GET THE APP

Clinical Relevance of Cathelicidin in Infectious Disease
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2013) Volume 0, Issue 0

Clinical Relevance of Cathelicidin in Infectious Disease

Annika Linde, Gerald H. Lushington, Javier Abello and Tonatiuh Melgarejo*
omi_departments of Human Nutrition and Anatomy & Physiology, Kansas State University, USA, E-mail: alinde@ksu.edu
*Corresponding Author: Tonatiuh Melgarejo, omi_departments of Human Nutrition and Anatomy & Physiology, Kansas State University, USA Email:

Abstract

The human body is subjected to constant microbial exposure from both a resident microbiome as well as the surrounding environment. Staying healthy in a world filled with “dangers” as such necessitates gene-encoded tools to enable an immediate and effective immunological shield. Antimicrobial host defense peptides are imperative to an apt innate immune response in which they serve both regulatory as well as executorial roles to eliminate infectious pathogens, control inflammation, and support healing of injured tissue. Cathelicidin peptides were originally isolated from bone marrow and neutrophils, although their pattern of expression is now known to span a broader spectrum. The clinical importance of an effective host defense peptide repertoire is best illustrated by data from patient groups with a deficient expression pattern and increased disease susceptibility, as well as experimental work with relevant animal models. This review paper is focused on the human cathelicidin LL37 and its clinical implications in infectious disease.

Keywords: Host defense peptides; Innate immunity; Antimicrobial; LL-37; hCAP18; Human microbiome; Cationic peptides

Abbreviations

ACES: Azithromycin and Coronary Events Study; AZACS: AZithromycin in Acute Coronary Syndromes; ICAM: Intercellular adhesion molecule-1; IFN: Interferon; MCP-1: Monocyte Chemoattractant Protein 1; MIC: Minimal Inhibitory Concentration; NFkB: Nuclear Factor Kappa Beta; NOD2: Nucleotide-binding Oligomerization Domain (NOD) protein. The NOD-like receptors, NLRs are a group of pattern recognition receptor, PRRs; PROVEIT: PRavastatin or atorVastatin Evaluation and Infection Therapy; TLRs: Toll-like receptors; WIZARD: Weekly Intervention with Zithromax for Atherosclerosis and its Related Disorders; 1,25(OH)2D3: 1,25-dihydroxy-vitamin D3

Introduction

Host defense peptides (HDPs) are expressed ubiquitously within an individual organism as well as across different species. The cathelicidin and defensin peptide families constitute the major groups of HDPs in mammals, where they perform a principal role on the stage of the innate immune response [1]. Our focus in this review is directed towards the cathelicidins, which have been documented broadly in nature, including numerous different animal species[2].

Cathelicidins are cationic amphipathic peptides that span a functionally diverse repertoire to enable homeostasis in response to pathogen-, disease- and danger-associated-molecular patterns [3,4]. The group serves as natural antimicrobials against bacterial, fungal, viral, and parasitic pathogens, while also contributing to the orchestration of the immune response, and elimination of abnormal host cells. The peptides’ broad pattern of distribution and efficacy make them attractive as templates for development of new antibiotics [5], and of particular interest in context of a species-spanning approach to health and medicine - “Zoobiquity” [6]. In this review we seek to provide an overview of the clinical relevance of the naturally occurring human cathelicidin in context of infectious disease, including the potential application of synthetic analogues as novel biotherapeutics.

Distribution and Biological Characteristics

The term “cathelicidin” was introduced twenty years ago to describe a subset of HDPs that was first isolated from bovine neutrophils [7]. Much research has since been dedicated to the field of innate immunity, and members of the cathelicidin peptide family have been documented in a variety of species across the animal kingdom. In example, the antimicrobial sequence database (AMSD) at University of Trieste yields sixty entries in response to the search term “cathelicidin”, while GenBank lists a total of 538 nucleotide entries, including 148 hits using Ref Seqa.

Examples of cathelicidin peptides identified in animal species examined to date are shown in Figure 1 and Table 1. The sequence homology between cathelicidins from different &ndash closely as well as more distantly related &ndash animal species is shown in Figure 2, while Figure 3 exemplifies the structural similarity between three of the mammalian cathelicidins.

Species (Order, Family or Subfamily) Common Name Peptide(s) Reference(s)
Bos taurus(Artiodactyla/Bovidae) Cow BMAP-27. BMAP-28. BMAP-34. Bac5. Bac7. Dodecapeptide. Indolicidin PMID: 9067433
PMID: 12960280
AMSDb
Ovis aries(Artiodactyla/Bovidae) Sheep SMAP-29. SMAP-34. oaBac6. oaBac11. sBAc7.5
sBac5. s-dodecapeptide.
PMID: 7498547
PMID: 12960280
AMSDb
*Capra hircus(Artiodactyla/Bovidae) Goat chMAP-28. chMAP-34. chBac5. chBac7.5 PMID: 10417180
PMID: 12960280
Sus scrofa(Artiodactyla/Suidae) Pig PMAP-23. PMAP- 36. PMAP-37. PR-39. Prophenin1, 2. Protegrin1-5 PMID: 1765098
PMID: 16053249
AMSDb
Homo sapiens(Primate) Human LL-37 (hCAP-18) PMID: 7890387
Pan troglodytes(Primate) Chimpanzee ptrLL-37 PMID: 16720578
Pan paniscus(Primate) Bonobo 1 cathelicidin predicted XM_003818426
Gorilla gorilla gorilla(Primate) Gorilla 1 cathelicidin predicted XM_004034062
Pongo pygmaeus(Primate) Orangutan ppyLL-37 PMID: 16720578
Papio anubis(Primate) Baboon 1 cathelicidin predicted XM_003894490
Macaca mulatta(Primate) Rhesus macaque mmuRL-37 PMID: 16720578
Callithrix jacchus(Primate) Marmoset cjaRL-37 PMID: 16720578
Saimiri boliviensis boliviensis(Primate) Squirrel monkey 1 cathelicidin predicted XM_003926293
Hylobates moloch (Primate) Gibbon hmdSL-37 PMID: 16720578
Otolemur garnettii(Primate) Bush baby 1 cathelicidin predicted XM_003800549
*Trachypithecus obscurus(Primate) Leaf monkey pobRL-37 PMID: 16720578
Mus musculus(Rodentia) Mouse mCRAMP PMID: 9148921
Rattus norvegicus(Rodentia) Rat rCRAMP PMID: 12737313
Cavia porcellus(Rodentia) Guinea pig CAP-11 PMID: 9278433
Cricetulus griseus(Rodentia) Hamster 1 cathelicidin predicted XM_003504259
Felis catus(Carnivora) Cat feCATH PMID: 21533281
Canis lupus familiaris(Carnivora) Dog K9CATH PMID: 17462733
Ailuropoda melanoleuca(Carnivora) Panda Cathelicidin-AM PMID: 22101189
Equus caballus(Perissodactyla/ Equidae) Horse eCATH-1, 2, 3 PMID: 11181349
Oryctolagus cuniculus(Lagomorpha) Rabbit CAP-18 PMID: 1883348
Loxodonta Africana(Proboscidae) Elephant 1 cathelicidin predicted XM_003409891
Sarcophilus harrisii(Dasyuromorphia) Tasmanian devil 1 cathelicidin predicted XM_003762945
Monodelphis domestica(Didelphimorphia) Opossum 12 cathelicidin genes PMID: 17495011
*Macropus eugenii(Diprotodontia) Wallaby 14 cathelicidin genes PMID: 21912615
Ornithorhynchus anatinus(Monotremata) Platypus 8 cathelicidin genes PMID: 21912615
Gallus gallus(Galliformes) Chicken Fowlicidin-1, 2, 3 PMID: 16326712
Meleagris gallopavo(Meleagridinae) Turkey 2 cathelicidins predicted XM_003206909/10
Salmo salar(Salmonidae) Atlantic salmon asCATH-1, 2 PMID: 16377685
Oncorhynchus mykiss(Salmonidae) Rainbow trout rtCATH-1, 2 PMID: 16377685
*Ophiophagus Hannah(Elapidae) King cobra OH-CATH PMID: 18620012
*Indicates species not included in the tree shown in Figure 1

Table 1: An Overview of Cathelicidin Host Defense Peptides in Animal Species.

clinical-cellular-immunology-Cathelicidin-Sequences

Figure 1: Distribution of Cathelicidin Sequences across Taxonomic Groups
The tree was constructed based on GenBank data (RefSeq search, January 2013). The species listed in the tree are outlined in Table 1, which includes the corresponding cathelicidin peptides.

clinical-cellular-immunology-Antimicrobial-Proteins

Figure 2: Multiple Sequence Alignment of Cationic Antimicrobial Proteins
Conserved domains are indicated with squares, including the Cathelicidins superfamily (red), and CAP18_C (blue). Amino acid residues that present 100% identity are shown in aquamarine (tyrosine, Y), royal blue (valine, V), and pink (cysteine, C). Residues with 93% identity are shown in yellow (leucine, L), light purple (phenylalanine, F), sky-blue (threonine, T) and orange (glycine, G). Accession numbers were obtained from NCBI GenBank. Sequence alignment and graphic made with Clustal Omega (Ver.1.1.0) and Jalview (Ver.2.8.). Species key: human (H. sapiens), chimpanzee (P. troglodytes), orangutan (P. pygmaeus), rhesus macaque (M. mulatta), cat (F. catus), dog (C. familiaris), rat (R. norvegicus), rabbit (O. cuniculus), mouse (M. musculus), panda (A. melanoleuca), guinea pig (C. porcellus), platypus (O. anatinus), wallaby (M. eugenii), opossum (M. domestica), chicken (G. gallus), cobra (N. atra).

clinical-cellular-immunology-Cathelicidin-Structures

Figure 3: Cathelicidin Structures from Human, Mouse and Dog
Computational modeling of alpha-helical cathelicidin structures from three mammalian species. Ribbon diagrams made with PyMol Molecular Graphics System (Ver.1.5.), and sequence data from Current Protein and Peptide Science, 2005, 6, 23-34.

The size and structural characteristics of cathelicidins vary broadly with peptides ranging from a dozen to about a hundred amino acid residues, and spanning both cyclic and linear structures (Figure 4). Cyclic peptides are typically characterized by a loop structure (beta-sheet) stabilized by di-sulphide bridges, while linear peptides tend to form amphipathic alpha helices or structures with repeat motifs of amino acids such as proline or tryptophan [8]. The common feature that ties this heterogeneous peptide family together is a conserved (100 residues) pro-sequence at the N-terminal, the so-called cathelinb domain (Figures 2 and 4).

clinical-cellular-immunology-Structural-Classes

Figure 4: Gene Organization and Major Structural Classes of Cathelicidins
Cathelicidins are ancient gene-encoded (four exons) immune compounds produced in the bone marrow as pre-pro-peptides with a conserved signaland cathelin - domain, stored as pro-peptides in neutrophil granules, prior to extracellular release and cleavage into mature (variable) cathelicidin HDPs. AA: amino acid. The cathelicidins listed here are human (LL37), and porcine (PR39, Protegrin-3). Figure content also based on: Scand. J. Infect. Dis. 35: 670-6, 2003.

The number of cathelicidins varies from a single peptide in humans to several within artiodactyl species (Table 1), and the potency and functional spectrum of individual cathelicidin peptides therefore also varies [1,9]. Many of the peptides are inducible in response to various molecular patterns (e.g. bacterial fragments or chemical “danger” signals released upon tissue injury), which signal through innate pattern recognition receptors and downstream pathways (such as TLRs and NFkB signaling), so the pattern of HDP expression fluctuates over time [1]. Elite members of the group exhibit broad-spectrum antimicrobial activity at sub-micromolar concentrations, including killing of pathogens that are otherwise resistant to commercially available antibiotics.

Microbial resistance to HDPs is rare in nature because the peptides utilize a fast-acting and relatively non-specific mechanism of action, which relies on the difference in lipid composition between normal eukaryotic versus prokaryotic (or transformed eukaryotic) cell membranes. One of the generally accepted mechanisms of action for HDPs, schematized in the Shai-Matsuzaki-Huang model [10] (Figure 5), involves bacterial killing via peptide-induced pore formation in the cell membrane, membrane destabilization and lysis (Figure 6). Synergism has moreover been observed between different HDPs, as well as between cathelicidins and conventional antibiotics [8,11,12].

clinical-cellular-immunology-Shai-Matsuzaki-Huang

Figure 5: The Shai-Matsuzaki-Huang Model for HDP Antimicrobial Activity
Cationic amphipathic HDPs connect to the bacterial surface via strong electrostatic and hydrophobic interactions made possible by the acidic phospholipid content (anionic) in the prokaryotic cell membrane. This is distinct from normal eukaryotic cell membranes that are electro-neutral consequent to the cholesterol content in their phospholipid bilayer. Figure based on: Nature 2002 Jan 24;415(6870):389-95.

clinical-cellular-immunology-Electron-Microscopic

Figure 6: Electron Microscopic Images of M. tuberculosis after Cathelicidin Exposure
Canine cathelicidin (K9CATH) exposure creates significant and increasing vacuolization (A.) of the Mycobacterium tuberculosis (TB) bacteria, followed by membrane disruption (B), and bacterial lysis – as compared to untreated Control (C). Photos republished (with permission) from: African Journal of Microbiology Research (2012) 6: 38; 6726-29.

Mechanisms of microbial resistance towards HDPs might involve bacterial modification of the (anionic) surface composition to diminish the attraction of the cationic HDPs, peptide removal from the bacterial interior via an efflux pump, as well as down-regulation, neutralization or proteolytic degradation of the cathelicidin peptides [13]. However, unlike commercially available antibiotics – which on an evolutionary scale “only” date back to 1929 with Alexander Fleming’s discovery of penicillin &ndash natural antimicrobial (host defense) peptides are ancient effector molecules of innate immunity [14]. HDPs have enjoyed long-lasting success in nature, and been very effective from an evolutionary perspective. While mutant (bacterial) strains can manipulate surface charge to a point where killing by HDPs is avoided under laboratory conditions, such change is likely to also impact bacterial viability and limit existence in nature. The notion of pathogens developing broad-scale resistance against natural HDPs is therefore, relatively speaking, less of a concern [1].

Our current knowledge on cathelicidins stems from a compilation of in vitro studies, animal models, and clinical investigations, which combined position these peptides at the core of the mammalian innate immune response. The distinction between maintenance of health versus development of disease depends first and foremost on the aptitude of the host’s innate immune response. The following sections serve to outline the association between human infectious disease susceptibility and the pattern of cathelicidin expression.

Human Cathelicidin

The innate immune defense repertoire in humans includes a single cathelicidin, which was first identified as FALL-39 (human counterpart of the proline-rich porcine cathelicidin PR-39) based on cDNA isolated from bone marrow [15,16]. This term was subsequently adjusted to FA-LL-37 [17], where LL-37 is the mature human cathelicidin peptide– highlighting the two first amino acids (leucine) and the overall length (amino acids) of the peptide. Meanwhile, hCAP18 refers to the 18kDa form of the “human cationic antimicrobial protein” [18,19], which maps to chromosome 3p21, involving four exons and three introns [20] (Figure 4).

The full-length hCAP18 (holoprotein) includes a signal peptide (N-terminal), a conserved cathelin-like domain (CLD), and the antimicrobial LL37 peptide domain (C-terminal). The hCAP18 precursor is synthesized in neutrophil progenitor cells in the bone marrow and stored in unprocessed form in specific granules of neutrophils [21]. hCAP18 is moreover expressed in epithelium throughout the body either constitutively or in response to molecular patterns from the host or microbial pathogens [20]. Following exocytosis, the holoprotein undergoes extracellular cleavage via protease 3 from azurophil granules to yield the active LL37 peptide (5 kDa) [22].

The peptide’s spectrum of biological activity spans broadly, and includes direct pathogen killing, promotion of angiogenesis in wound healing, mast cell histamine release, as well as chemo-attraction of monocytes, neutrophils, and T cells, and modification of dendritic cell differentiation [20,23]. This further highlights that the role of human cathelicidin as such spans beyond the immediate immune response. The mouse cathelicidin (mCRAMP) shares several similarities with the human LL37, and in vitro studies as well as infection models in CRAMP-deficient mice have received substantial research attention [24,25]. mCRAMP also regulate B- and T-cell function, which implicates this cathelicidin as a role player within the adaptive immune response [26]. The LL-37/CAP18 molecule has been studied in non-human primates, and the high inter-species homology makes these valuable model organisms for further study of the innate immune response [27].

Factors such as peptide concentration, ion composition and pH influence the biological activity and structural conformation of the LL37 peptide, while antimicrobial activity against Gram-negative and -positive bacteria correlates with factors such as degree of alpha-helicity [28]. The peptide exerts broad antimicrobial activity (in vitro) against clinically relevant pathogens such as Escherichia coli, Listeria monocytogenes, Pseudomonas aeruginosa, Salmonella typhimurium, Staphylococcus aureus, Staph. epidermis, and Vancomycin-resistant enterococci at MICs29].

LL37 can exhibit cytotoxic effects towards eukaryotic cells (in vitro) at a concentration of 25 μM, although this effect is attenuated by the presence of human serum [28]. The plasma level of hCAP18 is around 1.2 μg/ml with a significant amount of hCAP18 bound to lipoproteins [22]. Cathelicidin’s broad-ranging biological activities are also explained by the peptide’s interaction with different cell types in the body. The peptide impacts neutrophil function to increase the cellular uptake of bacteria, and can decrease release of pro-inflammatory mediators (TNF-alpha, IL-1beta, IL-6, and IL-8), thereby preventing a cytokine storm [24]. Other functions are highlighted in context of the examples below.

The Human Microbiome

The microbial collection hosted within the human ecosystem constitutes approximately 10,000 species, which contribute about 8 million (unique) protein-coding gene sequences – in other words, more than 300 times the number of (protein-coding) sequences contained in the human genomecd. With trillions of microorganisms present in the body, human cells are outnumbered by a factor 10 at any given time [30]. Fortunately microorganisms stay true to their name and take up relatively little space – although this still translates into 1-3% of our total body weightd. Consequently, if a 70 kg adult hosts about 2 kg of microorganisms (commensal species and potential pathogens) the importance of a functional and well-orchestrated immune system becomes self-evident. Figure 7 serves to visualize the key players in this complex act, as well as the distribution (%) of bacterial species in the human microbiome across body sites.

clinical-cellular-immunology-Biological-Dataset

Figure 7: The Human Composition from a Biological Dataset Perspective
The terms genotype and phenotype were introduced in 1903 by Danish geneticist Wilhelm Johannsen, and their interrelationship has classically been conceptualized as: Genotype (G)+Environment (E) → Phenotype (P). Fast-forward in time to the “era of -omics”, a plethora of biological datasets is available that also warrant consideration. The ubiquitous microbiota housed in/on the body is here represented by the green “humanoid shadow”, while percentages indicate distribution of bacterial species across body sites (Genome Res. 2009;19(12):2317-23). The body enables homeostasis via intrinsic production (“prescription”, Rx) of HDPs. Now, the physical traits (phenome) becomes a matrix reflecting the sum of the individual’s genetic layout (genome), exposures from the environment (exposome) and the resident microflora (microbiome), as well as the combined interactions. Figure made with images from 123RF.com.

The Human Microbiome Project (HMP), launched by the National Institutes of Health in 2007, provides novel insight on the composition of the microflora in a healthy Western population. Depending on body site, anywhere from 20% to 60% of the human-associated microbiome is uncultivable, which stress the value of new high-throughput technologies [30]. Below, we reference data in context of the five major body sites targeted by the HMP: oral cavity, gastrointestinal tract, skin, respiratory tract and urogenital/vaginal tract [31].

Oral cavity

Swedish pediatrician Rolf Kostmann (1909-1982) was the first to report on infantile genetic agranulocytosis – now known as morbus Kostmann – in a 1956 article in Acta Paediatrica Scandinavia. While the mechanism underlying this recessive genetic disorder was still unknown, Kostmann speculated on a missing serum factor [32,33]. The disease renders patients more susceptible to pathogens consequent to abnormal myelopoiesis, and clinical findings are characterized by chronic skin and mucous membrane infections that necessitate repeated treatment with antibiotics. Overall life-quality for patients was improved after introduction of recombinant human granulocyte colony-stimulating factor (G-CSF) in the late 1980s. While G-CSF works to restore neutrophil levels, problems with chronic infections – particularly severe periodontitis and chronic gingivitis – persisted despite treatment [34,35]. In the decades to follow, research in immunology focused increasingly on effector molecules of innate immunity, leading also to evaluation of HDPs in this patient category. In 2002, Hans Boman’s group reported that human cathelicidin is absent in plasma and saliva in morbus Kostmann patients, while individuals subjected to bone marrow transplantation have essentially normal LL-37 concentrations [36]. The study as such established accordance between the occurrence of severe periodontal disease in morbus Kostmann patients and deficient LL-37 production.

The role of LL37 in oral immunocompetency has been further investigated, and high LL37 levels in saliva have been correlated with resistance to caries. LL37 is present in gingival crevicular fluid, where it may contribute as a marker for oral health status [37]. Data from children show that the LL37 saliva concentration is higher in girls versus boys, increases with age, and is higher in those with mixed or permanent dentition, while levels are diminished in individuals with high caries activity [38].

Gastrointestinal system

The commensal gut flora creates a biological barrier, which together with the thick mucus layer (mechanical barrier) and secretion of immune defense molecules (chemical barrier) serve to maintain gastrointestinal health [39,40]. Cathelicidin is expressed by macrophages and epithelial cells of the colon to form part of the barrier system pathogens must bypass to invade the intestinal tissue [39]. Breach of this lining enables attachment of Shiga-toxin-producing bacteria such as enterohemorrhagic Escherichia coli O157:H7 (EHEC) to epithelial cells and secretion of virulence factors into the circulatory system. Hemolytic uremic syndrome with acute renal failure from EHEC infection is a major complication seen in about 15% of clinical cases [39]. Cathelicidin deficiency enhances host susceptibility to pathogenic E. coli O157:H7 in a mouse model through thinning of the mucus layer, which facilitate bacterial attachment to the epithelium, and systemic toxin release [39].

Helicobacter pylori can colonize the human gastric epithelium on a chronic basis, and the hCAP18/LL37 levels in gastric epithelial cells and gastric secretions are significantly increased in infected patients, which is not the case with gastric inflammation from other causes such as neoplasia or hyperplastic polyps [11]. Inflammatory bowel diseases, such as ulcerative colitis and Crohn’s disease, involve an abnormal immune response to bacterial pathogens [41]. Cathelicidin is upregulated in colonic macrophages and epithelium in a murine colitis model, while mCRAMP knockout mice develop more severe colitis supporting that cathelicidin also plays an important role in induction of colitis [42]. On the other hand, colonic epithelial expression of hCAP18 is not upregulated in patients with Crohn’s disease [43].

Cathelicidins exert a broad spectrum of antimicrobial activity against enteropathogenic bacteria, and mice are used extensively to model human disease, although differences exist. In humans, Salmonellosis is caused by Salmonella enterica serovar Typhimurium, which can also cause a typhoid-like response in mice [44]. However, while mCRAMP can inhibit growth of S. typhimurium [45], LL37 does not seem to be a key player in intracellular clearance of this pathogen [44].

Amebiasis is a significant health concern that affects about 10% of the global population, and cause potentially fatal amebic colitis and hepatic abscesses [46]. Studies on human intestinal epithelial cells and a mouse model of amebic colitis have shown that E. histolytica trophozoites upregulate intestinal cathelicidin expression, although amebic cysteine proteases cleave the cathelicidin into fragments without antiparasitic activity [46]. This enzymatic processing enables protozoan evasion of the innate immune response, although the peptides’ antibacterial activity remains intact [46]. Another recent study found that while the antiparasitic activity of LL37 is limited, shorter analogues (KR-20 and KS-30) are able to adversely affect the growth and integrity of E. histolytica trophozoites [47].

Expression of cathelicidin has also been documented in human breast milk, as well as in murine mammary gland tissue [48]. This suggests that cathelicidin serves to protect the mammary epithelium during lactation while also providing the neonatal gut with a natural antibiotic [49]. Importantly, the lactose component in human breast milk induces expression of the cathelicidin gene (CAMP) in colonic epithelium, and acts synergistically with other cathelicidin-inducing factors such as butyrate and phenylbutyrate [50].

Like other HDPs, the biological activity of LL37 spans beyond that of a simple antibiotic, and the peptide in example exerts a tumor-suppressing mechanism in colon tumorigenesis [51].

Integumentary system

Normal-weight adults have on average 2 m2 of body surface area, which is covered by a 2 mm thick layer of skin that effectively constitutes the first line of defense [52,53]. HDPs are expressed constitutively in normal skin, where they create a biochemical shield against external threats, ensure homeostasis with a diverse cutaneous microflora, and help to promote wound healing [54,55]. The principal (stationary) sources of HDPs in human skin are keratinocytes, eccrine glands, sebocytes, and mast cells, while circulatory cells such as neutrophils and NK-cells make significant HDP contributions also [56]. In keratinocytes, cathelicidin is stored in granules (lamellar bodies), and production is increased during times of cell differentiation [57]. Additionally, HDP production (bacteriocins) from the commensal skin flora (e.g. Staphylococcus epidermidis) acts synergistically with the host’s defense repertoire [58-60]. Imbalance in this expression pattern and collapse of the symbiotic arrangement correspond with clinical diseases such as psoriasis, atopic dermatitis, rosacea, as well as other skin conditions [59]. Cathelicidin protect the cutaneous surface via direct antimicrobial action as well as immunomodulation, including release of cytokines, inflammation, angiogenesis, and re-epithelialization [61].

In addition to its role in bone metabolism and calcium homeostasis, the focus on vitamin D3 has been directed increasingly towards immunoregulation in context of both the innate and adaptive arms of the system. Vitamin D3 is central to the cutaneous immune response, and involved in regulation of cathelicidin expression at epithelial surfaces [56,62]. Vitamin D3 deficiency is common in the US, with an overall prevalence of 42% in a recent study [63], and generally caused by either insufficient intake or a mainly indoor lifestyle. Also, the recommended daily allowance for vitamin D3 was historically geared towards bone health more so than optimization of the body’s immunological aptitude [56]. Keratinocyte development and function rely on the presence of vitamin D3, and deficiency leads to abnormal skin barrier formation and a deficient immune defense [56].

Atopic dermatitis (AD) is the most prevalent inflammatory skin disorder in humans, affecting up to 25% of children and about 3% of adults in the US [64]. The epidermal elements responsible for maintenance of skin homeostasis (physical, chemical, microbial, and immunological) are compromised to varying extents in this patient category [64]. The disease is associated with suppression of HDP induction, mediated by IL4 and IL13, and a diminished antimicrobial barrier at the cutaneous level [56]. Patients with AD are therefore also more susceptible to bacterial (e.g. Staphylococcus aureus) as well as viral, (such as herpes simplex) infections [65-67]. Meanwhile, oral vitamin D3 supplementation administered to AD patients at 4,000 IU/day for 21 days is able to boost the cutaneous cathelicidin expression by more than a factor six [67].

Psoriasis is another highly prevalent inflammatory disease of the skin, which affects between 1% to 5% of the population depending on geographical location and ethnicity [68]. The maturation of keratinocytes is accelerated from the usual 30 days to about 3 days in psoriasis patients, and this dramatically shortened skin turnover leads to accumulation of excess cells and plaque formation on the body surface [68]. The disease is characterized by a genetic predisposition, an autoimmune component, as well as abnormal HDP activity [56]. LL37 from psoriatic skin converts inert self-DNA into a ‘danger-signal’, leading to aberrant interferon production from plasmacytoid dendritic cells (pDCs), and local T-cell-mediated autoimmune inflammation, which exacerbates the injury to the skin [69]. In normal skin, plasmacytoid dendritic cells sense “danger signals” released from common skin wounds and play a critical role in the early inflammatory response and re-epithelialization of the cutaneous injury [70].

Rosacea is an inflammatory skin disorder with vascular abnormalities that typically involves the central part of the face. Also known as the “curse of the Celts” (due to the higher prevalence in people of Northern European descent) it affects an estimated 16 million adults (predominantly middle-aged) in the US [71,72]. The diagnosis is based solely on clinical manifestations, and subtypes of rosacea are erythematotelangiectatic, papulopustular, phymatous, and ocular, as well as a non-inflammatory (granulomatous) variant of the disease [71]. The skin’s innate immune response to environmental factors is exaggerated in affected individuals, where the vascular reactivity and abnormal inflammatory response also inspired the study of patients’ cathelicidin expression profiles [71]. Normal post-translational processing generally renders shorter forms of LL37 on the cutaneous surface of healthy individuals [48]. However, rosacea patients have abnormally high levels of unprocessed LL37, and the enzymatically-processed forms are furthermore different (more pro-inflammatory) compared to controls [61,73].

Data from dermatology centers in North America indicate that infections of the nail bed are a significant epidemiological problem, and that the prevalence of onychomycosis is about 14% [74]. The nail environment is isolated from direct cell-mediated immunity due to lack of a vascular supply, however, LL37 is expressed abundantly in epithelium across the nail bed unit. LL37 inhibits growth of Candida albicans at 25 μM, which shows that cathelicidin also serves a role in the immune defense against fungi and nail pathogens [74]. The role of LL37 in fungal skin infections, e.g. dermatophytosis (Epidermophyton, Microsporum, and Trichophyton species) and tinea versicolor (Malassezia furfur), has also been explored. Findings support that cathelicidin is key to an efficacious innate immune defense against fungal infections of the cutaneous surface [75].

Cathelicidin is moreover able to limit dissemination of cutaneous Leishmaniasis. The disease (cutaneous, mucocutaneous and visceral forms) affects approximately 12 million people in 88 countries, and is as such a significant health problem globally [76]. Data from a murine model support that cathelicidin is able to kill the parasitic protozoan through both indirect (enhanced parasite uptake by macrophages and neutrophils) as well as direct (membrane disruption) measures. Without cathelicidin (CAMP knockout model), the parasite grows uncontrolled and metastasizes to visceral organs [76].

Respiratory system

The internal surface area of the human lung covers a prodigious 50 square meters, which in context of a total lung volume of approximately 5 liters enable sufficient gas exchange [77]. Effective host defense of the airway system rely on mechanisms such as mucociliary clearance, reflex coughing and sneezing as well as cellular components and different innate immune factors [78]. Airway epithelium, neutrophils and alveolar macrophages impact the efficacy of pathogen clearance in part through secretion of HDPs into the airway surface fluid (ASF) [78].

While the airways historically were considered a sterile environment, more recent data has documented a commensal lung microbiota (consisting primarily of Proteobacteria, Firmicutes, and Bacteroidetes) in clinically healthy individuals [79]. It has furthermore become evident that the gastrointestinal microbiota impacts that of the lungs, and can tip the balance from lung health to disease [79]. LL37 serves an important role to protect the airway epithelium both through direct antimicrobial activity as well as via activation of epithelial cells [80]. Upregulation of LL37 in human alveolar macrophages utilizes both NLR- (NOD2) as well as TLR-mediated signaling (TLR-2, -4, and -9) [81]. Studies on newborns support that lung mucosal host defense peptides, including LL37, are expressed in both mature and preterm individuals, and that concentrations increase in the presence of systemic or pulmonary infection [82].

Cystic fibrosis (CF) is caused by a genetic defect in the chloride conductance of the airway epithelium, which is associated with chronic airway infection and inflammation [83]. Importantly, the activity of HDPs is influenced to varying extents by the salt concentration in the tissue environment, which in the case of CF patients is outside the normal reference range. Data from a human bronchial xenograft model support that the endogenous LL37 concentration is similar (about 2 μg/ml) in ASF from normal versus CF xenografts. Correction of the defect in CFTR (cystic fibrosis transmembrane conductance regulator) via adenovirus mediated gene transfer to surface epithelial cells is able to correct the defect in bacterial killing, without change in the LL37 concentration [83].

Vitamin D deficiency is a common finding in patients with both non-cystic-fibrosis and CF-associated bronchiectasis [84]. Vitamin D receptors are expressed ubiquitously on epithelial and immune cells, including neutrophils, macrophages and lymphocytes [62,84]. The vitamin is converted to its active form 1,25(OH)2D3 at a cellular level, and helps to support the body’s immune competency via facilitation of HDP production and regulation of the inflammatory response [62]. Supplementation with Vitamin D suggests that some level of protection against infection, including development of bronchiectasis, can be achieved at a clinical level [84].

Pneumonia from Pseudomona aeruginosa is the second most prevalent type of lung infection in hospital settings, with mortality rates reaching up to 90% in mechanically ventilated patients [85]. In a mouse pneumonia model, compartmentalized pre-exposure to P. aeruginosa flagellin can upregulate cathelicidin (via TLR5 mediated signaling) and boost protective mucosal immunity in the lungs [85]. CRAMP is similarly required for an apt lung mucosal immune response in pneumonia due to Klebsiella pneumonia [86]. This data may be exploited further to advance treatment options for pneumonia caused by Gram-negative pathogens [85].

Approximately 1/3 of the world’s population has latent tuberculosis (TB) caused by the intracellular bacteria Mycobacterium tuberculosis, while the mortality from clinical TB in 2011 was 1.4 million globally according to WHO statisticse. Although the disease is currently preventable and treatable, the increase in multi-drug resistant TB creates cause for concern. Several studies have therefore also assessed the role of LL37 in killing of mycobacteria, and found that cathelicidin plays a role in controlling this infection. In vitro studies support that LL37 and truncated versions can directly kill pathogenic mycobacteria including M. bovis and M. tuberculosis, as well as M. smegmatis (non-pathogenic) [87]. Mycobacteria target host macrophages where they rely on maturation-arrest of the phagosome for their survival. Regardless, LL37 is able to effectively reduce the intracellular survival of bacteria [87]. The thought is that LL37 after endocytosis targets the maturation-arrested mycobacterial phagosome to kill off the bacteria, and subsequently enable fusion with lysosomes to eliminate the intracellular pathogen [87].

TB is causative of 25% of all deaths amongst HIV+ patientse. Levels of vitamin D are markedly low in bronchoalveolar lavage fluid from patients with HIV, which results in further impairment of critical immune responses and renders these individuals more susceptible to opportunistic infections [88,89]. Exogenous vitamin D3 supplementation of alveolar macrophages from HIV+ patients can rescue this impaired (innate) response through TLR-mediated signaling [88]. The active form of vitamin D also induces autophagy to inhibit the replication of HIV, while the availability of cathelicidin is central to both the suppression of HIV replication and inhibition of mycobacterial growth [89].

While the anti-viral capacity of cathelicidin has been studied less as compared to defensins, recent reports support that LL37 is an active player with respect to ridding the body of seasonal influenza. Unlike the antiviral activity exerted by other HDPs, LL37 seemingly interacts directly with the virus to cause surface disruption [90]. The antiviral activity of LL37 against different strains of influenza A virus is dose-dependent, and additive with other innate components like surfactant proteins and human neutrophil defensin peptides (HNPs). The virus neutralizing effect is reduced, however, in the presence of human serum due to LL37’s binding with high-density lipoprotein (HDL) – the key serum lipoprotein that binds to cathelicidin [90].

Urogenital system

The HMP has contributed significant new knowledge on the human body’s endogenous microbial ecosystem, and more than one clinical dogma has had to face rejection as a result. Urine from clinically normal individuals was historically also thought to be sterile, however, recent high-throughput screening and metaproteomics analysis have since characterized the “healthy urine microbiome”, which is dominated by Corynebacterium in men and Lactobacillales in women [91]. While urinary tract infection (UTI) is a significant health problem [92], the presence of uncultivated bacteria in normal urine of both genders speaks for reassessment of the current use of conventional antibiotics.

Similar to other body system, homeostasis in the urogenital tract correlates with the combined efficacy of mechanical (urine flow & bladder emptying) and biochemical (HDP production and immune cell recruitment) defense measures [39,93]. Acute UTI is often caused by uropathogenic Escherichia coli that is also the host’s most abundant fecal strain (prevalence hypothesis) [94], which highlights the continuous balancing act that takes place between compartments of the human microbiome and the host’s innate defense repertoire. The LL37 peptide is produced constitutively by resident uroepithelial and tubular renal epithelial cells as well as neutrophils, and is measureable in normal urine at a median concentration of 0.3 ng/ml – versus an eight-fold up-concentration in cases with cystitis or pyelonephritis [93]. Cathelicidin kills clinical isolates of uropathogenic E. coli at a concentration of about 30 μg/ml (MIC 8 μM) with the greatest efficacy against isolates from lower urinary tract infections (cystitis) [93].

Improved understanding of the genital tract microbiota and local host defense mechanisms will help to better define human reproductive health, and optimize therapeutic strategies in the field. Culture-independent methodologies have enabled mapping of the vaginal microbiota and revealed that Lactobacillus species, as anticipated in human females, constitute the predominant finding in most women. However, a diverse microflora dominated by facultative and strict anaerobic species exists in a significant number of clinically normal females [95]. The dynamics in the spectrum of the vaginal flora in light of life span transitions and between different ethnicities has also been elucidated through HMP-funded research [95].

Research focused on the male reproductive tract microbiota is more limited, although a recent review outlines current knowledge in the field [96]. While the upper male genital tract is seemingly germ-free, the lower tract hosts a microbiota dominated by Actinobacteria, Bacteroidetes, Firmicutes, Fusobacteria, and Proteobacteria. Corynebacterium (Actinobacteria) is often identified as a commensal in the male reproductive tract, while it may also be associated with disease [96]. The interaction between the microbiota in the male versus female reproductive tract is similarly understudied, although post-coital increase in coliforms and a shift towards a vaginosis-type microflora has been reported [96].

Human cationic antimicrobial protein expression has been documented in tissues and body fluid from the male and female reproductive tract, including testes, epididymis, spermatozoa, seminal plasma, vagina and cervix [97-99]. The concentration of hCAP18 in seminal plasma is e.g. 70-fold higher than in blood, while 6.6×106 hCAP18 molecules are associated with each spermatozoa based on ELISA analysis [99]. When exposed to a pH equivalent to that of the vagina, the prostate-derived protease gastricsin is able to cleave the inactive hCAP18 into a 38 AAs antimicrobial peptide ALL-38, which represents one protective mechanism working to support reproductive tract health [100].

Other Clinical Considerations for Cathelicidin

Circulatory system

Morbidity and mortality from cardiovascular disease, in particular coronary artery disease (CAD), place a major burden on the healthcare system in industrialized countries. Atherosclerosis refers to inflammation of the vessel wall, which involves a network of cellular and molecular components. Well-established cardiovascular risk factors include hypertension, hypercholesterolemia, smoking, and diabetes mellitus, all of which exert potent pro-inflammatory effects on the vasculature [101].

LL37 cathelicidin is expressed at a six-fold increase in atherosclerotic lesions, and mainly by macrophages and endothelial cells. The peptide induces expression of the adhesion molecule ICAM-1 and the MCP-1 chemokine in endothelial cells – suggesting an immunoregulatory role – and may impact neovascularization [102]. In a murine arteriosclerosis model neutrophil-derived cathelicidin (CRAMP) promotes disease via recruitment of inflammatory monocytes [103]. CRAMP released from neutrophils is transported across the endothelium, and promote cell adhesion of classical (but not non-classical)f monocytes [104].

Percutaneous coronary intervention with stent implantation is a common treatment option used to dilate arterial narrowing caused by atherosclerotic plaque formation [105]. Drug-eluting stents serve to reduce the occurrence of restenosis through release of anti-inflammatory and anti-proliferative compounds. Interestingly, experimental studies suggest that a biofunctionalized LL37-coated stent can limit neointima formation to reduce in-stent stenosis in mice [105].

Seroprevalence studies suggest that infectious agents such as Chlamydia pneumonia (Cpn), Helicobacter pylori, enteroviruses, cytomegalovirus (CMV), herpes simplex virus (HSV-1, and -2), and hepatitis A virus are implicated in the atherosclerotic process [106]. The human respiratory pathogen Chlamydia pneumonia has received much research attention in an effort to establish a causative link to atherosclerosis. The presence of Cpn–specific DNA in peripheral monocytes from patients with CAD suggests that this is the pathogen’s mode of transportation from the lungs into the systemic circulation [107]. Animal studies indicate that Cpn can start the atherosclerotic process, or accelerate lipid-induced atherosclerosis in ApoE-deficient mice [106].

While experimental studies support the Cpn-CAD link, large-scale clinical trials like WIZARD, AZACS, ACES and PROVE-IT have, however, all been unable to establish a clinical benefit of antibiotics in prevention and treatment of CAD [101,108-110]. Furthermore, Cpn appears unaffected by LL37’s antimicrobial action [102].

Indeed, infection as a causative factor in human CAD remains controversial. Nevertheless, neutrophil-derived LL37 is central to host protection in bacterial invasion of the circulatory system, and the peptide’s multifunctional properties make it an important template for the development of novel treatment solutions for sepsis [111]. Data from a murine sepsis model suggest that administration of LL37 in combination with G-CSF is the most effective treatment strategy in reducing bacterial growth and lethality in neutropenic sepsis [112].

Conclusion

In many ways, cathelicidins set the stage for the clinical outcome in several infectious disease processes and inflammatory disorders studied to date. Knowledge on the mechanisms that impact cathelicidin expression patterns in the body is important, because it holds the potential of offering new and improved treatment strategies. Meanwhile, the omnipresence of the human microbiota provides food for thought in light of how conventional antibiotics are administered.

The pipeline for research and development of a new drug - antibiotic or otherwise – easily spans a decade or more. Conventional antibiotics have specific targets that define their mechanism of action, while also rendering them more vulnerable to pathogen countermeasures. Antimicrobials with novel modi operandi are consequently in high demand in a world faced with an increasing number of multi-drug resistant pathogens, as well as a changing panorama of infections.

Our emphasis in this review has been on the naturally occurring human cathelicidin, and the ways the peptide operate to preserve human health across and within organ systems. Evolution has modified and optimized the biological function of cathelicidins in animal species over time. “Immunological inspiration” attained from domestic and exotic species can likely provide us with superior templates for use in development of new immunomodulating and antimicrobial drugs.

While the One Health movement has placed emphasis on zoonotic/vector-borne infections, the more recent Zoobiquity initiative takes a broader, species-spanning, approach to health and medicine viewed in light of evolution. Molecular solutions utilized by more robust and (evolutionarily-speaking) longer-lived species can also help guide the design of new drugs that are more fit-to-fight challenging infections and possibly other debilitating diseases. It is through this “zoobiquitous” research approach that the search for new drugs is most likely to excel.

Acknowledgements

The authors would like to acknowledge the NIH Center of Biomedical Research Excellence (COBRE) award P20-RR017686, and KSU Program Director Daniel Marcus for continued support of our research. We also thank Emma Gordon for excellent technical assistance.

Conflict of Interest

The authors declare no conflicts of interest.

aThe NCBI Reference Sequence (RefSeq) database is a non-redundant set of reference standards derived from the INSDC databases, while the International Nucleotide Sequence Database Collaboration (INSDC) represents an archival repository of all sequences submitted to GenBank, the European Nucleotide Archive, and the DNA Data Bank of Japan. Sequence data is exchanged daily between collaborators to ensure current worldwide coverage (http://www.ncbi.nlm.nih.gov/books; Bookshelf ID: NBK50679. Accessed 01.09.13).

bHomologous to cathelin, which is a cathepsin L protease inhibitor from pig leukocytes (FEBS Lett. 1989 Sep 25;255(2):211-4).

cNational Institutes of Health (NIH) Human Microbiome Project, http://www.hmpdacc.org

dNIH News, http://www.nih.gov/news/health/jun2012/nhgri-13.htm (retrieved 02.05.13).

eTuberculosis, Fact Sheet No 104, http://www.who.int/mediacentre/factsheets/fs104/en/#, World Health Organization (retrieved 02.22.13)

fClassical monocytes are linked to inflammatory processes, e.g. the early response in infections, as well as sterile inflammatory conditions, including atherosclerosis or myocardial infarction. Non-classical monocytes are involved in phagocytosis, tissue remodeling, and wound healing. Monocyte subsets are divided according to CD14 and CD16 expression profiles (Circ Res. 2013 Jan 2. [Epub ahead of print], PMID: 23283724)

References

  1. Boman HG (2003) Antibacterial peptides: basic facts and emerging concepts. J Intern Med 254: 197-215.
  2. Tomasinsig L, Zanetti M (2005) The cathelicidins--structure, function and evolution. Curr Protein Pept Sci 6: 23-34.
  3. Matzinger P (2002) The danger model: a renewed sense of self. Science 296: 301-305.
  4. Zanetti M (2005) The role of cathelicidins in the innate host defenses of mammals. Curr Issues Mol Biol 7: 179-196.
  5. Hancock RE, Sahl HG (2006) Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nat Biotechnol 24: 1551-1557.
  6. Natterson-Horowitz B, Bowers K (2012) Zoobiquity: What Animals Can Teach Us About Health and the Science of Healing. (1st edn) Knopf, New York.
  7. Zanetti M, Gennaro R, Romeo D (1995) Cathelicidins: a novel protein family with a common proregion and a variable C-terminal antimicrobial domain. FEBS Lett 374: 1-5.
  8. Zanetti M (2004) Cathelicidins, multifunctional peptides of the innate immunity. J Leukoc Biol 75: 39-48.
  9. Zanetti M, Gennaro R, Skerlavaj B, Tomasinsig L, Circo R (2002) Cathelicidin peptides as candidates for a novel class of antimicrobials. Curr Pharm Des 8: 779-793.
  10. Zasloff M (2002) Antimicrobial peptides of multicellular organisms. Nature 415: 389-395.
  11. Hase K, Murakami M, Iimura M, Cole SP, Horibe Y, et al. (2003) Expression of LL-37 by human gastric epithelial cells as a potential host defense mechanism against Helicobacter pylori. Gastroenterology 125: 1613-1625.
  12. Yan H, Hancock RE (2001) Synergistic interactions between mammalian antimicrobial defense peptides. Antimicrob Agents Chemother 45: 1558-1560.
  13. Nizet V, Gallo RL (2003) Cathelicidins and innate defense against invasive bacterial infection. Scand J Infect Dis 35: 670-676.
  14. Linde A, Wachter B, Horner OP, Dib L, Ross C, Tamayo AR et al. (2009). Natural History of Innate Host Defense Peptides. Probiotics & Antimicro Prot 1: 97-112.
  15. Agerberth B, Lee JY, Bergman T, Carlquist M, Boman HG, et al. (1991) Amino acid sequence of PR-39. Isolation from pig intestine of a new member of the family of proline-arginine-rich antibacterial peptides. Eur J Biochem 202: 849-854.
  16. Agerberth B, Gunne H, Odeberg J, Kogner P, Boman HG, et al. (1995) FALL-39, a putative human peptide antibiotic, is cysteine-free and expressed in bone marrow and testis. Proc Natl Acad Sci U S A 92: 195-199.
  17. Gudmundsson GH, Agerberth B, Odeberg J, Bergman T, Olsson B, et al. (1996) The human gene FALL39 and processing of the cathelin precursor to the antibacterial peptide LL-37 in granulocytes. Eur J Biochem 238: 325-332.
  18. Cowland JB, Johnsen AH, Borregaard N (1995) hCAP-18, a cathelin/pro-bactenecin-like protein of human neutrophil specific granules. FEBS Lett 368: 173-176.
  19. Larrick JW, Hirata M, Balint RF, Lee J, Zhong J, et al. (1995) Human CAP18: a novel antimicrobial lipopolysaccharide-binding protein. Infect Immun 63: 1291-1297.
  20. Elloumi HZ, Holland SM (2008) Complex regulation of human cathelicidin gene expression: novel splice variants and 5'UTR negative regulatory element. Mol Immunol 45: 204-217.
  21. Sørensen O, Arnljots K, Cowland JB, Bainton DF, Borregaard N (1997) The human antibacterial cathelicidin, hCAP-18, is synthesized in myelocytes and metamyelocytes and localized to specific granules in neutrophils. Blood 90: 2796-2803.
  22. Sørensen OE, Follin P, Johnsen AH, Calafat J, Tjabringa GS, et al. (2001) Human cathelicidin, hCAP-18, is processed to the antimicrobial peptide LL-37 by extracellular cleavage with proteinase 3. Blood 97: 3951-3959.
  23. Davidson DJ, Currie AJ, Reid GS, Bowdish DM, MacDonald KL, et al. (2004) The cationic antimicrobial peptide LL-37 modulates dendritic cell differentiation and dendritic cell-induced T cell polarization. J Immunol 172: 1146-1156.
  24. Alalwani SM, Sierigk J, Herr C, Pinkenburg O, Gallo R, et al. (2010) The antimicrobial peptide LL-37 modulates the inflammatory and host defense response of human neutrophils. Eur J Immunol 40: 1118-1126.
  25. Gallo RL, Kim KJ, Bernfield M, Kozak CA, Zanetti M, et al. (1997) Identification of CRAMP, a cathelin-related antimicrobial peptide expressed in the embryonic and adult mouse. J Biol Chem 272: 13088-13093.
  26. Kin NW, Chen Y, Stefanov EK, Gallo RL, Kearney JF (2011) Cathelin-related antimicrobial peptide differentially regulates T- and B-cell function. Eur J Immunol 41: 3006-3016.
  27. Bals R, Lang C, Weiner DJ, Vogelmeier C, Welsch U, et al. (2001) Rhesus monkey (Macaca mulatta) mucosal antimicrobial peptides are close homologues of human molecules. Clin Diagn Lab Immunol 8: 370-375.
  28. Johansson J, Gudmundsson GH, Rottenberg ME, Berndt KD, Agerberth B (1998) Conformation-dependent antibacterial activity of the naturally occurring human peptide LL-37. J Biol Chem 273: 3718-3724.
  29. Turner J, Cho Y, Dinh NN, Waring AJ, Lehrer RI (1998) Activities of LL-37, a cathelin-associated antimicrobial peptide of human neutrophils. Antimicrob Agents Chemother 42: 2206-2214.
  30. NIH HMP Working Group, Peterson J, Garges S, Giovanni M, McInnes P, et al. (2009) The NIH Human Microbiome Project. Genome Res 19: 2317-2323.
  31. Human Microbiome Jumpstart Reference Strains Consortium, Nelson KE, Weinstock GM, Highlander SK, Worley KC, et al. (2010) A catalog of reference genomes from the human microbiome. Science 328: 994-999.
  32. Kostman R (1975) Infantile genetic agranulocytosis. A review with presentation of ten new cases. Acta Paediatr Scand 64: 362-368.
  33. KOSTMANN R (1956) Infantile genetic agranulocytosis; agranulocytosis infantilis hereditaria. Acta Paediatr Suppl 45: 1-78.
  34. Aytekin C, Germeshausen M, Tuygun N, Tanir G, Dogu F, et al. (2010) Eponym. Kostmann disease. Eur J Pediatr 169: 657-660.
  35. Carlsson G, Fasth A (2001) Infantile genetic agranulocytosis, morbus Kostmann: presentation of six cases from the original "Kostmann family" and a review. Acta Paediatr 90: 757-764.
  36. Pütsep K, Carlsson G, Boman HG, Andersson M (2002) Deficiency of antibacterial peptides in patients with morbus Kostmann: an observation study. Lancet 360: 1144-1149.
  37. Kido J, Bando M, Hiroshima Y, Iwasaka H, Yamada K, et al. (2012) Analysis of proteins in human gingival crevicular fluid by mass spectrometry. J Periodontal Res 47: 488-499.
  38. Davidopoulou S, Diza E, Menexes G, Kalfas S (2012) Salivary concentration of the antimicrobial peptide LL-37 in children. Arch Oral Biol 57: 865-869.
  39. Chromek M, Arvidsson I, Karpman D (2012) The antimicrobial peptide cathelicidin protects mice from Escherichia coli O157:H7-mediated disease. PLoS One 7: e46476.
  40. Guarner F, Malagelada JR (2003) Gut flora in health and disease. Lancet 361: 512-519.
  41. Ho S, Pothoulakis C, Koon HW (2013) Antimicrobial peptides and colitis. Curr Pharm Des 19: 40-47.
  42. Koon HW, Shih DQ, Chen J, Bakirtzi K, Hing TC, et al. (2011) Cathelicidin signaling via the Toll-like receptor protects against colitis in mice. Gastroenterology 141: 1852-1863.
  43. Schauber J, Rieger D, Weiler F, Wehkamp J, Eck M, et al. (2006) Heterogeneous expression of human cathelicidin hCAP18/LL-37 in inflammatory bowel diseases. Eur J Gastroenterol Hepatol 18: 615-621.
  44. Strandberg KL, Richards SM, Gunn JS (2012) Cathelicidin antimicrobial peptide expression is not induced or required for bacterial clearance during salmonella enterica infection of human monocyte-derived macrophages. Infect Immun 80: 3930-3938.
  45. Rosenberger CM, Gallo RL, Finlay BB (2004) Interplay between antibacterial effectors: a macrophage antimicrobial peptide impairs intracellular Salmonella replication. Proc Natl Acad Sci U S A 101: 2422-2427.
  46. Cobo ER, He C, Hirata K, Hwang G, Tran U, et al. (2012) Entamoeba histolytica induces intestinal cathelicidins but is resistant to cathelicidin-mediated killing. Infect Immun 80: 143-149.
  47. Rico-Mata R, De Leon-Rodriguez LM, Avila EE (2013) Effect of antimicrobial peptides derived from human cathelicidin LL-37 on Entamoeba histolytica trophozoites. Exp Parasitol 133: 300-306.
  48. Murakami M, Lopez-Garcia B, Braff M, Dorschner RA, Gallo RL (2004) Postsecretory processing generates multiple cathelicidins for enhanced topical antimicrobial defense. J Immunol 172: 3070-3077.
  49. Ménard S, Förster V, Lotz M, Gütle D, Duerr CU, et al. (2008) Developmental switch of intestinal antimicrobial peptide expression. J Exp Med 205: 183-193.
  50. Cederlund A, Kai-Larsen Y, Printz G, Yoshio H, Alvelius G, et al. (2013) Lactose in human breast milk an inducer of innate immunity with implications for a role in intestinal homeostasis. PLoS One 8: e53876.
  51. Ren SX, Cheng AS, To KF, Tong JH, Li MS, et al. (2012) Host immune defense peptide LL-37 activates caspase-independent apoptosis and suppresses colon cancer. Cancer Res 72: 6512-6523.
  52. Laurent A, Mistretta F, Bottigioli D, Dahel K, Goujon C, et al. (2007) Echographic measurement of skin thickness in adults by high frequency ultrasound to assess the appropriate microneedle length for intradermal delivery of vaccines. Vaccine 25: 6423-6430.
  53. Verbraecken J, Van de Heyning P, De Backer W, Van Gaal L (2006) Body surface area in normal-weight, overweight, and obese adults. A comparison study. Metabolism 55: 515-524.
  54. Carretero M, Escámez MJ, García M, Duarte B, Holguín A, et al. (2008) In vitro and in vivo wound healing-promoting activities of human cathelicidin LL-37. J Invest Dermatol 128: 223-236.
  55. Grice EA, Kong HH, Conlan S, Deming CB, Davis J, et al. (2009) Topographical and temporal diversity of the human skin microbiome. Science 324: 1190-1192.
  56. Schauber J, Gallo RL (2008) The vitamin D pathway: a new target for control of the skin's immune response? Exp Dermatol 17: 633-639.
  57. Braff MH, Di Nardo A, Gallo RL (2005) Keratinocytes store the antimicrobial peptide cathelicidin in lamellar bodies. J Invest Dermatol 124: 394-400.
  58. Cogen AL, Yamasaki K, Muto J, Sanchez KM, Crotty Alexander L, et al. (2010) Staphylococcus epidermidis antimicrobial delta-toxin (phenol-soluble modulin-gamma) cooperates with host antimicrobial peptides to kill group A Streptococcus. PLoS One 5: e8557.
  59. Gallo RL, Nakatsuji T (2011) Microbial symbiosis with the innate immune defense system of the skin. J Invest Dermatol 131: 1974-1980.
  60. Wang Z, MacLeod DT, Di Nardo A (2012) Commensal bacteria lipoteichoic acid increases skin mast cell antimicrobial activity against vaccinia viruses. J Immunol 189: 1551-1558.
  61. Schauber J, Gallo RL (2008) Antimicrobial peptides and the skin immune defense system. J Allergy Clin Immunol 122: 261-266.
  62. Wang TT, Nestel FP, Bourdeau V, Nagai Y, Wang Q, et al. (2004) Cutting edge: 1,25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. J Immunol 173: 2909-2912.
  63. Forrest KY, Stuhldreher WL (2011) Prevalence and correlates of vitamin D deficiency in US adults. Nutr Res 31: 48-54.
  64. Kuo IH, Yoshida T, De Benedetto A, Beck LA (2013) The cutaneous innate immune response in patients with atopic dermatitis. J Allergy Clin Immunol 131: 266-278.
  65. Crack LR, Jones L, Malavige GN, Patel V, Ogg GS (2012) Human antimicrobial peptides LL-37 and human β–defensin-2 reduce viral replication in keratinocytes infected with varicella zoster virus. Clin Exp Dermatol 37: 534-543.
  66. Hata TR, Gallo RL (2008) Antimicrobial peptides, skin infections, and atopic dermatitis. Semin Cutan Med Surg 27: 144-150.
  67. Hata TR, Kotol P, Jackson M, Nguyen M, Paik A, et al. (2008) Administration of oral vitamin D induces cathelicidin production in atopic individuals. J Allergy Clin Immunol 122: 829-831.
  68. Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, et al. (2007) Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 449: 564-569.
  69. Gregorio J, Meller S, Conrad C, Di Nardo A, Homey B, et al. (2010) Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med 207: 2921-2930.
  70. Baldwin HE (2012) Diagnosis and treatment of rosacea: state of the art. J Drugs Dermatol 11: 725-730.
  71. Bevins CL, Liu FT (2007) Rosacea: skin innate immunity gone awry? Nat Med 13: 904-906.
  72. Yamasaki K, Di Nardo A, Bardan A, Murakami M, Ohtake T, et al. (2007) Increased serine protease activity and cathelicidin promotes skin inflammation in rosacea. Nat Med 13: 975-980.
  73. Dorschner RA, Lopez-Garcia B, Massie J, Kim C, Gallo RL (2004) Innate immune defense of the nail unit by antimicrobial peptides. J Am Acad Dermatol 50: 343-348.
  74. López-García B, Lee PH, Gallo RL (2006) Expression and potential function of cathelicidin antimicrobial peptides in dermatophytosis and tinea versicolor. J Antimicrob Chemother 57: 877-882.
  75. Kulkarni MM, Barbi J, McMaster WR, Gallo RL, Satoskar AR, et al. (2011) Mammalian antimicrobial peptide influences control of cutaneous Leishmania infection. Cell Microbiol 13: 913-923.
  76. Hasleton PS (1972) The internal surface area of the adult human lung. J Anat 112: 391-400.
  77. Bals R, Weiner DJ, Wilson JM (1999) The innate immune system in cystic fibrosis lung disease. J Clin Invest 103: 303-307.
  78. Beck JM, Young VB, Huffnagle GB (2012) The microbiome of the lung. Transl Res 160: 258-266.
  79. Tjabringa GS, Aarbiou J, Ninaber DK, Drijfhout JW, Sørensen OE, et al. (2003) The antimicrobial peptide LL-37 activates innate immunity at the airway epithelial surface by transactivation of the epidermal growth factor receptor. J Immunol 171: 6690-6696.
  80. Juárez E, Carranza C, Hernández-Sánchez F, León-Contreras JC, Hernández-Pando R, et al. (2012) NOD2 enhances the innate response of alveolar macrophages to Mycobacterium tuberculosis in humans. Eur J Immunol 42: 880-889.
  81. Schaller-Bals S, Schulze A, Bals R (2002) Increased levels of antimicrobial peptides in tracheal aspirates of newborn infants during infection. Am J Respir Crit Care Med 165: 992-995.
  82. Bals R, Weiner DJ, Meegalla RL, Wilson JM (1999) Transfer of a cathelicidin peptide antibiotic gene restores bacterial killing in a cystic fibrosis xenograft model. J Clin Invest 103: 1113-1117.
  83. Bartley J, Garrett J, Grant CC, Camargo CA Jr (2013) Could Vitamin D Have a Potential Anti-Inflammatory and Anti-Infective Role in Bronchiectasis? Curr Infect Dis Rep .
  84. Yu FS, Cornicelli MD, Kovach MA, Newstead MW, Zeng X, et al. (2010) Flagellin stimulates protective lung mucosal immunity: role of cathelicidin-related antimicrobial peptide. J Immunol 185: 1142-1149.
  85. Kovach MA, Ballinger MN, Newstead MW, Zeng X, Bhan U, et al. (2012) Cathelicidin-related antimicrobial peptide is required for effective lung mucosal immunity in Gram-negative bacterial pneumonia. J Immunol 189: 304-311.
  86. Sonawane A, Santos JC, Mishra BB, Jena P, Progida C, et al. (2011) Cathelicidin is involved in the intracellular killing of mycobacteria in macrophages. Cell Microbiol 13: 1601-1617.
  87. Anandaiah A, Sinha S, Bole M, Sharma SK, Kumar N, et al. (2013) Vitamin D rescues impaired Mycobacterium tuberculosis-mediated tumor necrosis factor release in macrophages of HIV-seropositive individuals through an enhanced Toll-like receptor signaling pathway in vitro. Infect Immun 81: 2-10.
  88. Campbell GR, Spector SA (2012) Vitamin D inhibits human immunodeficiency virus type 1 and Mycobacterium tuberculosis infection in macrophages through the induction of autophagy. PLoS Pathog 8: e1002689.
  89. Tripathi S, Tecle T, Verma A, Crouch E, White M, et al. (2013) The human cathelicidin LL-37 inhibits influenza A viruses through a mechanism distinct from that of surfactant protein D or defensins. J Gen Virol 94: 40-49.
  90. Fouts DE, Pieper R, Szpakowski S, Pohl H, Knoblach S, et al. (2012) Integrated next-generation sequencing of 16S rDNA and metaproteomics differentiate the healthy urine microbiome from asymptomatic bacteriuria in neuropathic bladder associated with spinal cord injury. J Transl Med 10: 174.
  91. Säemann MD, Hörl WH, Weichhart T (2007) Uncovering host defences in the urinary tract: cathelicidin and beyond. Nephrol Dial Transplant 22: 347-349.
  92. Chromek M, Slamová Z, Bergman P, Kovács L, Podracká L, et al. (2006) The antimicrobial peptide cathelicidin protects the urinary tract against invasive bacterial infection. Nat Med 12: 636-641.
  93. Moreno E, Andreu A, Pigrau C, Kuskowski MA, Johnson JR, et al. (2008) Relationship between Escherichia coli strains causing acute cystitis in women and the fecal E. coli population of the host. J Clin Microbiol 46: 2529-2534.
  94. Ma B, Forney LJ, Ravel J (2012) Vaginal microbiome: rethinking health and disease. Annu Rev Microbiol 66: 371-389.
  95. Mändar R (2013) Microbiota of male genital tract: impact on the health of man and his partner. Pharmacol Res 69: 32-41.
  96. Doussau M, Lasserre A, Hammami-Hamza S, Massaad C, Gasc JM, et al. (2008) Testicular and epididymal dual origin of hCAP-18/SOB3, a human sperm protein. Fertil Steril 90: 853-856.
  97. Frohm Nilsson M, Sandstedt B, Sørensen O, Weber G, Borregaard N, et al. (1999) The human cationic antimicrobial protein (hCAP18), a peptide antibiotic, is widely expressed in human squamous epithelia and colocalizes with interleukin-6. Infect Immun 67: 2561-2566.
  98. Malm J, Sørensen O, Persson T, Frohm-Nilsson M, Johansson B, et al. (2000) The human cationic antimicrobial protein (hCAP-18) is expressed in the epithelium of human epididymis, is present in seminal plasma at high concentrations, and is attached to spermatozoa. Infect Immun 68: 4297-4302.
  99. Sørensen OE, Gram L, Johnsen AH, Andersson E, Bangsbøll S, et al. (2003) Processing of seminal plasma hCAP-18 to ALL-38 by gastricsin: a novel mechanism of generating antimicrobial peptides in vagina. J Biol Chem 278: 28540-28546.
  100. Neumann FJ (2002) Chlamydia pneumoniae-atherosclerosis link: a sound concept in search for clinical relevance. Circulation 106: 2414-2416.
  101. Edfeldt K, Agerberth B, Rottenberg ME, Gudmundsson GH, Wang XB, et al. (2006) Involvement of the antimicrobial peptide LL-37 in human atherosclerosis. Arterioscler Thromb Vasc Biol 26: 1551-1557.
  102. Döring Y, Drechsler M, Wantha S, Kemmerich K, Lievens D, et al. (2012) Lack of neutrophil-derived CRAMP reduces atherosclerosis in mice. Circ Res 110: 1052-1056.
  103. Wantha S, Alard JE, Megens RT, van der Does AM, Döring Y, et al. (2013) Neutrophil-derived cathelicidin promotes adhesion of classical monocytes. Circ Res 112: 792-801.
  104. Soehnlein O, Wantha S, Simsekyilmaz S, Döring Y, Megens RT, et al. (2011) Neutrophil-derived cathelicidin protects from neointimal hyperplasia. Sci Transl Med 3: 103ra98.
  105. Leinonen M, Saikku P (2002) Evidence for infectious agents in cardiovascular disease and atherosclerosis. Lancet Infect Dis 2: 11-17.
  106. Boman J, Söderberg S, Forsberg J, Birgander LS, Allard A, et al. (1998) High prevalence of Chlamydia pneumoniae DNA in peripheral blood mononuclear cells in patients with cardiovascular disease and in middle-aged blood donors. J Infect Dis 178: 274-277.
  107. Cercek B, Shah PK, Noc M, Zahger D, Zeymer U, et al. (2003) Effect of short-term treatment with azithromycin on recurrent ischaemic events in patients with acute coronary syndrome in the Azithromycin in Acute Coronary Syndrome (AZACS) trial: a randomised controlled trial. Lancet 361: 809-813.
  108. Grayston JT, Kronmal RA, Jackson LA, Parisi AF, Muhlestein JB, et al. (2005) Azithromycin for the secondary prevention of coronary events. N Engl J Med 352: 1637-1645.
  109. O'Connor CM, Dunne MW, Pfeffer MA, Muhlestein JB, Yao L, et al. (2003) Azithromycin for the secondary prevention of coronary heart disease events: the WIZARD study: a randomized controlled trial. JAMA 290: 1459-1466.
  110. Cirioni O, Giacometti A, Ghiselli R, Bergnach C, Orlando F, et al. (2006) LL-37 protects rats against lethal sepsis caused by gram-negative bacteria. Antimicrob Agents Chemother 50: 1672-1679.
  111. Cirioni O, Ghiselli R, Tomasinsig L, Orlando F, Silvestri C, et al. (2008) Efficacy of LL-37 and granulocyte colony-stimulating factor in a neutropenic murine sepsis due to Pseudomonas aeruginosa. Shock 30: 443-448.
Citation: Linde A, Lushington GH, Abello J, Melgarejo T (2013) Clinical Relevance of Cathelicidin in Infectious Disease. J Clin Cell Immunol S13:003.

Copyright: © 2013 Linde A, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top