inhibitor Lactoperoxidase (Salivary Peroxidase) Myeloperoxidase Possible (14) 0.35.5 (185, 195) Peroxidase identied in gingival crevicular uid is most likely myeloperoxidase (162) No change in aggressive or chronic periodontitis (195). 660 lg ml in >5 mm pockets. Concentration decreased twofold after therapy (121) Increased in juvenile periodontitis (70) SP concentration was similar in healthy, gingivitis or periodontitis (148). Others found that concentration was decreased sevenfold post periodontal treatment (156) Concentration 20-fold lower in gingivitis, not detected in periodontitis (160)

Lysozyme C Substance P

(70, 112) 0.0610.11 (15) (148)

Calcitonin Gene Related Peptide

0.0130.7 (15, 160)

Common name, presence in gingival crevicular uid (concentration in lg ml is given when known) from healthy subjects and changes in periodontitis patients. hBD, human b-defensin; HNP, human neutrophil peptide, a-defensin; MS, proteins detected by mass spectrometry of gingival crevicular uid (159, 193).

disintegration of the plasma membrane and release of ATP, nucleotides and proteins up to 40 kDa in size (54). Histatin 1 and 3 Histatin 1 and 3 are histidine-rich, cationic peptide precursors for multiple peptides that exhibit bactericidal and candidacidal activity. Histatin 5 is derived from histatin 3. Unlike LL-37, histatin causes only minor disruption of the Candida plasma membrane and accumulates intracellularly. Thus, C. albicans is killed by histatin 5 in a mechanism that involves the nonlytic release of ATP from the fungal cells (134). However, the efux of nucleotides is similar with the two peptides and may play a signicant role for candidacidal activity (54). Histatins are found in saliva (55, 242) and are only expressed in salivary glands. The histatin genes and the statherin gene map to the same area of chromosome 4q13 as aggressive (juvenile) periodontitis and dentinogenesis imperfecta (239). Statherin The 5.4 kDa peptide, statherin, belongs to the histatin statherin family. Statherin and a basic histidine-rich peptide may have evolved from a common ancestral gene (59). Statherin is found in gingival crevicular uid (193) and saliva (55, 242, 260). The peptide is secreted by the parotid and

submandibular glands and inhibits the crystallization of calcium phosphate but also inhibits growth of anaerobic bacteria isolated from the oral cavity. It is the C-terminal peptide of statherin that exhibits the antibacterial effect (131). Statherin has recently emerged as a potential biomarker for oral infections through proteomic analysis of saliva from patients with high and low scores for bacterial adhesion and agglutination (206). C-C motif chemokine 28 This 128-amino acid peptide is preferentially expressed in various epithelial cells, including salivary glands, and is found in saliva (55). The peptide acts both as a chemokine and a broad-spectrum antimicrobial peptide. Indeed, a C-terminal 28-amino acid peptide is similar to histatin 5. This peptide induces membrane permeability and is salt sensitive, as noted for other cationic antimicrobial peptides (102). Azurocidin Azurocidin (a 37 kDa cationic antimicrobial protein, CAP37) is expressed in azurophil granules of neutrophils and was identied in human saliva by proteomic analysis (55, 264). This 251-amino acid protein is antibacterial to gram-negative bacteria, presumably because of a strong afnity for lipopolysacchaide. Two cysteine residues in positions 52 and 68 are necessary for antibacterial activity.

158

Antimicrobial peptides of the oral cavity

Table 4A. Antimicrobial activities of peptides that are directly bactericidal or bacteriostatic
Antimicrobial peptide Adrenomedullin Targets P. gingivalis S. mutans HNP-1 S. mutans P. aeruginosa C. albicans Enveloped viruses HIV-1 A. actinomycetemcomitans P. gingivalis HNP-2 P. gingivalis A. actinomycetemcomitans C. albicans Herpes simplex virus-1 HIV-1 HNP-3 P. gingivalis A. actinomycetemcomitans C. albicans Herpes simplex virus-1 HIV-1 HNP-4 E. coli C. albicans hBD-1 P. gingivalis A. actinomycetemcomitans C. albicans hBD-2 P. gingivalis S. mutans C. albicans HIV-1 hBD-3 P. gingivalis A. actinomycetemcomitans S. mutans C. albicans HIV-1 LL-37 P. gingivalis A. actinomycetemcomitans S. gordonii C. albicans Activity MIC 7.75 10)4 lg ml MIC 12.5 lg ml MIC 4.1 lg ml MIC 10.3 lg ml MIC 52.5 lg ml Inactivation Inhibits replication No activity (>500 lg ml) No activity (>200 lM) No activity (>200 lM) No activity (>500 lg ml) EI90 65 lM Inactivation Inhibits replication No activity (>200 lM) No activity (>500 lg ml) No activity Inactivation Inhibits replication LD50 0.085 lg ml LD50 0.5 lg ml MIC 50 lg ml MIC 50 lg ml MFC 7 lM MIC 34.6>250 lg ml MIC 48 lg ml MIC 559 lg ml IC50 919 lg ml MIC 42.1 lg ml MIC 45.6 lg ml MIC 35 lg ml MIC 37 lg ml IC50 2040 lg ml MIC >125 lg ml MIC 37.8 lg ml MIC 102.6 lg ml LD50 0.8 lM 113 113 113 54 113 113 117 117 157 157 157 48 267 171 197 197 171 197 48 267 197 171 143 48 267 258 258 187 187 138 117 Reference 6

159

Gorr

Table 4A. Continued


Antimicrobial peptide Histatin 5 Targets A. actinomycetemcomitans C. albicans Statherin Oral anaerobes C. albicans CCL28 S. mutans C. albicans Azurocidin CAP37 E. coli C. albicans Substance P P. aeruginosa S. mutans C. albicans Calcitonin gene related peptide Neuropeptide Y P. aeruginosa S. mutans C. albicans P. aeruginosa S. mutans C. albicans Vasoactive intestinal peptide Psoriasin Lysozyme C Peptidoglycan recognition protein 1 Peptidoglycan recognition protein 3 P. aeruginosa S. mutans C. albicans E. coli Gram-positive bacteria S. aureus E. coli C. albicans S. aureus Gram-negative bacteria C. albicans Peptidoglycan recognition protein 4 S. aureus Gram-negative bacteria C. albicans SKALP Elan Secretory leukocyte protease inhibitor P. aeruginosa P. aeruginosa Activity Neutralizes leukotoxin LD50 1.6 lM MIC <12.5 lg ml to >100 lg ml Aggregation IC50 1.7 lM IC50 0.7 lM LD50 1.3 lg ml MIC 40 lg ml MIC 15.7 lg ml MIC 171.6 lg ml MIC 8.1 lg ml MIC 5.9 lg ml MIC >500 lg ml MIC 63.1 lg ml MIC 134.3 lg ml MIC 210.9 lg ml MIC 243.2 lg ml MIC 4.1 lg ml MIC 150.7 lg ml MIC 46.5 lg ml MBC 100 lg ml Cell wall lysis LD99 60 lg ml LD99 30 No activity LD99 45 lg ml LD99 30 lg ml No activity LD99 45 lg ml LD99 200 lg ml No activity LD96 2.5 lM LF95 2.5 lM 245 245 153 245 245 153 245 245 153 213 213 168 63 63 63 8 165 63 Reference 178 54 131 115 102

Antimicrobial peptide name, microbe targets, antimicrobial activity and representative references are shown. hBD, human b-defensin; HNP, human neutrophil peptide, a-defensin; LDxx, concentration that kills XX% of bacteria; MBC, minimal bactericidal concentration; MFC, minimal fungicidal concentration; MIC, minimal inhibitory concentration.

160

Antimicrobial peptides of the oral cavity

Table 4B. Antimicrobial activities of peptides that modulate microbial adhesion or activity
AMP Mucin 7 Targets Oral streptococci A. actinomycetemcomitans C. albicans HIV GP340 salivary agglutinin DMBT1 Oral streptococci A. actinomycetemcomitans HIV-1 Inuenza A virus Surfactant protein-A Bacteria Inuenza A virus Beta-2-microglobulin Proline rich proteins S. mutans Oral bacteria C. albicans HIV-1 Fibronectin P. gingivalis S. mutans Calprotectin P. gingivalis C. albicans S. aureus Lactoferrin P. gingivalis A. actinomycetemcomitans C. albicans Cystatins Cystatin C Bacterial or host proteases P. gingivalis Herpes simplex virus-1 Cystatin S Secretory leukocyte protease inhibitor P. gingivalis P. gingivalis S. aureus HIV C. albicans Myeloperoxidase A. actinomycetemcomitans Oral streptococci P. gingivalis Bactericidal OI) 105 Bactericidal OCl) Blocks virus entry 166 173 Activity Bacterial binding and agglutination Bacterial binding Binding and clearance Prevent cell entry Agglutination Binding Blocks cell entry Inactivation by agglutination Binding and clearance Agglutination Agglutination in presence of Ca2+ Bacterial adhesion Adhesion Inhibits infectivity Binding to mbrillin Adhesion and agglutination Protects epithelial cells from infection Candidastatic by Zn chelation; MIC 18 lg ml Zn chelation, MIC 64 lg ml 35% growth inhibition at 2 mg ml apoLf 1.9 lM apoLf (iron-free) kills 99.9% in 3 h Induces apoptosis Cysteine protease inhibitor Growth inhibition Inhibits viral replication Growth inhibition Degraded by gingipain References 11 84 104 86 145 84 263 97 133 256 66 139 115 202 177 149 181 220 34 34 4 120 12 32 31 29 31 108 259

161

Gorr

Table 4B. Continued


AMP Lactoperoxidase (salivary peroxidase) Targets A. actinomycetemcomitans Oral streptococci P. gingivalis Activity Bactericidal I H2O2 Bacteriostatic OSCN Bactericidal OI) Bactericidal OI)
) )

References 107 234 105

These proteins are typically not bactericidal, with the exception of the peroxidases that produce bactericidal oxidation products. Antimicrobial peptide name, microbe targets, antimicrobial activity and representative references are shown. I), iodide; H2O2, hydrogen peroxide; MIC, minimal inhibitory concentration; OCl), hypochlorite; OI), hypoiodite; OSCN), hypothiocyanite.

Neuropeptides
The neuropeptides calcitonin gene related peptide and substance P are found in gingival crevicular uid (15). These peptides, and neuropeptide Y and vasoactive intestinal peptide, are also found in saliva (52). However, the concentrations ranging from 245 pg ml are several orders of magnitude lower than the minimal inhibitory concentrations against bacteria and C. albicans (63).

Staphylococcus aureus but not with Pseudomonas aeruginosa (119), indicating that different host-defense proteins act on different microorganisms. DMBT1 also acts as an opsonin receptor to promote bacterial clearance. The highly glycosylated protein acts as a mucin that binds inuenza virus hemagglutinin via sialic acid residues and inactivates the virus (96, 97). Entry of HIV-1 into epithelial cells is also blocked by DMBT1 (263). Surfactant protein-A This 248-amino acid protein was detected in saliva and sputum (214) and is up-regulated in patients with chronic sialadenitis (141). Surfactant protein-A acts like mucins by providing sialic acid residues that bind bacteria and the inuenza virus. In the latter, binding of hemagglutinin causes inactivation of the virus (256). Beta-2-microglobulin This 11.8 kDa protein is found in saliva (55, 242) and in gingival crevicular uid. In gingival biopsies, beta2-microglobulin is absent in most samples (71%) from normal controls and in specimens from patients with chronic severe periodontitis, but is detected in most (82%) biopsies from patients with juvenile periodontitis (224). Similarly, beta-2-microglobulin levels are higher in gingival crevicular uid from patients with periodontal than in gingival crevicular uid from healthy controls (225). However, this difference was not noted in a study of juvenile periodontitis saliva, where the beta-2-microglobulin level did not differ between patients and controls, while serum levels were higher in periodontitis patients than in controls (5). Beta-2-microglobulin causes agglutination of S. mutans in the presence of 1.4 mM Ca2+ (66). Proline-rich proteins These proteins are found in saliva (55) and bind to the tooth surface and affect bacterial adhesion. Thus, while the larger proline-rich proteins promote

Bacterial agglutination and adhesion


Mucin-7 (MUC7) The small salivary mucin MUC7 (MG2) is expressed in the mucous acinar cells of the sublingual and submandibular glands and is found in saliva (55). The 377-amino acid protein contains four potential Nglycosylation sites. Two isoforms, MG2a and MG2b, differ in their sialic acid and fucose contents (201). Mucin-7 promotes bacterial agglutination. Binding of mucin-7 to clinical isolates of A. actinomycetemcomitans depends on sialic acid residues on the mucin (84). The levels of MUC7 in stimulated saliva are twofold lower in patients with periodontitis than in healthy subjects (16.7 lg ml vs. 30.6 lg ml) (85). In addition to interacting with bacteria, MUC7 and the larger mucin, Muc5b, prevent entry of human immunodeciency virus (HIV) into host cells. Salivary agglutinin GP340 deleted in malignant brain tumors-1 (DMBT1) This large glycoprotein contains multiple scavenger receptor cysteine-rich repeats. The protein is found in saliva (55, 260, 264) and is expressed in salivary glands, ocular mucosal tissue, lacrimal glands, lung, trachea, gastointestinal tract and macrophages. DMBT1 polymorphisms have been associated with a high incidence of caries (118). The protein acts in binding and agglutination of bacteria, including oral streptococci (145). Thus, DMBT1 associates with

162

Antimicrobial peptides of the oral cavity

bacterial attachment to the pellicle, it has been suggested that smaller proline-rich proteins reduce attachment (139). Similarly, proteolytic processing of proline-rich proteins by commensal bacteria can produce peptides that limit bacterial adhesion (144). Adhesion of Streptococcus mutans correlates with high caries incidence and the presence of the Db allele of PRH1 (222). Binding of bacterial mbrillin to proline-rich protein 1 is inhibited by C-terminal mbrillin peptides corresponding to amino acid residues 266286 and 318337 (10). Proline-rich proteins, bronectin and other proteins that affect bacterial adhesion and biolm formation can delay or prevent bacterial colonization of oral surfaces. Fibronectin Fibronectin is a large glycoprotein (2386 amino acids in size) that is expressed in hepatocytes, epithelial cells and other cells, and is present in saliva (150). The protein induces bacterial agglutination and plays a role in reducing bacterial adhesion to oral surfaces (149). Fibronectin also binds directly to mbrillin from P. gingivalis and thereby inhibits the mbrillininduced expression of inammatory cytokines in macrophages (177). Low levels of bronectin are correlated with high levels of S. mutans in children (149) and periodontitis is associated with a relative lack of bronectin in adults (177).

Lactoferrin lactotransferrin This 80 kDa iron-binding glycoprotein acts as a scavenger of Fe3+ ions. Lactoferrin binds two Fe3+ ions in association with bicarbonate or another anion. It is produced by mucosal epithelial cells and is found in their secretions, including saliva (250) and gingival crevicular uid, which is a signicant source of oral lactoferrin (60). Decreased levels of lactoferrin in granulocytes have been associated with periodontal disease (219), while the saliva levels of lactoferrin appear to be increased in periodontitis (85). Gene polymorphisms of lactoferrin have recently been associated with aggressive periodontitis (262), suggesting that the antibacterial activity of this protein plays a role in the development of the disease. Lactoferrin acts on bacteria, viruses, fungi and parasites. Lactoferrin kills A. actinomycetemcomitans (120), inhibits the growth of P. gingivalis but has no effect on the growth of Prevotella intermedia and Prevotella nigrescens (4). However, lactoferrin inhibits the adhesion of P. intermedia, P. nigrescens and A. actinomycetemcomitans to broblasts (9). In addition to its effects on periodontal pathogens, the protein induces apoptosis in C. albicans in a process that depends on cellular K+-channels (12). Lactoferrin also binds to the lipid A part of lipopolysaccharide, resulting in both anti-inammatory activity and bactericidal activity as a result of increased membrane permeability (184).

Metal ion chelators


Calgranulin A (S100A8) and calgranulin B (S100A9); calprotectin These metal ion-binding proteins inhibit bacterial growth by acting as divalent cation scavengers. The dimer of calgranulin A and B, termed calprotectin, is expressed in the cytosol of neutrophils, monocytes and keratinocytes. The expression levels of calgranulin A and B are up-regulated in oral epithelial H400 cells exposed to P. gingivalis or Fusobacterium nucleatum (170). Accordingly, calprotectin is upregulated in periodontal disease and is detected at increased levels in the gingival crevicular uid of patients with periodontitis (125). This is probably a defense mechanism because the expression of calprotectin in KB (HeLa) cells protects the cells from invasion with P. gingivalis (181). Calprotectin levels are also increased in saliva from patients with candidiasis (129). Calprotectin inhibits the growth of S. aureus by chelation of Mn2 + and Zn2 + ions, which leads to reprogramming of the bacterial transcriptome (47).

Protease inhibitors
Cystatins The human cystatin gene family contains 14 genes and two pseudogenes. Seven of these genes are expressed in saliva (Table 2). The cystatins are cysteine protease inhibitors that block the action of bacterial proteases on target tissues (58). Based on structural similarities it has recently been proposed that cathelicidin gradually evolved from cystatins (270). Indeed, Cystatins C and S inhibit the growth of P. gingivalis (31). Secretory leukocyte protease inhibitor This 11.7 kDa cationic, nonglycosylated protein contains 16 cysteine residues that form eight disulde bridges. The protein is expressed in mucosal epithelial cells and secretions, including gingival keratinocytes and saliva. Secretory leukocyte protease inhibitor acts as a bacterial serine protease inhibitor and is an antibacterial and anti-inammatory protein

163

Gorr

(252). The N-terminal cationic domain of secretory leukocyte protease inhibitor is antimicrobial with activity against P. aeruginosa, S. aureus and C. albicans (259). Secretory leukocyte protease inhibitor expression in oral epithelial cells is induced by HIV-1 (110) and the protein blocks the entry of HIV-1 by interaction with the host cell. This activity appears to be independent of the protease inhibitor activity. Secretory leukocyte protease inhibitor expression is decreased in chronic periodontitis, and the gingival crevicular uid levels of secretory leukocyte protease inhibitor are lower in P. gingivalis-infected sites, possibly because of secretory leukocyte protease inhibitor degradation by arginine-specic gingipains from P. gingivalis (108). SKALP Elan Skin-derived antileukoproteinase (SKALP) Elan is a 12 kDa protein expressed in the human submandibular gland (142) and in saliva (142, 235). The protein has an N-terminal domain that acts as a transglutaminase substrate and a C-terminal domain that exhibits anti-elastase activity. In addition, the protein kills both gram-negative and gram-positive bacteria; this activity depends on the presence of both peptide domains (213). Peroxidases Lactoperoxidase (salivary peroxidase) is a heme peroxidase that is found in saliva and milk. Lactoperoxidase and myeloperoxidase form the principal components of the peroxidase system of saliva (106). The enzymes catalyse the oxidation of thiocyanate ions (SCN)) by hydrogen peroxide, and the reaction product hypothiocyanite (OSCN)) is bactericidal. (14). Hydrogen peroxide-mediated oxidation of chloride and iodide produces further bactericidal reaction products (105, 173). Having a similar action to lactoperoxidase, myeloperoxidase (a heme peroxidase) is expressed in neutrophilic polymorphonuclear leukocytes and catalyses the oxidation of thiocyanate ions by hydrogen peroxide to produce hypothiocyanite. In addition, myeloperoxidase can produce hypochlorite (OCl)), which is a stronger oxidant implicated in inammatory tissue damage (14). Myeloperoxidase is found in gingival crevicular uid at a concentration of about 5 lg ml but no signicant differences were found between the levels of myeloperoxidase found in chronic periodontitis, aggressive periodontitis and healthy controls (195). However, the myeloperoxidase levels in gingival crevicular uid were signicantly reduced in periodontal patients after antibiotic treatment for 3 months (121).

Activity against bacterial cell walls


Lysozyme 1,4-Beta-N-acetylmuramidase (lysozyme) is a bacteriolytic enzyme that hydrolyses the (1, 4)-betalinkages between N-acetylmuramic acid and N-acetyl-D-glucosamine residues in peptidoglycans. The enzyme is mainly active against the cell wall of grampositive bacteria. The 14 kDa protein is expressed widely in mucosal epithelia and is found in saliva, milk and tears. Lysozyme is found in gingival crevicular uid. The lysozyme levels do not appear to differ between adult periodontitis and healthy controls, (225) but are signicantly increased in juvenile periodontitis (70). Peptidoglycan recognition proteins 3 and 4 These large host-defense proteins (89115 kDa disulde-linked homodimers or heterodimers) are found in species ranging from insects to mammals (61). Interestingly, different peptidoglycan-recognition proteins have diversied to carry out different antimicrobial functions (205). Four peptidoglycanrecognition proteins are found in humans: peptidoglycan recognition proteins 3 and 4 are expressed in skin and mucosal epithelia, including salivary glands, and peptidoglycan recognition protein 1 is found in saliva. Peptidoglycan recognition protein 2 is an amidase that is secreted by the liver and epithelial cells. The proteins exert their bactericidal effect by binding to cell wall peptidoglycans, but do not permeabilize bacterial membranes (153). The proteins are bactericidal for pathogenic and nonpathogenic gram-positive bacteria but not for normal ora bacteria. Moreover, the proteins are bacteriostatic for most gram-positive and gram-negative bacteria but not for C. albicans (153).

The role of antimicrobial proteins in periodontal disease


Striking differences in susceptibility to the antimicrobial peptides LL-37 and hBD3 have been observed between different species of oral bacteria and different strains of the same species (113). As an example, Streptococcus gordonii M5 is weakly susceptible to both antimicrobial peptides, while S. gordonii 10558 is highly susceptible. P. gingivalis 33277, by contrast, is weakly susceptible to LL-37 but highly susceptible to killing by hBD3 (113) (Table 4A). Indeed, different oral bacteria elicit distinct transcription proles in oral epithelial cells (90), thus further diversifying the

164

Antimicrobial peptides of the oral cavity

antimicrobial peptide response to bacteria. The production of beta-defensins, and LL-37, in particular, has been analyzed after exposure of gingival epithelial cells to bacteria associated with periodontitis (Table 1). Approximately 20 genetic diseases connected with periodontitis have been identied (92). Some of these diseases are associated with changes in antimicrobial peptide expression, which may play a role in the increased susceptibility to bacterial infection. Morbus Kostman disease is a severe congenital neutropenia that is associated with severe periodontitis (196). Patients with morbus Kostmann are decient in LL-37 in neutrophils, plasma and saliva. Patients are also decient in alpha-defensins (30% of normal) while the plasma lactoferrin content is normal (196). Patients treated with granulocytecolony stimulating factor exhibit normal numbers of neutrophils but lack LL-37 and continue to show periodontal disease (40, 196). In a single patient, a bone marrow transplant restored both LL-37 and neutrophils to normal levels and no dental problems were noted. While this study suggests that a lack of LL-37 is associated with periodontal disease, normal levels of LL-37 are not sufcient to prevent the disease. Thus, periodontal disease is prevalent in children with Downs syndrome (trisomy 21) (183). However, this increased occurrence of periodontitis is not correlated with a decrease in LL-37 levels (16). Other antimicrobial peptides associated with periodontal disease are mucin-7 and lactoferrin. The level of mucin-7 (MG2) is decreased threefold in patients with A. actinomycetemcomitans-associated periodontitis compared with healthy controls. Lactoferrin levels were normal but the protein was iron saturated, suggesting that its antimicrobial properties are reduced in patients with periodontitis (85). ` vre syndrome and HaimMunk synPapillonLefe drome are characterized by palmoplantar keratoderma and severe periodontitis. Both diseases are caused by allelic mutations of the cathepsin C gene, CTSC (94). The lysosomal enzyme cathepsin C is responsible for the activation of neutrophil serine proteases, which are antibacterial to S. aureus and A. actinomycetemcomitans (93, 95, 192, 223). While ` vre syndrome express patients with PapillonLefe normal levels of the cathelicidin precursor, little is processed to the mature LL-37 peptides. As in morbus Kostman, it is likely that the low levels of LL-37 contribute to the development of periodon` vre syndrome titis in patients with PapillonLefe (53).

Antimicrobial proteins and other oral infections


Oral infections other than periodontitis have also been linked to the levels of expression of antimicrobial peptides. Oral candidiasis is associated with low levels of several antimicrobial peptides, including lactoferrin, and beta-defensins 1 and 2. By contrast, the concentration of alpha-defensin 1 increased in patients, whereas the concentration of transferrin did not differ between patients and healthy controls (228). The correlation of antimicrobial peptide expression levels and caries incidence has been difcult to establish. Caries incidence in children has been associated with the expression of a low-level of alphadefensins (human neutrophil peptides 13) (229). However, because caries is seen at a wide range of alpha-defensin expression levels, it is not clear if alpha-defensin expression is predictive for future caries development (50). Furthermore, the hostdefense proteins lysozyme, salivary peroxidase and lactoferrin were not correlated with caries incidence in studies of 13-year-old children (128) and navy recruits (82). The difculty in linking single point analysis of antimicrobial peptides with oral disease (231) may be a result of the wide variety of antimicrobial peptide expression levels between subjects. Salivary peptide levels can vary about 100-fold between subjects, even when normalized to total salivary protein (229). Thus, it is difcult to dene normal values for individual antimicrobial peptides in saliva. Instead, it may be possible to use multiplex analysis of antimicrobial protein expression in health and disease to identify antimicrobial peptide signatures that will have predictive or diagnostic power.

New antimicrobial proteins


Many types of antimicrobial peptides have been identied in the oral cavity, as described in the preceding sections. With the advent of saliva and gingival crevicular uid proteomics it is predicted that additional antimicrobial peptides will be identied. It should be noted that not all of these peptides will be present at biologically signicant concentrations. Nevertheless, identication of antimicrobial peptides in saliva provides a starting point for the study of their potential role in host defense. In one study, proteomic analysis of labial salivary gland secretions identied 56 proteins, 32% of which were characterized as host-defense proteins (215). The

165

Gorr

antimicrobial peptides listed in the preceding sections were extracted from a data set of 633 known salivary proteins described in PubMed. A recent comprehensive analysis of the human glandular salivary proteome, supported by the National Institute of Dental and Craniofacial Research, at the US National Institutes of Health, has identied 1166 total proteins: 914 were found in parotid saliva and 917 were found in submandibular sublingual saliva (55). About 25% of these proteins were hypothetical and lacked annotations. It is possible that additional antimicrobial peptides will be detected in this data set. As in the case of the palate, lung, nasal epithelium clone (PLUNC) proteins, described below, sequencesequence analyses, structural prediction (75, 81) and analysis for antimicrobial consensus motifs in peptide sequences (265) can be used to identify potential new antimicrobial peptides.

Bactericidal permeabilityincreasing protein-like proteins


Recent proteomic analyses of saliva have identied bactericidal permeability-increasing protein and several potential antimicrobial proteins related to bactericidal permeability-increasing protein (Table 5). Bactericidal permeability-increasing protein is a 55 kDa cationic protein expressed in polymorphonuclear leukocytes where it is stored in azurophilic granules (38, 65). The protein is also expressed in mucosal epithelia (39), and its presence in saliva suggests that it may have a functional role in the oral cavity. Bactericidal permeability-increasing protein acts as an anti-inammatory protein by inhibiting the binding of lipopolysaccharide to lipopolysaccharidebinding protein. As both bactericidal permeabilityincreasing protein and lipopolysaccharide-binding protein bind lipopolysaccharide, it is probable that common structures in these proteins interact with lipopolysaccharide, although with opposing effects (109). Bactericidal permeability-increasing protein is active against gram-negative bacteria and neutralizes their lipopolysaccharides (65). Bactericidal permeability-increasing protein contains two functional domains, namely BPI1 and BPI2. The protein exhibits opsonic activity via binding of the N-terminal BPI1 domain to bacteria and binding of the Cterminal BPI2 domain to neutrophils and monocytes (38).

Bactericidal permeability-increasing protein belongs to the bactericidal permeability-increasing protein superfamily, which consists of lipopolysaccharide-binding protein, cholesteryl ester transport protein and phospholipid transport protein. More recently, 10 genes related to bactericidal permeability-increasing protein have been identied on human chromosome 20q11 (nine genes) and chromosome 22q12 (one gene; BPIL2). These are termed the PLUNC proteins after an early identied member (2628, 126, 175). Several of these proteins are found in saliva (Table 5). The PLUNC proteins are divided into the short PLUNCs (SPLUNC), which are similar to the N-terminal BPI1 antibacterial domain of bactericidal permeability-increasing protein, and the long PLUNCs (LPLUNC), which exhibit similarity to both BPI1 and BPI2 domains (126). The sequence identity of bactericidal permeability-increasing protein and the PLUNC proteins is low (<25%), consistent with observations for other proteins related to the bactericidal permeability-increasing protein family (20). However, bactericidal permeability-increasing protein and the PLUNC proteins (except for BASE) contain conserved Cys residues that form a disulde bridge in bactericidal permeability-increasing protein. The disulde bridge has not been veried in parotid secretory protein, but a parotid secretory protein mutant lacking the Cys residues is not secreted, suggesting that its structure is signicantly altered (C. Geetha and S.-U. Gorr, unpublished data). Several of these human proteins have homologs in other species, including rat and mouse parotid secretory protein (17, 161), bovine Bsp30, PLUNC and Von Ebner minor salivary gland protein (198, 254), and mouse PLUNC (253). Although the existence of some of these proteins has been known for over 20 years, it was the identication of mouse PLUNC (253) and the sequencing of human chromosome 20 that triggered the current interest in these proteins (27). The PLUNC proteins are primarily expressed in oral and airway epithelia and it is thought that they play antimicrobial roles in these tissues. The difference in expression between different PLUNC proteins suggests that each occupies an epithelial niche (126). Latherin (breast cancer and salivary gland expression gene) is a member of the PLUNC protein family (27) that is specically expressed in normal salivary glands, as determined by messenger RNA hybridization of 61 human tissues (62). Surprisingly, latherin was not detected in proteomic studies of saliva (55,

166

Antimicrobial peptides of the oral cavity

Table 5. Bactericidal permeability-increasing protein-like proteins in saliva


Protein Protein Plunc Bactericidal permeabilityincreasing protein-like 1 Bactericidal permeabilityincreasing protein-like 2 Bactericidal permeabilityincreasing protein-like 3 Long palate, lung and nasal epithelium carcinomaassociated protein 1 Long palate, lung and nasal epithelium carcinomaassociated protein 3 Long palate, lung and nasal epithelium carcinomaassociated protein 4 Short palate, lung and nasal epithelium carcinomaassociated protein 2 (parotid secretory protein) (PSP) Short palate, lung and nasal epithelium carcinomaassociated protein 3 Latherin (BASE) Gene PLUNC BPIL1 Accession Q9NP55 Q8N4F0 Parotid saliva X X SMG SLG saliva X X Whole saliva X1 X1,2

BPIL2

Q8NFQ6

BPIL3

Q8NFQ5

LPLUNC1

Q8TDL5

X1,2

LPLUNC3

P59826

LPLUNC4

P59827

SPLUNC2

Q96DR5

X1,2

SPLUNC3

Q9BQP9

LATH

Q86YQ2

Protein names, Gene symbols and accession numbers are from the UniProt database. Parotid and submandibular gland (SMG) sublingual gland (SLG) saliva data are from a previous publication (55). Whole saliva data are from 1(264); and 2(260). X indicates proteins that are found in saliva. None of the proteins listed have been found in gingival crevicular uid.

264). It is possible that the unusual surface-adhesion properties of latherin prevented its detection (23). To date, none of the PLUNC proteins have been detected in gingival crevicular uid (193).

The predicted structural similarity of PLUNC proteins and bactericidal permeability-increasing protein, suggested that the PLUNC proteins could play a role as antibacterial and or anti-inammatory

167

Gorr

host-defense proteins (26, 76). Consistent with this proposal, we found that parotid secretory protein expression in human gingival epithelial cells is induced by P. gingivalis, tumor necrosis factor-alpha and estradiol (210). Mouse parotid secretory protein binds bacteria (203), and human parotid secretory protein exhibits moderate antibacterial activity to P. aeruginosa (76). Similarly, PLUNC shows moderate activity against Mycoplasma pneumoniae (6070% killing in 2 h) (41) and one group reported 6070% killing of P. aeruginosa (268, 269), while others failed to detect killing of P. aeruginosa, Escherichia coli or Listeria monocytogenes in 3-h kill assays based on colony-forming unit counts (19). Importantly, none of these experiments report more than 90% (one order of magnitude) killing. Thus, it remains to be determined if the bactericidal permeabilityincreasing protein-like proteins function as bactericidal proteins in vivo. Indeed, we have recently found that a parotid secretory protein-derived peptide causes bacterial agglutination and lowers colonyforming unit counts in bacterial killing assays (81). However, the peptide did not result in decreased bacterial viability in biochemical assays, suggesting that the reduced colony-forming unit counts could be attributed to bacterial agglutination. Based on these results, we speculate that parotid secretory protein is functionally related to salivary agglutinin (DMBT1) and to surfactant protein D. As such, parotid secretory protein could play a role in bacterial binding, agglutination and clearance by phagocytic cells (81). Consistent with the proposed bacterial binding function of bactericidal permeability-increasing protein-like proteins, the members of this family bind lipids and lipopolysaccharide. Similarly, human PLUNC (77) and parotid secretory protein (S.-U. Gorr et al., unpublished data) bind immobilized lipopolysaccharide. Immobilized BSP30, by contrast, does not bind soluble lipopolysaccharide (87) but it is not clear if this is a result of structural or methodological differences. While PLUNC does not compete with lipopolysaccharide-binding protein for lipopolysaccharide binding (37), the identication of anti-inammatory peptides in the parotid secretory protein sequence (75) suggests that parotid secretory protein may play a role in neutralizing lipopolysaccharide at mucosal surfaces. The relatively recent identication of human parotid secretory protein and its related proteins in human saliva, and the large number of unknown proteins present in saliva, suggest that more potential antimicrobial proteins can be identied in this biouid.

Peptides derived from host-defense proteins


Antimicrobial peptides constitute an interesting new class of compounds that can augment a decreasing arsenal of effective antibiotics to many bacterial species and fungi. Microbial resistance to antibiotics was recognized early and the problem was pointed out in Flemings 1945 Nobel lecture (68). As resistance to known antibiotics is on the rise, the search for new classes of antibiotics continues to be of importance (80). The development of new antibiotics based on host-defense proteins is a particularly attractive option (88). Antimicrobial peptides have functioned in host defense for millions of years and it is thought that the co-evolution of antimicrobial peptides and pathogens is a factor in the relatively low resistance of bacteria to these compounds (191). To date, over 1000 antimicrobial peptides have been identied and are accessible in online databases (33, 42, 211, 244, 247) (http://www.bbcm.univ.trieste. it/~tossi/pag1.htm). A comprehensive list of known and potential antimicrobial peptides has also been reported: http://aps.unmc.edu/AP/main.php. This list contains over 1100 peptides. These databases are a rich resource for the identication of clinically useful peptides that could exhibit both high in vivo efcacy and low host toxicity. Both factors have been concerns in clinical trials of antimicrobial peptides (80). While naturally occurring host-defense proteins are effective endogenous antimicrobial agents, often their size, folding and other physicochemical properties make them less ideal as synthetic therapeutics. Instead, the active regions of several host-defense proteins have been identied and peptides based on these regions have been designed. Examples of such antimicrobial peptides derived from human salivary proteins are provided below. The sequences of representative peptides from each parent protein are listed in Table 6.

Bactericidal permeability-increasing protein


Bactericidal permeability-increasing protein exhibits both antibacterial and anti-inammatory activities (251). A recombinant N-terminal fragment (rBPI21) retains the biological activity of bactericidal permeability-increasing protein and has been evaluated

168

Antimicrobial peptides of the oral cavity

Table 6. Examples of antimicrobial peptides derived from salivary host-defense proteins (parent protein) (see the text for functional details and references)
Parent protein BPI MUC7 Histatin Defensin Cathelicidin Lactoferrin DMBT1 PSP Peptide XOMA 629 12-mer-L Hsn5 12-mer 2Abz S
23 29

Sequence KLFR-[3-(1-naphthyl)-A]-QAK-[3-(1naphthyl)-A]-NH2 RKSYKCLHKRCR AKRHHGYKRKFH RYGTC(Acm)IYQ-2Abz-RLWAFS KLFKRIVKRILKFLRKLV TKCFQWQRNMRKVRGPPVSCIKR QGRVEVLYRGSWGTVC GQIINLKASLDLL-NH2

LL-37 K-18-mer Lfpep SRCRP2 GL13-NH2

BPI, bactericidal permeability increasing protein; DMBT1, deleted in malignant brain tumor 1,salivary agglutinin; MUC7, mucin 7; PSP, parotid secretory protein.

in clinical trials (78). The bactericidal permeabilityincreasing protein molecule has also served as a template for the design of anti-inammatory peptides. Chimeric peptides consisting of bactericidal permeability-increasing protein and the Limulus anti-lipopolysaccharide factor were especially effective in blocking lipopolysaccharide action in vitro and in vivo (51).

exhibits candidacidal activity (204) and is bactericidal against several species of oral bacteria (248).

Defensin
The defensin family of antimicrobial peptides consists of alpha and beta (and theta) defensins, which are characterized by the location of three characteristic disulde bonds (72). Chemical synthesis of defensins is complicated by the correct formation of disulde pairs. Truncated alpha-defensins have been produced to overcome this obstacle. While linear peptides were not antimicrobial (163), the incorporation of a 2-aminobenzoyl derivative restored antimicrobial activity (157). The most promising of these truncated peptides, 2Abz23S29, killed both grampositive and gram-negative bacteria with minimal inhibitory concentrations of 210-fold those of the native alpha-defensin 1 peptide (157).

Mucins
The small salivary mucin MUC7 (MG2, 357 amino acid residues) is a host-defense protein with a role in binding and clearance of microorganisms. Peptides (1250 amino acid residues) from the N-terminal domain of MUC7 exhibit antifungal activities (216). The optimal 12-mer peptide also exhibits activity against oral bacteria in either planktonic growth or in biolms (248, 249). A 23-amino acid prolinerich tandem repeat, when used at concentrations of 1.53.0 lM, exhibits bactericidal activity to the oral bacteria A. actinomycetemcomitans, P. gingivalis, S. gordonii and S. mutans (13).

Lactoferrin
Proteolytic digestion of lactoferrin produces lactoferricin, which has been used as a template to model pharmacologically effective antimicrobial peptides with high antibacterial activity (98). Two additional antimicrobial peptides derived from lactoferrin have recently been described. Lfpep (Lf 1840) has a net charge of +7, while Kaliocin-1 (Lf 153183) has a net charge of +1. The latter peptide is highly similar to the frog antimicrobial peptide brevinin-1Sa (241). Both peptides kill C. albicans, but only Lfpep is able to permeabilize the cell membrane (241). Lactoferricin B induced resistance in S. aureus (208), raising discussion over the use of this or similar peptides in human therapy (24, 89).

Histatins
The histatin family is a naturally rich source of antimicrobial peptides found in the oral cavity but not in the airways (57). Histatins are active against C. albicans and other fungi, which they kill by nonlytic release of ATP (243). Proteomic analysis of glandular saliva has identied a vast array of histatin peptides (91). Several histatin 3 peptides were newly identied and one of these also exhibited anticandidal activity. A histatin 5 12-mer peptide (P113)

169

Gorr

Salivary agglutinin (DMBT1)


A consensus peptide based on the SRCR domain of DMBT1 was found to cause bacterial agglutination and block the agglutination induced by DMBT1. Thus, this peptide captures an important biological activity of the native protein. The bacteria agglutinated by this peptide include several oral bacteria such as S. gordonii (25).

Parotid secretory protein


Based on the predicted structural similarity of parotid secretory protein and bactericidal permeabilityincreasing protein, and the location of active peptides in the bactericidal permeability-increasing protein sequence, we identied potential antimicrobial sequences in parotid secretory protein. The peptides designed initially inhibited the binding of lipopolysaccharide to lipopolysaccharide-binding protein and inhibited lipopolysaccharide-stimulated secretion of tumor necrosis factor-alpha from macrophages (75). Recent results suggest that a modied parotid secretory protein peptide is also active against lipopolysaccharide in a mouse model of sepsis (S. U. Gorr, unpublished data). The peptide GL-13 caused agglutination of both gram-positive and gram-negative bacteria in a dose-dependent manner. Agglutination was not affected by salt concentration, but nonionic detergent reduced agglutination, suggesting that hydrophobic interactions play a role. Agglutination limited the spread of P. aeruginosa infection in Romaine lettuce leaves and increased the uptake of P. aeruginosa by macrophages (81).

siderable challenge, as evidenced by the limited number of antimicrobial peptides in clinical trials. A promising alternative to natural or synthetic peptides is the use of peptide mimetics that capture the biological activity of host-defense peptides but in a chemical format that is more readily produced and delivered (232). Mimetics based on the structure of defensin have shown a high therapeutic index in preclinical studies (22). Similarly, XOMA 629 is a modied D-enantiomer peptide derived from functional domain II of human bactericidal permeability-increasing protein (amino acid residues 6599) and is highly active against a wide variety of bacteria and fungi (146). Given the large variety of antimicrobial peptides, it may be possible to design both broad-range mimetics and some that are limited to specic infectious agents without the unintended elimination of benecial commensals.

Clinical applications
The promise of antimicrobial peptides lies in their broad efcacy and potentially low rates of induced resistance resulting from the co-evolution of pathogens and host antimicrobial peptides. Indeed, a number of antimicrobial peptides or synthetic derivatives have reached clinical trials (Table 7). However, a recent review indicates that the results have been mixed and none has been approved by the US Food and Drug Administration (80). The most promising antimicrobial peptide appears to be the histatin 5 12-mer P113 (Demegen), which completed Phase II clinical trials as a mouth rinse for oral candidiasis in patients with HIV. The bactericidal permeability-increasing protein derivative XOMA 629 (XOMA) is in Phase 2a trials for the skin infection impetigo, while the defensin mimetic PMX-30063 (Polymedix Inc.) has passed Phase I safety evaluation.

Antimicrobial peptide mimetics


The synthesis, purication, delivery and in vivo efcacy of antimicrobial peptides remain areas of conTable 7. Clinical trials of antimicrobial peptides
Type BPI peptide Histatin 5 Defensin mimetic Lactoferrin Short fatty acid Compound XOMA629 P113 PMX-30063 hLf1-11 Sodium butyrate

Function Antibacterial Antifungal Antibacterial Antimicrobial Stimulate LL-37 and hBD-1 expression

Condition Impetigo Candidiasis Infection Infections in transplant recipients Shigellosis

Reference XOMA Demegen Polymedix NCT00430469 NCT00800930

Antimicrobial peptides discussed in the text that have been or are in clinical trials are listed. The References are the ClinicalTrials.com ID number or the name of the company developing the drug. BPI, bactericidal permeability-increasing protein.

170

Antimicrobial peptides of the oral cavity

While the initial promise of this class of antimicrobials remains to be conrmed, continued efforts in this area appear justied as resistance to existing antibiotics and emerging infections are an ongoing problem (80, 226). As an alternative to antimicrobial peptide therapy, the stimulation of expression of endogenous antimicrobial peptides may be used to ght infections. As an example, reduced intestinal levels of LL-37 and betadefensin-1 are associated with shigellosis. In a rabbit model of the disease, induction of CAP-18 (the rabbit homologue of LL-37) with sodium butyrate reduced clinical illness and the bacterial load in the stool (200). A clinical trial is underway to determine if butyrate treatment is effective in human shigellosis (Table 7). By using similar approaches it may be possible to regulate the expression of antimicrobial peptides in oral tissues, saliva and gingival crevicular uid.

References
1. Abiko Y, Saitoh M, Nishimura M, Yamazaki M, Sawamura D, Kaku T. Role of b-defensins in oral epithelial health and disease. Med Mol Morphol 2007: 40: 179184. 2. Adonogianaki E, Mooney J, Kinane DF. Detection of stable and active periodontitis sites by clinical assessment and gingival crevicular acute-phase protein levels. J Periodontal Res 1996: 31: 135143. 3. Adonogianaki E, Moughal NA, Kinane DF. Lactoferrin in the gingival crevice as a marker of polymorphonuclear leucocytes in periodontal diseases. J Clin Periodontol 1993: 20: 2631. 4. Aguilera O, Andres MT, Heath J, Fierro JF, Douglas CW. Evaluation of the antimicrobial effect of lactoferrin on Porphyromonas gingivalis, Prevotella intermedia and Prevotella nigrescens. FEMS Immunol Med Microbiol 1998: 21: 2936. 5. Akalin FA, Bulut S, Yavuzyilmaz E beta 2-Microglobulin levels in serum and saliva of patients with juvenile periodontitis. J Nihon Univ Sch Dent 1993: 35: 230234. 6. Allaker RP, Zihni C, Kapas S. An investigation into the antimicrobial effects of adrenomedullin on members of the skin, oral, respiratory tract and gut microora. FEMS Immunol Med Microbiol 1999: 23: 289293. 7. Allgrove JE, Gomes E, Hough J, Gleeson M. Effects of exercise intensity on salivary antimicrobial proteins and markers of stress in active men. J Sports Sci 2008: 26: 653 661. 8. Almeida RP, Vanet A, Witko-Sarsat V, Melchior M, McCabe D, Gabay JE. Azurocidin, a natural antibiotic from human neutrophils: Expression, antimicrobial activity, and secretion. Prot Expr Purif 1996: 7: 355366. 9. Alugupalli KR, Kalfas S. Characterization of the lactoferrindependent inhibition of the adhesion of Actinobacillus actinomycetemcomitans, Prevotella intermedia and Prevotella nigrescens to broblasts and to a reconstituted basement membrane. APMIS 1997: 105: 680688. 10. Amano A, Sharma A, Lee JY, Sojar HT, Raj PA, Genco RJ. Structural domains of Porphyromonas gingivalis recombinant mbrillin that mediate binding to salivary proline-rich protein and statherin. Infect Immun 1996: 64: 16311637. 11. Amerongen AV, Bolscher JGM, Veerman ECI. Salivary mucins: protective functions in relation to their diversity. Glycobiology 1995: 5: 733740. 12. Andres MT, Viejo-Diaz M, Fierro JF. Human lactoferrin induces apoptosis-like cell death in Candida albicans: Critical role of K+-channel-mediated K+-efux. Antimicrob Agents Chemother 2008: 52: 40814088. 13. Antonyraj KJ, Karunakaran T, Raj PA. Bactericidal activity and poly-L-proline II conformation of the tandem repeat sequence of human salivary mucin glycoprotein (MG2). Arch Biochem Biophys 1998: 356: 197206. 14. Ashby MT. Inorganic chemistry of defensive peroxidases in the human oral cavity. J Dent Res 2008: 87: 900 914. 15. Awawdeh LA, Lundy FT, Linden GJ, Shaw C, Kennedy JG, Lamey P-J. Quantitative analysis of substance P, neurokinin A and calcitonin gene-related peptide in gingival

Conclusions
A broad array of antimicrobial proteins and peptides with diverse functions has been identied in oral tissues and uids. Most of the antimicrobial peptides found in gingival crevicular uid are also found in saliva, while saliva contains additional (as yet undetected) antimicrobial peptides in gingival crevicular uid. These antimicrobial peptides play a protective role, and their diversity is thought to protect against the many microbes entering the oral cavity and to combine effectively to ght individual organisms. Several antimicrobial peptides are active against the main bacteria associated with periodontal disease, and the absence of antimicrobial peptides has been linked with inherited forms of periodontitis. It is predicted that additional new antimicrobial peptides will be detected in oral tissue and uids and serve as templates for the design of effective antibiotics against oral microbes.

Acknowledgments
The authors work on parotid secretory protein was supported by grant number 2 R01 DE012205 from the National Institute for Dental and Craniofacial Research at the National Institutes of Health. Work on antimicrobial PSP peptides was supported by a Proof of Concept Grant from the Ofce of the Executive Vice President for Research of the University of Louisville. The University of Louisville School of Dentistry is gratefully acknowledged for support.

171

Gorr
crevicular uid associated with painful human teeth. Eur J Oral Sci 2002: 110: 185191. Bachrach G, Chaushu G, Zigmond M, Yefenof E, Stabholz A, Shapira J, Merrick J, Chaushu S. Salivary LL-37 secretion in individuals with Down syndrome is normal. J Dent Res 2006: 85: 933936. Ball WD, Hand AR, Moreira JE, Iversen JM, Robinovitch MR. The B1-immunoreactive proteins of the perinatal submandibular gland: similarity to the major parotid gland protein, RPSP. Crit Rev Oral Biol Med 1993: 4: 517524. Baron AC, Gansky SA, Ryder MI, Featherstone DB. Cysteine protease inhibitory activity and levels of salivary cystatins in whole saliva of periodontally diseased patients. J Periodontal Res 1999: 34: 437444. Bartlett JA, Hicks BJ, Schlomann JM, Ramachandran S, Nauseef WM, McCray PB Jr. PLUNC is a secreted product of neutrophil granules. J Leukoc Bid 2008: 83: 12011206. Beamer LJ, Fischer D, Eisenberg D. Detecting distant relatives of mammalian LPS-binding and lipid transport proteins. Protein Sci 1998: 7: 16431646. Becerra L, Soares RV, Bruno LS, Siqueira CC, Oppenheim FG, Offner GD, Troxler RF. Patterns of secretion of mucins and non-mucin glycoproteins in human submandibular sublingual secretion. Arch Oral Biol 2003: 48: 147154. Beckloff N, Laube D, Castro T, Furgang D, Park S, Perlin D, Clements D, Tang H, Scott RW, Tew GN, Diamond G. Activity of an antimicrobial peptide mimetic against planktonic and biolm cultures of oral pathogens. Antimicrob Agents Chemother 2007: 51: 41254132. Beeley JG, Eason R, Snow DH. Isolation and characterization of latherin, a surface-active protein from horse sweat. Biochem J 1986: 235: 645650. Bell G, Gouyon P-H. Arming the enemy: the evolution of resistance to self-proteins. Microbiology 2003: 149: 1367 1375. Bikker FJ, Ligtenberg AJM, Nazmi K, Veerman ECI, vant Hof W, Bolscher JGM, Poustka A, Amerongen AVN, Mollenhauer J. Identication of the nacteria-binding peptide domain on salivary agglutinin (gp-340 DMBT1), a member of the scavenger receptor cysteine-rich superfamily. J Biol Chem 2002: 277: 3210932115. Bingle CD, Craven CJ. PLUNC: a novel family of candidate host defence proteins expressed in the upper airways and nasopharynx. Hum Mol Genet 2002: 11: 937943. Bingle CD, Gorr S-U. Host defense in oral and airway epithelia: chromosome 20 contributes a new protein family. Intern J BiochemCell Biol 2004: 36: 21442152. Bingle CD, LeClair EE, Havard S, Bingle L, Gillingham P, Craven CJ. Phylogenetic and evolutionary analysis of the PLUNC gene family. Protein Sci 2004: 13: 422430. Bjorck L, Grubb A, Kjellen L. Cystatin C, a human proteinase inhibitor, blocks replication of herpes simplex virus. J Virol 1990: 64: 941943. Blankenvoorde MF, Henskens YM, van der Weijden GA, van den Keijbus PA, Veerman EC, Nieuw Amerongen AV. Cystatin A in gingival crevicular uid of periodontal patients. J Periodontal Res 1997: 32: 583588. Blankenvoorde MF, vant Hof W, Walgreen-Weterings E, van Steenbergen TJ, Brand HS, Veerman EC, Nieuw Amerongen AV. Cystatin and cystatin-derived peptides have antibacterial activity against the pathogen Porphyromonas gingivalis. Biol Chem 1998: 379: 13711375. 32. Bobek LA, Levine MJ. Cystatins inhibitors of cysteine proteinases. Crit Rev Oral Biol Med 1992: 3: 307332. 33. Brahmachary M, Krishnan SPT, Koh JLY, Khan AM, Seah SH, Tan TW, Brusic V, Bajic VB. ANTIMIC: a database of antimicrobial sequences. Nucl Acids Res 2004: 32: D586 D589. 34. Brandtzaeg P, Gabrielsen TO, Dale I, Muller F, Steinbakk M, Fagerhol MK. The leucocyte protein L1 (calprotectin): a putative nonspecic defence factor at epithelial surfaces. Adv Exp Med Biol 1995: 371A: 201206. 35. Brissette CA, Pham TT, Coats SR, Darveau RP, Lukehart SA. Treponema denticola does not induce production of common innate immune mediators from primary gingival epithelial cells. Oral Microbiol Immunol 2008: 23: 474481. 36. Brogden KA. Antimicrobial peptides: pore formers or metabolic inhibitors in bacteria? Nat Rev Microbiol 2005: 3: 238250. 37. Campos MA, Abreu AR, Nlend MC, Cobas MA, Conner GE, Whitney PL. Purication and characterization of PLUNC from human tracheobronchial secretions. Am J Respir Cell Mol Biol 2004: 30: 184192. 38. Canny G, Levy O. Bactericidal permeability-increasing protein (BPI) and BPI homologs at mucosal sites. Trends Immunol 2008: 29: 541547. 39. Canny G, Levy O, Furuta GT, Narravula-Alipati S, Sisson RB, Serhan CN, Colgan SP. Lipid mediator-induced expression of bactericidal permeability-increasing protein (BPI) in human mucosal epithelia. Proc Natl Acad Sci U S A 2002: 99: 39023907. 40. Carlsson G, Wahlin YB, Johansson A, Olsson A, Eriksson T, Claesson R, Hanstrom L, Henter JI. Periodontal disease in patients from the original Kostmann family with severe congenital neutropenia. J Periodontol 2006: 77: 744751. 41. Chu HW, Thaikoottathil J, Rino JG, Zhang G, Wu Q, Moss T, Refaeli Y, Bowler R, Wenzel SE, Chen Z, Zdunek J, Breed R, Young R, Allaire E, Martin RJ. Function and regulation of SPLUNC1 protein in Mycoplasma infection and allergic inammation. J Immunol 2007: 179: 39954002. 42. Chugh JK, Wallace BA. Peptaibols: models for ion channels. Biochem Soc Trans 2001: 29: 565570. 43. Chung WO, Dale BA. Innate immune response of oral and foreskin keratinocytes: utilization of different signaling pathways by various bacterial species. Infect Immun 2004: 72: 352358. 44. Cole AM, Liao H-I, Stuchlik O, Tilan J, Pohl J, Ganz T. Cationic polypeptides are required for antibacterial activity of human airway uid. J Immunol 2002: 169: 6985 6991. 45. Colombo AV, Silva CM, Haffajee A, Colombo APV Identication of oral bacteria associated with crevicular epithelial cells from chronic periodontitis lesions doi: 10.1099/jmm.0.46417-0. J Med Microbiol 2006: 55: 609 615. 46. Contucci AM, Inzitari R, Agostino S, Vitali A, Fiorita A, Cabras T, Scarano E, IMessana I. Statherin levels in saliva of patients with precancerous and cancerous lesions of the oral cavity: a preliminary report. Oral Dis 2005: 11: 9599. 47. Corbin BD, Seeley EH, Raab A, Feldmann J, Miller MR, Torres VJ, Anderson KL, Dattilo BM, Dunman PM, Gerads R, Caprioli RM, Nacken W, Chazin WJ, Skaar EP. Metal

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

172

Antimicrobial peptides of the oral cavity


chelation and inhibition of bacterial growth in tissue abscesses. Science 2008: 319: 962965. Daher KA, Selsted ME, Lehrer RI. Direct inactivation of viruses by human granulocyte defensins. J Virol 1986: 60: 10681074. Dale BA, Fredericks LP. Antimicrobial peptides in the oral environment: expression and function in health and disease. Curr Issues Mol Biol 2005: 7: 119133. Dale BA, Tao R, Kimball JR, Jurevic RJ. Oral antimicrobial peptides and biological control of caries. BMC Oral Health 2006: 6: S13. Dankesreiter S, Hoess A, Schneider-Mergener J, Wagner H, Miethke T. Synthetic endotoxin-binding peptides block endotoxin-triggered TNF-{alpha} production by macrophages in vitro and in vivo and prevent endotoxin-mediated toxic shock. J Immunol 2000: 164: 48044811. Dawidson I, Blom M, Lundeberg T, Theodorsson E, Angmar-Mansson B. Neuropeptides in the saliva of healthy subjects. Life Sci 1997: 60: 269278. de Haar SF, Hiemstra PS, van Steenbergen MTJM, Everts V, Beertsen W. Role of polymorphonuclear leukocyte-derived serine proteinases in defense against Actinobacillus actinomycetemcomitans. Infect Immun 2006: 74: 52845291. den Hertog AL, van Marle J, van Veen HA, Vant Hof W, Bolscher JG, Veerman EC, Nieuw Amerongen AV. Candidacidal effects of two antimicrobial peptides: histatin 5 causes small membrane defects, but LL-37 causes massive disruption of the cell membrane. Biochem J 2005: 388: 689695. Denny P, Hagen FK, Hardt M, Liao L, Yan W, Arellanno M, Bassilian S, Bedi GS, Boontheung P, Cociorva D, Delahunty CM, Denny T, Dunsmore J, Faull KF, Gilligan J, Gonzalez-Begne M, Halgand F, Hall SC, Han X, Henson B, Hewel J, Hu S, Jeffrey S, Jiang J, Loo JA, Ogorzalek Loo RR, Malamud D, Melvin JE, Miroshnychenko O, Navazesh M, Niles R, Park SK, Prakobphol A, Ramachandran P, Richert M, Robinson S, Sondej M, Souda P, Sullivan MA, Takashima J, Than S, Wang J, Whitelegge JP, Witkowska HE, Wolinsky L, Xie Y, Xu T, Yu W, Ytterberg J, Wong DT, Yates JR 3rd, Fisher SJ. The Proteomes of human parotid and submandibular sublingual gland salivas collected as the ductal secretions. J Proteome Res 2008: 7: 19942006. Diamond DL, Kimball JR, Krisanaprakornkit S, Ganz T, Dale BA. Detection of beta-defensins secreted by human oral epithelial cells. J Immunol Methods 2001: 256: 6576. Diamond G, Beckloff N, Ryan LK. Host defense peptides in the oral cavity and the lung: similarities and differences. J Dent Res 2008: 87: 915927. Dickinson DP. Salivary(SD-type) cystatins: over one billion years in the making but to what purpose? Crit Rev Oral Biol Med 2002: 13: 485508. Dickinson DP, Ridall AL, Levine MJ. Human submandibular gland statherin and basic histidine-rich peptide are encoded by highly abundant mRNAs derived from a common ancestral sequence. Biochem Biophys Res Commun 1987: 149: 784790. DiPaola C, Herrera MS, Mandel ID. Immunochemical study of host proteins in human supragingival compared with denture plaque. Arch Oral Biol 1984: 29: 161163. Dziarski R, Gupta D. Mammalian PGRPs: novel antibacterial proteins. Cell Microbiol 2006: 8: 10591069. Egland KA, Vincent JJ, Strausberg R, Lee B, Pastan I. Discovery of the breast cancer gene BASE using a molecular approach to enrich for genes encoding membrane and secreted proteins. Proc Natl Acad Sci U S A 2003: 100: 10991104. El Karim IA, Linden GJ, Orr DF, Lundy FT. Antimicrobial activity of neuropeptides against a range of micro-organisms from skin, oral, respiratory and gastrointestinal tract sites. J Neuroimmunol 2008: 200: 1116. Elkaim R, Werner S, Kocgozlu L, Tenenbaum H P. gingivalis regulates the expression of Cathepsin B and Cystatin C. J Dent Res 2008: 87: 932936. Elsbach P, Weiss J. The bactericidal permeabilityincreasing protein (BPI), a potent element in host-defense against gram-negative bacteria and lipopolysaccharide. Immunobiology 1993: 187: 417429. Ericson D. Agglutination of Streptococcus mutans by low-molecular-weight salivary components: effect of beta 2-microglobulin. Infect Immun 1984: 46: 526530. Ericson T, Rundegren J. Characterization of a salivary agglutinin reacting with a serotype c strain of Streptococcus mutans. Eur J Biochem 1983: 133: 255261. Fleming A Penicillin, Nobel Lectures, Physiology or Medicine 19421962. Amsterdam, NL: Elsevier Publishing Company, 1964. Freije J, Balbin M, Abrahamson M, Velasco G, Dalbge H, pez-Ot n C. Human cystatin D. cDNA cloning, Grubb A, Lo characterization of the Escherichia coli expressed inhibitor, and identication of the native protein in saliva. J Biol Chem 1993: 268: 1573715744. Friedman SA, Mandel ID, Herrera MS. Lysozyme and lactoferrin quantitation in the crevicular uid. J Periodontol 1983: 54: 347350. Fujii G, Selsted ME, Eisenberg D. Defensins promote fusion and lysis of negatively charged membranes. Protein Sci 1993: 2: 13011312. Ganz T. Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol 2003: 3: 710720. Ganz T. Defensins and other antimicrobial peptides: a historical perspective and an update. Comb Chem High Throughput Screen 2005: 8: 209217. Gardner MS, Rowland MD, Siu AY, Bundy JL, Wagener DK, Stephenson JL. Comprehensive defensin assay for saliva. Ana Chem 2009: 81: 557566. Geetha C, Venkatesh SG, Bingle L, Bingle CD, Gorr SU. Design and validation of anti-inammatory peptides from human parotid secretory protein. J Dent Res 2005: 84: 149 153. Geetha C, Venkatesh SG, Fasciotto Dunn BH, Gorr S-U. Expression and anti-bacterial activity of human parotid secretory protein (PSP). Biochem Soc Trans 2003: 31: 815 818. Ghafouri B, Kihlstrom E, Tagesson C, Lindahl M. PLUNC in human nasal lavage uid: multiple isoforms that bind to lipopolysaccharide. Biochim Biophys Acta 2004: 1699: 57 63. Giroir BP, Scannon PJ, Levin M. Bactericidal permeability-increasing proteinlessons learned from the phase III, randomized, clinical trial of rBPI21 for adjunctive treatment of children with severe meningococcemia. Crit Care Med 2001: 29: S130S135. Goebel C, Mackay L, Vickers E, Mather L. Determination of defensin HNP-1, HNP-2, and HNP-3 in human saliva by using LC MS. Peptides 2000: 21: 757765.

48.

63.

49.

64.

50.

65.

51.

66.

52.

67.

53.

68.

54.

69.

55.

70.

71.

72. 73.

74.

56.

75.

57.

76.

58.

59.

77.

78.

60.

61. 62.

79.

173

Gorr
80. Gordon YJ, Romanowski EG, McDermott AM. A review of antimicrobial peptides and their therapeutic potential as anti-infective drugs. Curr Eye Res 2005: 30: 505515. 81. Gorr S-U, Sotsky JB, Shelar AP, Demuth DR. Design of bacteria-agglutinating peptides derived from parotid secretory protein, a member of the bactericidal permeability increasing-like protein family. Peptides 2008: 29: 21182127. 82. Grahn E, Tenovuo J, Lehtonen OP, Eerola E, Vilja P. Antimicrobial systems of human whole saliva in relation to dental caries, cariogenic bacteria, and gingival inammation in young adults. Acta Odontol Scand 1988: 46: 6774. 83. Grifths GS, Wilton JMA, Curtis MA. Contamination of human gingival crevicular uid by plaque and saliva. Arch Oral Biol 1992: 37: 559564. 84. Groenink J, Ligtenberg AJ, Veerman EC, Bolscher JG, Nieuw Amerongen AV. Interaction of the salivary low-molecular-weight mucin (MG2) with Actinobacillus actinomycetemcomitans. Antonie Van Leeuwenhoek 1996: 70: 7987. 85. Groenink J, Walgreen-Weterings E, Nazmi K, Bolscher JG, Veerman EC, van Winkelhoff AJ, Nieuw Amerongen AV. Salivary lactoferrin and low-Mr mucin MG2 in Actinobacillus actinomycetemcomitans-associated periodontitis. J Clin Periodontol 1999: 26: 269275. 86. Habte H, Mall A, de Beer C, Lotz Z, Kahn D. The role of crude human saliva and puried salivary MUC5B and MUC7 mucins in the inhibition of Human Immunodeciency Virus type 1 in an inhibition assay. Virol J 2006: 3: 99. 87. Haigh B, Hood K, Broadhurst M, Medele S, Callaghan M, Smolenski G, Dines M, Wheeler T. The bovine salivary proteins BSP30a and BSP30b are independently expressed BPI-like proteins with anti-Pseudomonas activity. Mol Immunol 2008: 45: 19441951. 88. Hancock RE, Chapple DS. Peptide antibiotics. Antimicrob Agents Chemother 1999: 43: 13171323. 89. Hancock REW. Concerns regarding resistance to self-proteins. Microbiol 2003: 149: 33433344. 90. Handeld M, Mans JJ, Zheng G, Lopez MC, Mao S, Progulske-Fox A, Narasimhan G, Baker HV, Lamont RJ. Distinct transcriptional proles characterize oral epithelium-microbiota interactions. Cell Microbiol 2005: 7: 811823. 91. Hardt M, Thomas LR, Dixon SE, Newport G, Agabian N, Prakobphol A, Hall SC, Witkowska HE, Fisher SJ. Toward dening the human parotid gland salivary proteome and peptidome: Identication and characterization using 2D SDS-PAGE, ultraltration, HPLC, and mass spectrometry. Biochemistry 2005: 44: 28852899. 92. Hart TC, Atkinson JC. Mendelian forms of periodontitis. Periodontol 2000 2007: 45: 95112. 93. Hart TC, Hart PS, Bowden DW, Michalec MD, Callison SA, Walker SJ, Zhang Y, Firatli E. Mutations of the cathepsin C ` vre syndrome. J Med gene are responsible for Papillon-Lefe Genet 1999: 36: 881887. 94. Hart TC, Hart PS, Michalec MD, Zhang Y, Firatli E, Van Dyke TE, Stabholz A, Zlotogorski A, Shapira L, Soskolne ` vre synWA. Haim-Munk syndrome and Papillon-Lefe drome are allelic mutations in cathepsin C. J Med Genet 2000: 37: 8894. ` vre syndrome. Period95. Hart TC, Shapira L. Papillon-Lefe ontol 2000 1994: 6: 88100. 96. Hartshorn KL, Ligtenberg A, White MR, Van Eijk M, Hartshorn M, Pemberton L, Holmskov U, Crouch E. Salivary agglutinin and lung scavenger receptor cysteine-rich glycoprotein 340 have broad anti-inuenza activities and interactions with surfactant protein D that vary according to donor source and sialylation. Biochem J 2006: 393: 545 553. 97. Hartshorn KL, White MR, Mogues T, Ligtenberg T, Crouch E, Holmskov U. Lung and salivary scavenger receptor glycoprotein-340 contribute to the host defense against inuenza A viruses. Am J Physiol Lung Cell Mol Physiol 2003: 285: L1066L1076. 98. Haug BE, Strom MB, Svendsen JS. The medicinal chemistry of short lactoferricin-based antibacterial peptides. Curr Med Chem 2007: 14: 118. 99. Hay DI, Smith DJ, Schluckebier SK, Moreno EC. Basic biological sciences relationship between concentration of human salivary statherin and inhibition of calcium phosphate precipitation in stimulated human parotid saliva. J Dent Res 1984: 63: 857863. 100. Heft MW, Baum BJ. Basic biological sciences unstimulated and stimulated parotid salivary ow rate in individuals of different ages. J Dent Res 1984: 63: 11821185. 101. Henskens YM, van den Keijbus PA, Veerman EC, Van der Weijden GA, Timmerman MF, Snoek CM, Van der Velden U, Nieuw Amerongen AV. Protein composition of whole and parotid saliva in healthy and periodontitis subjects. Determination of cystatins, albumin, amylase and IgA. J Periodont Res 1996: 31: 5765. 102. Hieshima K, Ohtani H, Shibano M, Izawa D, Nakayama T, Kawasaki Y, Shiba F, Shiota M, Katou F, Saito T, Yoshie O. CCL28 has dual roles in mucosal immunity as a chemokine with broad-spectrum antimicrobial activity. J Immunol 2003: 170: 14521461. 103. Hirsch T, Jacobsen F, Steinau HU, Steinstraesser L. Host defense peptides and the new line of defence against multiresistant infections. Protein Pept Lett 2008: 15: 238 243. 104. Hoffman MP, Haidaris CG. Analysis of Candida albicans adhesion to salivary mucin. Infect Immun 1993: 61: 1940 1949. 105. Ihalin R, Loimaranta V, Lenander-Lumikari M, Tenovuo J. The sensitivity of Porphyromonas gingivalis and Fusobacterium nucleatum to different (pseudo)halide-peroxidase combinations compared with mutans streptococci. J Med Microbiol 2001: 50: 4248. 106. Ihalin R, Loimaranta V, Tenovuo J. Origin, structure, and biological activities of peroxidases in human saliva. Arch Biochem Biophys 2006: 445: 261268. kkinen K, Lenander M, Tenovuo J, Jousi107. Ihalin R, Pieniha mies-Somer H. Susceptibilities of different Actinobacillus actinomycetemcomitans strains to lactoperoxidase-iodidehydrogen peroxide combination and different antibiotics. Int J Antimicrob Agents 2003: 21: 434440. 108. Into T, Inomata M, Kanno Y, Matsuyama T, Machigashira M, Izumi Y, Imamura T, Nakashima M, Noguchi T, Matsushita K. Arginine-specic gingipains from Porphyromonas gingivalis deprive protective functions of secretory leucocyte protease inhibitor in periodontal tissue. Clin Experiment Immunol 2006: 145: 545554. 109. Iovine NM, Elsbach P, Weiss J. An opsonic function of the neutrophil bactericidal permeability-increasing protein

174

Antimicrobial peptides of the oral cavity


depends on both its N- and C-terminal domains. Proc Natl Acad Sci U S A 1997: 94: 1097310978. Jana NK, Gray LR, Shugars DC. Human immunodeciency virus type 1 stimulates the expression and production of secretory leukocyte protease inhibitor (SLPI) in oral epithelial cells: a role for SLPI in innate mucosal immunity. J Virol 2005: 79: 64326440. Jensen JL. Salivary acidic proline-rich proteins in rheumatoid arthritis. Ann New York Acad Sci 1998: 842: 209 211. cke R. Lactoferrin and other Jentsch H, Sievert Y, Go markers from gingival crevicular uid and saliva before and after periodontal treatment. J Clin Periodontol 2004: 31: 511514. Ji S, Hyun J, Park E, Lee BL, Kim KK, Choi Y. Susceptibility of various oral bacteria to antimicrobial peptides and to phagocytosis by neutrophils. J Periodontal Res 2007: 42: 410419. Ji S, Kim Y, Min BM, Han SH, Choi Y. Innate immune responses of gingival epithelial cells to nonperiodontopathic and periodontopathic bacteria. J Periodontal Res 2007: 42: 503510. Johansson I, Bratt P, Hay DI, Schluckebier S, Stromberg N. Adhesion of Candida albicans, but not Candida krusei, to salivary statherin and mimicking host molecules. Oral Microbiol Immunol 2000: 15: 112118. Johnson DA, Yeh CK, Dodds MWJ. Effect of donor age on the concentrations of histatins in human parotid and submandibular sublingual saliva. Arch Oral Biol 2000: 45: 731740. Joly S, Maze C, McCray PB Jr, Guthmiller JM. Human {beta}-defensins 2 and 3 demonstrate strain-selective activity against oral microorganisms. J Clin Microbiol 2004: 42: 10241029. Jonasson A, Eriksson C, Jenkinson HF, Kallestal C, Johansson I, Stromberg N. Innate immunity glycoprotein gp-340 variants may modulate human susceptibility to dental caries. BMC Infect Dis 2007: 7: 57. Jumblatt M, Imbert Y, Young W, Foulks G, Steele P, Demuth D. Glycoprotein 340 in normal human ocular surface tissues and tear lm. Infect Immun 2006: 74: 40584063. Kalmar JR, Arnold RR. Killing of Actinobacillus actinomycetemcomitans by human lactoferrin. Infect Immun 1988: 56: 25522557. Kaner D, Bernimoulin J, Kleber B, Heizmann W, Friedmann A. Gingival crevicular uid levels of calprotectin and myeloperoxidase during therapy for generalized aggressive periodontitis. J Periodontal Res 2006: 41: 132139. Kapas S, Bansal A, Bhargava V, Maher R, Malli D, HagiPavli E, Allaker RP. Adrenomedullin expression in pathogen-challenged oral epithelial cells. Peptides 2001: 22: 14851489. Kapas S, Pahal K, Cruchley AT, Hagi-Pavli E, Hinson JP. Expression of adrenomedullin and its receptors in human salivary tissue. J Dent Res 2004: 83: 333337. Keijser BJF, Zaura E, Huse SM, van der Vossen JMBM, Schuren FHJ, Montijn RC, ten Cate JM, Crielaard W. Pyrosequencing analysis of the oral microora of healthy adults. J Dent Res 2008: 87: 10161020. Kido J, Nakamura T, Kido R, Ohishi K, Yamauchi N, Kataoka M, Nagata T. Calprotectin in gingival crevicular uid correlates with clinical and biochemical markers of periodontal disease. J Clin Periodontol 1999: 26: 653 657. Kinane DF, Demuth DR, Gorr SU, Hajishengallis GN, Martin MH. Human variability in innate immunity. Periodontol 2000 2007: 45: 1434. Kinane DF, Galicia J, Gorr SU, Stathopoulou P, Benakanakere MM. P. gingivalis interactions with epithelial cells. Front Biosci 2008: 13: 966984. Kirstila V, Hakkinen P, Jentsch H, Vilja P, Tenovuo J. Longitudinal analysis of the association of human salivary antimicrobial agents with caries increment and cariogenic micro-organisms: a two-year cohort study. J Dent Res 1998: 77: 7380. Kleinegger CL, Stoeckel DC, Kurago ZB. A comparison of salivary calprotectin levels in subjects with and without oral candidiasis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2001: 92: 6267. Klimiuk A, Waszkiel D, Jankowska A, Zelazowska-Rutkowska B, Choromanska M. The evaluation of lysozyme concentration and peroxidase activity in non-stimulated saliva of patients infected with HIV. Adv Med Sci 2006: 51(Suppl 1): 4951. Kochanska B, Kedzia A, Kamysz W, Mackiewicz Z, Kupryszewski G. The effect of statherin and its shortened analogues on anaerobic bacteria isolated from the oral cavity. Acta Microbiol Pol 2000: 49: 243251. Kojima T, Andersen E, Sanchez JC, Wilkins MR, Hochstrasser DF, Pralong WF, Cimasoni G. Human gingival crevicular uid contains MRP8 (S100A8) and MRP14 (S100A9), two calcium-binding proteins of the S100 family. J Dent Res 2000: 79: 740747. Korfhagen TR. Surfactant Protein A (SP-A)-mediated bacterial clearance. SP-A and cystic brosis. Am J Respir Cell Mol Biol 2001: 25: 668672. Koshlukova SE, Araujo MWB, Baev D, Edgerton M. Released ATP is an extracellular cytotoxic mediator in salivary Histatin 5-induced killing of Candida albicans. Infect Immun 2000: 68: 68486856. Krisanaprakornkit S, Chotjumlong P, Kongtawelert P, Reutrakul V. Involvement of phospholipase D in regulating expression of anti-microbial peptide human -defensin-2. Int Immunol 2008: 20: 2129. Krisanaprakornkit S, Kimball JR, Weinberg A, Darveau RP, Bainbridge BW, Dale BA. Inducible expression of human beta-defensin 2 by Fusobacterium nucleatum in oral epithelial cells: multiple signaling pathways and role of commensal bacteria in innate immunity and the epithelial barrier. Infect Immun 2000: 68: 29072915. Krisanaprakornkit S, Weinberg A, Perez CN, Dale BA. Expression of the peptide antibiotic human beta-Defensin 1 in cultured gingival epithelial cells and gingival tissue. Infect Immun 1998: 66: 42224228. Krishnakumari V, Rangaraj N, Nagaraj R. Antifungal activities of human beta-Defensins HBD-1 to HBD-3 and their C-terminal analogs Phd1 to Phd3. Antimicrob Agents Chemother 2009: 53: 256260. Lamkin MS, Oppenheim FG. Structural features of salivary function. Crit Rev Oral Biol Med 1993: 4: 251259. Laube DM, Dongari-Bagtzoglou A, Kashleva H, Eskdale J, Gallagher G, Diamond G. Differential regulation of innate immune response genes in gingival epithelial cells

110.

126.

127.

111.

128.

112.

113.

129.

114.

130.

115.

131.

116.

132.

117.

133.

118.

134.

119.

135.

120.

136.

121.

122.

137.

123.

138.

124.

139. 140.

125.

175

Gorr
stimulated with Aggregatibacter actinomycetemcomitans. J Periodontal Res 2008: 43: 116123. Lee HM, Park IH, Woo JS, Chae SW, Kang HJ, Hwang SJ. Up-regulation of surfactant protein A in chronic sialadenitis. Arch Otolaryngol Head Neck Surg 2005: 131: 1108 1111. Lee SK, Lee SS, Hirose S, Park SC, Chi JG, Chung SI, Mori M. Elan expression in human fetal and adult submandibular glands. Histochem Cell Biol 2002: 117: 423 430. Lehrer RI, Lichtenstein AK, Ganz T. Defensins: antimicrobial and cytotoxic peptides of mammalian cells. Ann Rev Immunol 1993: 11: 105128. Li T, Bratt P, Jonsson AP, Ryberg M, Johansson I, Grifths WJ, Bergman T, Stromberg N. Possible release of an ArgGlyArgProGln pentapeptide with Innate Immunity properties from acidic proline-rich proteins by proteolytic activity in commensal Streptococcus and Actinomyces species. Infect Immun 2000: 68: 54255429. Ligtenberg AJ, Veerman EC, Nieuw Amerongen AV, Mollenhauer J. Salivary agglutinin glycoprotein-340 DMBT1: a single molecule with variable composition and with different functions in infection, inammation and cancer. Biol Chem 2007: 388: 12751289. Lim E, Ammons S, Mohler V, Killian D, Dedrick R, Gikonyo K, Lin J XMP.629, a peptide derived from functional domain II of BPI, demonstrates broad-spectrum antimicrobial and endotoxin-neutralizing properties in vitro and in vivo, 41st Interscience conference on antimicrobial agents and chemotherapy. Chicago, IL: American Society for Microbiology, 2001. Lin AL, Johnson DA, Stephan KT, Yeh C-K. Salivary secretory leukocyte protease inhibitor increases in HIV infection*. J Oral PathMed 2004: 33: 410416. Linden GJ, McKinnell J, Shaw C, Lundy FT. Substance P and neurokinin A in gingival crevicular uid in periodontal health and disease. J Clin Periodontol 1997: 24: 799803. Llena-Puy MC, Montanana-Llorens C, Forner-Navarro L. Fibronectin levels in stimulated whole-saliva and their relationship with cariogenic oral bacteria. Int Dent J 2000: 50: 5759. Llena-Puy MC, Montanana-Llorens C, Forner-Navarro L. Optimal assay conditions for quantifying bronectin in saliva. Med Oral 2004: 9: 191196. Loe H, Anerud A, Boysen H, Morrison E. Natural history of periodontal disease in man. Rapid, moderate and no loss of attachment in Sri Lankan laborers 14 to 46 years of age. J Clin Periodontol 1986: 13: 431445. Lopatin DE, Caffesse ER, Bye FL, Caffesse RG. Concentrations of bronectin in the sera and crevicular uid in various stages of periodontal disease. J Clin Periodontol 1989: 16: 359364. Lu X, Wang M, Qi J, Wang H, Li X, Gupta D, Dziarski R. Peptidoglycan recognition proteins are a new class of human bactericidal proteins. J Biol Chem 2006: 281: 5895 5907. Lundy FT, Chalk R, Lamey P-J, Shaw C, Linden GJ. Quantitative analysis of MRP-8 in gingival crevicular uid in periodontal health and disease using microbore HPLC. J Clin Periodontol 2001: 28: 11721177. Lundy FT, Chalk R, Lamey PJ, Shaw C, Linden GJ. Identication of MRP-8 (calgranulin A) as a major responsive protein in chronic periodontitis. J Pathol 2000: 192: 540 544. Lundy FT, Mullally BH, Burden DJ, Lamey PJ, Shaw C, Linden GJ. Changes in substance P and neurokinin A in gingival crevicular uid in response to periodontal treatment. J Clin Periodontol 2000: 27: 526530. Lundy FT, Nelson J, Lockhart D, Greer B, Harriott P, Marley JJ. Antimicrobial activity of truncated [alpha]-defensin (human neutrophil peptide (HNP)-1) analogues without disulphide bridges. Mol Immunol 2008: 45: 190 193. Lundy FT, OHare MMT, McKibben BM, Fulton CR, Briggs JE, Linden GJ. Radioimmunoassay quantication of adrenomedullin in human gingival crevicular uid. Arch Oral Biol 2006: 51: 334338. Lundy FT, Orr DF, Shaw C, Lamey PJ, Linden GJ. Detection of individual human neutrophil alpha-defensins (human neutrophil peptides 1, 2 and 3) in unfractionated gingival crevicular uid a MALDI-MS approach. Mol Immunol 2005: 42: 575579. Lundy FT, Shaw C, McKinnell J, Lamey PJ, Linden GJ. Calcitonin gene-related peptide in gingival crevicular uid in periodontal health and disease. J Clin Periodontol 1999: 26: 212216. Madsen HO, Hjorth JP. Molecular cloning of mouse PSP mRNA. Nucleic Acids Res 1985: 13: 113. Makinen KK, Tenovuo J. Chromatographic separation of human salivary peroxidases. Acta Odontol Scand 1976: 34: 141150. Mandal M, Nagaraj R. Antibacterial activities and conformations of synthetic alpha-defensin HNP-1 and analogs with one, two and three disulde bridges. J Peptide Res 2002: 59: 95104. Mathews M, Jia HP, Guthmiller JM, Losh G, Graham S, Johnson GK, Tack BF, McCray PB Jr, . Production of beta-defensin antimicrobial peptides by the oral mucosa and salivary glands. Infect Immun 1999: 67: 27402745. McCabe D, Cukierman T, Gabay JE. Basic residues in azurocidin HBP contribute to both heparin binding and antimicrobial activity. J Biol Chem 2002: 277: 27477 27488. McNeely TB, Shugars DC, Rosendahl M, Tucker C, Eisenberg SP, Wahl SM. Inhibition of human immunodeciency virus type 1 infectivity by secretory leukocyte protease inhibitor occurs prior to viral reverse transcription. Blood 1997: 90: 11411149. Messana I, Cabras T, Pisano E, Sanna MT, Olianas A, Manconi B, Pellegrini M, Paludetti G, Scarano E, Fiorita A, Agostino S, Contucci AM, Calo L, Picciotti PM, Manni A, Bennick A, Vitali A, Fanali C, Inzitari R, Castagnola M. Trafcking and postsecretory events responsible for the formation of secreted human salivary peptides: A proteomics approach. Mol Cell Proteomics 2008: 7: 911926. der Michalek M, Gelhaus C, Hecht O, Podschun R, Schro tzinger J. The human antimicrobial JM, Leippe M, Gro protein psoriasin acts by permeabilization of bacterial membranes. Dev Comp Immunol 2009: 33: 740746. Michelis R, Sela S, Ben-Zvi I, Nagler RM. Salivary b2-microglobulin analysis in chronic kidney disease and hemodialyzed patients. Blood Purif 2007: 25: 505509. Milward MR, Chapple IL, Wright HJ, Millard JL, Matthews JB, Cooper PR. Differential activation of NF-kappaB and

141.

156.

142.

157.

143.

158.

144.

159.

145.

160.

146.

161. 162.

163.

147.

164.

148.

149.

165.

150.

166.

151.

167.

152.

153.

168.

154.

169.

155.

170.

176

Antimicrobial peptides of the oral cavity


gene expression in oral epithelial cells by periodontal pathogens. Clin Exp Immunol 2007: 148: 307324. Miyasaki KT, Bodeau AL, Ganz T, Selsted ME, Lehrer RI. In vitro sensitivity of oral, gram-negative, facultative bacteria to the bactericidal activity of human neutrophil defensins. Infect Immun 1990: 58: 39343940. Miyasaki KT, Voganatsi A, Huynh T, Marcus M, Underwood S. Calprotectin and lactoferrin levels in the gingival crevicular uid of children. J Periodontol 1998: 69: 879883. Miyasaki KT, Wilson ME, Genco RJ. Killing of Actinobacillus actinomycetemcomitans by the human neutrophil myeloperoxidase-hydrogen peroxide-chloride system. Infect Immun 1986: 53: 161165. Mogi M, Otogoto J, Ota N, Inagaki H, Minami M, Kojima K. Interleukin 1[beta], interleukin 6, [beta]2-microglobulin, and transforming growth factor-[alpha] in gingival crevicular uid from human periodontal disease. Arch Oral Biol 1999: 44: 535539. Mulero JJ, Boyle BJ, Bradley S, Bright JM, Nelken ST, Ho TT, Mize NK, Childs JD, Ballinger DG, Ford JE, Rupp F. Three new human members of the lipid transfer lipopolysaccharide binding protein family (LT LBP). Immunogenetics 2002: 54: 293300. Murakami M, Ohtake T, Dorschner RA, Gallo RL. Cathelicidin antimicrobial peptides are expressed in salivary glands and saliva. J Dent Res 2002: 81: 845850. Murakami Y, Hanazawa S, Tanaka S, Iwahashi H, Kitano S, Fujisawa S. Fibronectin in saliva inhibits Porphyromonas gingivalis mbria-induced expression of inammatory cytokine gene in mouse macrophages. FEMS Immunol Med Microbiol 1998: 22: 257262. Murakami Y, Xu T, Helmerhorst EJ, Ori G, Troxler RF, Lally ET, Oppenheim FG. Inhibitory effect of synthetic histatin 5 on leukotoxin from Actinobacillus actinomycetemcomitans. Oral Microbiol Immunol 2002: 17: 143149. Nakamura-Minami M, Furuichi Y, Ishikawa K, MitsuzonoTofuku Y, Izumi Y. Changes of alpha1-protease inhibitor and secretory leukocyte protease inhibitor levels in gingival crevicular uid before and after non-surgical periodontal treatment. Oral Dis 2003: 9: 249254. Nakamura T, Kido J, Kido R, Ohishi K, Yamauchi N, Kataoka M, Nagata T. The association of calprotectin level in gingival crevicular uid with gingival index and the activities of collagenase and aspartate aminotransferase in adult periodontitis patients. J Periodontol 2000: 71: 361 367. Nisapakultorn K, Ross KF, Herzberg MC. Calprotectin expression in vitro by oral epithelial cells confers resistance to infection by Porphyromonas gingivalis. Infect Immun 2001: 69: 42424247. Oppenheim FG, Salih E, Siqueira WL, Zhang W, Helmerhorst EJ. Salivary proteome and its genetic polymorphisms. Ann N Y Acad Sci 2007: 1098: 2250. Orner G. Periodontal disease among children with Downs syndrome and their siblings. J Dent Res 1976: 55: 778782. Orsi N. The antimicrobial activity of lactoferrin: current status and perspectives. Biometals 2004: 17: 189196. Ortiz GC, Rahemtulla B, Tsurudome SA, Chaves E, Rahemtulla F. Quantication of human myeloperoxidase in oral uids. Eur J Oral Sci 1997: 105: 143152. 186. Ouhara K, Komatsuzawa H, Shiba H, Uchida Y, Kawai T, Sayama K, Hashimoto K, Taubman MA, Kurihara H, Sugai M. Actinobacillus actinomycetemcomitans outer membrane protein 100 triggers innate immunity and production of {beta}-defensin and the 18-kilodalton cationic antimicrobial protein through the bronectin-integrin pathway in human gingival epithelial cells. Infect Immun 2006: 74: 52115220. 187. Ouhara K, Komatsuzawa H, Yamada S, Shiba H, Fujiwara T, Ohara M, Sayama K, Hashimoto K, Kurihara H, Sugai M. Susceptibilities of periodontopathogenic and cariogenic bacteria to antibacterial peptides, {beta}-defensins and LL37, produced by human epithelial cells. J Antimicrob Chemother 2005: 55: 888896. 188. Paster BJ, Olsen I, Aas JA, Dewhirst FE. The breadth of bacterial diversity in the human periodontal pocket and other oral sites. Periodontol 2000 2006: 42: 80 87. 189. Payment SA, Liu B, Soares RV, Offner GD, Oppenheim FG, Troxler RF. The effects of duration and intensity of stimulation on total protein and mucin concentrations in resting and stimulated whole saliva. J Dent Res 2001: 80: 15841587. 190. Periodontics WWoC. Consensus report. Periodontal diseases: pathogenesis and microbial factors. Ann Periodontol 1996: 1: 926932. 191. Peschel A, Sahl HG. The co-evolution of host cationic antimicrobial peptides and microbial resistance. Nat Rev Microbiol 2006: 4: 529536. 192. Pham CTN, Ivanovich JL, Raptis SZ, Zehnbauer B, Ley TJ. ` vre syndrome: correlating the molecular, Papillon-Lefe cellular, and clinical consequences of cathepsin C dipeptidyl peptidase I deciency in humans. J Immunol 2004: 173: 72777281. 193. Pisano E, Cabras T, Montaldo C, Piras V, Inzitari R, Olmi C, Castagnola M, Messana I. Peptides of human gingival crevicular uid determined by HPLC-ESI-MS. Eur J Oral Sci 2005: 113: 462468. 194. Prime SS, Nixon SV, Crane IJ, Stone A, Matthews JB, Maitland NJ, Remnant L, Powell SK, Game SM, Scully C. The behaviour of human oral squamous cell carcinoma in cell culture. J Pathol 1990: 160: 259269. 195. Puklo M, Guentsch A, Hiemstra PS, Eick S, Potempa J. Analysis of neutrophil-derived antimicrobial peptides in gingival crevicular uid suggests importance of cathelicidin LL-37 in the innate immune response against periodontogenic bacteria. Oral Microbiol Immunol 2008: 23: 328335. 196. Putsep K, Carlsson G, Boman HG, Andersson M. Deciency of antibacterial peptides in patients with morbus Kostmann: an observation study. The Lancet 2002: 360: 11441149. 197. Raj PA, Antonyraj KJ, Karunakaran T Large-scale synthesis and functional elements for the antimicrobial activity of defensins. Biochem J 2000: 347 (Pt 3): 633641. 198. Rajan GH, Morris CA, Carruthers VR, Wilkins RJ, Wheeler TT. The relative abundance of a salivary protein, bSP30, is correlated with susceptibility to bloat in cattle herds selected for high or low bloat susceptibility. Anim Genet 1996: 27: 407414. 199. Ramachandran P, Boontheung P, Xie Y, Sondej M, Wong DT, Loo JA. Identication of N-linked glycoproteins in

171.

172.

173.

174.

175.

176.

177.

178.

179.

180.

181.

182.

183.

184. 185.

177

Gorr
human saliva by glycoprotein capture and mass spectrometry. J Proteome Res 2006: 5: 14931503. Raqib R, Sarker P, Bergman P, Ara G, Lindh M, Sack DA, Nasirul Islam KM, Gudmundsson GH, Andersson J, Agerberth B. Improved outcome in shigellosis associated with butyrate induction of an endogenous peptide antibiotic. Proc Natl Acad Sci U S A 2006: 103: 9178 9183. Reddy MS, Bobek LA, Haraszthy GG, Biesbrock AR, Levine MJ Structural features of the low-molecular-mass human salivary mucin. Biochem J 1992: 287 (Pt 2): 639 643. Robinovitch MR, Ashley RL, Iversen JM, Vigoren EM, Oppenheim FG, Lamkin M. Parotid salivary basic proline-rich proteins inhibit HIV-I infectivity. Oral Dis 2001: 7: 8693. Robinson CP, Bounous DI, Alford CE, Nguyen KH, Nanni JM, Peck AB, Humphreys-Beher MG. PSP expression in murine lacrimal glands and function as a bacteria binding protein in exocrine secretions. Am J Physiol 1997: 272: G863G871. Rothstein DM, Spacciapoli P, Tran LT, Xu T, Roberts FD, Dalla Serra M, Buxton DK, Oppenheim FG, Friden P. Anticandida activity is retained in P-113, a 12-amino-acid fragment of histatin 5. Antimicrob Agents Chemother 2001: 45: 13671373. Royet J, Dziarski R. Peptidoglycan recognition proteins: pleiotropic sensors and effectors of antimicrobial defences. Nat Rev Micro 2007: 5: 264277. Rudney J, Staikov R, Johnson J. Potential biomarkers of human salivary function: a modied proteomic approach. Arch Oral Biol 2009: 54: 91100. Rudney JD, Smith QT. Relationships between levels of lysozyme, lactoferrin, salivary peroxidase, and secretory immunoglobulin A in stimulated parotid saliva. Infect Immun 1985: 49: 469475. mer M, Samuelsen , Haukland HH, Jenssen H, Kra Sandvik K, Ulvatne H, Vorland LH. Induced resistance to the antimicrobial peptide lactoferricin B in Staphylococcus aureus. FEBS Lett 2005: 579: 34213426. Schroder J-M, Harder J. Antimicrobial peptides in skin disease. Drug Discov Today: Therap Strategies 2006: 3: 93100. Shiba H, Venkatesh SG, Gorr SU, Barbieri G, Kurihara H, Kinane DF. Parotid secretory protein is expressed and inducible in human gingival keratinocytes. J Periodontal Res 2005: 40: 153157. Shtatland T, Guettler D, Kossodo M, Pivovarov M, Weissleder R. PepBank - a database of peptides based on sequence text mining and public peptide data sources. BMC Bioinformatics 2007: 8: 280. Shugars DC, Watkins CA, Cowen HJ. Salivary concentration of secretory leukocyte protease inhibitor, an antimicrobial protein, is decreased with advanced age. Gerontology 2001: 47: 246253. Simpson AJ, Maxwell AI, Govan JR, Haslett C, Sallenave JM. Elan (elastase-specic inhibitor) has anti-microbial activity against gram-positive and gram-negative respiratory pathogens. FEBS Lett 1999: 452: 309313. Simpson JL, Wood LG, Gibson PG. Inammatory mediators in exhaled breath, induced sputum and saliva. Clin Exp Allergy 2005: 35: 11801185. 215. Siqueira WL, Salih E, Wan DL, Helmerhorst EJ, Oppenheim FG. Proteome of human minor salivary gland secretion. J Dent Res 2008: 87: 445450. 216. Situ H, Wei G, Smith CJ, Mashhoon S, Bobek LA. Human salivary MUC7 mucin peptides: effect of size, charge and cysteine residues on antifungal activity. Biochem J 2003: 375: 175182. 217. Slots J, Ting M. Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis in human periodontal disease: occurrence and treatment. Periodontol 2000 1999: 20: 82 121. 218. Socransky SS, Haffajee AD, Cugini MA, Smith C, Kent RL Jr. Microbial complexes in subgingival plaque. J Clin Periodontol 1998: 25: 134144. 219. Soderstrom T, Wikstrom M. Decreased lactoferrin content in granulocytes from subjects with Actinobacillus actinomycetemcomitans associated periodontal diseases. J Parodontol 1990: 9: 195199. 220. Sohnle PG, Hunter Michael J, Hahn B, Chazin Walter J. Zinc-reversible antimicrobial activity of recombinant calprotectin (migration inhibitory factor-related proteins 8 and 14). J Infect Dis 2000: 182: 12721275. 221. Sorensen OE, Borregaard N, Cole AM. Antimicrobial peptides in innate immune responses. Contrib Microbiol 2008: 15: 6177. 222. Stenudd C, Nordlund A, Ryberg M, Johansson I, Kallestal C, Stromberg N. The association of bacterial adhesion with dental caries. J Dent Res 2001: 80: 20052010. 223. de Haar SF, Jansen DC, Schoenmaker T, De Vree H, Everts V, Beertsen W. Loss-of-function mutations in cathepsin C ` vre syndrome are assoin two families with Papillon-Lefe ciated with deciency of serine proteinases in PMNs. Hum Mutation 2004: 23: 524. nen S, Markkanen H, Syrja nen K. Gingival beta 2224. Syrja microglobulin in juvenile and chronic periodontitis. Acta Odontol Scand 1985: 43: 133138. nen SM, Alakuijala P, Markkanen SO, Markkanen H. 225. Syrja Gingival uid, beta 2-microglobulin and protein levels as indicators of periodontal disease. Scand J Dent Res 1989: 97: 500504. 226. Talbot GH, Bradley J, Edwards JE Jr, Gilbert D, Scheld M, Bartlett JG. Bad bugs need drugs: an update on the development pipeline from the Antimicrobial Availability Task Force of the Infectious Diseases Society of America. Clin Infect Dis 2006: 42: 657668. 227. Talonpoika J, Heino J, Larjava H, Hakkinen L, Paunio K. Gingival crevicular uid bronectin degradation in periodontal health and disease. Scand J Dent Res 1989: 97: 415421. 228. Tanida T, Okamoto T, Okamoto A, Wang H, Hamada T, Ueta E, Osaki t, . Decreased excretion of antimicrobial proteins and peptides in saliva of patients with oral candidiasis. J Oral Path Med 2003: 32: 586594. 229. Tao R, Jurevic RJ, Coulton KK, Tsutsui MT, Roberts MC, Kimball JR, Wells N, Berndt J, Dale BA. Salivary antimicrobial peptide expression and dental caries experience in children. Antimicrob Agents Chemother 2005: 49: 3883 3888. 230. Teles RP, Haffajee AD, Socransky SS. Microbiological goals of periodontal therapy. Periodontol 2000 2006: 42: 180218.

200.

201.

202.

203.

204.

205.

206.

207.

208.

209. 210.

211.

212.

213.

214.

178

Antimicrobial peptides of the oral cavity


231. Tenovuo J, Grahn E, Lehtonen OP, Hyyppa T, Karhuvaara L, Vilja P. Antimicrobial factors in saliva: ontogeny and relation to oral health. J Dent Res 1987: 66: 475479. 232. Tew GN, Clements D, Tang H, Arnt L, Scott RW. Antimicrobial activity of an abiotic host defense peptide mimic. Biochim Biophys Acta (BBA) Biomembranes 2006: 1758: 13871392. 233. Thomas EL, Jefferson MM, Joyner RE, Cook GS, King CC. Leukocyte myeloperoxidase and salivary lactoperoxidase: identication and quantitation in human mixed saliva. J Dent Res 1994: 73: 544555. 234. Thomas EL, Milligan TW, Joyner RE, Jefferson MM. Antibacterial activity of hydrogen peroxide and the lactoperoxidase-hydrogen peroxide-thiocyanate system against oral streptococci. Infect Immun 1994: 62: 529535. 235. Tjabringa GS, Vos JB, Olthuis D, Ninaber DK, Rabe KF, Schalkwijk J, Hiemstra PS, Zeeuwen PL. Host defense effector molecules in mucosal secretions. FEMS Immunol Med Microbiol 2005: 45: 151158. 236. Tynelius-Bratthall G, Ericson D, Araujo HM. Fibronectin in saliva and gingival crevices. J Periodontal Res 1986: 21: 563568. 237. Ulker AE, Tulunoglu O, Ozmeric N, Can M, Demirtas S. The evaluation of cystatin C, IL-1beta, and TNF-alpha levels in total saliva and gingival crevicular uid from 11- to 16-year-old children. J Periodontol 2008: 79: 854860. 238. van Gils PC, Brand HS, Timmerman MF, Veerman ECI, van der Velden U, van der Weijden GA. Salivary cystatin activity and cystatin C in experimental gingivitis in nonsmokers. J Clin Periodontol 2003: 30: 882886. 239. vanderSpek JC, Wyandt HE, Skare JC, Milunsky A, Oppenheim FG, Troxler RF. Localization of the genes for histatins to human chromosome 4q13 and tissue distribution of the mRNAs. Am J Hum Genet 1989: 45: 381387. 240. Vankeerberghen A, Nuytten H, Dierickx K, Quirynen M, Cassiman JJ, Cuppens H. Differential induction of human beta-defensin expression by periodontal commensals and pathogens in periodontal pocket epithelial cells. J Periodontol 2005: 76: 12931303. 241. Viejo-Diaz M, Andres MT, Fierro JF. Different anti-Candida activities of two human lactoferrin-derived peptides, Lfpep and kaliocin-1. Antimicrob Agents Chemother 2005: 49: 25832588. 242. Vitorino R, Lobo MJ, Ferrer-Correira AJ, Dubin JR, Tomer KB, Domingues PM, Amado FM. Identication of human whole saliva protein components using proteomics. Proteomics 2004: 4: 11091115. 243. Vylkova S, Sun JN, Edgerton M. The role of released ATP in killing Candida albicans and other extracellular microbial pathogens by cationic peptides. Purinergic Signal 2007: 3: 9197. 244. Wade D, Englund J. Synthetic antibiotic peptides database. Protein Pept Lett 2002: 9: 5357. 245. Wang M, Liu L-H, Wang S, Li X, Lu X, Gupta D, Dziarski R. Human peptidoglycan recognition proteins require zinc to kill both gram-positive and gram-negative bacteria and are synergistic with antibacterial peptides. J Immunol 2007: 178: 31163125. 246. Wang PL, Azuma Y, Shinohara M, Ohura K. Effect of Actinobacillus actinomycetemcomitans protease on the proliferation of gingival epithelial cells. Oral Dis 2001: 7: 233237. Wang Z, Wang G. APD: the Antimicrobial Peptide Database. Nucleic Acids Res 2004: 32: D590D592. Wei G-X, Campagna AN, Bobek LA. Effect of MUC7 peptides on the growth of bacteria and on Streptococcus mutans biolm. J Antimicrob Chemother 2006: 57: 1100 1109. Wei GX, Campagna AN, Bobek LA. Factors affecting antimicrobial activity of MUC7 12-mer, a human salivary mucin-derived peptide. Ann Clin Microbiol Antimicrob 2007: 6: 14. Weinberg ED. Antibiotic properties and applications of lactoferrin. Curr Pharm Des 2007: 13: 801811. Weiss J, Elsbach P, Shu C, Castillo J, Grinna L, Horwitz A, Theofan G. Human bactericidal permeability-increasing protein and a recombinant NH2-terminal fragment cause killing of serum-resistant gram-negative bacteria in whole blood and inhibit tumor necrosis factor release induced by the bacteria. J Clin Invest 1992: 90: 11221130. Weldon S, McGarry N, Taggart CC, McElvaney NG. The role of secretory leucoprotease inhibitor in the resolution of inammatory responses. Biochem Soc Trans 2007: 035: 273276. Weston WM, LeClair EE, Trzyna W, McHugh KM, Nugent P, Lafferty CM, Ma L, Tuan RS, Greene RM. Differential display identication of plunc, a novel gene expressed in embryonic palate, nasal epithelium, and adult lung. J Biol Chem 1999: 274: 1369813703. Wheeler TT, Haigh BJ, McCracken JY, Wilkins RJ, Morris CA, Grigor MR. The BSP30 salivary proteins from cattle, LUNX PLUNC and von Ebners minor salivary gland protein are members of the PSP LBP superfamily of proteins. Biochim Biophys Acta 2002: 1579: 92100. Wheeler TT, Hood KA. The mammalian innate immune system: potential targets for drug development. Curr Drug Targets Immune Endocr Metabol Disord 2005: 5: 237247. White MR, Crouch E, van Eijk M, Hartshorn M, Pemberton L, Tornoe I, Holmskov U, Hartshorn KL. Cooperative antiinuenza activities of respiratory innate immune proteins and neuraminidase inhibitor. Am J Physiol Lung Cell Mol Physiol 2005: 288: L831L840. Whitelegge J, Zabrouskov V, Halgand F, Souda P, Bassilian S, Yan W, Wolinsky L, Loo J, Wong D, Faull K. Proteinsequence polymorphisms and post-translational modications in proteins from human saliva using top-down Fourier-transform ion cyclotron resonance mass spectrometry. Int J Mass Spectrom 2007: 268: 190197. Wilde C, Grifth J, Marra M, Snable J, Scott R. Purication and characterization of human neutrophil peptide 4, a novel member of the defensin family. J Biol Chem 1989: 264: 1120011203. Williams SE, Brown TI, Roghanian A, Sallenave J-M. SLPI and elan: one glove, many ngers. Clin Sci 2006: 110: 21 35. Wilmarth P, Riviere M, Rustvold D, Lauten J, Madden T, David L. Two-dimensional liquid chromatography study of the human whole saliva proteome. J Proteome Res 2004: 3: 10171023. Wolff A, Begleiter A, Moskona D. A novel system of human submandibular sublingual saliva collection. J Dent Res 1997: 76: 17821786.

247. 248.

249.

250. 251.

252.

253.

254.

255.

256.

257.

258.

259.

260.

261.

179

Gorr
262. Wu Y-M, Juo S-H, Ho Y-P, Ho K-Y, Yang Y-H, Tsai C-C. Association between lactoferrin gene polymorphisms and aggressive periodontitis among Taiwanese patients. J Periodontal Res 2009: 44: 418424. 263. Wu Z, Van Ryk D, Davis C, Abrams WR, Chaiken I, Magnani J, Malamud D. Salivary agglutinin inhibits HIV type 1 infectivity through interaction with viral glycoprotein 120. AIDS Res Hum Retroviruses 2003: 19: 201209. 264. Xie H, Rhodus NL, Grifn RJ, Carlis JV, Grifn TJ. A catalogue of human saliva proteins identied by free ow electrophoresis-based peptide separation and tandem mass spectrometry. Mol Cell Proteomics 2005: 4: 18261830. 265. Yount NY, Yeaman MR. Multidimensional signatures in antimicrobial peptides. Proc Natl Acad Sci U S A 2004: 101: 73637368. 266. Zasloff M. Innate immunity, antimicrobial peptides, and protection of the oral cavity. The Lancet 2002: 360: 1116 1117. 267. Zhang L, Yu W, He T, Yu J, Caffrey RE, Dalmasso EA, Fu S, Pham T, Mei J, Ho JJ, Zhang W, Lopez P, Ho DD. Contribution of human alpha-defensin 1, 2, and 3 to the antiHIV-1 activity of CD8 antiviral factor. Science 2002: 298: 9951000. 268. Zhou HD, Li XL, Li GY, Zhou M, Liu HY, Yang YX, Deng T, Ma J, Sheng SR. Effect of SPLUNC1 protein on the Pseudomonas aeruginosa and Epstein-Barr virus. Mol Cell Biochem 2008: 309: 191197. 269. Zhou HD, Wu MH, Shi L, Zhou M, Yang YX, Zhao J, Deng T, Li XL, Sheng SR, Li GY. [Effect of growth inhibition of the secretory protein SPLUNC1 on Pseudomonas aeruginosa]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2006: 31: 464469. 270. Zhu S. Did cathelicidins, a family of multifunctional hostdefense peptides, arise from a cysteine protease inhibitor? Trends Microbiol 2008: 16: 353360.

180

You might also like