Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 May 23.
Published in final edited form as: Curr Pharm Des. 2013;19(1):40–47. doi: 10.2174/13816128130108

Antimicrobial Peptides and Colitis

Samantha Ho 1, Charalabos Pothoulakis 1, Hon Wai Koon 1,*
PMCID: PMC3662473  NIHMSID: NIHMS468154  PMID: 22950497

Abstract

Antimicrobial peptides (AMPs) are important components of innate immunity. They are often expressed in response to colonic inflammation and infection. Over the last several years, the roles of several antimicrobial peptides have been explored. Gene expression of many AMPs (beta defensin HBD2-4 and cathelicidin) is induced in response to invasion of gut microbes into the mucosal barrier. Some AMPs are expressed in a constitutive manner (alpha defensin HD 5-6 and beta defensin HBD1), while others (defensin and bactericidal/permeability increasing protein BPI) are particularly associated with Inflammatory Bowel Disease (IBD) due to altered defensin expression or development of autoantibodies against Bactericidal/permeability increasing protein (BPI). Various AMPs have different spectrum and strength of antimicrobial effects. Some may play important roles in modulating the colitis (cathelicidin) while others (lactoferrin, hepcidin) may represent biomarkers of disease activity. The use of AMPs for therapeutic purposes is still at an early stage of development. A few natural AMPs were shown to be able to modulate colitis when delivered intravenously or intracolonically (cathelicidin, elafin and SLPI) in mouse colitis models. New AMPs (synthetic or artificial non-human peptides) are being developed and may represent new therapeutic approaches against colitis. This review discusses the latest research developments in the AMP field with emphasis in innate immunity and pathophysiology of colitis.

Keywords: Antimicrobial peptides, colitis, infection, microflora, protein, Crohn’s disease, ulcerative colitis

INTRODUCTION

Antimicrobial peptides (AMPs) are endogenous antibiotics with antimicrobial activities. They are generally expressed in the intestinal lining in close contact with the gut microflora. AMPs are expressed in a constitutive or inducible manner in intestinal epithelial cells, and Paneth and immune cells, respectively. Over the last few years, many endogenous AMPs have been studied for their expression during intestinal inflammation in Crohn’s disease (CD) or Ulcerative colitis (UC) as well as during infections such as E. Coli., C. difficile or Amoeba.

Many AMPs, in addition to their antimicrobial effects, can also modulate immune responses. One AMP, hepcidin, can act as a hormone and regulate iron metabolism. Many AMPs are expressed during inflammatory processes and can serve as disease biomarkers, as in the case of Inflammatory Bowel Disease (IBD). Since the pathophysiology of colitis involves interactions between the gut microflora and the host mucosa, recent reports suggested that AMP induction is associated with dysfunctional gut barrier and involvement of bacterial components.

This review article summarizes recent findings of several major antimicrobial peptides found in the ileum and colon and discusses their role in the pathophysiology of intestinal inflammation of different etiologies.

DEFENSIN FAMILY

Defensin is a large group of 10 peptides in humans. Defensins represent an important part of the gut’s innate immune response and they are secreted from Paneth cells, epithelial cells, as well as immune cells. Defensins are classified as alpha defensin and beta-defensin based on their molecular distribution of cysteine amino acids and the resulting disulfide bonds [1]. Defensins are further classified as constitutive (expression remains unchanged during inflammation or infection) and inducible (increased expression during inflammation or infection) [1, 2].

Human Alpha Defensin (HNP1-4)

Human alpha defensins 1-4 (HNP1-4), also called human neutrophil peptides, are primarily secreted from neutrophils [3]. They appear to contribute to innate immunity at the systemic level as neutrophils can circulate around the whole body with a broad spectrum of antibacterial activity against many pathogens [4]. HNP-1 had been shown to block LPS induced IL-1β release from monocytes, suggesting anti-inflammatory effects against this endotoxin [5]. But another study showed that intraperitoneal administration of HNP-1 to mice with DSS-induced colitis leads to more severe colitis with higher colonic cytokine levels compared to controls, suggesting a potential pro-inflammatory role for HNP-1 in colitis [6]. On the other hand, HNP-1 and HNP-3 had been shown to inhibit cytotoxicity and Rho glucosylation in Caco-2 cells caused by C. difficile toxin B, but not toxin, A while beta defensin had no such protective effect [7]. Interestingly, HNP1-3 protein is expressed in active IBD mucosa and this response may be associated with increased neutrophil infiltration into IBD tissues [8]. Plasma concentrations of HNP1-3 are also significantly increased in IBD patients, but not normal subjects, possibly due to increased HNP expression from circulating neutrophils [9, 10]. Up to now, there is no report indicating a role for HNP-4 in IBD or any other form of colitis, although its anti-bacterial effects are stronger compared to HNP1-3 [11].

Human Alpha Defensin (HD5 and HD6)

Another group of human alpha defensin (HD5 and HD6) is expressed only in Paneth cells of the human duodenum, jejunum and ileum [3]. HD-5 and HD-6 are not expressed in normal adult colon, possibly due to the lack of Paneth cells [12]. But in ileal CD patients, expression of HD-5 and HD-6 is reduced, compared to ileum of control subjects [13, 14]. Although one study suggested that NOD2 mutations may be associated with reduced expression of alpha defensin (HD-5 and HD-6) [15], another study did not find a mechanistic link between NOD2 and Paneth cell alpha defensin expression [16]. Interestingly, HD-5 is also expressed in metaplastic Paneth cells in the colons of IBD patients [17, 18], which may represent a self-defense response to bacterial challenges during colitis.

Paneth cell alpha defensin has multiple roles in infection and inflammation. Mature HD-5 shows bactericidal activities to all bacterial strains. It also induces IL-8 expression in intestinal epithelial cells and injection of exogenous HD-5 reduces mortality in mice with DSS colitis [19]. Transgenic mice overexpressing HD-5 are highly resistant to enteric Salmonella infection [20]. HD-5 can also inhibit C. difficile toxin B cytotoxicity in intestinal epithelial Caco2 cell monolayers by inhibiting toxin B-catalyzed Rho glucosylation [7].

Mouse Alpha Defensin (cryptdin)

Mouse cryptdins (Crps), like HD-5 and HD-6, are expressed in Paneth cells. The precursor forms of Crps are localized in Paneth cell granules and processed into their microbicidal form by interacting with matrix metalloproteinase-7 (MMP-7) [21, 22]. Thus, Crps preparations from MMP7 −/− mice have decreased antimicrobial activity 21, while MMP-7 deficient mice are more susceptible to colitis [5]. Crps, like other alpha defensins, are resistant to proteolysis and may remain functional in the ileum to colon [23, 24]. There are 6 isoforms of cryptdin with cryptdin-4 being the most bactericidal against many gut bacteria species [25, 26]. Although Salmonella infection inhibit Cryptdin expression in gut Paneth cells [27], cryptdin-2 is effective in treating Salmonella Typhimurium infection in mice [28].

Human beta-defensin 1 (HBD1)

Human beta defensin 1 (HBD-1) is constitutively expressed in colonic and ileal epithelium of humans regardless of colitis [29, 30]. Even challenges with IL-1α and/or E.coli still fail to alter the mRNA expression of hBD-1 in human colonic epithelial Caco-2 and HT-29 cells. A study showed that Peroxisome proliferator-activated receptor gamma (PPARγ) directly regulates DEFB1 expression in human colonic Caco-2. PPARγ deficient mice express much less beta-defensin mDefB10 in the colonic mucosa, with defective killing of Candida albicans, Bacteroides fragilis, Enterococcus faecalis and E. coli [31], suggesting that this defensin may play a role in colonic infection and inflammation.

INDUCIBLE human beta-defensin (HBD-2, -3, -4)

Human beta-defensin 2 (HBD-2) is barely expressed in normal colon but its expression is significantly increased in inflamed colonic epithelium of IBD patients [30]. Despite this increase, however, plasma levels of HBD-2 in IBD patients remain unchanged [9]. Unlike constitutive HBD-1, exposure of human colonic epithelial Caco-2 and HT-29 cells to proinflammatory IL-1α and/or enteroin-vasive E.coli (O29:NM) significantly increase HBD-2 expression, suggesting an important role for HBD-2 in the pathophysiology of colitis and colitis-associated microflora. Fahlgren et al also found that HBD-3 and HBD-4 expression are similarly increased in colonic crypts of UC, but not CD patients [32]. Mouse beta defensin-3 (analogue of HBD-2 in humans) is strongly up-regulated in the colonic epithelium of mice with chronic experimental, DSS-induced colitis [33]. However, unlike HD-5, beta defensin has no protective effect against cytotoxicity of C. difficile toxin B [7].

Theta Defensin (Not Existing Naturally in Humans)

Theta defensin protein is not expressed in humans because of a stop codon at the human theta defensin DNA [34]. But synthetically modified theta defensin (retrocyclin) possesses remarkable antibacterial activity and excellent antiviral activity against HIV [35]. There is no report showing a therapeutic role for theta defensin in colitis in vivo. However, compared to alpha defensin HNP-1, another modified theta defensin (RC-1) exerts an even greater inhibition of intracellular growth of the gut pathogen Listeria monocytogenes within macrophages [36].

CATHELICIDIN

Cathelicidins are a family of peptides with established antibacterial, anti-viral and anti-fungal effects [37, 38]. LL-37 and mCRAMP are the only forms of cathelicidin in humans and mice, respectively. Cathelicidin is located on chromosome 3 and contains 4 exons [39, 40]. Although the exact antimicrobial mechanism of cathelicidin is not fully understood, it is established that LL-37 is able to form transmembrane pores which can penetrate bacteria [41]. Cathelicidin permeabilizes the bacterial cell membrane and inhibits cell wall biogenesis leading to inhibition of bacterial cell growth, as in the case of E. coli [42].

Cathelicidin peptides are secreted from surfaces exposed to the external environment, including the gut. In addition, cathelicidins are present in amniotic fluid and breast milk. Western blot analysis of human milk showed that the mature form of LL-37 is present in human milk. LL-37 also possesses antimicrobial activity against Staphylococcus aureus, group A Streptococcus and enteroinvasive E. coli O29, common pediatric pathogens [43]. This indicates that LL-37 can confer immune protection to fetuses and newborns before full autonomous immunity is established.

In a total 89 normal and IBD patients, colonic cathelicidin Camp mRNA expression is significantly increased, but only in UC, not CD patients [44]. There is also no significant difference of cathelicidin expression levels among different NOD2 gene polymorphism status or severity of inflammation in CD patients [44]. In the colonic mucosa, cathelicidin is typically expressed at the top of colonic crypts but not in deeper crypts and this expression is similar among normal and IBD patients. However, such cathelicidin induction is unrelated to the presence of pro-inflammatory cytokines in IBD patients, since TNFα, IFNγ, LPS, IL-12, IL-4 and IL-13 are not able to induce cathelicidin expression in human colonic epithelial HT-29 cells [44].

There is some evidence showing that the mechanism of cathelicidin expression involves stimulation by bacterial components. For example, the short-chain fatty acid butyrate, a bacteria metabolite, is a well established inducer of cathelicidin. Sodium butyrate belongs to HDAC inhibitors family and therefore is not surprising that another HDAC inhibitor, Trichostatin A induces expression of cathelicidin [45]. In addition, the transcription factor PU.1 of the Ets family binds to Camp promoter segments and triggers Camp gene expression in HT-29 cells [46]. Upon cell stimulation by vitamin D, butyrate, or the secondary bile acid lithocholic acid, both Vitamin D receptor and PU.1 are recruited to the Camp promoter thereby enhancing cathelicidin gene transcription [46]. Another study showed that vitamin D, but not lithocholic acid induces cathelicidin mRNA and protein expression in human colonic Caco-2 cells [47], although another group failed to reproduce this response [48].

Toll-like receptors (TLRs) are sensors of pathogen-associated molecular patterns. Several TLR ligands stimulate cathelicidin in several different cell types, including macrophages [49]. Koon et al recently found that cathelicidin deficient Camp−/− mice develop worse experimental acute colitis than wild-type mice [50]. Administration of bacterial DNA induces colonic cathelicidin expression in normal mice as well as in mice with DSS-induced colitis. Bacterial DNA, a known TLR9 ligand, stimulates LL-37 expression in primary human monocytes via an ERK1/2-dependent mechanism. Further, bone marrow transplantation experiments demonstrated that expression of cathelicidin from bone-marrow derived immune cells plays an important role in the development of DSS-induced colitis in mice [50].

Therapeutic Effects of Cathelicidin

As shown by Koon et al, endogenous cathelicidin plays an anti-inflammatory role in the development of DSS colitis in mice [50]. Tai et al used intracolonic administration of the cathelicidin mCRAMP to treat mouse colitis in the same model [51]. Intrarectal administration of mCRAMP to mice with DSS colitis significantly ameliorated several colonic inflammatory parameters, restored colonic mucus thickness through increased expression of mucin genes (MUC1-4), and suppressed colonic apoptosis with negligible effects on mucosal healing. Importantly, mCRAMP administration reduced the total population of fecal microflora, suggesting significant antimicrobial effects. In vitro experiments showed that LL-37 has no effects on cell proliferation, but exerts anti-apoptotic and wound healing effects in human intestinal epithelial HT-29 and Caco-2 cells [52]. One of the putative LL-37 receptors, P2X7 is expressed in primary intestinal epithelial cells and Caco-2 cells but not HT-29 cells [52]. LL-37 induces mucin gene expression via P2X7-dependent pathway [52].

Induction of endogenous cathelicidin may also produce similar therapeutic effects in models of infection. Oral administration of butyrate or phenylbutyrate to a rabbit model of shigellosis significantly increased cathelicidin mRNA and protein expression in colonic and rectal mucosa, a response associated with reduced clinical signs of infection [53, 54]. Despite these positive findings, however, both human and mouse cathelicidin failed to kill Entamoeba histolytica and did not ameliorate colitis in response to this pathogen in a mouse model. Entamoeba histolytica releases a cysteine protease that cleaves cathelicidin, leading to degradation of this antimicrobial peptide and resistance to killing [55].

In conclusion, cathelicidin may be a potential therapeutic solution for colitis, at least during acute colitis states, while its role in chronic colitis remains to be evaluated. Pro-angiogenic effects, alterations of the gut microflora, and participation of the putative cathelicidins receptors FPRL1 or P2X in the development of colitis remain to be evaluated.

PROTEASE INHIBITORS: ELAFIN AND SECRETORY LEUKOCYTE PEPTIDASE INHIBITOR (SLPI)

During colitis, a delicate balance between proteases and anti-protease responses determines in part the development of inflammation [56]. In inflammatory states, proteases damage tissues while protease inhibitors stabilize tissue damage and facilitate healing. Elafin is a protease inhibitor with anti-bacterial effects [57] that modulates inflammation via its anti-protease activity [58]. In a microarray study of human colonic biopsies, UC patients expressed approximately 30 times more elafin mRNA compared to the healthy controls [59]. Such elevation of elafin occurs in rectal epithelium, including goblet cells and enterocytes in patients with total colon UC [59]. Another report also showed similar results as elafin was expressed in active, but not inactive inflamed colonic mucosa of UC patients [60]. Surprisingly, increased elafin expression is not evident in CD patients [60]. Expression of another anti-protease equivalent, SLPI, was also elevated in inflamed UC mucosa, but not in non-inflamed UC mucosa or colon of CD patients [60]. It is possible that low expression of the anti-protease elafin and SLPI match with high expression of MMPs in CD which leads to high risk of fistula. In UC patients, elafin and SLPI levels are high and may tend to act as a self-protective mechanism against colitis [60]. SLPI is expressed in human jejunum and colonic biopsies as well as human colonic epithelial Caco-2-BBE, T84, and HT29-Cl.19A cells [61]. Although SLPI exerts direct antimicrobial effects including against Salmonella typhimurium, it does not affect epithelial barrier integrity [61].

In both the DSS and the TNBS colitis models, overexpression of elafin by adenovirus delivery significantly ameliorates colitis that is associated with reduced colonic proteolytic activity, and diminished cytokine levels and NF-κB activation [62]. Overexpression of elafin also significantly reduced TNFα mediated increased permeability of Caco-2 cells, suggesting a beneficial effect in epithelial barrier integrity [62]. Elafin can decrease IL-8 secretion and NF-κB luciferase activity induced by TNFα or LPS in HT-29 cells, indicating potent anti-inflammatory effects [62]. SLPI may also promote healing during colitis. Thymic stromal lymphopoetin (Tslp) deficient mice had reduced expression of SLPI [63]. Interestingly, these deficient mice developed colitis during the inflammatory stage to a similar degree to wild-type mice, but failed to recover from colitis, resulting in increased mortality [63]. Administration of recombinant SLPI to Tslp deficient mice reduced DSS colitis- associated mortality, indicating an important role for SLPI for mucosal healing following colitis.

BACTERICIDAL/PERMEABILITY INCREASING PROTEIN (BPI)

BPI can kill gram negative bacteria via binding to LPS and inhibit LPS-induced host cell toxic responses [64]. Its SNP (GLU216Lys) genotype is associated with CD but not UC [65], suggesting that it may be involved in impaired defense against gram-negative bacteria in CD patients. BPI expression is increased in the colonic mucosa of UC patients, compared to controls [66]. BPI is generally localized in PMN cells in the mucosa and stroma of colons and its concentration is correlated with disease activity in UC patients [66]. Several human intestinal epithelial cells (Caco-2, T84 and SW480) also express BPI mRNA, while over-expression of BPI in Caco-2 cells reduces Salmonella induced IL-8 secretion, suggesting anti-inflammatory effects [67].

Some IBD patients develop anti-neutrophil cytoplasmic auto-antibodies (ANCA) which target BPI [68]. Most IgG of UC and CD patients can neutralize BPI resulting in reduced antibiotic function of BPI [68]. BPI-targeting auto-antibodies in IBD patients is associated with greater mucosal damage and extent of disease [69] and thus may contribute to development of IBD.

HEPATOCARCINOMA-INTESTINE-PANCREAS (HIP) / PANCREATITIS-ASSOCIATED PROTEIN (PAP)

HIP/PAP belongs to the Rag family and RagIII subfamily. HIP/PAP is expresses in colorectal cancer [70]. In adults, HIP/PAP is expressed in Paneth cells at the base of intestinal pits and endocrine cells around the epithelial lining of the jejunum, ileum and ascending colon [71]. These cells coexpress the endocrine cell marker chromogranin A and synaptophysin [71]. HIP/PAP expression is significantly induced in colonic epithelial cells upon exposure to bacteria in germ free or colitic mice exposed to DSS [72]. HIP/PAP mRNA is also induced in the colonic epithelial cells of IBD patients [72]. HIP/PAP, as a C-type lectin, can kill bacteria by binding to peptidoglycan carbohydrate [73]. Although HIP/PAP possesses antibacterial activity [74], its role in the development of colitis is not fully understood.

LYSOZYME

Lysozyme is a common antimicrobial protein which damages bacterial cell wall by hydrolysis of peptidoglycans [75]. Lysozyme is secreted from polymorphonuclear (PMN) cells and exists in a wide range of host secreted products, such as mucus [76], tears [77] and milk [78]. Colonic epithelial cells of UC patients express significantly higher lysozyme mRNA than controls [18], but fecal lysozyme may not be the best indicator of UC, compared to other fecal IBD biomarkers [79]. In CD patients, lysozyme mRNA is also found in colonic, but not ileal epithelial cells and chronic colonic inflammation results in increased lysozyme expression [18]. In a model of colitis in pigs, hen egg lysozyme infusion reduces DSS-induced colitis, a response associated with reduced colonic TNFα and IL-6 expression [80]. Like cathelicidin, hen egg lysozyme also increases mucin gene expression which promotes colonic barrier integrity [80].

LACTOFERRIN

Lactoferrin is also called lactotransferrin as it belongs to transferring family which consists of protein and iron. Lactoferrin is abundant in milk, earning its lacto- prefix [78]. With its iron binding property, lactoferrin exert its multiple antibacterial effects via deprivation of iron from pathogens [81]. Lactoferrin binds to the LPS layer of bacterial cell wall and causes increased membrane permeability and bacterial cell lysis [82]. Iron in the bacterial cell may kill the cell via peroxide formation [83]. Alternatively, lactoferrin may also stimulate phagocytosis in immune cells [84].

Multiple reports suggest that fecal lactoferrin is a non-invasive biomarker of IBD as its levels are significantly increased in IBD, but not IBS patients [85]. Like another IBD marker, calprotectin, both are neutrophil derived-indicators of disease activity [86]. Lactoferrin is simple and inexpensive to detect and has excellent stability in feces for an extended period of time. One report showed that a decrease of fecal lactoferrin may be correlated to mucosal healing and response to therapy [87]. Apart from IBD, fecal lactoferrin levels are also increased along with levels of IL-1β and IL-8 in patients with severe C. difficile colitis [88], as well as patients with Enterohemorrhagic E. coli infection [89].

The therapeutic value of oral lactoferrin administration is underscored by its ability to dose-dependently ameliorate DSS-induced colitis in rats [90]. After oral bovine lactoferrin treatment, colonic expression of anti-inflammatory cytokines, IL-4 and IL-10 are increased, while expression of proinflammatory cytokines TNFα, IL-1β and IL-6, histology damage and MPO levels are improved [90]. Such oral bovine lactoferrin treatment has similar beneficial effects to TNBS colitis in rats [91]. Lactoferrin-derived lactoferricin and lactoferrampin had been shown to kill Entamoeba histolytica in and this may help reduce use of the antibiotic metronidazole that is associated with several side effects [92]. Bovine Lactoferrin also exert anti-inflammatory effects as it reduces IL-8 secretion from Caco-2 cells infected with E. coli HB101 [93].

HEPCIDIN

Hepcidin plays an important regulatory role in iron homeostasis while it also possesses antimicrobial properties [94]. It limits the availability of iron to bacteria, thus withholding the growth of invading pathogens [95]. Hepcidin can also control intracellular Salmonella and Mycobacteria by modulating iron availability within macrophages. Prohepcidin is a precursor form of hepcidin. Acting as a hormone, hepcidin is generally secreted from the liver and inhibits iron absorption from the gut. Although hepcidin has antibacterial effects [81], no anti-bacterial role in colitis has been reported.

One study showed that serum hepcidin levels are significantly higher in both UC and CD patients, compared to healthy subjects [96]. Serum hepcidin was also correlated with disease activity and C-reactive protein levels in UC patients [96]. Serum hepcidin is negatively correlated, while serum prohepcidin is positively correlated with hemoglobin levels [96], suggesting that hepcidin and prohepcidin may be related to anemia associated with IBD. However, another group found that hepcidin expression to be dependent on bone morphogenetic protein (BMP) and IL-6. Using the T-cell transfer mouse model of colitis, an anti-BMP reagent could inhibit hepcidin expression, thus increasing serum iron levels in mice with colitis [97]. Therefore, inhibition of hepcidin may correct IBD associated anemia and help reduce colonic inflammation.

NOVEL OR ARTIFICIAL AMPS IN INFLAMMATION

Apart from natural endogenous antimicrobial peptides, there are many non-host and artificial ant-microbial peptides possessing anti-inflammatory effects in various kinds of colitis. The nine amino acid peptide coprisin (LLCIALRKK), that derives from Korean dung beetle, exerts antimicrobial activity and prevents mice from C. difficile -associated inflammation and mucosal damage in mice [98]. A modified coprisin analogue does not affect commensal bacteria such as lactobacillus and bifidobacterium, but inhibits colonization of C. difficile in mice via a mechanism that involves disruption of the bacterial membrane [98]. Two semi-synthetic glycopeptides, telavancin and dalbavancin with antibacterial activity against Gram-positive bacteria have been evaluated in clinical trials for their oral effectiveness in C. difficile colitis and digestive tract decontamination [99]. A novel antimicrobial peptide (wrwycr) was recently found to inhibit bacterial DNA repair –associated mechanisms [100]. This peptide significantly reduces survival of Shiga toxin producing O157-H7 E. coli in acid stress [100]. Although data from animal experiments or clinical trials are not available, this peptide may be another potential candidate for prevention of intestinal bacterial infection.

One difficulty in developing new antimicrobial peptides relates to the lack of consensus about the essential structure contributing to their antimicrobial and/or anti-inflammatory activity. Among all of the antimicrobial peptides discussed above, their amino acid sequence, molecular size and structure are largely different [101]. It is possible to alter individual amino acid(s) in a short AMP sequence to understand how the peptide sequence affects its antimicrobial activity, but this approach is very difficult for larger protein molecules [102, 103]. As cathelicidin is short (37 amino acids) and easy to manipulate, there are many reports studying antimicrobial effects of altered forms of cathelicidin [104106]. Up to now, however, there is no clear way to predict how to design a novel antimicrobial peptide based on our current knowledge of AMPs.

CONCLUDING REMARKS

Expression of most of endogenous antimicrobial peptides and proteins are increased during colitis or colonic infection (Table 1). Some AMPs act as disease markers of colitis that predicts disease activity or response to therapy. The intimate association of AMP, microflora and immune regulation in the gut is still being extensively investigated. A few of AMPs had shown potential therapeutic effects in animal models by administration of exogenous antimicrobial peptides or protein. Development of new antimicrobial peptides for treating colitis is at an early stage and more information on peptide/protein stability, delivery method, efficacy and safety is much needed. Since many AMPs are unstable in blood, AMP gene therapy via viral vectors may be another possible way for prolonged expression of AMP in the host for long-term treatment. But safety of AMPs and its delivery methods still require more research as no human clinical trial data of AMP in colitis is available. Alternatively, induction of endogenous AMPs may be another interesting future research direction of innate immunity and protection of the host. The associated anti-inflammatory effects of antimicrobial peptides and proteins represent an important correlative advantage in developing new AMPs for pharmaceutical purposes.

Table 1.

Overview of Antimicrobial Peptides in Colitis

AMP(s) Expression Roles in IBD/colitis Roles in bacterial infection Roles in C. difficile toxin inflammation
HNP1-4 Neutrophils:
Increase possibly due to neutrophil infiltration in colons.
HNP-1 is protective against DSS colitis in mice. Protective against many strains of bacteria and LPS HNP-1 and HNP-3 are protective against toxin B
HD-5, HD-6 Constitutive expression in ileal Paneth cells.
Decreased in CD, further reduced with NOD2 mutation.
Protective against DSS colitis in mice. Against all strains HD-5 is protective against toxin B.
Cryptdin Ileal Paneth cells in mice only Not known Against Salmonella Not known
HBD-1 Constitutive in colonic epithelium Not changed in colitis Against many strains of bacteria Not known
HBD-2-4 Colonic epithelium, increase in colitis. Stimulated by IL-1α Stimulated by LPS Not protective against toxin B
Theta defensin Not expressed in humans Not known Against Listeria monocytogenes by retrocyclin Not known
Cathelicidin Colon epithelium, increase in UC but not CD Inhibit DSS colitis, Camp−/− mice have more serious DSS colitis.
Anti-inflammatory.
Bactericidal against many strains of bacteria including Shigella but not Entamoeba histolytica Not protective against toxin B
Elafin/SLPI Colonic epithelium, increase in UC but not CD Protective against DSS colitis, possibly anti- inflammatory. At least bactericidal against Salmonella Not known
BPI Colonic epithelium, increase in UC Compromised by ANCA antibody, possibly anti- inflammatory. Kill gram negative bacteria. Not known
HIP/PAP Paneth cells, endocrine cells Increased in IBD/colitis Against many strains of bacteria Not known
Lyzozyme PMN cells, increased in IBD Protective against DSS colitis Against many strains of bacteria Not known
Lactoferrin Neutrophils, Increased in IBD, E. coli and C. difficile infection Protective against DSS and TNBS colitis Protective against Entamoeba histolytica and E. coli Not known
Hepcidin From liver, act like hormone, serum hepcidin level increases in IBD. Associated with IBD anemia May be not important Not known

Acknowledgments

ACKNOWLEDGEMENT OF FUNDING

NIH funding DK084256, CCFA career development grant to Hon Wai Koon.

Footnotes

Send Orders of Reprints at reprints@benthamscience.org

CONFLICT OF INTEREST

The authors confirm that this article content has no conflicts of interest.

References

  • 1.Cederlund A, Gudmundsson GH, Agerberth B. Antimicrobial peptides important in innate immunity. FEBS J. 278:3942–51. doi: 10.1111/j.1742-4658.2011.08302.x. [DOI] [PubMed] [Google Scholar]
  • 2.Ramasundara M, Leach ST, Lemberg DA, Day AS. Defensins and inflammation: the role of defensins in inflammatory bowel disease. J Gastroenterol Hepatol. 2009;24:202–8. doi: 10.1111/j.1440-1746.2008.05772.x. [DOI] [PubMed] [Google Scholar]
  • 3.Cunliffe RN. Alpha-defensins in the gastrointestinal tract. Mol Immunol. 2003;40:463–7. doi: 10.1016/s0161-5890(03)00157-3. [DOI] [PubMed] [Google Scholar]
  • 4.Ganz T, Selsted ME, Szklarek D, Harwig SS, Daher K, Bainton DF, Lehrer RI. Defensins. Natural peptide antibiotics of human neutrophils. J Clin Invest. 1985;76:1427–35. doi: 10.1172/JCI112120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Shi J, Aono S, Lu W, Ouellette AJ, Hu X, Ji Y, Wang L, Lenz S, van Ginkel FW, Liles M, Dykstra C, Morrison EE, Elson CO. A novel role for defensins in intestinal homeostasis: regulation of IL-1beta secretion. J Immunol. 2007;179:1245–53. doi: 10.4049/jimmunol.179.2.1245. [DOI] [PubMed] [Google Scholar]
  • 6.Hashimoto S, Uto H, Kanmura S, Sakiyama T, Oku M, Iwashita Y, Ibusuki R, Sasaki F, Ibusuki K, Takami Y, Moriuchi A, Oketani M, Ido A, Tsubouchi H. Human neutrophil peptide-1 aggravates dextran sulfate sodium-induced colitis. Inflamm Bowel Dis. doi: 10.1002/ibd.21855. [DOI] [PubMed] [Google Scholar]
  • 7.Giesemann T, Guttenberg G, Aktories K. Human alpha-defensins inhibit Clostridium difficile toxin B. Gastroenterology. 2008;134:2049–58. doi: 10.1053/j.gastro.2008.03.008. [DOI] [PubMed] [Google Scholar]
  • 8.Cunliffe RN, Kamal M, Rose FR, James PD, Mahida YR. Expression of antimicrobial neutrophil defensins in epithelial cells of active inflammatory bowel disease mucosa. J Clin Pathol. 2002;55:298–304. doi: 10.1136/jcp.55.4.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Yamaguchi N, Isomoto H, Mukae H, Ishimoto H, Ohnita K, Shikuwa S, Mizuta Y, Nakazato M, Kohno S. Concentrations of alpha- and beta-defensins in plasma of patients with inflammatory bowel disease. Inflamm Res. 2009;58:192–7. doi: 10.1007/s00011-008-8120-8. [DOI] [PubMed] [Google Scholar]
  • 10.Kanmura S, Uto H, Numata M, Hashimoto S, Moriuchi A, Fujita H, Oketani M, Ido A, Kodama M, Ohi H, Tsubouchi H. Human neutrophil peptides 1–3 are useful biomarkers in patients with active ulcerative colitis. Inflamm Bowel Dis. 2009;15:909–17. doi: 10.1002/ibd.20854. [DOI] [PubMed] [Google Scholar]
  • 11.Ericksen B, Wu Z, Lu W, Lehrer RI. Antibacterial activity and specificity of the six human {alpha}-defensins. Antimicrob Agents Chemother. 2005;49:269–75. doi: 10.1128/AAC.49.1.269-275.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mallow EB, Harris A, Salzman N, Russell JP, DeBerardinis RJ, Ruchelli E, Bevins CL. Human enteric defensins. Gene structure and developmental expression. J Biol Chem. 1996;271:4038–45. doi: 10.1074/jbc.271.8.4038. [DOI] [PubMed] [Google Scholar]
  • 13.Wehkamp J, Salzman NH, Porter E, Nuding S, Weichenthal M, Petras RE, Shen B, Schaeffeler E, Schwab M, Linzmeier R, Feathers RW, Chu H, Lima H, Jr, Fellermann K, Ganz T, Stange EF, Bevins CL. Reduced Paneth cell alpha-defensins in ileal Crohn’s disease. Proc Natl Acad Sci U S A. 2005;102:18129–34. doi: 10.1073/pnas.0505256102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Elphick D, Liddell S, Mahida YR. Impaired luminal processing of human defensin-5 in Crohn’s disease: persistence in a complex with chymotrypsinogen and trypsin. Am J Pathol. 2008;172:702–13. doi: 10.2353/ajpath.2008.070755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wehkamp J, Harder J, Weichenthal M, Schwab M, Schaffeler E, Schlee M, Herrlinger KR, Stallmach A, Noack F, Fritz P, Schroder JM, Bevins CL, Fellermann K, Stange EF. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal alpha-defensin expression. Gut. 2004;53:1658–64. doi: 10.1136/gut.2003.032805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Simms LA, Doecke JD, Walsh MD, Huang N, Fowler EV, Radford-Smith GL. Reduced alpha-defensin expression is associated with inflammation and not NOD2 mutation status in ileal Crohn’s disease. Gut. 2008;57:903–10. doi: 10.1136/gut.2007.142588. [DOI] [PubMed] [Google Scholar]
  • 17.Cunliffe RN, Rose FR, Keyte J, Abberley L, Chan WC, Mahida YR. Human defensin 5 is stored in precursor form in normal Paneth cells and is expressed by some villous epithelial cells and by metaplastic Paneth cells in the colon in inflammatory bowel disease. Gut. 2001;48:176–85. doi: 10.1136/gut.48.2.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fahlgren A, Hammarstrom S, Danielsson A, Hammarstrom ML. Increased expression of antimicrobial peptides and lysozyme in colonic epithelial cells of patients with ulcerative colitis. Clin Exp Immunol. 2003;131:90–101. doi: 10.1046/j.1365-2249.2003.02035.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ishikawa C, Tanabe H, Maemoto A, Ito T, Watari J, Kono T, Fujiya M, Ashida T, Ayabe T, Kohgo Y. Precursor processing of human defensin-5 is essential to the multiple functions in vitro and in vivo. J Innate Immun. 2:66–76. doi: 10.1159/000242114. [DOI] [PubMed] [Google Scholar]
  • 20.Salzman NH, Ghosh D, Huttner KM, Paterson Y, Bevins CL. Protection against enteric salmonellosis in transgenic mice expressing a human intestinal defensin. Nature. 2003;422:522–6. doi: 10.1038/nature01520. [DOI] [PubMed] [Google Scholar]
  • 21.Wilson CL, Ouellette AJ, Satchell DP, Ayabe T, Lopez-Boado YS, Stratman JL, Hultgren SJ, Matrisian LM, Parks WC. Regulation of intestinal alpha-defensin activation by the metalloproteinase matrilysin in innate host defense. Science. 1999;286:113–7. doi: 10.1126/science.286.5437.113. [DOI] [PubMed] [Google Scholar]
  • 22.Ayabe T, Wulff H, Darmoul D, Cahalan MD, Chandy KG, Ouellette AJ. Modulation of mouse Paneth cell alpha-defensin secretion by mIKCa1, a Ca2+-activated, intermediate conductance potassium channel. J Biol Chem. 2002;277:3793–800. doi: 10.1074/jbc.M107507200. [DOI] [PubMed] [Google Scholar]
  • 23.Mastroianni JR, Ouellette AJ. Alpha-defensins in enteric innate immunity: functional Paneth cell alpha-defensins in mouse colonic lumen. J Biol Chem. 2009;284:27848–56. doi: 10.1074/jbc.M109.050773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ouellette AJ. Paneth cell alpha-defensins in enteric innate immunity. Cell Mol Life Sci. 68:2215–29. doi: 10.1007/s00018-011-0714-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ouellette AJ, Satchell DP, Hsieh MM, Hagen SJ, Selsted ME. Characterization of luminal paneth cell alpha-defensins in mouse small intestine. Attenuated antimicrobial activities of peptides with truncated amino termini. J Biol Chem. 2000;275:33969–73. doi: 10.1074/jbc.M004062200. [DOI] [PubMed] [Google Scholar]
  • 26.Masuda K, Sakai N, Nakamura K, Yoshioka S, Ayabe T. Bactericidal activity of mouse alpha-defensin cryptdin-4 predominantly affects noncommensal bacteria. J Innate Immun. 3:315–26. doi: 10.1159/000322037. [DOI] [PubMed] [Google Scholar]
  • 27.Salzman NH, Chou MM, de Jong H, Liu L, Porter EM, Paterson Y. Enteric salmonella infection inhibits Paneth cell antimicrobial peptide expression. Infect Immun. 2003;71:1109–15. doi: 10.1128/IAI.71.3.1109-1115.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Preet S, Verma I, Rishi P. Cryptdin-2: a novel therapeutic agent for experimental Salmonella Typhimurium infection. J Antimicrob Chemother. 65:991–4. doi: 10.1093/jac/dkq066. [DOI] [PubMed] [Google Scholar]
  • 29.O’Neil DA, Porter EM, Elewaut D, Anderson GM, Eckmann L, Ganz T, Kagnoff MF. Expression and regulation of the human beta-defensins hBD-1 and hBD-2 in intestinal epithelium. J Immunol. 1999;163:6718–24. [PubMed] [Google Scholar]
  • 30.Wehkamp J, Fellermann K, Herrlinger KR, Baxmann S, Schmidt K, Schwind B, Duchrow M, Wohlschlager C, Feller AC, Stange EF. Human beta-defensin 2 but not beta-defensin 1 is expressed preferentially in colonic mucosa of inflammatory bowel disease. Eur J Gastroenterol Hepatol. 2002;14:745–52. doi: 10.1097/00042737-200207000-00006. [DOI] [PubMed] [Google Scholar]
  • 31.Peyrin-Biroulet L, Beisner J, Wang G, Nuding S, Oommen ST, Kelly D, Parmentier-Decrucq E, Dessein R, Merour E, Chavatte P, Grandjean T, Bressenot A, Desreumaux P, Colombel JF, Desvergne B, Stange EF, Wehkamp J, Chamaillard M. Peroxisome proliferator-activated receptor gamma activation is required for maintenance of innate antimicrobial immunity in the colon. Proc Natl Acad Sci U S A. 107:8772–7. doi: 10.1073/pnas.0905745107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Fahlgren A, Hammarstrom S, Danielsson A, Hammarstrom ML. beta-Defensin-3 and -4 in intestinal epithelial cells display increased mRNA expression in ulcerative colitis. Clin Exp Immunol. 2004;137:379–85. doi: 10.1111/j.1365-2249.2004.02543.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rahman A, Fahlgren A, Sundstedt C, Hammarstrom S, Danielsson A, Hammarstrom ML. Chronic colitis induces expression of beta-defensins in murine intestinal epithelial cells. Clin Exp Immunol. 163:123–30. doi: 10.1111/j.1365-2249.2010.04282.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Selsted ME. Theta-defensins: cyclic antimicrobial peptides produced by binary ligation of truncated alpha-defensins. Curr Protein Pept Sci. 2004;5:365–71. doi: 10.2174/1389203043379459. [DOI] [PubMed] [Google Scholar]
  • 35.Penberthy WT, Chari S, Cole AL, Cole AM. Retrocyclins and their activity against HIV-1. Cell Mol Life Sci. 68:2231–42. doi: 10.1007/s00018-011-0715-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Arnett E, Lehrer RI, Pratikhya P, Lu W, Seveau S. Defensins enable macrophages to inhibit the intracellular proliferation of Listeria monocytogenes. Cell Microbiol. 13:635–51. doi: 10.1111/j.1462-5822.2010.01563.x. [DOI] [PubMed] [Google Scholar]
  • 37.Bucki R, Leszczynska K, Namiot A, Sokolowski W. Cathelicidin LL-37: a multitask antimicrobial peptide. Arch Immunol Ther Exp (Warsz) 58:15–25. doi: 10.1007/s00005-009-0057-2. [DOI] [PubMed] [Google Scholar]
  • 38.Doss M, White MR, Tecle T, Hartshorn KL. Human defensins and LL-37 in mucosal immunity. J Leukoc Biol. 87:79–92. doi: 10.1189/jlb.0609382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Larrick JW, Lee J, Ma S, Li X, Francke U, Wright SC, Balint RF. Structural, functional analysis and localization of the human CAP18 gene. FEBS Lett. 1996;398:74–80. doi: 10.1016/s0014-5793(96)01199-4. [DOI] [PubMed] [Google Scholar]
  • 40.Gudmundsson GH, Agerberth B, Odeberg J, Bergman T, Olsson B, Salcedo R. The human gene FALL39 and processing of the cathelin precursor to the antibacterial peptide LL-37 in granulocytes. Eur J Biochem. 1996;238:325–32. doi: 10.1111/j.1432-1033.1996.0325z.x. [DOI] [PubMed] [Google Scholar]
  • 41.Lee CC, Sun Y, Qian S, Huang HW. Transmembrane pores formed by human antimicrobial peptide LL-37. Biophys J. 100:1688–96. doi: 10.1016/j.bpj.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sochacki KA, Barns KJ, Bucki R, Weisshaar JC. Real-time attack on single Escherichia coli cells by the human antimicrobial peptide LL-37. Proc Natl Acad Sci U S A. 108:E77–81. doi: 10.1073/pnas.1101130108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Murakami M, Dorschner RA, Stern LJ, Lin KH, Gallo RL. Expression and secretion of cathelicidin antimicrobial peptides in murine mammary glands and human milk. Pediatr Res. 2005;57:10–5. doi: 10.1203/01.PDR.0000148068.32201.50. [DOI] [PubMed] [Google Scholar]
  • 44.Schauber J, Rieger D, Weiler F, Wehkamp J, Eck M, Fellermann K, Scheppach W, Gallo RL, Stange EF. Heterogeneous expression of human cathelicidin hCAP18/LL-37 in inflammatory bowel diseases. Eur J Gastroenterol Hepatol. 2006;18:615–21. doi: 10.1097/00042737-200606000-00007. [DOI] [PubMed] [Google Scholar]
  • 45.Schauber J, Iffland K, Frisch S, Kudlich T, Schmausser B, Eck M, Menzel T, Gostner A, Luhrs H, Scheppach W. Histone-deacetylase inhibitors induce the cathelicidin LL-37 in gastrointestinal cells. Mol Immunol. 2004;41:847–54. doi: 10.1016/j.molimm.2004.05.005. [DOI] [PubMed] [Google Scholar]
  • 46.Termen S, Tollin M, Rodriguez E, Sveinsdottir SH, Johannesson B, Cederlund A, Sjovall J, Agerberth B, Gudmundsson GH. PU. 1 and bacterial metabolites regulate the human gene CAMP encoding antimicrobial peptide LL-37 in colon epithelial cells. Mol Immunol. 2008;45:3947–55. doi: 10.1016/j.molimm.2008.06.020. [DOI] [PubMed] [Google Scholar]
  • 47.Peric M, Koglin S, Dombrowski Y, Gross K, Bradac E, Ruzicka T, Schauber J. VDR and MEK-ERK dependent induction of the antimicrobial peptide cathelicidin in keratinocytes by lithocholic acid. Mol Immunol. 2009;46:3183–7. doi: 10.1016/j.molimm.2009.08.010. [DOI] [PubMed] [Google Scholar]
  • 48.Lagishetty V, Chun RF, Liu NQ, Lisse TS, Adams JS, Hewison M. 1alpha-hydroxylase and innate immune responses to 25-hydroxyvitamin D in colonic cell lines. J Steroid Biochem Mol Biol. 121:228–33. doi: 10.1016/j.jsbmb.2010.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Rivas-Santiago B, Hernandez-Pando R, Carranza C, Juarez E, Contreras JL, Aguilar-Leon D, Torres M, Sada E. Expression of cathelicidin LL-37 during Mycobacterium tuberculosis infection in human alveolar macrophages, monocytes, neutrophils, and epithelial cells. Infect Immun. 2008;76:935–41. doi: 10.1128/IAI.01218-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Koon HW, Shih DQ, Chen J, Bakirtzi K, Hing TC, Law I, Ho S, Ichikawa R, Zhao D, Xu H, Gallo R, Dempsey P, Cheng G, Targan SR, Pothoulakis C. Cathelicidin Signaling via the Toll-like Receptor Protects Against Colitis in Mice. Gastroenterology. doi: 10.1053/j.gastro.2011.06.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tai EK, Wong HP, Lam EK, Wu WK, Yu L, Koo MW, Cho CH. Cathelicidin stimulates colonic mucus synthesis by up-regulating MUC1 and MUC2 expression through a mitogen-activated protein kinase pathway. J Cell Biochem. 2008;104:251–8. doi: 10.1002/jcb.21615. [DOI] [PubMed] [Google Scholar]
  • 52.Otte JM, Zdebik AE, Brand S, Chromik AM, Strauss S, Schmitz F, Steinstraesser L, Schmidt WE. Effects of the cathelicidin LL-37 on intestinal epithelial barrier integrity. Regul Pept. 2009;156:104–17. doi: 10.1016/j.regpep.2009.03.009. [DOI] [PubMed] [Google Scholar]
  • 53.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;103:9178–83. doi: 10.1073/pnas.0602888103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Sarker P, Ahmed S, Tiash S, Rekha RS, Stromberg R, Andersson J, Bergman P, Gudmundsson GH, Agerberth B, Raqib R. Phenylbutyrate counteracts Shigella mediated downregulation of cathelicidin in rabbit lung and intestinal epithelia: a potential therapeutic strategy. PLoS One. 6:e20637. doi: 10.1371/journal.pone.0020637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cobo ER, He C, Hirata K, Hwang G, Tran U, Eckmann L, Gallo RL, Reed SL. Entamoeba histolytica Induces Intestinal Cathelicidins but Is Resistant to Cathelicidin-Mediated Killing. Infect Immun. 80:143–9. doi: 10.1128/IAI.05029-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Medina C, Radomski MW. Role of matrix metalloproteinases in intestinal inflammation. J Pharmacol Exp Ther. 2006;318:933–8. doi: 10.1124/jpet.106.103465. [DOI] [PubMed] [Google Scholar]
  • 57.Wiesner J, Vilcinskas A. Antimicrobial peptides: the ancient arm of the human immune system. Virulence. 1:440–64. doi: 10.4161/viru.1.5.12983. [DOI] [PubMed] [Google Scholar]
  • 58.Williams SE, Brown TI, Roghanian A, Sallenave JM. SLPI and elafin: one glove, many fingers. Clin Sci (Lond) 2006;110:21–35. doi: 10.1042/CS20050115. [DOI] [PubMed] [Google Scholar]
  • 59.Flach CF, Eriksson A, Jennische E, Lange S, Gunnerek C, Lonnroth I. Detection of elafin as a candidate biomarker for ulcerative colitis by whole-genome microarray screening. Inflamm Bowel Dis. 2006;12:837–42. doi: 10.1097/01.mib.0000232469.23574.11. [DOI] [PubMed] [Google Scholar]
  • 60.Schmid M, Fellermann K, Fritz P, Wiedow O, Stange EF, Wehkamp J. Attenuated induction of epithelial and leukocyte serine antiproteases elafin and secretory leukocyte protease inhibitor in Crohn’s disease. J Leukoc Biol. 2007;81:907–15. doi: 10.1189/jlb.0906581. [DOI] [PubMed] [Google Scholar]
  • 61.Si-Tahar M, Merlin D, Sitaraman S, Madara JL. Constitutive and regulated secretion of secretory leukocyte proteinase inhibitor by human intestinal epithelial cells. Gastroenterology. 2000;118:1061–71. doi: 10.1016/s0016-5085(00)70359-3. [DOI] [PubMed] [Google Scholar]
  • 62.Motta JP, Magne L, Descamps D, Rolland C, Squarzoni-Dale C, Rousset P, Martin L, Cenac N, Balloy V, Huerre M, Frohlich LF, Jenne D, Wartelle J, Belaaouaj A, Mas E, Vinel JP, Alric L, Chignard M, Vergnolle N, Sallenave JM. Modifying the protease, antiprotease pattern by elafin overexpression protects mice from colitis. Gastroenterology. 140:1272–82. doi: 10.1053/j.gastro.2010.12.050. [DOI] [PubMed] [Google Scholar]
  • 63.Reardon C, Lechmann M, Brustle A, Gareau MG, Shuman N, Philpott D, Ziegler SF, Mak TW. Thymic stromal lymphopoetin-induced expression of the endogenous inhibitory enzyme SLPI mediates recovery from colonic inflammation. Immunity. 35:223–35. doi: 10.1016/j.immuni.2011.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Elsbach P. The bactericidal/permeability-increasing protein (BPI) in antibacterial host defense. J Leukoc Biol. 1998;64:14–8. doi: 10.1002/jlb.64.1.14. [DOI] [PubMed] [Google Scholar]
  • 65.Akin H, Tahan G, Ture F, Eren F, Atug O, Tahan V, Hamzaoglu I, Imeryuz N, Tozun N, Hamzaoglu HO. Association between bactericidal/permeability increasing protein (BPI) gene polymorphism (Lys216Glu) and inflammatory bowel disease. J Crohns Colitis. 5:14–8. doi: 10.1016/j.crohns.2010.08.008. [DOI] [PubMed] [Google Scholar]
  • 66.Haapamaki MM, Haggblom JO, Gronroos JM, Pekkala E, Alanen K, Nevalainen TJ. Bactericidal/permeability-increasing protein in colonic mucosa in ulcerative colitis. Hepatogastroenterology. 1999;46:2273–7. [PubMed] [Google Scholar]
  • 67.Canny G, Cario E, Lennartsson A, Gullberg U, Brennan C, Levy O, Colgan SP. Functional and biochemical characterization of epithelial bactericidal/permeability-increasing protein. Am J Physiol Gastrointest Liver Physiol. 2006;290:G557–67. doi: 10.1152/ajpgi.00347.2005. [DOI] [PubMed] [Google Scholar]
  • 68.Schultz H. From infection to autoimmunity: a new model for induction of ANCA against the bactericidal/permeability increasing protein (BPI) Autoimmun Rev. 2007;6:223–7. doi: 10.1016/j.autrev.2006.08.005. [DOI] [PubMed] [Google Scholar]
  • 69.Schinke S, Fellermann K, Herlyn K, Reichel PH, Fundke R, Stange EF, Gross WL, Schultz H. Autoantibodies against the bactericidal/permeability-increasing protein from inflammatory bowel disease patients can impair the antibiotic activity of bactericidal/permeability-increasing protein. Inflamm Bowel Dis. 2004;10:763–70. doi: 10.1097/00054725-200411000-00011. [DOI] [PubMed] [Google Scholar]
  • 70.Zheng HC, Sugawara A, Okamoto H, Takasawa S, Takahashi H, Masuda S, Takano Y. Expression profile of the REG gene family in colorectal carcinoma. J Histochem Cytochem. 59:106–15. doi: 10.1369/jhc.2010.956961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hervieu V, Christa L, Gouysse G, Bouvier R, Chayvialle JA, Brechot C, Scoazec JY. HIP/PAP, a member of the reg family, is expressed in glucagon-producing enteropancreatic endocrine cells and tumors. Hum Pathol. 2006;37:1066–75. doi: 10.1016/j.humpath.2006.03.005. [DOI] [PubMed] [Google Scholar]
  • 72.Ogawa H, Fukushima K, Naito H, Funayama Y, Unno M, Takahashi K, Kitayama T, Matsuno S, Ohtani H, Takasawa S, Okamoto H, Sasaki I. Increased expression of HIP/PAP and regenerating gene III in human inflammatory bowel disease and a murine bacterial reconstitution model. Inflamm Bowel Dis. 2003;9:162–70. doi: 10.1097/00054725-200305000-00003. [DOI] [PubMed] [Google Scholar]
  • 73.Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science. 2006;313:1126–30. doi: 10.1126/science.1127119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lehotzky RE, Partch CL, Mukherjee S, Cash HL, Goldman WE, Gardner KH, Hooper LV. Molecular basis for peptidoglycan recognition by a bactericidal lectin. Proc Natl Acad Sci U S A. 107:7722–7. doi: 10.1073/pnas.0909449107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Callewaert L, Michiels CW. Lysozymes in the animal kingdom. J Biosci. 35:127–60. doi: 10.1007/s12038-010-0015-5. [DOI] [PubMed] [Google Scholar]
  • 76.Widdicombe J. Relationships among the composition of mucus, epithelial lining liquid, and adhesion of microorganisms. Am J Respir Crit Care Med. 1995;151:2088–92. doi: 10.1164/ajrccm.151.6.7767562. discussion 2092–3. [DOI] [PubMed] [Google Scholar]
  • 77.McClellan KA. Mucosal defense of the outer eye. Surv Ophthalmol. 1997;42:233–46. doi: 10.1016/s0039-6257(97)00090-8. [DOI] [PubMed] [Google Scholar]
  • 78.Clare DA, Catignani GL, Swaisgood HE. Biodefense properties of milk: the role of antimicrobial proteins and peptides. Curr Pharm Des. 2003;9:1239–55. doi: 10.2174/1381612033454874. [DOI] [PubMed] [Google Scholar]
  • 79.Langhorst J, Elsenbruch S, Mueller T, Rueffer A, Spahn G, Michalsen A, Dobos GJ. Comparison of 4 neutrophil-derived proteins in feces as indicators of disease activity in ulcerative colitis. Inflamm Bowel Dis. 2005;11:1085–91. doi: 10.1097/01.mib.0000187980.08686.18. [DOI] [PubMed] [Google Scholar]
  • 80.Lee M, Kovacs-Nolan J, Yang C, Archbold T, Fan MZ, Mine Y. Hen egg lysozyme attenuates inflammation and modulates local gene expression in a porcine model of dextran sodium sulfate (DSS)-induced colitis. J Agric Food Chem. 2009;57:2233–40. doi: 10.1021/jf803133b. [DOI] [PubMed] [Google Scholar]
  • 81.Johnson EE, Wessling-Resnick M. Iron metabolism and the innate immune response to infection. Microbes Infect. doi: 10.1016/j.micinf.2011.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Drago-Serrano ME, de la Garza-Amaya M, Luna JS, Campos-Rodriguez R. Lactoferrin-lipopolysaccharide (LPS) binding as key to antibacterial and antiendotoxic effects. Int Immunopharmacol. doi: 10.1016/j.intimp.2011.11.002. [DOI] [PubMed] [Google Scholar]
  • 83.Yen CC, Shen CJ, Hsu WH, Chang YH, Lin HT, Chen HL, Chen CM. Lactoferrin: an iron-binding antimicrobial protein against Escherichia coli infection. Biometals. 24:585–94. doi: 10.1007/s10534-011-9423-8. [DOI] [PubMed] [Google Scholar]
  • 84.Kanyshkova TG, Buneva VN, Nevinsky GA. Lactoferrin and its biological functions. Biochemistry (Mosc) 2001;66:1–7. doi: 10.1023/a:1002817226110. [DOI] [PubMed] [Google Scholar]
  • 85.Sidhu R, Wilson P, Wright A, Yau CW, D’Cruz FA, Foye L, Morley S, Lobo AJ, McAlindon ME, Sanders DS. Faecal lactoferrin--a novel test to differentiate between the irritable and inflamed bowel? Aliment Pharmacol Ther. 31:1365–70. doi: 10.1111/j.1365-2036.2010.04306.x. [DOI] [PubMed] [Google Scholar]
  • 86.Sipponen T, Karkkainen P, Savilahti E, Kolho KL, Nuutinen H, Turunen U, Farkkila M. Correlation of faecal calprotectin and lactoferrin with an endoscopic score for Crohn’s disease and histological findings. Aliment Pharmacol Ther. 2008;28:1221–9. doi: 10.1111/j.1365-2036.2008.03835.x. [DOI] [PubMed] [Google Scholar]
  • 87.Gisbert JP, McNicholl AG, Gomollon F. Questions and answers on the role of fecal lactoferrin as a biological marker in inflammatory bowel disease. Inflamm Bowel Dis. 2009;15:1746–54. doi: 10.1002/ibd.20920. [DOI] [PubMed] [Google Scholar]
  • 88.Steiner TS, Flores CA, Pizarro TT, Guerrant RL. Fecal lactoferrin, interleukin-1beta, and interleukin-8 are elevated in patients with severe Clostridium difficile colitis. Clin Diagn Lab Immunol. 1997;4:719–22. doi: 10.1128/cdli.4.6.719-722.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Iida T, Naka A, Suthienkul O, Sakaue Y, Guerrant RL, Honda T. Measurement of fecal lactoferrin for rapid diagnosis of enterohemorrhagic Escherichia coli infection. Clin Infect Dis. 1997;25:167. doi: 10.1086/516898. [DOI] [PubMed] [Google Scholar]
  • 90.Togawa J, Nagase H, Tanaka K, Inamori M, Nakajima A, Ueno N, Saito T, Sekihara H. Oral administration of lactoferrin reduces colitis in rats via modulation of the immune system and correction of cytokine imbalance. J Gastroenterol Hepatol. 2002;17:1291–8. doi: 10.1046/j.1440-1746.2002.02868.x. [DOI] [PubMed] [Google Scholar]
  • 91.Togawa J, Nagase H, Tanaka K, Inamori M, Umezawa T, Nakajima A, Naito M, Sato S, Saito T, Sekihara H. Lactoferrin reduces colitis in rats via modulation of the immune system and correction of cytokine imbalance. Am J Physiol Gastrointest Liver Physiol. 2002;283:G187–95. doi: 10.1152/ajpgi.00331.2001. [DOI] [PubMed] [Google Scholar]
  • 92.Lopez-Soto F, Leon-Sicairos N, Nazmi K, Bolscher JG, de la Garza M. Microbicidal effect of the lactoferrin peptides lactoferricin17-30, lactoferrampin265-284, and lactoferrin chimera on the parasite Entamoeba histolytica. Biometals. 23:563–8. doi: 10.1007/s10534-010-9295-3. [DOI] [PubMed] [Google Scholar]
  • 93.Berlutti F, Schippa S, Morea C, Sarli S, Perfetto B, Donnarumma G, Valenti P. Lactoferrin downregulates pro-inflammatory cytokines upexpressed in intestinal epithelial cells infected with invasive or noninvasive Escherichia coli strains. Biochem Cell Biol. 2006;84:351–7. doi: 10.1139/o06-039. [DOI] [PubMed] [Google Scholar]
  • 94.Ganz T. Hepcidin and iron regulation, 10 years later. Blood. 117:4425–33. doi: 10.1182/blood-2011-01-258467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Collins HL. Withholding iron as a cellular defence mechanism--friend or foe? Eur J Immunol. 2008;38:1803–6. doi: 10.1002/eji.200838505. [DOI] [PubMed] [Google Scholar]
  • 96.Oustamanolakis P, Koutroubakis IE, Messaritakis I, Malliaraki N, Sfiridaki A, Kouroumalis EA. Serum hepcidin and prohepcidin concentrations in inflammatory bowel disease. Eur J Gastroenterol Hepatol. 23:262–8. doi: 10.1097/MEG.0b013e328343b885. [DOI] [PubMed] [Google Scholar]
  • 97.Wang L, Trebicka E, Fu Y, Ellenbogen S, Hong CC, Babitt JL, Lin HY, Cherayil BJ. The bone morphogenetic protein-hepcidin axis as a therapeutic target in inflammatory bowel disease. Inflamm Bowel Dis. 18:112–9. doi: 10.1002/ibd.21675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kang JK, Hwang JS, Nam HJ, Ahn KJ, Seok H, Kim SK, Yun EY, Pothoulakis C, Lamont JT, Kim H. The insect peptide coprisin prevents Clostridium difficile-mediated acute inflammation and mucosal damage through selective antimicrobial activity. Antimicrob Agents Chemother. 55:4850–7. doi: 10.1128/AAC.00177-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Van Bambeke F. Glycopeptides and glycodepsipeptides in clinical development: a comparative review of their antibacterial spectrum, pharmacokinetics and clinical efficacy. Curr Opin Investig Drugs. 2006;7:740–9. [PubMed] [Google Scholar]
  • 100.Lino M, Kus JV, Tran SL, Naqvi Z, Binnington B, Goodman SD, Segall AM, Foster DB. A novel antimicrobial peptide significantly enhances acid-induced killing of Shiga toxin-producing Escherichia coli O157 and non-O157 serotypes. Microbiology. 157:1768–75. doi: 10.1099/mic.0.047365-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pasupuleti M, Schmidtchen A, Malmsten M. Antimicrobial peptides: key components of the innate immune system. Crit Rev Biotechnol. doi: 10.3109/07388551.2011.594423. [DOI] [PubMed] [Google Scholar]
  • 102.Johansson J, Gudmundsson GH, Rottenberg ME, Berndt KD, Agerberth B. Conformation-dependent antibacterial activity of the naturally occurring human peptide LL-37. J Biol Chem. 1998;273:3718–24. doi: 10.1074/jbc.273.6.3718. [DOI] [PubMed] [Google Scholar]
  • 103.Travis SM, Anderson NN, Forsyth WR, Espiritu C, Conway BD, Greenberg EP, McCray PB, Jr, Lehrer RI, Welsh MJ, Tack BF. Bactericidal activity of mammalian cathelicidin-derived peptides. Infect Immun. 2000;68:2748–55. doi: 10.1128/iai.68.5.2748-2755.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Pathan FK, Venkata DA, Panguluri SK. Recent patents on antimicrobial peptides. Recent Pat DNA Gene Seq. 4:10–6. doi: 10.2174/187221510790410831. [DOI] [PubMed] [Google Scholar]
  • 105.Burton MF, Steel PG. The chemistry and biology of LL-37. Nat Prod Rep. 2009;26:1572–84. doi: 10.1039/b912533g. [DOI] [PubMed] [Google Scholar]
  • 106.Chen C, Wu S, Li X, Zhang X, Yan M. Structure, function and molecular design strategies of antibacterial peptide SMAP-29: a review. Sheng Wu Gong Cheng Xue Bao. 27:846–59. [PubMed] [Google Scholar]

RESOURCES