* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download daniela tesi on line 3 - Padis
Polyclonal B cell response wikipedia , lookup
Hygiene hypothesis wikipedia , lookup
Adaptive immune system wikipedia , lookup
Drosophila melanogaster wikipedia , lookup
Cancer immunotherapy wikipedia , lookup
Psychoneuroimmunology wikipedia , lookup
Molecular mimicry wikipedia , lookup
Adoptive cell transfer wikipedia , lookup
Antimicrobial peptides wikipedia , lookup
Innate immune system wikipedia , lookup
Immunosuppressive drug wikipedia , lookup
Hepatitis B wikipedia , lookup
α -DEFENSINS EXPRESSION IN PERIPHERAL BLOOD MONONUCLEAR CELLS FROM PATIENTS WITH HEPATITIS C VIRUS INFECTIONS Dottorando: DANIELA FIOCCO Dottorato di Ricerca in Biochimica- XVII Ciclo Dipartimento di Scienze Biochimiche “A. Rossi Fanelli” Università degli Studi di Roma “La Sapienza” Coordinatore: Prof. PAOLO SARTI Dipartimento di Scienze Biochimiche “A. Rossi Fanelli” Università degli Studi di Roma “La Sapienza” Docente guida: Prof. DONATELLA BARRA Dipartimento di Scienze Biochimiche “A. Rossi Fanelli” Università degli Studi di Roma “La Sapienza” Docenti esaminatori: Prof. MAURIZIO PACI - Dip. di Scienze e Tecnologie Chimiche Università di Roma “Tor Vergata” Prof. GIOVANNI ANTONINI - Dip. di Biologia Università di Roma 3 Prof. NAZZARENO CAPITANIO - Dip. Scienze Biomediche Università di Foggia ABSTRACT Hepatitis C virus (HCV) is the major causative agent of chronic liver diseases. Some reports indicate that, besides hepatocytes, the virus can also infect lymphocytes. HCV core protein (HCV C) was recently reported to activate IL-2 gene transcription through the NFAT pathway. α-Defensins (HNPs) are antimicrobial peptides representing key components of the innate immune system in humans. The presence of an NFAT putative binding site in the HNPs promoter led us to investigate whether HCV infection could affect the expression of HNPs. HNPs expression, in peripheral blood mononuclear cells (PBMC) from HCV-infected patients, was studied and found significantly higher than that of healthy controls; a strong correlation was observed between HNPs level and liver fibrosis. By monitoring for some months patients undergoing INF-α- therapy, a drop in HNPs expression level was found. In vitro incubation of PBMC with recombinant HCV C, triggered HNP expression, while no induction was detected when stimulating with HBV core antigen; moreover, transcriptional activation by HCV C was reduced by pre-incubation with an inhibitor of NFAT cascade. Our data suggest that HCV might directly or indirectly affect HNPs expression, possibly through the participation of NFAT; furthermore, our results support the hypothesis that HNPs might be involved in HCV pathogenesis. i INDEX 1 1.1 1.1.1 1.5 1.6 1.6.1 1.6.2 1.6.3 1.6.4 INTRODUCTION……………………………………………………… 1 Antimicrobial peptides…………………………………………………… 1 Structural features and classification of antimicrobial peptides………………………………………………….1 Mechanism of action of antimicrobial peptides………………………….. 3 Innate immunity………………………………………………………….. 5 The role of antimicrobial peptides in innate immunity…………………... 6 Human antimicrobial peptides: defensins and cathelicidins……………... 7 Defensins: structure and tissue distribution……………………………… 7 Defensin genes and regulation of their expression…………………….. 10 Activity of human defensins…………………………………………... 11 Cathelicidins…………………………………………………………... 12 Roles of defensins and cathelicidins in the human immune system…………………………………………. 13 NFAT………………………………………………………………….. 13 Hepatitis C virus………………………………………………………. 16 HCV structure…………………………………………………………. 17 HCV diagnosis and therapy…………………………………………… 17 HCV pathogenesis…………………………………………………….. 18 HCV core protein……………………………………………………… 19 2 AIMS OF THE RESEARCH………………………………………... 21 3 3.1 3.1.1 MATERIAL AND METHODS……………………………………… 22 MATERIALS…………………………………………………………. 22 Patients. Study population: epidemiologic data, biochemical and virological measurements…………………………... 22 Microorganisms……………………………………………………….. 24 Cells……………………………………………………………………. 24 Growth media …………………………………………………………. 24 Drugs and solutions…………………………………………………… 24 Oligonucleotides for RT-PCR…………………………………………. 26 METHODS …………………………………………………………… 27 PBMC preparation…………………………………………………….. 27 Granulocytes preparation ……………………………………………… 27 In vitro treatment of PBMC……………………………………………. 27 Peptide extraction……………………………………………………….28 Antibacterial activity…………………………………………………... 28 ELISA …………………………………………………………………. 28 Mass spectrometry……………………………………………………... 29 RNA preparation………………………………………………………. 29 Purification of DNA fragments from agarose gel……………………... 31 Quantitative real time RT-PCR ……………………………………….. 31 Statistical analysis…………………………………………………….. 35 1.1.2 1.2 1.3 1.4 1.4.1 1.4.2 1.4.3 1.4.4 1.4.5 3.1.2 3.1.3 3.1.4 3.1.5 3.1.6 3.2 3.2.1 3.2.2 3.2.3 3.2.4 3.2.5 3.2.6 3.2.7 3.2.8 3.2.9 3.2.10 3.2.11 4 4.1 RESULTS AND DISCUSSION …………………………………….. 36 Expression profiling of HNPs in HCV patients……………………….. 36 Quantitative real time reverse transcription PCR (qRT-PCR): validation of the housekeeping genes ………………………………... 36 ii Quantitative real time RT-PCR analysis of HNP genes ………………. 39 ELISA ………………………………………………………………….. 41 Antibacterial activity ………………………………………………….. 42 MALDI ToF analysis …………………………………………………. 43 4.2 4.3 4.8. HNPs expression in other hepatic pathologies …………………………44 Expression profiles of other immune-related genes in HCV-patients ……………………………………………………….. 46 HNPs expression in HIV patients ……………………………………... 48 HNPs expression in selected cell subsets ………………………………49 Correlation between HNP level and liver damage ……………………. 50 In vitro experiments on PBMC ………………………………………... 52 Effect of HCV C, cyclosporin A and the core protein of hepatitis B virus …………………………………………………… 52 Effect of phorbol 12-myristate 13-acetate, ionomycin and phytohemagglutinin ……………………………………………... 54 Selectivity of HCV C induction ……………………………………….. 56 HNP expression profile in HCV patients under antiviral therapy ……. 56 5 CONCLUSIONS …………………………………………………….. 58 6 ACKNOWLEDGMENTS …………………………………………... 60 7 REFERENCES ………………………………………………………. 61 8 PUBLICATIONS ……………………………………………………. 69 4.4 4.5 4.6 4.7 iii 1 INTRODUCTION 1.1 Antimicrobial peptides For the survival of an organism, an efficient defence for protection against microorganisms is primarily important. Antimicrobial peptides (AMPs) are essential effector molecules of the natural defence system of most living organisms. AMPs are extraordinarily widespread in nature, as they have been found in all forms of life, including bacteria, plants and animals, both vertebrates and invertebrates. Therefore, they constitute one of the most conserved theme in nature’s struggle to control pathogens. Antimicrobial peptides are gene encoded molecules of 12-50 amino acid residues, that are produced by the host organism and have the capacity to kill microbes. Although it represents an ancient defence mechanism, the importance of the production of gene-encoded peptides in the animal defence was recognised only recently. In fact, most known AMPs have been identified and characterized during the past 20 years. Insect cecropins, mammalian defensins and frog magainins were among the first antimicrobial peptides to be thoroughly characterized and clearly related to bacteria-induced immunity in animals (Boman and Hultmark, 1987; Selsted et al., 1985; Zasloff, 1987). Since then, a large number of antimicrobial peptides have been identified in many different organisms, so that they are currently counted in the hundreds. By studying AMPs, insight has been gained into fundamental host defence systems and into mechanisms of membrane-protein interaction. The interest in AMPs has been growing even due to the exciting perspective to develop new antimicrobial agents, in face of the declining efficacy of conventional antibiotics (Hancock and Leher, 1998). 1.1.1 Structural features and classification of antimicrobial peptides While classical antibiotics are produced by microorganisms through a series of reactions catalysed by different enzymes, AMPs are gene-encoded and hence ribosomally synthesised from an RNA template. AMPs are usually synthesized as inactive, larger, precursor molecules, which are then processed by specific proteases to release the mature, active peptide. Most of the precursors consist of a signal sequence, for targeting to the endoplasmic reticulum, and an anionic pro-region masking the activity of the peptide until it is secreted (Hancock, 1997). AMPs are typically composed of 12 to 50 amino acids and have molecular masses around 5 kDa. Although the AMPs vary considerably in length, sequence and secondary structure, some common structural features can be recognized: - they are cationic at physiological pH values, since they possess amino acids, such as arginine and lysine, which are positively charged at neutral pH; - they can fold in such a way as to have both a hydrophobic and a hydrophilic face of polar and positively charged residues. This amphipatic nature is presumably the key to their biological activity, enabling them to associate with the negatively charged phospholipids of the bacterial membranes and disrupt normal membrane functions. According to their chemical characteristics and 3D structure, AMPs can be classified into three major groups (Boman, 1995): 1) linear peptides without cysteines, forming amphipatic helices; 2) linear peptides with a high proportion of one or more residues (often proline, arginine or tryptophan); 3) peptides with an even number of cysteines intralinked by disulfide bonds; this class can be further divided into a sub-group of peptides with one disulfide bond only and 1 α-helix loop structure, and a larger sub-group characterized by two or more disulfide bonds and a predominant β–sheet structure. Certain AMPs undergo post-translational modifications such as C-terminal amidation (insect cecropins, amphibian temporins or bombinins), glycosylation (drosocin), amino acid isomerization (D amino acids-containing peptides from amphibia), etc. These modifications are likely to influence the biological properties of the mature peptide. Table 1.1 outlines structure, biological source and activity of some common antimicrobial peptides. Table 1.1. Structural class, source and activity of representative antimicrobial peptides STRUCTURE Linear, cysteine-free α-helix PEPTIDE cecropins dermaseptins magainins temporins bombinins LL37 Linear with high content of certain residues 1 disulfide bond, cyclic apidaecins drosocin PR-39 indolicidin Bac -5, Bac-7 brevinins esculentins 2 or more α -defensins disulfides with predominant β-defensins β–sheet structure defensins θ-defensins SOURCE insect hemolymph REFERENCE Boman and Hultmark, 1987 frog skin antibacterial and Mor A, et al., 1991 antifungal Xenopus laevis skin antibacterial Zasloff, 1987 Moore et al., 1991 and stomach Rana species skin antibacterial Simmaco et al., 1996 Bombina species skin antibacterial Simmaco et al., 1991 Human leukocytes antibacterial, Agerberth et al., 1995 chemotactic and skin bee hemolymph antibacterial Casteels et al., 1989 Drosophila antibacterial Bulet et al., 1993 hemolymph Pig intestine antibacterial Agerberth et al., 1991 Bovine neutrophils antibacterial Selsted et al., 1992 Mammalian antibacterial Frank et al., 1990 leukocytes Rana species skin antibacterial, Morikawa et al., 1992 hemolytic Rana species skin antibacterial, Simmaco et al., 1994 antifungal Mammalian antibacterial, Selsted et al., 1983; 1985 phagocytes, mouse antifungal, and human intestinal antiviral, Paneth cells cytotoxic Mammalian epithelia, antibacterial, Selsted et al., 1993 bovine neutrophils, chemotactic chicken leukocytes Insect hemolymph, antibacterial Hoffmann and Hetru, plant tissue 1992, Brokaert et al., 1995 Monkey leukocytes antiviral Tang et al., 1999 2 ACTIVITY antibacterial 1.1.2 Mechanism of action of antimicrobial peptides AMPs usually have broad activity spectra and may kill both Gram-negative and Grampositive bacteria, as well as fungi. Some AMPs have also antiviral activity and cytotoxic effects on eukaryotic cells. Susceptible microorganisms are killed in vitro by AMPs at minimum inhibitory concentration (MIC) in the micromolar range (0.5-8 µg/mL). The killing rate is usually much faster than that observed with conventional antibiotics; moreover, AMPs do not induce resistant mutants so frequently as the conventional antibiotics do. Both the above characteristics may be ascribed to their physical mechanism of action: most of the AMPs appear to function as membrane-active agents disturbing its permeability and causing its disgregation (Hancock et al., 1995) Different approaches in in vitro studies have helped in elucidating the mode of action of the cationic antimicrobial peptides, including the well studied peptides melittin, magainin, cecropin and defensin (Christensen et al. 1988; Agawa et al., 1991; Kagan et al., 1990; Matsuzaki et al., 1989). The use of convenient assay bacteria or common animal pathogens have defined the spectra of action, while experiments on artificial membranes, lipid vesicles and different spectroscopic methods, as well as the use of synthetic peptides, have been applied to understand the lytic mechanism of the antibacterial peptides. These studies support the formation of channels in some cases, and a general collapse of the membrane in other. By contrast to the detailed and numerous in vitro studies, there are only a few in vivo experiments to test the biological function of AMPs. Therefore, it is likely that the gap of complexity between an artificial membrane and the envelope of a bacterium is too large to provide an exhaustive, definitive conclusion about the mechanism of lysis occurring in vivo. As a result of the extensive in vitro studies, a general mechanism of action can be outlined (Hancock, 1997; Shai, 2002). The positively charged AMPs interact first with the negatively charged sites on lipopolysaccharides (LPS), on the surface of Gram-negative bacteria, and are subsequently taken up through a process called “self-promoted uptake”: the affected membrane develops transient “cracks” allowing passage of a variety of molecules, including the perturbing peptide itself. The killing event for both Gram-positive and Gram-negative bacteria is then the chemical attack on the cytoplasmic (inner) membrane (Fig. 1.1): the peptides interact with the negatively charged membranes and, as a consequence of this interaction and of the high electrical potential of the membrane, they re-orient and undergo a transition from an unstructured to a structured form. Thus, they aggregate into clusters, directing their hydrophobic faces towards the membrane core, while pointing their hydrophilic faces either inwards, to form channels, or towards the phospholipid headgroups of the membrane. In both cases, either by the formation of transmembrane pores (barrel-stave model) or by a general, progressive destabilization of the membrane structure (carpet model), membrane collapses, and the free ions flux dissipates its potential causing cell lysis and death. 3 CARPET MODEL BARRELSTAVE MODEL Fig. 1.1. Schematic mechanism of interaction between the bacterial membrane and antimicrobial peptides; hydrophilic faces are in red. The selectivity for microbial cells compared to host cells is favoured by: i) the unique high content of surface anionic lipids, ii) the large transmembrane potentials and iii) the lack of cholesterol, that are all peculiar characteristics of the bacterial membrane. This mechanism of action explains some of the clinically desirable properties of AMPs. First of all, the difficult selection of mutant resistant strains: unlike classical antibiotics, which have a specific single target (e.g. a key enzyme for the metabolism of the pathogen), AMPs are directed against a fundamental, conserved structure as complex as the cytoplasmic membrane is, and, therefore, only simultaneous, multiple genes mutations in the bacterial genome could prevent their destructive action. Moreover, while most of the traditional antibiotics promote endotoxinemia by releasing LPS (endotoxins) during cell killing, some AMPs can bind to LPS, hence neutralizing their toxic effects for the host organism (Gough et al., 1996). The protective mechanism appears to be the binding of LPS in such a way that it fails to induce tumour necrosis factor. This protective role confers to these peptides a great advantage over other antimicrobial agents. Another intriguing property of the AMPs is that they can enhance the activity of other antibiotics, facilitating their penetration into the target cell in a synergic effect. 4 1.2 Innate immunity Metazoans have evolved two basic host defence mechanisms against invading microorganisms: the innate and adaptive immune systems. Innate, non adaptive immunity represents a first line of defence and involves both a cellular response by specialized blood cells and the rapid synthesis of proteins with a wide range of activities (opsonisation of microbes, inhibition of proteases, intercellular signalling or direct antimicrobial action) (Medzihtof and Janeway, 1997). Whereas the innate immune response is present in all classes of the animal kingdom, adaptive immunity is restricted to gnatostome vertebrates. Distinguishing features of each type of immunity are given in Table 1.2. Summarizing, the innate immune system relies on a limited number of receptors, able to recognize the vast variety of the molecular structures associated with pathogens, and on defensive mechanisms that can counteract the broadest spectrum of potential pathogens. Thereby, the innate immunity provides a selective but not highly specific recognition system and a rapid initial response. By contrast, the adaptive immunity lacks both rapidity and broader activity, while gaining extraordinary efficiency and specificity. From an evolutionary point of view, the innate immune system has probably predated the adaptive immune response: first, innate host defences are found in all multicellular organisms, while adaptive immunity is found only in higher vertebrates; second, the innate recognition systems distinguish self from non-self perfectly; third, the innate immunity uses receptors and signalling pathways which are strikingly conserved throughout the animal kingdom and seem rather ancient in their lineage. Table 1.2. Main characteristics of innate and adaptive immunity Type of response Mechanisms of recognition Distribution INNATE IMMUNITY Antigen independent Not antigen specific Immediate and maximal Exposure results in no immunologic memory Pathogen recognized by receptors encoded in the germ line Receptors have broad specificity: recognize many related molecular structures called PAMPs (pathogen-associated molecular patterns) PAMPs are essential polysaccharides and polynucleotides that differ little from one pathogen to another, but are not found in the host Receptors are PRR (pattern recognition receptor), such as the Toll-like receptor family In all metazoan 5 ADAPTIVE IMMNUNITY Antigen-dependent Antigen specific Slow (because clones of responding cells need to develop) Exposure results in immunologic memory Pathogen recognized by receptors generated randomly by somatic rearrangement of genes Receptors have narrow specificity; recognize a particular epytope Most epytopes are derived from polypeptides and reflect the individuality of the pathogen Receptors are B-cell (BCR) and T-cell (TCR) receptors for antigen In jawed vertebrates only The innate immune response is based on all the pre-existing (pre-encoded) anti-infection tools of the host. These include anatomical and physiological barriers, chemical factors and cellular components, as summarised in Table 1.3. Table 1.3. Defence tools of the innate immunity Anatomical barriers Physiological barriers Chemical factors Cellular components skin, saliva, tears, intestinal movements, oscillation of broncho-pulmonary cilia, etc. body temperature and pH values which may inhibit microbial growth lysozyme; cytokines (interferons and TNF); complement; transferrin and lactoferrin; peptide antibiotics, surfactants proteins A and D, fibronectin, lectins, etc. neutrophils, macrophages, monocytes, NK cells The chemical factors, mostly secretory molecules, play a fundamental protective role: lysozyme, in serum and tears, breaks down the bacterial cell walls; cytokines such as interferons are secreted by virus-infected cells and prevent further viral replication; tumour necrosis factor alpha (TNF-α) suppresses viral replication and triggers phagocytes; activated complement is a group of serum proteins that participate in an enzymatic cascade, whose ultimate goal is damaging the membrane of the pathogen; transferrin and lactoferrin deprive invading organism of iron; antimicrobial compounds, such as peptide antibiotics, directly damage microbes membrane, and surfactants proteins A and D enhance phagocytosis; lectins can bind sugar molecules on the cell surface making them sticky and causing them to clump. Also cellular components greatly contribute to host protection: neutrophils and macrophages do so in virtue of their chemotactic and phagocytic capacity. Innate immunity has long been considered as a separate entity from the adaptive immune response and has been regarded to be of secondary importance in the hierarchy of immune functions. In the past years, however, interest in innate immunity has grown considerably. Recent studies demonstrate that the two systems are intimately correlated: the innate system controls the initiation of the adaptive immune response and instructs it to develop a particular effector response (Fearon and Locksley, 1996). The function of lymphocytes bearing clonally rearranged receptors is dependent on signals provided by the innate recognition system (Romagnani, 1992). Thus, innate and adaptive immune responses are integrated as a single immune system, with the innate response preceding, and being necessary for, the adaptive immune response. 1.3 The role of antimicrobial peptides in innate immunity In animals, AMPs have been identified in several physiological contexts: secreted onto mucosal surfaces and skin (e.g. human epithelial defensins and amphibian skin bioactive peptides) or into internal body fluids (e.g. cecropins in the haemolymph of insects); accumulated inside circulating fagocytic cells (e.g. the human neutrophil defensins). Both their powerful antimicrobial properties and their “strategic” location in specialised immune cells, or at the interface, between the organism and its environment, provide an indirect but strong evidence that AMPs participate in resistance to pathogen attack. Moreover, the induction of their expression in response to bacterial infection and tissue injury, highlights their central role in what is generally indicated as the innate immune system. Because host defence mechanisms are multiple and complex, it is difficult to demonstrate the contribution of any single peptide or mechanism. Nevertheless, a compelling experimental 6 cue of the physiological importance of AMPs has come from studies on cystic fibrosis: the salt inactivation of the airway epithelial AMPs (β-defensins) determines colonization and, sometimes, fatal infections, by Pseudomonas aeruginosa, which are characteristic in patients affected by this genetic disease (Goldman et al., 1997). More recently, a strong association was shown between oral diseases and lack of measurable AMPs (LL37) in the saliva of patients with Kostmann syndrome, a severe congenital neutropenia (Putsep et al., 2002). According to Hans G. Boman (1996, 2003), AMPs represent an ideal first line of defence and can be considered key effector molecules in innate immunity, particularly in mounting an early and immediate response against invading micro-organisms. AMPs production is much faster than that of immunoglobulins and this feature is not negligible when considering that the growth rate of most microorganisms enormously overcomes that of a typical animal lymphocyte. The adaptive immune system alone would not be able to efficiently counteract a microbial infection. A peptide-based defence is not only necessary, it is easy to control and is more energy- and information-saving, when compared to a clonally-based defence. All these considerations underline the great survival value of peptide antibiotics in the immune system of animals. Innate immunity and, specifically, AMPs probably play a major role even in controlling the natural symbiotic flora. In animals, including humans, normal flora of bacteria exists on the skin, in the mouth, in the gastro-intestinal tract and in the airways, as well as in part of the reproductive organs. New bacteria continuously colonize these organs, yet microbial populations are kept in a quasi-steady state. It is likely that the control of the natural flora is due to the continuative action of peptide antibiotics, rather than to the secretion of immunoglobulins. 1.4 Human antimicrobial peptides: defensins and cathelicidins In humans and other mammals, the two main antimicrobial peptide families are defensins and cathelicidins. Although structurally and evolutionarily distinct, these two families are similar for distribution, abundance and function, both classes being implied in antimicrobial activity of phagocytes, inflammatory body fluids and epithelial secretions. 1.4.1 Defensins: structure and tissue distribution Among all the AMPs, defensins are particularly prominent in humans, as evidenced by the large number of expressed genes, the various forms present in human tissues and their occurrence in inflamed or infected tissues (Lehrer, 2004). After their original isolation from mammalian leukocytes (Lehrer et al., 1983; Ganz et al., 1985), defensins were also found to be produced by various epithelial cells (Ouelette and Selsted, 1996; Diamond et al., 1991). Besides mammals, distantly related defensins have been found also in insects and plants (Hoffmann and Hetru , 1992; Brokaert et al., 1995) Defensins are arginine-rich cationic peptides characterized by a β-sheet fold and a framework of six disulfide-linked cysteines. The two main defensin subfamilies are α and βdefensins, differing in the spacing and connectivity of their cysteine residues, which are typically conserved among members of each subfamily. α-Defensins comprise the group of human neutrophil peptides (HNP-1 to 4) and human defensins 5 and 6 (HD-5 and HD-6). βDefensins include the group of human β-defensins (HBD-1 to 4). Their sequences are reported in Fig. 1.2. 7 α-defensins HNP-1 HNP-2 HNP-3 HD-5 HD-6 HNP-4 ACYCRIPACIAGERRYGTCIYQGRLWAFCC CYCRIPACIAGERRYGTCIYQGRLWAFCC DCYCRIPACIAGERRYGTCIYQGRLWAFCC ARATCYCRTGRCATRESLSGVCEISGRLYRLCCR RAFTCHCRR-SCYSTEYSYGTCTVMGN-HRFCCL VCSCRLVFCRRTELRVGNCLIGGVSFTYCCTRV * ** * * * * * ** 30 29 30 34 32 33 β-defensins HBD-1 DHYNCVSSGGQCLYSACPIFTKIQGTCYRGKAKCCK HBD-2 GGIG--DPVTCLKSGAICKPVFCPRRYKQIGTCGLPGTKCCKKP HBD-3 GIINTLQKYYCRVRGGRCAVLSCLPKEEQIGKCSTRGRKCCRRKK HBD-4 RSEFELDRICGIGTARCR-KKCRSQEYRIGRCPN-TYACCLRKPWDESLLNRTK * . * * * * ** 36 42 45 52 Fig. 1.2. Sequence alignment and cysteine bonding of human α- and β-defensins. Conserved residues in each subgroups are indicated by asterisks. Underlined amino acids may or may not be part of mature peptides. Gaps (-) were inserted to maximize the identities. The number of residues is indicated for each peptide. 8 α-Defensins are synthesized as 90-100 amino acids-long pre-pro-peptide precursors, consisting of an N-terminal signal sequence and an anionic propiece followed by the mature peptide at the C-terminus (~ 30 amino acids); in many cases, the charge of the propiece counterbalances that of the mature peptide, and this might be relevant both for proper folding and for preventing intracellular damage to cell membranes. β-Defensins are longer (~ 40 aa), with a stronger cationic nature, and their precursor has a short or absent propiece. As determined by X-ray-crystallography and 2D NMR (Hill et al., 1991), both α and βdefensins consist of a triple stranded β-sheet structure (Fig. 1.3). Fig. 1.3. Structure of HNP-3 and HBD-3. The essential structure is a triple-stranded β-sheet. Cysteine disulfides are in orange, basic and acidic regions are blue and red, respectively. In mammals, the pattern of defensin tissue distribution is species-dependent, and varies considerably even when comparing closely related species. The main sites of α-defensins expression in humans are neutrophils and intestinal Paneth cells. - The primary (azurophil) granules of neutrophils contain high amounts of four distinct αdefensins also referred to as human neutrophil peptides (HNP-1 to 4). The HNPs are stored as fully processed mature peptides (~ 3 kDa). They constitute 30-50% of the total proteins of these organelles, greatly contributing to the oxygen-independent killing of invading pathogens. HNP-1, 2 and 3 are the most abundant, while HNP-4 concentration is about 100-fold lower. - The Paneth cells of the small intestinal crypts express the human α-defensins HD-5 and 6, and store them in specialized secretory granules as pro-peptides. Homologous defensins (cryptidins) are expressed also in the mouse intestine. The presence of AMPs in the 9 gastrointestinal tracts of insects, amphibians and mammals supports the hypothesis that the expression of epithelial defensins serves to limit the proliferation of intraluminal flora. α-Defensins are expressed also in some epithelia, such as that of the female uro-genital tract and of the nasal and bronchial airways; sensitive analytical techniques have demonstrated the expression of HNP-1 and 3 also by monocytes, macrophages, B and NKcells (Agerberth et al., 2000). Human β-defensins (HBD) have been isolated from many cell types, mainly epithelial, confirming that, in addition to many other biological functions, these cells actively participate in host defence, not simply constituting a barrier for this purpose. So far four β-defensins (HBD-1 to 4) have been described, but genomic analyses have recently found additional βdefensin genes, whose corresponding peptide products have yet to be characterized. HBD-1 is mainly expressed in the epidermis and in the epithelia of pancreas, kidney and urinary tract; HBD-2 and 3 are found in the skin and in airway epithelia; HBD-4 is expressed in testis, stomach, lung and neutrophils. HBD-2 is stored as mature peptide in the lamellar bodies of the skin cells. 1.4.2 Defensin genes and regulation of their expression Since polymorphisms in copy number has been observed for both HNP and HBD genes, the number of human defensin genes cannot be exactly defined; the main cluster of α and βdefensin genes maps on chromosome 8p22-23, with other minor clusters, more recently characterized, in other genomic regions (Liu et al., 1997; Schutte et al., 2003). The structural organization of defensin genes and the close proximity of α and β-defensin genes in the main cluster suggest a common evolutionary origin of the two defensin families. From a common ancestral gene, events of genomic duplication end insertions have led to a multiplicity of defensins, differing markedly in amino acid sequence, net charge and quaternary structure, capable to cope successfully with different types of pathogens. Despite defensin genes share a common ancestor, control mechanisms and pattern of their expression differ markedly. As a general rule, α-defensins are expressed constitutively and their synthesis is closely linked to cell differentiation processes, whereas most β-defensins are inducible and their synthesis seems to be triggered by infectious signals. α-Defensins HNPs are synthesized constitutively by the bone marrow precursors, during specific differentiation stages of neutrophil development: transcription of the genes is likely to be massive and brief in the stage of granulocyte precursor, while little or no transcription is observed at the stage of mature neutrophil. The promoter region of HNPs appears well conserved and contains transcription factor binding sites, such as Ets-like elements, C/EPB and c-Myb sites, which are also involved in the transcriptional regulation of many myeloidspecific genes. These sites resulted essential for transcription of HNP genes in the HL60 myeloid cell line (Ma et al., 1998; Tsutsumi-Ishii et al., 2000). Having completed their maturation, and having accumulated in their granules high and definite levels of HNPs, together with several other antimicrobial proteins (including lactoferrin, bactericidal permeability ìncreasing protein, BPI, lysozyme, etc), neutrophils are released into the blood and enter the tissues. Plasma concentration of HNP-1 and 3 was found significantly higher during severe bacterial infections (Ihi et al., 1997): this increased level might reflect an enhanced neutrophil activation at sites of infection. It even raises the possibility that HNP biosynthesis might be stimulated in response to infections, by means of signalling pathways as yet unknown, 10 therefore implying a mode of expression for HNP genes that is not only constitutive. Recent findings support that peripheral leukocytes retain substantial ability to transcribe HNP1-3 genes, although the expression level was not affected by ex vivo treatment with bacterial components such as LPS (Fang et al., 2003). Paneth cell α-defensin genes, similarly to the hematopoietic defensins, have a constitutive level of transcription that is part of a developmental program of the highly differentiated cells in which they are expressed. Nonetheless, the presence of several nuclear factor interleukin-6 (NF-IL6) recognition sites, in the promoter of both HD-5 and 6, does not rule out that these genes might be up-regulated in response to inflammatory stimuli (Mallow et al., 1996). Once accumulated in specialized granules, the enteric defensins are secreted into the lumen in response to bacteria by a process involving receptors and transduction pathways that have not been characterized yet. β-Defensins HBD genes are characterized by both constitutive and inducible expression. HBD-1 gene promoter contains NF-IL6 and γ-interferon consensus sites; HBD-2 gene has several nuclear factor-κB (NF-κB) and NF-IL6 consensus sites. The presence of these regulatory cis-elements is consistent with the inducibility of these genes, either directly by the infection or by the production of inflammatory cytokines. HBD-1 seems to be constitutively synthesized by many epithelial cells, whereas HBD-2 and 3 are expressed when epithelial tissues are stimulated with bacteria. HBD-2 gene transcription in the skin is induced by bacterial contact through the intermediate synthesis of interleukin-1 (IL-1) by myeloid cells (Liu et al., 2003). Transcriptional activation of HBD-2 gene in airway epithelia results upon treating with bacterial molecules such as lipoteichoic acids (LTA) and is mediated by an epithelial expressed toll like receptor (TLR) that recognizes components of the Gram-positive bacteria, thereby activating the NF-κB pathway (Wang et al., 2003). NF-κB binding sites in the promoter region of HBD-2 gene, confer responsiveness to both LTA and proinflammatory cytokines such as IL-1. Transcription of HBD-1 and 2 is also strongly induced in LPS-treated peripheral leukocytes (Fang et al., 2003). HBD-3 and 4 genes are regulated by NF-κB–independent mechanisms, that remain to be characterized. In bovine tracheal epithelial cells, LPS exposure or bacterial contact induce transcription of a β-defensin, the tracheal antimicrobial peptide (TAP), and transcription is accompanied by NF-κB activation and binding to the TAP promoter sequence. Involvement of the well known transcription factors of the Rel/NF-κB family seems quite a conserved theme in the activation of genes which are infection- and inflammation-induced, and not only in mammals: these proteins, in facts, have been implied in the activation of several inducible AMP genes, in quite a broad range of phylogenetically distant organisms, including insects, amphibians and mammals (Boman, 1998). 1.4.3 Activity of human defensins Most defensins have antimicrobial activity against Gram-positive, Gram-negative bacteria and fungi especially when tested under low concentration of salts and plasma proteins. In these conditions, the activity is in the low µM range (1-10 µg/mL). Antiviral properties have been ascribed to HNPs: in vitro incubation with these defensins inactivate several types of enveloped viruses such as cytomegalovirus, herpes simplex (HSV) 1 and 2, vesicular stomatitis and influenza A virus (Daher et al., 1986). HNPs inhibit also human immunodeficiency virus 1 (HIV-1) replication (Nakashima et al., 1993). It has been recently suggested that HNPs could be major components of the CD8+ 11 T cell antiviral factor (CAF) and contribute to its anti-HIV activity (Zhang et al., 2002). CAF is thought to play an important role in HIV-non-progressor patients, that is HIV-infected individuals, whose infection remains stable for a long time without developing the distinctive symptoms of AIDS. The involvement of α-defensins in CAF activity has been argued and finally ruled out, since no mRNAs for HNPs could be detected in highly purified CD8+ T cells. Yet the anti-HIV property of HNPs remains undisputed and represents an intriguing subject to understand how the immune system can prevent HIV infection. Interestingly, members of the most recently identified defensin subfamily, the θ-defensins, were found to possess a potent anti-HIV activity. These cyclic octadecapeptides have been isolated from leukocytes of the rhesus monkey and seem to arise from post-translational splicing of two α-defensin-like nonapeptides (Tang et al., 1999). The θ-defensins apparently evolved in all the primates, but were subsequently lost in humans, due to mutations in the corresponding and still functional genes. Artificially created human θ-defensins (named “retrocyclins”) exert substantial anti-HIV activity (Cole and Lehrer, 2002). Phylogenetic evidence indicates that loss of retrocyclin production might date back to the hominid stage: this evolutionary loss could account for the HIV-1 susceptibility in modern humans. Many diverse biological functions, other than a direct antimicrobial effect, have been ascribed to mammalian and human defensins; most of them somehow contribute to host defence system. These activities can be summarized as follows. - - Both α- and β-defensins chemoattract several types of immune cells (monocytes, dendritic cells, lymphocytes and cytokine-activated neutrophils), influencing and enhancing antimicrobial immunity (Yang et al., 1999; 2000). Some defensin stimulate and/or modify cytokines production (Yang et al., 2002). HNPs interact with complement (van den Berg et al., 1998). Some defensins, such as rabbit NP-3A, act as corticostatin by binding the ACHT receptor (Zhu et al., 1989) HNPs participate in wound repair, by enhancing extracellular matrix deposition, and promote cell growth (Murphy et al., 1993; Oono et al., 2002). HNPs can have cytotoxic effects and contribute to lysis of tumour cells (Lichtestein et al., 1986). Many α- and θ-defensins are capable of binding membrane glycoproteins: this lectin-like behaviour could be important for their antiviral properties as well as for their antibacterial activity, since polysaccharides are integral components of fungal and bacterial cell walls (Wang et al., 2003). 1.4.4 Cathelicidins Cathelicidins constitute a unique mammalian gene family. They are structurally organized as an N-terminal signal peptide, a highly conserved prosequence - the cathelin domain, after which the family takes its name - and a variable cationic peptide at the C-terminus (Zanetti et al., 1995; Bals and Wilson, 2003) The conservation of the cathelin domain is striking between species and indicates that the diverse members of this family evolved from a common ancestor gene. Unlike most mammals, in humans the cathelicidin family is restricted to a single gene product, the LL37 peptide (elsewhere indicated as hCAP18). Both halves of the human cathelicidin are active: the cathelin domain is antimicrobial and acts as a protease inhibitor; LL37 peptide, characterized by an α-helical conformation, is active against Gram-positive and Gram-negative bacteria. Like defensins, also LL37 is a multifunctional AMP. It participates in host defence not only by direct antimicrobial activity, but also by: i) recruiting cellular defence (LL37 is 12 chemotactic for monocytes, neutrophils and T cells) and ii) promoting wound healing and tissue repair (LL37 stimulates angiogenesis). LL37 was originally identified in specific granules of neutrophils, where it is stored as propeptide, being processed and released upon appropriate stimulation; subsequently, it has been found in various epithelial tissues (skin, gastrointestinal tract, airways, etc.), in different body fluids, as well as in macrophages, B cells, NK cells, and γδ T cells. The promoter region of the gene encoding LL37 contains several putative binding sites for many transcription factors, including NF-κB and NF-IL6, acute-phase response factor and C/EPB. All these sites suggest that transcription of LL37 gene may be regulated by infection and inflammatory cytokines. LL37 is induced in inflamed skin and its serum concentration rises during infection, whereas, in intestinal cells, its synthesis seems rather differentiationdependent. To date, little is known about constitutive or regulated expression of LL37 gene. 1.4.5 Roles of defensins and cathelicidins in the human immune system The role of defensins and cathelicidins in innate immune system of mammals and humans is being established by recent experiments on transgenic mice, but additional studies in these and related models would be desirable to document the function of these AMPs in in vivo contexts (Nizet et al.,2001; Morrison et al., 2002; Salzman et al., 2003). Nonetheless, there is already an expanding body of experimental researches pointing to the relevance of defensins and cathelicidins in human diseases: several clinical studies have begun to associate alterations in AMP synthesis or function with human infectious diseases, inflammatory syndromes, or immune deficiencies. AMP production increases in response to specific infections and acute damage to the epithelial barrier (e.g. psoriasis, Helicobacter pylori gastritis, etc.) in agreement with the expected protective role of these molecules. By contrast, increased and dysregulated AMP synthesis is also associated with some chronic inflammatory disorder (e.g. lung fibrosis) reflecting the dual function of AMPs in the immune system (Gallo and Nizet, 2003). 1.5 NFAT NFAT (nuclear factor of activated T cells) was originally described as a putative transcription factor present in the nuclear extracts of activated Jurkat T cells that binds to human interleukin-2 (IL-2) promoter (Shaw et al., 1988). The NFAT family contains at least four major isoforms, named NFATC1-4, each of which exhibits a characteristic pattern of cell type specificity. Transcription factors of the NFAT family regulate the expression of several immunerelated genes, such as those encoding cytokines and their receptors, in response to antigenic stimulation of cells of the immune system (Crabtree and Clipstone, 1994; Rao et al.,1997). NFAT proteins are also involved in heart valve development, angiogenesis of peripheral vessels, myogenesis and signalling mechanisms in the hippocampus (Horsley and Pavlath, 2002). NFAT consists of two components: a pre-existing cytosolic component (NFATc), whose activation is regulated by Ca2+ concentration and blocked by cyclosporin A (CsA), and a nuclear component, NFATn, whose induction requires de novo protein synthesis upon T cell stimulation. Both components are required for binding to the NFAT site, a purine-rich core motif, (A/T)GGAAAA, with surrounding bases varying according to the specific nuclear partner of NFATc. NFATn is expressed under the control of the p21ras pathway (Woodrow et al., 1993); members of the AP-1 (activator protein-1) family (such as Fra-1, Jun B, and c-Fos) have been 13 demonstrated to be able to functionally substitute for NFATn (Northrop et al., 1993). Recent evidence indicates that several other proteins, such as the zinc finger proteins GATA4 and cMAF, can also cooperate with NFATc. NFATc proteins share some cardinal functional and structural features: - their cytoplasmic location; - their induction via a peculiar calcium/calcineurin-activated pathway; - their inhibition by clinically important immunosuppressive drugs CsA and FK506 (Mattila, et al. 1990); - the presence of a conserved regulatory SP repeated motif and serine rich domain; - the conserved Rel homology DNA-binding domain; - a conserved calcineurin binding site. The schematic structure of NFATc family members is depicted in Fig.1.4. Rel-homology DNA-binding domain NFAT homology region SRR Cn NLS Cn NLS SP-repeats motifs Fig.1.4. Structural features of a typical NFATc protein. Cn, calcineurin binding site; SRR, serine-rich region; NLS, nuclear localization signal. The signalling pathway leading to NFAT activation has been mainly characterized in T cells and is illustrated schematically in Fig. 1.5. A key step in NFAT activation is its nuclear import: in resting T cells, NFAT is phosphorylated and resides in the cytoplasm; upon T cell stimulation, which elicits a sustained rise of intracellular Ca2+ concentration, NFAT is dephosphorylated by the Ca2+-activated Ser/Thr phosphatase calcineurin, which leads to its nuclear import, likely as a result of the exposure of the nuclear localization signal (NLS). Once accumulated in the nucleus, dephosphorylated NFATc recruits the nuclear partner, NFATn, to form a complex on the ARRE-2 (antigen receptor response element) site of the IL2 promoter (Timmerman et al., 1996). Thus, the Ca2+-dependent and p21ras-dependent pathways, that emanate in parallel from the T cell receptor upon stimulation, are reintegrated through NFAT-mediated assembly of a transcriptional complex on lymphokine gene promoters to initiate the immune response. The activation of NFAT can be reversed by several constitutively active kinases which have been implicated in the rephosphorylation of NFAT and its nuclear export (Beals et al., 1997). In addition, phosphorylation of NFAT in vitro inhibits its ability to bind DNA. Surprisingly, in the animal kingdom, relatively few biochemical pathways appear to transmit all signals from receptors into the nucleus and virtually all of these pathways are present in both vertebrates and invertebrates. Conversely, NFAT signalling seems to have arisen in vertebrates only, since, so far, no invertebrate homolog of NFATc has ever been found. This observation supports the suggestive hypothesis that NFATc arose just to suit vertebrates’ specialized and distinctive needs, such as certain aspects of the nervous system and a complex adaptive immune response. NFATc genes might have derived from a process of exon shuffling between a Rel domain and a precursor gene with a calcineurin-sensitive Ca2+ region. This process could have provided a new link between Ca 2+ signals and the nucleus, useful for the control of genes primarily dedicated to cellular interactions. 14 Fig.1.5. Schematic illustration of intracellular signalling pathways resulting in activation of IL2 transcription. Antigen recognition by TCR initiates two pathways: one, p21rasdependent, stimulates de novo synthesis of NFATn; the other promotes an increase of intracellular Ca 2+ levels. This activates calcineurin that dephosphorylates NFATc allowing it to enter the nucleus, where it cooperates with NFATn to bind specific sites on the promoter of various cytokine genes, thus activating transcription of the corresponding genes. The process is reversed by kinases that re-phosphorylate NFAT, inducing its nuclear export. 15 1.6 Hepatitis C virus Hepatitis C virus (HCV) is the major aetiological agent of blood-borne non-A, non-B hepatitis and a leading cause of liver cirrhosis and hepatocellular carcinoma (Kuo et al. 1989; Purcell, 1994; Houghton et al., 1991). HCV represents one of the most significant health problems since it affects an estimated 170 million people worldwide. The prevalence of HCV infections varies in different parts of the world: for example, in Scandinavia, it is less than 0.5% of the population; whereas, in Egypt, over 20% of the population is infected. In the USA and Western Europe, the complications of HCV chronic hepatitis and cirrhosis are the most common reasons for liver transplantation. One of the major problems with HCV infections is that up to 85% of individuals initially infected become chronically infected, usually for decades. The other 15-20% have an acute infection that is resolved spontaneously in a few weeks or months. The propensity of HCV to cause chronic, persistent infections is explained by its extraordinary ability to escape destruction by the host immune system. Once established, chronic HCV infection causes an inflammation of the liver called chronic hepatitis; this condition can further progress to scarring of the liver, called fibrosis, or more advanced scarring, called cirrhosis. Some patients with cirrhosis can develop liver failure and complications such as hepatocellular carcinoma (HCC). HCV is spread most efficiently trough the blood, thereby it is transmitted by infected blood or blood products, transplantation of infected solid organs and the sharing of contaminated needles among intravenous drug users. In retrospect, HCV was the most common cause of transfusion-associated hepatitis in the 1980s: at that time, HCV had not been identified yet, and post-transfusion cases of hepatitis were called non-A non-B hepatitis. In the mid 1980s, when the practice of using paid blood donors was stopped and blood started to be screened for HIV, the risk of HCV transmission fell to about 5%. Isolation and identification of the virus, and subsequent development of specific serologic tests, have dramatically lowered the risk of acquiring HCV through blood transfusion. At present, the most common cause of HCV transmission is by intravenous drug abuse (Fig. 1.6). Fig 1.6. Risk factors for infection with hepatitis C virus 16 Despite the striking reduction in the number of new cases of hepatitis, the number of deaths and the need for liver transplantation, due to complications of chronic infections, are expected to increase within the next two decades: this is because of the large number of individuals who became infected 10-20 years ago. 1.6.1 HCV structure The identification of HCV is relatively recent (Choo et al, 1989). HCV, unrelated to the other common hepatitis viruses (A, B, D and E), is an enveloped, single-stranded positivesense RNA virus belonging to the Hepacivirus genus of the Flaviviridae family (Fig. 1.7). Its 9,6-kb genome consists of a single RNA molecule with conserved terminal 5’ and 3’ noncoding elements, flanking an open reading frame encoding a single large polyprotein, which is cleaved by host and viral proteases into three structural (core, E1, E2) and at least six nonstructural (NS) proteins (Grakoui et al., 1993) (see Fig. 1.8). Structural proteins contribute to build up the envelop, while non structural proteins are involved in RNA replication. HCV infects mainly hepatocytes; its genome does not integrate into that of the infected cell, for it is directly translated and replicated by the host machinery. Based on sequence diversity, HCV has been categorized into 6 major genotypes and many more subtypes. Different genotypes show a distinct geographic distribution. The influence of genotype on the long-term prognosis of HCV is still unclear, however, it seems that patients infected with genotype 1 (particularly genotype 1b) are more likely to develop chronic infection and do not respond well to pharmacological therapy. core Fig. 1.7. Morphology of hepatitis C virus. 1.6.2 HCV diagnosis and therapy Viral hepatitis may develop without clinical signs, or nonspecific symptoms may appear for a short time, with or without jaundice. These symptoms may vary from non-specific flulike symptoms to liver failure. Diagnosis of hepatitis often depends on accumulation of findings considered together. HCV infection can be diagnosed and monitored by evaluating biochemical and histological parameters. Abnormally high serum levels of hepatic enzymes (ALT and AST) can be symptomatic of liver injury and are detected by routine blood analysis. Immunoblot and ELISA tests are specifically used to detect anti-HCV antibodies in the blood and indicate whether an individual has been exposed to the virus. Detection of viral RNA, through sensitive molecular tests (such as RT-PCR), can confirm that a positive anti-HCV result 17 reflects an active infection and, although viral load does not correlate with the severity of disease, may be helpful to monitor patient response to therapy. Liver biopsy assesses the extent of fibrosis and the level of inflammation; though invasive, this type of analysis provides important information about the severity, and thus the prognosis, of the disease. The pharmacological antiviral therapy to treat hepatitis C is currently based on pegylated IFN-α (PEG INF-α), alone or in combination with ribavirin (Fried et al., 2002). These drugs enhance immune reaction to viral infection: PEG IFN-α, like other naturally occurring interferons, has antiviral, antiproliferative and immunomodulating functions; ribavirin, a nucleoside analogue, interferes with the RNA metabolism of the viral genome. Three types of responses to the antiviral therapy have been described: - sustained virological response is the optimal response: viral RNA becomes undetectable 6 months after therapy is stopped; most of sustained responders will remain in remission (no signs of disease) indefinitely; - relapse: relapser patients initially respond, but virus becomes again detectable within 3-6 months after stopping therapy; - non-response: virus RNA remains or becomes detectable during therapy. No vaccine is currently available for preventing HCV infection. 1.6.3 HCV pathogenesis A deep knowledge of HCV pathogenesis is not yet clear. Since there is no definite experimental evidence for a direct cytopatic effect of HCV, the liver damage in chronic infection is probably caused by the interplay between the virus and the host immune reaction. Since HCV infects only humans and chimpanzees, and replicates inefficiently in cell cultures, studies aimed at assessing abilities of the virus to modulate host response have been difficult to perform. Cell-mediated specific response seems pivotal both for a successful viral clearance and in mediating hepatic injury. Individuals who experience complete virological recovery, show vigorous, multispecific CD4+ T helper 1 responses, while poorer CD4+ T helper 1 responses are typical of chronic hepatitis (Cerny and Chisari, 1999). Induction of virus-specific CTLs (cytotoxic T lymphocytes) is a well established mechanism for virus elimination during infection; however, in the case of HCV, the lymphocytes may not be sufficient to eliminate the virus completely. Conversely, cytotoxic response may cause extensive necrosis of both infected cells and adjacent healthy cells, contributing to tissue damage. The role of the humoral response is still controversial, but mostly considered of minor relevance; indeed, it is documented that chronicization is associated to an unbalanced cytokine production which enhances the humoral rather than the cell-mediated response (Cooper et al., 1999). The reasons why the host immune system often fails to eradicate the virus are not known. HCV has probably evolved mechanisms to elude the host response. One proposed mechanism of immune evasion is the generation, during the infection, of viral variants that could circumvent antibody and CTL recognition. This is consistent with the high error rate of the viral polymerase, which generates multiple virus quasi-species within the same host (Mc Michael and Phillips, 1997). Alternatively, as noted for several other viruses, HCV may encode products that act to inhibit viral clearance by the host and can lead to progressive, persistent infection. Besides hepatocytes, HCV infects lymphocytes and dendritic cells: by affecting these immune cells the virus might compromise host immune functions (Pavio, 2003). The viral product that is more likely to participate in this immune-interference is the core protein. 18 1.6.4 HCV core protein HCV core protein (HCV C) derives from the N-terminal 191 amino acids of the large precursor polyprotein and is highly conserved among all isolates, with a sequence identity in the range of 85-100% (Fig. 1.8). HCV C is highly basic, consistently with its RNA-binding activity; it has two potential nuclear localization signals and a conserved putative DNAbinding motif (DBD), which support the hypothesis that HCV C also functions as a gene regulatory protein; a predominantly hydrophobic domain is present in the C-terminal region and is probably involved in protein-protein interactions during assembly of the viral nucleocapsid (Bukh et al., 1994). HCV C has a packaging function, as it is the major component of the viral nucleocapsid (Fig 1.7); however, emerging evidence suggests that this protein is multifunctional and several of its biological properties might have implications in HCV distinctive pathogenesis. HCV C localizes both in the cytoplasm, where it has been found to associate with the endoplasmic reticulum, lymphotoxin-β receptor and TNF receptor 1, and in the nucleus, where, especially its C-terminal-truncated forms, seems to participate in transcriptional regulation. 3’ C E1 E2 p7 5’ NS2 NS3 NS4A/B NS5A/B MSTnPKPQRkTkRNTnrRPqDvKFPGGGQIVGGVYILPRRGPR IGVRatRKtSERQPRGRRQPIPkaRrpeGrsWaqPGyPWPIYg nEGcgWAGWLLSPrGSrPsWGptDPRrrSRNlGkVIDTlTCgf ADLMGYiPlVGaPlGGvArALAHGVRvlEDGvNyATGNlPGCs FSIFlLALlSCltvPasa Fig.1.8. Diagram of the HCV genome and consensus amino acid sequence of the core protein. The lines indicate the 5’ and 3’ non-coding regions, while the boxes stand for the reading frame. Regions encoding structural proteins are coloured. NS, non structural proteins. In the sequence, capital letters indicate conserved amino acids. Three basic regions, probably involved in RNA interaction, are marked in yellow; two putative nuclear localization signals are underlined; a potential DNA-binding domain is dotunderlined. HCV C has been implicated in cellular proliferation and there are several experimental studies ascribing to this protein an oncogenic potential (Ray et al., 1996; Tsuchinhara et al., 1999); the tumorigenic properties of HCV C has been associated to its capability to bind and inactivate the endogenous LZIP factor, which functions as a cellular tumour suppressor (Jin et al., 2000). The oncogenic nature of HCV C is noteworthy, since it could directly contribute to the onset of HCC. Another relevant biological function of HCV C is its well documented, anti-apoptotic action: this effect might be advantageous for the virus, since it would allow the infected hepatocytes to avoid the apoptosis, induced by the host immune system as a protective mechanism, thereby allowing persisting and spreading infection (Marusawa et al., 1999). HCV C has the potential to affect host immune gene expression through the activation of the NFAT pathway: in fact, a recombinant core protein was found to activate NFAT-mediated transcription from IL2-promoter in Jurkat cells (Bergqvist and Rice, 2001). The potential of 19 this effect in vivo is even more intriguing when considering that several other cytokines and many immune-related genes are NFAT-regulated. Dysregulation of cytokine expression could be part of the strategy used by the virus to interfere with the host immune system, thereby hampering an effective defence response. Studies on animal models (mice infected with a panel of recombinant vaccinia, expressing the various HCV proteins) confirm that HCV core is sufficient to induce immune suppression (Large et al., 1999). In addition to cytoplasmic and nuclear localization, HCV C is secreted from stably transfected cell lines, and likely from HCV-infected hepatocytes in vivo; indeed, circulating core protein is detectable in the blood stream of infected patients, where it may provide the virus with an indirect mechanism of immune dysregulation, influencing even host cells not directly infected (Kanto et al., 1995; Sabile et al., 1999). Consistently with this immunomodulatory action, exogenous addition of HCV C was shown to affect the expression of various cytokines implicated in CTLs activation, such as IL12 and INF-γ (Eisen-Vandervelde et al., 2004). 20 2 AIMS OF THE RESEARCH The aim of this research was to investigate the immuno-modulatory effect of HCV, studying the expression of some immune genes in peripheral blood mononuclear cells (PBMC) from HCV-infected patients. In this regard, we focussed our attention particularly on the expression of HNPs. In fact, taking into account that: i) the expression of a recombinant HCV core protein was found to induce the NFAT pathway (Bergqvist and Rice, 2001), and ii) a putative NFAT core binding site is present in the promoter of HNP-1 and 3 genes (Fig 2.1), we intended to find out whether, during the viral infection, the expression of these antimicrobial peptides could be affected. The study was carried out analyzing the profile of expression in cells directly extracted from healthy and diseased individuals; another part of the study concerned with a set of in vitro experiments to assess the potential effect of HCV core protein. The choice to use PBMC rather than hepatic tissue, that is actually the main target of HCV, is based on the following motivations: i) PBMC comprise relevant elements of the innate and adaptive immune systems; ii) they are part of a “moving tissue”, the blood, which can be informative about the status of the whole organism, as it reaches every district within the body; iii) PBMC can be easily collected from a blood sample, without any need for invasive and/or risky biopsy. ets -90 -80 -70 -60 -50 -40 TTTAATGGACCCAACAGAAAGTAACCCCGGAAATTAGGACACCTCATCCCA HNP promoter NFAT Fig. 2.1. Nucleotide sequence of the HNP promoter in the region -90/-40. The shaded sequence indicates the Ets binding site, while the putative NFAT recognition site is underlined. 21 3 MATERIALS AND METHODS 3.1 MATERIALS 3.1.1 Patients. Study population: epidemiologic data, biochemical and virological measurements We investigated a total of consecutive 117 patients with chronic HCV infection (64 men, 53 women, mean age 47.6+11.9, range 30-72 years). Patients had serum antibodies to HCV, detected by enzyme-linked immunosorbent assay and confirmed by recombinant immunoblot assay. All patients had high aminotransferase (: aspartate and alanine aminotransferases, AST and ALT) levels for at least 6 months. The epidemiologic characteristics, and the biochemical, virological and histological data of these patients are shown in Table 3.1. None of the patients had received or was receiving antiviral therapy at the time of the liver biopsy or at the time of blood tests, with the exception of those included in the specific study (see results). Exclusion criteria were coinfection with HIV, hepatitis B virus, other causes of liver disease diagnosed by standard clinical, serological and biochemical criteria. Patients with alcohol intake >80 g/day in men and >60 g/day in women or chronic drug intake, dyslipidemia, hyperglycemia and severe steatosis on liver biopsy were also excluded. As control group, we studied 22 sex- and age-matched normal healthy volunteers. Patients with acute C, A and B hepatitis, chronic and cirrhotic B hepatitis, hepatocellular carcinoma (HCV-), HIV infection, and various types of bacterial infections were also included in this study. The study was approved by the institutional review board of the Department of Infectious Diseases, Università La Sapienza, Roma. All patients and controls gave their written informed consent before inclusion. Evaluation of α-defensin levels and determination of histological score were done in a blind manner. 22 No. % 18 21 78 66 ± 48 15 18 67 HCV transmission route Intravenous drug use Transfusion Other AST level (mean ± SD) ALT level (mean ± SD) HCV-RNA level (mean ± SD) 95 ± 67 2567000 ± 4159000 HCV genotype Genotype 1 Genotype non-1 68 49 58 42 15 102 13 87 56 46 55 45 10 36 21 16 19 9.8 35.2 20.7 15.6 18.7 HISTOLOGY Hepatocellular carcinoma Chronic hepatitis HAI 1-6 >7 Fibrosis 0 1 2 3 4 Table 3.1. Epidemiologic characteristics and biochemical, virological, and histological data of 117 patients with HCV chronic infection. HAI, histological activity index. ALT, alanine aminotransferase. AST, aspartate aminotransferase. SD, standard deviation. Serum HCV-RNA detection, quantification and genotyping Serum HCV-RNA was detected by reverse transcriptase polymerase chain reaction (Amplicor Roche) and quantified by branched-DNA assay (b-DNA assay, Quantiplex TM, Chiron Co., Emmeryville, CA, USA). The detection limit of the test is 600 IU/ml. HCV genotype was done using the reverse hybridization line probe assay (INNO-LIPA TM HCV assay, Innogenetics NV, Ghent, Belgium), according to the manufacturer’s instructions. Histological evaluation The histological activity index (HAI) was quantified according to Knodell score. The index represents the sum of periportal + bridging necrosis (0-10), intralobular degeneration (0-4) and portal inflammation (0-4). Fibrosis is classified as follows: 0 = none; 1 = enlarged, fibrotic portal tracts; 2 = periportal or portal-portal septa but intact architecture; 3 = fibrosis with architectural distortion but no obvious cirrhosis; 4 = cirrhosis 23 3.1.2 Microorganisms Bacillus megaterium Bm11, Gram-positive, streptomycin resistant, was used in the antibacterial activity assays. 3.1.3 Cells CD4+ and CD8+ T cells, from peripheral blood of healthy donors, were a gift from IRCCS, Santa Lucia, Rome. Cells were obtained by magnetic cell sorting, with a level of purity >95%, as evaluated by flow cytometry. Peripheral blood mononuclear cells (PBMC) and granulocytes were purified by density gradient purification (see methods). 3.1.4 Growth media For PBMC, the medium used was RPMI 1640, supplemented with freshly added 10 % fetal bovine serum (all from Sigma). RPMI is stored at 4 °C; fetal bovine serum aliquots are stored at -80 °C. For bacteria Luria Bertani (LB) broth was prepared: Bacto-tryptone 10g/l Yeast extract. 5g/l NaCl 5g/l Solid LB is prepared by adding 15 g/l agar Soft agarose LB is prepared by adding 10 g/l agarose. Sterilize the media by autoclaving at 120 °C for 120 min. 3.1.5 Drugs and solutions Cyclosporin A (CsA) stock solution is prepared by dissolving the powder in ethanol at a concentration of 1mg/ml. Store aliquots at -20 °C. Streptomycin stock solution is prepared by dissolving lyophilized antibiotic salt in distilled H2O (dH2O) at a concentration of 100 mg/ml. Sterilize by filtration through a 0.22 µm filter and store aliquots at -20 °C. Ionomycin stock solution is prepared by dissolving the corresponding calcium salt in dimethyl sulfoxide, DMSO, at a concentration of 1mg/ml. Store aliquots at -20 °C, shielded from light. Phorbol 12-myriststate 13-acetate, PMA, is dissolved in DMSO at a concentration of 0.1 mg/ml. Aliquots stored at -20 °C, protected from light. Phytohemagglutinin, PHA, is dissolved in sterile PBS at a concentration of 0.5 mg/ml. Aliquots stored at -20 °C, and at 4 °C after first thawing. 24 Matrix for mass spectrometry: 3,5-dymethoxy-4-hydroxy-cinnamic acid (sinapinic acid) is dissolved in 0.1% trifluoroacetic acid, acetonitrile:water (1:1, v/v) solution, at a concentration of 30 g/l. Store at -20 °C. DEPC-treated H2O: RNAse free distilled water is prepared by adding 1µl/ml of diethylpyrocarbonate, DEPC. Shake bottle and let stand overnight under chemical hood before autoclaving 120 min at 120 °C. Phosphate-buffered saline (PBS) 137 mM NaCl 2,7 mM KCl 10 mM Na2HPO4 2 mM KH2PO4 Adjust pH to 7.4 with HCl. Sterilize by filtration. Store at room temperature Ethidium bromide stock solution (10mg/mL) is stored at 4 °C, protected from light. TE 20 mM 1mM pH 7.5 TAE 20X 800 mM Tris base 100 mM CH3COONa 20mM EDTA Adjust pH to 7.4 with CH3COOH Agarose gel loading buffer (6x) 0.25 % 0.25 % 30 % Tris-HCl EDTA Bromophenol blue Xylen cianol Glycerol Agarose gel for RNA and DNA electrophoresis 1% Agarose gel has been used routinely for RNA electrophoresis, while 1.5-2% agarose gels have been used for analysis of PCR products. The gel is prepared as follows: - Agarose 1-2 g - TAE 20x 5ml - dH2O to 100 ml - Melt in microwaves, let it cool and add 5µl ethidium bromide stock solution. - Pour the solution in the gel-caster, let it harden. Use TAE 1x as running buffer. Substitute dH2O with DEPC H2O for RNA electrophoresis. dNTPs 10 mM dATP, dCTP, dGTP,dTTP (100mM) 10 µl each and add dH2O to 100 µl. Sybr Green I (Diatech) is stocked as 1000-fold dilution in TE pH 7,6; aliquots are stored at 20°C; after thawing, they are kept at 4 °C, shielded from light. TaqMan probes and LUX primers are stored at -20 °C, protected from light, as mono or double-use aliquots. 25 HCV core protein, HCV C, was a kind gift from Penelope Mavromara and Urania Georgopoulou (Hellenic Pasteur Institute, Athens). The recombinant protein, genotype 1, has an N-terminal His tag and was purified, under native conditions, as a mixture of truncated products, with the highest molecular weight corresponding to the first 120 amino acids of the native HCV C (191 aa total length). Stock solution in PBS, at a concentration of 300ng/ml, is stored at -20 °C. Hepatitis B virus core antigen, HBcAg (from IBT, Germany). Stock solution, with a concentration of 0.75 mg/ml in 100mM NaCO3 pH 9.3, is stored at -20 °C. 3.1.6 Oligonucleotides for RT-PCR An oligo-(dT)18 has been used to prime first strand cDNA synthesis. Stock solution, 500 µg/ml, is stored at -20 °C. Sequences of the primers used for real time RT-PCR are given below. HNP forward : 5’-GCAGAATACCAGCGTGCATTGCAGGAG-3’ and HNP reverse: 5’-CAGCAGAATGCCCAGAGTCTTCCC-3;’ GAPDH for: 5’-TGGGCTACACTGAGCACCAG-3’ and GAPDH rev: 5’-CAGCGTCAAAGGTGGAGGAG-3; GAPDH-certified LUX primer set, JOE-labelled(Invitrogen). TBP for: 5’-GCACAGGAGCCAAGAGTGAAG-3’ TBP rev: 5’-TCACAGCTCCCCACCATGTTC-3’ LL37 for: 5’-CATCATTGCCCAGGTCCTCA-3’ LL37 rev FAM-labelled: 5’-caaccTCCGAGGACCGCTGGTTG-3’ IL-15 for: 5’-TGCAAAGAATGTGAGGAACTGG-3’ IL-15 rev FAM-labelled: 5’-cactcgCACATTTGAAATGAAATGCCGAGTG-3’ IL-2 for FAM-labelled: 5’-gacttagCCTGTCTTGCATTGCACTAAGTC-3’ IL-2 rev: 5’-TGAGCATCCTGGTGAGTTTGG-3’ The above LUX primer pairs, for LL37, IL-2 and IL-15, were designed using LUX-primer design programme, available at www.invitrogen.com; bold and underlined case indicates the fluorophore-marked nucleotide in the labelled primer; lower case letters indicate the 5’ nonannealing regions involved in the intramolecular hairpin structure. Primers and FAM-labelled TaqMan probes, produced by Diatech laboratories (Jesi), for amplification and detection of HNP and GAPDH transcripts, have also been used (sequences available on request). In part of the study, Diatech primers have been used also in combination with Sybr Green I dye, without TaqMan probe. 26 3.2 METHODS 3.2.1 PBMC preparation Peripheral blood mononuclear cells, PBMC, were isolated from EDTA-anticoagulated peripheral blood (~20ml) by Ficoll density gradient centrifugation according to the procedure given below. - Dilute freshly drawn blood with 2-4 volumes of PBS. - Carefully layer 35ml of the diluted blood over 15 ml Ficoll in a 50ml conical tube and centrifuge 30 min at 400g at 20 °C. - Transfer the interphase cells (lymphocytes and monocytes) to a new conical tube, fill it with PBS and centrifuge at 300g for 10 min at 20°C. Remove supernatant carefully. -Wash cells from platelets: resuspend pellet in 20 ml PBS. From this suspension a small aliquot is used for counting in a hemocytometer; the remaining is divided into two aliquots and centrifuged at 300g for 10 min at 20°C. Discard super. - Freeze one aliquot for subsequent peptide extraction; resuspend cells of the second aliquot in appropriate lysis buffer (TRIzol), for subsequent RNA extraction. Alternatively, PBMC are directly used for in vitro experiments. - Store samples at -80 °C. 3.2.2 Granulocytes preparation Use freshly drawn EDTA-anticoagulated blood. - Prepare the density gradient by layering 10 ml histopaque 1119 and 10 ml histopaque 1077 (Sigma) in a conical tube. - Layer the blood (15-20 ml) over the solution and centrifuge at 400 g for 30 min at 20°C. - Collect the white cell layer just above the red blood cells (that is the lowest cell ring), transfer to a new tube and add PBS to 30 ml; centrifuge 10 min at 300g for 10 min at 20 °C. Repeat this wash. - Resuspend in 5 ml PBS, use an aliquot for hemocytometer reading and centrifuge the remaining to recover the granulocytes as before. - Resuspend the cells in TRIzol, for subsequent RNA extraction, and store at -80 °C. 3.2.3 In vitro treatment of PBMC Freshly isolated PBMC were resuspended in RPMI 1640, supplemented with 10 % fetal bovine serum and 80µg/ml streptomycin. PBMC final concentration was in the range of 1-1.5 x106 cell/ ml. Cells were cultured in 18-well round-bottomed polystyrene plates, at 37°C in a humidified atmosphere with 5% CO2 in air, in the absence (unstimulated controls) or in the presence of: - 2 µg/ml of recombinant HCV C, or - 2,5 µg/ml of HBcAg. To analyse whether α-defensins expression is mediated by the transcription factor NFAT, part of the PBMC were pre-incubated with: 1 µg/ml cyclosporin A, CsA, at 37°C for 30 min, before HCV C addition. 27 PBMC have been incubated also with: - 0.05 µg/ml phorbol 12-myristate 13 acetate, PMA; - 1.5 µg/ml ionomycin; - 3 µg/ml phytohemagglutinin, PHA. These drugs were used alone or variously combined, with or without 30 min preincubation with CsA; control non-stimulated cells were incubated in RPMI alone, contemporarily. After 3 hours and 30 min incubation, PBMC were recovered by mild centrifugation and used for subsequent peptide and RNA extraction procedure. The HNP mRNA and peptide levels, detected in the differently stimulated cells, were normalized to those observed in the unstimulated control cells. 3.2.4 Peptide extraction Low molecular weight proteins of the granule fraction were obtained from PBMC, according to the following protocol. - Thaw PBMC and resuspend in 0.34 M sucrose (pH 7.4). - Disrupt cells by rapid sonication (cell breakage is confirmed by light microscopy) and centrifuge at 200g for 10 min to remove the cell debris. - Centrifuge supernatant at 27,000g for 30 min at 4 °C. Resuspend the pellet in 5% cold acetic acid, sonicate on ice, and let overnight at 4 °C for peptide extraction. - Clear the peptide extract by centrifugation at 27,000g for 30 min at 4 °C. - Dry the extract in a vacuum centrifuge, and dissolve in 20% ethanol for antibacterial activity determination. 3.2.5 Antibacterial activity The antibacterial activity of the PBMC peptide extracts was tested against Bacillus megaterium Bm11, using the inhibition zone assay (Hultmark et al., 1983). - Thin plates of 1% agarose in LB broth, containing approximately 4x105 cells are made: bacteria are grown in LB broth till an optical density of 1 at 590 nm, corresponding to a mid-logarithmic phase. The bacterial cells are hence diluted in LB medium and approximately 2x105 colony forming units are added to 6 ml LB/1% agarose and poured onto a Petri dish. - Small wells (3 mm diameter) are punched onto the LB-agar layer and 3 µl samples are loaded into the wells. - Incubate overnight at 30 °C; measure the diameters of the inhibition zones and express the resulting antibacterial activity as cecropin A units (CAU)/106 PBMC, using a standard curve. One cecropin A unit corresponds to the activity of 1 ng of cecropin A on B. megaterium Bm 11 (Simmaco et al., 1998). 3.2.6 ELISA HNP levels in PBMC peptide extract were measured using a human HNP ELISA test kit (Hbt, Walter Occhiena), based on the sandwich principle. Instructions from the manufacturer were followed and the main steps of the procedure are reported below: - incubation of standards and sample peptide extracts for 1hr at RT, followed by 3 washes; 28 - incubation with secondary biotinilated-antibody for 1 hr at RT, followed by 4 washes; - incubation with streptavidin-peroxidase conjugate for 1hr at RT, followed by 4 washes; - incubation with substrate reagent for 20-30 min in the dark, at RT; - stop of the reaction; - measurement of absorbance at 450 nm in a microplate reader. The sensitivity of this test ranges from 40 to 10,000 pg/ml. 3.2.7 Mass spectrometry Peptide extracts were analyzed by matrix assisted laser desorption/ionization time-offlight mass spectroscopy (MALDI-ToF MS) using a Voyager DE mass spectrometer (PerSeptive Biosystem), using external calibration. The calibrator mixture is composed by: angiotensin I (1,297.51 Da), adrenocorticotropic hormone (ACTH), fragment 1-17 (2,094.46 Da), ACTH, clip 18-39 (2,094.46), ACTH, fragment 7-38 (3,660.19) and bovine insulin (5,734.59 Da). Sample preparation is as follows: - mix 1 µl of peptide extract with 4 µl of the matrix solution (sinapinic acid solution); - deposit a droplet (0.5-1 µl) of the matrix/sample mixture on the stainless steel plate and let it dry. - analyse by MALDI ToF, using up to 500 shots per spectrum. 3.2.8 RNA preparation -Total RNA was purified from PBMC using TRIzol reagent (Invitrogen). The main steps of the procedure are reported below. - Homogenize up to 107 cells in 1ml TRIzol, by repetitive pipetting and vortexing, and incubate at RT for 5 min; - add 200 µl chloroform, shake vigorously and incubate at RT for 3 min; - centrifuge at 12,500 g for 15 min at 4 °C and transfer the upper aqueous phase to a new tube; precipitate by adding 0.5 ml isopropanol and keeping at -20 °C for 15 min. Centrifuge at 12,500 g for 15 min at 4 °C; - wash the pellet with 1 ml ice-cold 75% ethanol, centrifuge again at 8,000 g for 5 min at 4°C; discard supernatant, blot the excess of liquid and let the pellet dry for 10-15 min at RT. - Resuspend the RNA pellet in 20-50 µl of DEPC H2O and store at -80 °C. Part of the RNAs was extracted using Qiagen RNeasy mini kit, following the instructions on the accompanying handbook. RNA recovery and purity were controlled by spectrophotometric absorption measurements at 260 nm and 280 nm; RNA integrity is checked by electrophoresis and visualization on 1% agarose gel. Part of the RNA preparations was analysed by capillary electrophoresis on RNA 6000 Nano LabChip Kit using the Bioanalyser (Agilent Technologies), and following the manual instructions. This highly sensitive method is particularly useful when handling small amounts of RNA sample, in fact it allows qualitative 29 and quantitative analysis of as little as 50 ng of total RNA. Figure 3.1 shows representative conventional RNA gel electrophoresis and Bioanalyzer chip electrophoresis. A. B. Fig 3.1 Examples of total RNA analysis by gel electrophoresis (A) and by capillary electrophoresis on the Bioanalyzer chip (B). In A, the two major bands correspond to 28S and 18S ribosomal RNAs, a lower fainter band corresponds to 5S rRNA. In lane 5, an upper band due to genomic contamination is evident. B is the elecropherogram profile of an RNA sample : peaks at about 40 sec and 47 sec represent 18S and 28S, respectively, while the peak on the left is the marker; the sharper the peaks, the better the quality of the RNA. DNase I treatment In order to remove any genomic contamination, RNA was treated with RNase free DNase I, prior to retrotranscription. Major steps of the protocol are described below. - Incubate total RNA (5 µg) with DNase I (5 units), in the presence of 20 mM Tris HCl, pH 8.2, 50 mM KCl, 1 mM MnCl2, at 25 ˚C for 30 min, in a final volume of 50 µl; - stop the reaction by adding 2,5 mM EDTA (pH 8.0) and heating 5 min at 75˚C. RNA is subsequently re-extracted using TRIzol, and, after that, its integrity is checked again by gel electrophoresis and/or chip electrophoresis on the Bioanalyzer (Fig 3.1). 30 cDNA synthesis First strand cDNAs are synthesized from total RNA using SuperScript II RT (Invitrogen) with oligo-(dT)18, according to the manufacturer’s protocol: - preheat at 65 °C for 5 min the RT-reaction mixture, including up to 2 µg of total RNA, 25 µg/ul oligo-dT primer and 0.5 mM dNTPs; - add 0.01 M DTT and 1x First-Strand Buffer; incubate the mix at 42 °C for 1 min; - add 200 units of SuperScript II and incubate at 42 ˚C for 50 min; - inactivate reverse transcriptase by heating at 70 ˚C for 15 min. cDNAs are stored at -20 °C. 3.2.9 Purification of DNA fragments from agarose gel Specificity of PCR was checked by gel electrophoresis of the amplification products. PCR-bands are compared to those of a molecular weight standard. The band corresponding to the expected size is excised from the gel and purified using an agarose gel DNA extraction kit (Roche), based on a silica matrix with high affinity for DNA. DNA is eluted from the matrix using appropriate volumes of dH2O, usually 20-50 µl. DNA concentration is determined by spectrophotometer reading at 260 nm. The purified amplicon is then serially diluted, to be used as template and to generate the standard, reference curve. 3.2.10 Quantitative real time RT-PCR Instrumentations and principles Quantitative real-time PCR was performed in the Rotor-Gene 3000 (Corbett Research). In this real time PCR instrument, a thermal cycler and a fluorescence-detection system are controlled by a software which monitors real time product accumulation, by measuring the increase of fluorescence at each cycle; the product of amplification, or amplicon, is detected by specific fluorogenic probes and/or DNA binding dyes (see figure 3.2). This type of technique provides high sensitivity and allows quantitative analysis. FLUORESCENCE CYCLE Fig 3.2. Typical fluorescence profile during a real time PCR run in the Rotor Gene 3000. Generation and accumulation of the amplicon determines an exponential increase in fluorescence as the cycles progress. 31 Three different types of detection chemistries were used: Sybr Green I dye, TaqMan probe, and LUX- primers. Fig 3.3 illustrates their features. 2 1 A 3 1) Intercalating dye: Sybr Green I 2) Dual-labelled probe : TaqMan B 3) LUX primer Fig 3.3. Types of detection chemistries, and corresponding principles, used in real time PCR, in the present thesis. 1) Sybr Green I is a DNA-intercalating dye, whose fluorescence increases upon DNA binding. 2) TaqMan probe is an oligonucleotide with a fluorophore at its 5’ end and a quencher at its 3’ end: due to the exonuclease activity of Taq polymerase, the probe is degraded, the fluorophore released into the solution and separated from the quencher, allowing a higher fluorescence emission. 3) LUX primers are oligonucleotides labelled with a single fluorophore at their 3’ end: fluorescence is quenched by the intrinsic hairpin structure and increased upon primer extension (LUX: Light Upon eXtension). Fluorescence acquisition and calibration were performed on the following channels, according to the fluorophores used: - Sybr (source 470 nm, detector 585 nm); - FAM/Sybr (source 470 nm, detector 510nm); - JOE (source 530nm, detector 585 nm) Quantitation methods The two standard curves method was used to obtain either relative or absolute quantification of the genes of interest, comparing their level of expression to that of the housekeeping gene: fig 3.4 (Corbett Research user manual). 32 Fig 3.4. Representative fluorescence profile and corresponding standard curve obtained by amplifying a six log-range dilution series of a standard DNA, with known absolute concentration. Cycle threshold, Ct, values obtained for the unknown samples are related to the standard curve and so concentration can be calculated. For relative quantification, the standard curve is generated, by using, as template, serial dilutions of a cDNA sample; the units, used to describe the dilution series, are relative and based on the dilution factor of the standard curve. For absolute quantification, the standard curve is generated from serial dilutions of the purified specific amplicon, whose concentration has been spectrophotometrically determined; the units used to describe the dilution series are expressed as “copy number/µl”. The relative quantification method was adopted for a first part of the study on the expression of HNPs, and for IL-2 gene. The absolute quantitation method was used for HNP genes profile in most of the HCV-patients, including those in therapy, and the in vitro experiments, as well as for LL37 and IL-15. Assay conditions PCR mix and cycling conditions (such as oligonucleotides and fluorophore concentration, MgCl2 concentration, annealing and fluorescence acquisition temperatures, cycle number, etc.) were pre-optimized for each particular primer pair and target gene, so to improve efficiency and specificity of the reaction. A typical PCR reaction mix contained two-to-twenty ng of template cDNA, corresponding to 2-5 µl of diluted cDNA, in a final reaction volume of 20-25 µl. For Sybr Green I-detection, the PCR mix includes: -2.5 U of Taq polymerase (Diatech) -0.2 mM dNTPs 33 -1.5-2.5 mM MgCl2 -1x PCR buffer -Sybr Green I (1:40,000-1:20,000 dilution) -50-125 nM forward and reverse primers When using LUX primers, the mix is essentially the same as above, but it contains a Platinum hot start polymerase (Invitrogen) and no Sybr Green I. Concentrations of each component, in the mix used with TaqMan probes, have been optimized in Diatech laboratories. Typical cycling conditions in a Sybr Green or LUX primer-based reaction are: - initial denaturation at 95 °C for 5-3 min - 40-30 cycles comprising: 20 sec denaturation at 94 °C; 20-30 sec annealing at 55-61 °C; 20-30 sec extension at 72 °C ( with or without acquisition of fluorescence); 15 sec at 80-85 °C, for acquisition of fluorescence; - 1 min final extension at 72 °C; - melt analysis (see below). Typical cycling conditions in a TaqMan probe-based reaction are: - initial denaturation at 95 °C for 5-3 min; - 40 cycles comprising: 20 sec denaturation at 95 °C; 60 sec annealing and extension at 60 °C, with fluorescence acquisition. Fluorescence data are acquired either during the extension phases or in a subsequent phase at higher temperature, according to the melting point of the specific amplicons. When using Sybr Green I and/or LUX primers, a melt curve is generated after each PCR run, and subsequently analyzed to check for specificity of the amplification products: samples are heated at 70 °C for 2 min and then slowly heated at 0,2 °C /sec up to 99 °C with a continuous acquisition of the fluorescence. IL-2 IL-15 Fig 3.5. A representative melt curve analysis. In this example, two different melting temperatures were detected for IL-2 and IL-15 amplification products. PCR were run using specific LUX primers. Different amplification products will melt (=denature) at different temperatures, based on their lengths and G/C contents; the melting peaks result from the differential of the fluorescence (y axis) relatively to the temperature (x axis), as illustrated in figure 3.5. The melting peaks reflect the products amplified during the reaction, and are analogous to the 34 bands on an electrophoresis gel. Melting curve analysis allows qualitative monitoring of the reaction, and permits to adjust the fluorescence acquisition temperature so to exclude aspecific, primer-dimer signals. Each PCR assay included duplicates of each cDNA sample, no-template controls, and two standard curves, for the housekeeping gene (GAPDH), and for the gene of interest, respectively, constructed so to cover a 3 to 6 LOG-range (1-1,000,000). Data were analyzed using the Rotor-Gene Analysis Software V 4.6. 3.2.11 Statistical analysis Statistical analysis was performed using Student's t test for parametric data and Wilcoxon rank sum test for non parametric data. The correlation between α-defensin levels and the degree of hepatic fibrosis was assessed by the Pearson’s correlation coefficient. Any p value<0.05 was considered significant. 35 4 RESULTS AND DISCUSSION 4.1 Expression profiling of HNPs in HCV patients Quantitative real time reverse transcription PCR (qRT-PCR): validation of the housekeeping genes Quantitative gene expression assays require a parameter for normalization of the data: this can be either the number of cells, the RNA quantity, or an internal control gene. The approach based on an endogenous control gene, to which normalize the mRNA fraction, is the most frequently used, and it has been adopted even in this study. In a quantitative RT-PCR experiment, the reference gene is simultaneously amplified with the target gene or gene of interest (GOI): the quantitative values obtained for the GOI can then be normalised to this internal standard; this operation allows minimisation of errors due to sample variations (variations in the amount of starting material, samples obtained from different individuals, different efficiencies in enzymatic activities, etc.) and makes possible comparison of the data obtained from different samples and sets of experiments. The expression of an ideal internal standard, also referred to as house-keeping gene (HKG), should not vary among different individuals or tissues under investigation, nor in response to experimental treatment or pathological states (Bustin, 2002). In addition, the HKG should also be expressed at about the same level as the gene/s under study. Validation of the candidate reference genes, in order to confirm their presumed stability of expression, is an essential prerequisite to any serious quantitative real time RT-PCR experiment. In the following study, we considered two putative housekeeping genes: the moderately abundant level-expressed glyceraldehydes-3-phosphate-dehydrogenase (GAPDH) (accession number BC029618, NM_002046), coding for the fundamental glycolytic pathway enzyme, and the low level-expressed TATA box binding protein (TBP) (accession M55654, NM_003194), encoding the general RNA polymerase II transcription factor. Fixed amounts of template cDNA, corresponding to approximately 10-20 ng of starting RNA, were used to drive real time RT-PCR of both potential housekeeping genes; RNA samples were representative of a population including PBMC from both healthy and diseased individuals. The genes exhibited a marked difference in abundance: GAPDH transcript resulted far more abundant (Ct values ≈ 20) than TBP (Ct values ≈32). The Ct values obtained are conveniently represented on a graph as relative concentrations. Figures 4.1 and 4.2 show the results obtained for GAPDH and TBP genes, respectively. 36 Gapdh relative concentration GAPDH mRNA calculated concentration using 1020 ng of cDNA template in RT-PCR 120 100 80 60 40 20 A H cu IV C te hr on A ic ot C B he ir r rh in os fe is m ctio B ea n n s va lu e C C irr ho t H ic C C C hr on ic H ea lth y C co n tro ls 0 Fig. 4.1. Relative concentrations obtained for GAPDH transcripts using cDNA samples from healthy controls and different types of diseased patients. The mean value ± standard deviation is also shown. HCC, hepatocellular carcinoma. TBP mRNA calculated concentration using 10-20 ng of cDNA template in RT-PCR TBP mRNA relative concentration 100 80 60 40 20 0 H ea s C ol ic tr n n o co hr y C h lt C tic ho r ir C H CC H IV C ic on r h B A e te valu u c A an e m Fig. 4.2. Relative concentrations obtained for TBP transcripts using cDNA samples from healthy controls and different types of diseased patients. The mean value ± SD is also shown. HCC, hepatocellular carcinoma. The observed variability was evaluated using Student’s T test: no statistically significant variation was found (P> 0.05, comparing values from diseased patients, versus those from healthy controls). The variance observed among different samples may be compatible with intrinsic variability deriving from errors in spectrophotometric determinations of RNA concentration, different efficiencies in RT reactions, pipetting inaccuracies, differences in handling and storage of biological samples (cells, cDNA), etc. To further confirm this evaluation, the ratios between the observed RNA levels of GAPDH and TBP were calculated and plotted: if the two putative HKGs are truly invariant, this ratio should be constant for the 37 different samples examined. Fig. 4.3 shows the results: the medium value of the ratios is 1.2 ± 0.7 (mean ± SD) and the plot shows an acceptable level of inter-samples variability. Gapdh mRNA/TBP mRNA) Ratio between GAPDH and TBP mRNA levels in different patients and healthy controls 5 4 3 2 1 0 patients healthy controls m ean ratio Fig. 4.3. Values obtained dividing GAPDH mRNA level by TBP mRNA level for cDNAs from diseased patients and healthy controls. In order to prove that either of the above genes could be used as internal references, the transcriptional level of the gene of interest, that is HNP, was analysed in a limited set of samples using simultaneously both genes for the normalization of the data. The results, shown in Fig. 4.4, suggest that the same expression trend is maintained using either of the proposed HKGs. The graph indicates also that the variation in the level of expression of HNP, among different individuals, is much higher than the inter-sample variability observed when comparing the levels of the two candidate HKGs: this allows us to neglect GAPDH and TBP variability and to consider them as good HKGs. 38 HNP relative transcription using GAPDH and TBP as internal reference genes HNP/TBP HNP/GAPDH relative mRNA level 10000 1000 100 10 1 sample Fig. 4.4. HNP expression profile using two alternative housekeeping genes. The dotted bars refer to values obtained from healthy controls, all the other data are from diseased patients. Given that both genes are equally valid as internal standards, we decided to use GAPDH for the rest of the study: this choice is motivated by its higher expression level (thus producing a stronger and clear fluorescence signal, even at low template concentrations) and by the better efficiency in its RT-PCR reaction. Quantitative real time RT-PCR analysis of HNP genes The expression profile of HNP genes was studied at the transcriptional level, using two different real time qRT-PCR approaches. In a first set of HCV-patients the relative abundance of the HNP transcripts was determined using a Sybr Green detection based method. In this case the ratios HNP mRNA /GAPDH mRNA levels obtained from each analysed sample (both patients and healthy controls) were normalized to that of a calibrator sample arbitrarily designated among the healthy controls. The results of this approach are outlined in Fig. 4.5. 39 Healthy controls Acute Chronic Cirrhotic HCC 1 10 100 1000 Relative HNP mRNA level Fig. 4.5. HNP relative transcriptional level in PBMC from healthy controls and patients at different HCV infection stages. HCC, hepatocellular carcinoma. Relative quantitation and Sybr Green detection-based method. Bar charts show mean values ± SEM (standard error of the mean) for each subgroup of patients. The expression of HNP genes was further analysed in a larger group of patients and healthy individuals by a real time RT-PCR approach, based on a more specific and sensitive TaqMan detection system and using an absolute quantitation method. The results obtained by this approach are outlined in Fig. 4.6. Comparing the data obtained using these two alternative approaches a correlation factor of 0.82 was obtained, which is indicative of a good agreement. 40 Healthy controls Acute Chronic Cirrhotic HCC 1 10 100 1000 HNP mRNA level (HNP/GAPDH) Fig. 4.6. HNP transcriptional level in PBMC from healthy controls and patients at different HCV infection stages. HCC, hepatocellular carcinoma. Absolute quantitation method and TaqMan probe detection-based system. Mean values and standard errors for each subgroup of patients are shown. The transcriptional level of HNP genes was found to be significantly higher in all HCV infection stages with respect to the level observed in the healthy controls: p(healthy/acute) < 0.001; p(healthy/chronic) = 0.004, p(healthy/ cirrhotic) = 0.02; p(healthy/HCC) = 0.02. No significant correlation was found between HNP mRNA levels and percentage of contaminating granulocytes and/or monocytes in the starting PBMC preparation. ELISA The concentration of the HNPs in peptide extracts obtained from PBMC of healthy and diseased individuals was specifically determined by ELISA. Fig. 4.7 outlines the result of this analysis: the concentration values were normalized to the number of starting cells. The concentration of HNPs resulted significantly higher in PBMC from patients at every HCV infection stages when compared to the range of concentrations observed in PBMC from healthy individuals. Values are as follows: p(healthy/acute)<10-6; p(healthy/chronic)<0.001; p(healthy/cirrhotic)<10-5; p(healthy/HCC)<0.0005. 41 Healthy controls Acute Chronic Cirrhotic HCC 0,0 0,4 0,8 1,2 1,6 6 HNP level (ng/10 cell) Fig. 4.7. HNP level, as detected by ELISA, in peptide extracts from PBMC of healthy controls and patients at different HCV infection stages. Mean values and corresponding standard errors are reported for each class. HCC, hepatocellular carcinoma. HNP levels did not correlate with aspartate aminotransferase (AST) or alanine aminotransferase (ALT) serum concentrations; besides, higher amounts of α-defensins were detected in subjects with HCV-RNA>100,000 IU/mL, but there was no correlation with HCV viral load or different HCV genotypes. Antibacterial activity The presence of the α-defensins in the peptide extracts can be assessed by evaluating their distinctive biological activity, that is their antimicrobial property. To this aim, the antimicrobial capacity of the whole peptide extract was tested against a laboratory strain of Gram-positive bacteria, Bacillus megaterium Bm11, using an inhibition zone assay. The diameter of the zone where the growth of the microorganism has been inhibited is converted into cecropin A units. This type of test lacks the specificity of the ELISA, since also low molecular weight antimicrobial agents, other than defensins, might contribute to the antimicrobial capacity of the extract; nevertheless, results obtained with both these approaches were found to be in good agreement with each other (R=0.82); moreover, the antimicrobial assay provides a useful measure of the α-defensins which are present in a biologically active form. The mean values of antimicrobial activity found in PBMC extracts from healthy controls and HCV patients at different stages are reported in Fig. 4.8. 42 Healthy controls Acute Chronic Cirrhotic HCC 0 300 600 900 1200 6 Antimicrobial activity (CAU/10 cell) Fig. 4.8. Antimicrobial activity of peptide extracts from PBMC of healthy individuals and patients at different stages of HCV infection. The antimicrobial activity is expressed as cecropin A units normalized to cell number. Mean values and corresponding standard errors are reported for each class. HCC, hepatocellular carcinoma. The results indicate that in all stages of HCV infection, the level of antimicrobial activity is significantly greater than in healthy controls: P(healthy/acute)<10-5; -5 P(healthy/chronic)<0.01; P(healthy/cirrhotic)<10 ; P(healthy/HCC)<0.001. MALDI ToF analysis The mass spectral analysis of the peptide extracts from PBMC of healthy controls and HCV-patients revealed the presence of the three most abundant HNP isoforms. A representative mass spectrum is shown in Fig 4.9: a cluster of three peaks with the expected molecular masses for HNP-1, 2, and 3 was observed in each sample, including those from healthy controls. 43 3372 HNP-2 % Intensity 3443 HNP-1 3487 HNP-3 m/z Fig. 4.9. Mass spectral analysis. Representative MALDI-ToF mass spectrum of peptide extract from PBMC. The three major peaks correspond to molecular masses compatible with those expected for HNP-1, 2 and 3. The MALDI ToF analysis confirmed that the peptide extracts contained mostly defensins, since little or no signal of comparable intensity was observed in the spectral region spanning from 3 kDa to 10 kDa, where other antimicrobial peptides, such as cathelicidins, might be detected (see below). 4.2 HNPs expression in other hepatic pathologies The expression of HNP genes was studied also in PBMC from patients affected by liver pathologies other than hepatitis C. A quantitative real time RT-PCR analysis allowed us to compare the HNP transcriptional levels between patients in acute stages of infection by hepatitis C, A and B viruses (HCV, HAV and HBV) and in HCV and HBV chronic and cirrhotic stages. Fig. 4.10 outlines the results of this study: HNP expression level in acute A and chronic B stages is similar to that of the healthy controls, whereas it increases in acute and cirrhotic B patients, though the extent of the increase seems less relevant than that of the corresponding HCV stage. The HNP transcription was compared between patients with hepatocellular carcinoma associated to HCV infection and two patients with hepatic carcinoma of non-viral origin (Fig. 4.11). Due to the limited sample size, the result of this analysis may only suggest that the highest expression level is a distinguishing feature of HCV-originated HCC. All the above data, obtained by quantitative real time PCR, were confirmed by ELISA and antimicrobial assays. 44 45 30 15 cirrhosis B cirrhosis C chronic B chronic C acute B acute C acute A 0 healthy controls HNP transcriptional level (HNP/GAPDH) 60 Fig. 4.10. HNP transcriptional level in PBMC from healthy controls and from patients at different stages of infection by hepatitis C, A and B viruses. Mean values and corresponding standard errors for each class are indicated. Healthy controls HCC (HCV-) HCC (HCV+) 1 10 100 1000 relative HNP m RNA level Fig. 4.11. HNP trancriptional level in PBMC from healthy controls and from patients with hepatocellular carcinoma (HCC) of viral (HCV+) and non-viral (HCV-) origin. Data were obtained using relative quantitation method and Sybr Green detection. Overall, these result indicate that, though an increased HNP expression level may be associated to a generic pathological condition, a much more significant increase seems to be characteristic of HCV-linked pathologies. 45 4.3 Expression profile of other immune-related genes in HCV-patients To better understand the characteristics of the immune response to HCV, we decided to study the expression of the genes encoding the cathelicidin antimicrobial peptide LL37 and interleukin-15 (IL-15). LL37 gene was chosen as a kind of negative control: this gene codes for a peptide that shares with defensins the same fundamental role in human innate immunity, being an important component of both the phagocyte and the epithelial defences systems; yet, the structure of its promoter, led us to expect a rather different type of transcriptional regulation. IL-15, a novel cytokine sharing many immunological activities with IL-2, is critical in the defence against viral infections by activating NK cells (Waldmann and Tagaya Y, 1999). IL15 was a sort of positive control, as we actually expected an enhanced transcription for this gene: in fact, it is known that the plasma concentration of this cytokine rises in HCV infections and correlates strongly with disease progression; moreover, IL-15 promotes transcription of α-defensin genes in NK cells, in vitro (Kakumu et al., 1997; Obata-Onai, et al., 2002). A quantitative real time RT-PCR, based on sensitive LUX primer detection and absolute quantitation method, allowed us to study the expression of these genes at the transcriptional level. The results obtained for each class of HCV patients, together with the trend of expression already found for HNP genes, are shown in Figs. 4.12 and 4.13. HNP/Gapdh HNP or LL37 mRNA level . 120 (LL37/Gapdh)x100 100 80 60 40 20 0 healthy controls acute chronic cirrhotic HCC Fig 4.12. HNP and LL37 mean transcriptional levels in PBMC from healthy controls and HCV patients at different stages. 46 HNP/Gapdh (IL-15/Gapdh)x100 HNP or IL-15 mRNA level 120 100 80 60 40 20 0 healthy controls Fig. 4.13. chronic cirrhotic HCC HNP and IL-15 mean transcriptional levels in PBMC from healthy controls and HCV patients at different stages. No significant variation was found in LL37 expression (p>0.05 comparing healthy with each class of diseased subjects); conversely, a relevant increase in transcription of IL-15 gene was observed: p(healthy/chronic)<0.001, p(healthy/cirrhotic)<0.05, p(healthy/HCC)<0.05. Moreover, the trend of IL-15 transcription seems to reflect that of HNP genes. IL-15 transcription was found to be significantly higher also in chronic B hepatitis patients (data not shown), with a mean level that is very close to that found in HCV chronic subjects. It is noteworthy, however, that in the case of chronic B hepatitis, the higher IL-15 expression is not accompanied by a parallel increase of α-defensins. The absolute level of transcription of both LL37 and IL-15 was found to be about 100 fold lower than HNP genes: this feature can be partly explained considering that cytokines act at very low concentrations, resembling, for this aspect, the function of hormones. For LL37, the low level of expression can be due either to the lack of appropriate stimuli, or to very poor expression by the types of cells (mostly lymphocytes) contained in the PBMC preparation. The low expression of LL37 was confirmed also by the lack of any peak corresponding to its molecular mass (≈4.5 kDa), when analyzing the peptide extracts by MALDI ToF. The expression of HNP, LL37 and IL-15 was studied in an acute HCV patient undergoing spontaneous viral clearance: the greatest modulation was observed for HNP and IL-15 genes, with little or no variation for LL37 (Fig 4.14). This finding suggests that both IL-15 and HNP are somehow associated to HCV pathology, whereas LL37 is not. 47 mRNA level 50 HNP/Gapdh 40 (LL37/Gapdh)x100 IL-15/Gapdh 30 20 10 0 acute C aviremic acute C Fig. 4.14. Transcriptional levels of HNP, LL37 and IL-15 in an acute C patient before and after viral clearance. HNP or LL37 mRNA level A strong modulation for LL37 gene was found only when studying control patients with bacterial infections, such as meningitis (Fig. 4.15), suggesting the involvement of this AMP in the immune response to this kind of disease. In that case, transcription of both HNP and LL37 was found to rise significantly. This finding implies that HNP transcriptional enhancement is not specific of immune response to HCV infection. Moreover, it may explain the raise in the plasmatic level of defensins, observed during bacterial infections (Ihi et al., 1997), as the result of a de novo synthesis of these peptides, rather than as a consequence of a more intense neutrophil activity. 10000 HNP/Gapdh (LL37/Gapdh)x100 1000 100 10 1 healthy controls Fig. 4.15. bacterial infection HNP and LL37 transcriptional levels in PBMC from healthy controls and from patients with bacterial infections. 4.4 HNPs expression in HIV patients In this study, we included also a control group of ten patients with HIV infection. The expression of both HNP and LL37 genes was studied by qRT-PCR in PBMC isolated from these individuals: results are shown in Fig. 4.16. 48 HNP or LL37 transcriptional level HNP/GAPDH 1000 (LL37/GAPDH)X100 100 10 1 HIV patients healthy controls Fig. 4.16. Mean HNP and LL37 transcriptional levels in PBMC from HIV+patients and healthy controls. Mean HNP/GAPDH values and standard errors are 149±39 for HIV positive patients, and 4.4±0.9 for healthy controls; means for LL37/GAPDH are 0.125±0.03 and 0.036±0.02 for HIV patients and controls, respectively. Both HNP and LL37 transcriptional levels were found significantly higher in cells from HIV-infected patients when compared to the values of the healthy controls: p<10-6 for HNP, and p<0.05 for LL37. The overexpression of HNPs is of particular interest, since these peptides have been shown to have clear anti-HIV 1 properties; what is more, HNPs have been identified as components of the CD8+ T cell antiviral factor, which is thought to play a fundamental role in delaying AIDS symptoms in the so called HIV-non-progressor patients. More recently, Trabattoni et al. (2004) have examined α-defensin expression in PBMC and CD8+ T cells and found that the expression level was up to 10-fold higher in HIV-exposed seronegative patients than in low risk healthy controls. On the whole, our preliminary data appear to confirm that HNPs might have an important function in immune response to this kind of viral infection; besides, they suggest that αdefensin overexpression is an index of immune activation in response to exposure to HIV, rather than a genetically predetermined feature. More studies, are needed to understand HNP specific role in this pathology, as well as the mechanism/s that might elicit their expression. 4.5 HNPs expression in selected cell subsets Our PBMC preparations typically consisted of T and B lymphocytes, with a low percentage of monocytes (≈10%) and less than 5% of neutrophils. Sensitive techniques, such as double immuno-staining and RT-PCR, have already been used to demonstrate the expression of HNP-1 and 3 by monocytes, B cells and NK cells (Agerberth et al., 2000). We therefore sought to verify whether HNP are expressed also by the other cell types composing PBMC. By qRT-PCR, we assessed the expression of HNP and LL37 genes in CD4+ and CD8+ T lymphocytes (purity >95%), and in neutrophils (Fig. 4.17). The α-defensins are expressed by all these cell types with the lowest level found in neutrophils; conversely, LL37, whose transcriptional level is much lower than HNP, is mainly expressed in neutrophils, with very little mRNA in CD4+ and CD8+ T lymphocytes. 49 HNP/Gapdh HNP or LL37 mRNA level 8 (LL37/Gapdh)x100 7 6 5 4 3 2 1 0 PBMC Fig. 4.17. CD4+ CD8+ Neutrophils Transcriptional levels of HNP and LL37 genes in the whole PBMC and selected cellular subsets from healthy individuals. These results agree with our expectations: HNPs are massively expressed by neutrophils only over a limited period of time corresponding to the stage of granulocyte precursor; once differentiated, neutrophils contain high levels of defensins peptides but transcription of their genes, at least in healthy individuals, is low and comparable to that of T lymphocytes. Overall, these data support that peripheral leukocytes retain substantial ability to transcribe HNP genes and to synthesize HNP peptides, in accordance with other recent works ( Fang et al., 2003). On the other hand, LL37 is poorly expressed by T lymphocytes and the low level we have found in PBMC is likely to be largely due to contaminating neutrophils; with respect to the low expression level in T cells, our data agree with previous findings (Agerberth et al., 2000). Transcription of HNP in CD8+ cells has been questioned by many authors (Lehrer, 2004), though Levy and colleagues have recently excluded that this type of T lymphocytes might produce defensins (Mackewicz, et al. 2003). Yet, we are quite confident of our results: considering the purity of the CD8+ cells (>95%), the intense signals obtained in real time PCR, both using sensitive Taq Man probe and confirming specificity further by Sybr Green melt curve analysis, cannot be solely due to an original contamination of the cells. 4.6 Correlation between HNP level and liver damage In 90 patients with chronic HCV infection the HNP expression was studied in relation to the histological damage, as determined by liver biopsy. The antibacterial activity of peptide extracts from PBMC of patients with chronic C hepatitis was found to rise progressively in correlation with liver fibrosis (Fig. 4.18). In patients with moderate stage of fibrosis (stage 0-2), the median value of antibacterial activity was 612 CAU/106 cells (mean=627, SD=140, range=400-888), whereas in those with severe stage of fibrosis or cirrhosis (stage 3-4), the median value was 835 (mean=891, SD=221, range=426-1342). Moreover, in patients with hepatocellular carcinoma, the median value of antibacterial activity was 1104 CAU/106 (mean=1260, SD=560, range=630-2360). By using the Wilcoxon rank sum test, we found higher CAU levels in patients with stage 0-2 compared with controls (p=0.046, z=–1.992); in patients with stage 3-4 higher than in those with stage 0-2 (p<0.001, z=-3.563); in patients with hepatocellular carcinoma higher than in stage 3-4 (p=0.012, z=-2.51). 50 3000 6 CAU/10 cells 2000 1000 0 controls stage 0-2 stage 0-4 HCC Fig 4.18. Antibacterial activity in peptide extracts from PBMC of healthy controls and patients with a different stage of liver fibrosis. The antibacterial activity is expressed as cecropin A units per cell number (CAU/106 cells). The top and the bottom of boxes represent the 25th and 75th percentiles. The line in the middle of the box is the median. The whiskers represent the values within 1.5 times the interquartile range from the upper or lower quartile. Our data also showed a very strong linear correlation between the stage of fibrosis and the CAU values (r=0.706, p<10-6). A similarly strong correlation was found even when considering the HNP levels as detected by ELISA (Fig 4.19). HNP levels from healthy controls showed values ranging from 0.1 to 0.4 ng/106 cells (median value=0.2, mean value= 0.217, SD=0.09). In patients with stage of fibrosis 0-2, the median value was 0.6 ng/106 cells (mean=0.72, SD=0.45, range=0.25-2.5), whereas in those with stage of fibrosis 3-4, the median value was 1.1 (mean=1.29, SD=0.59, range=0.5-2.6). Moreover, in patients with hepatocellular carcinoma, the median value of HNP levels was 1.78 ng/106 cells (mean=1.94, SD=1.14, range=1-4). By using the Wilcoxon rank sum test, we found higher HNP levels in patients with stage 0-2 compared with controls (p=0.08, z=–2.669); in patients with stage 3-4 higher than in those with stage 0-2 (p<0.001, z=-3.580); in patients with hepatocellular carcinoma higher than in stage 3-4 (p=0.049, z=-1.961). We found a statistically significant linear correlation between HNP levels and stage of fibrosis (r=0.59, p<106). 51 5 HNP level ng/106 cells 4 3 2 1 0 -1 controls stage 0-2 stage 0-4 HCC Fig 4.19. α-Defensin levels, as detected by ELISA, in PBMC of healthy controls and patients with a different stage of liver fibrosis. The top and the bottom of boxes represent the 25th and 75th percentiles. The line in the middle of the box is the median. The whiskers represent the values within 1.5 times the interquartile range from the upper or lower quartile We also measured the α-defensin mRNAs in the same PBMC samples and found a direct correlation between the antimicrobial activity and the number of HNP transcripts (r=0.51, p<10-6) and between the HNP levels and the number of HNP transcripts (r=0.493, p<0.001). On the basis of this correlation between HNP levels and liver damage, α-defensins might be prospectively used as helpful biological markers to stage disease progression. 4.7 In vitro experiments on PBMC Effect of HCV C, Cyclosporin A and the core protein of hepatitis B virus In order to assess the potential effect of HCV core protein (HCV C) on enhancing the expression of HNPs, we performed a series of in vitro experiments in which freshly isolated PBMC were incubated in RPMI medium with or without the addition of a recombinant HCV C (2 µg/ml). To verify the possibility of an involvement of NFAT in the transcriptional activation of the α-defensin, HCV C stimulated PBMC were pre-treated with cyclosporin A (CsA, 1 µg/ml), an immunosuppressive drug that specifically inhibits the NFAT pathway, by preventing calcineurin-dependent dephosphorylation. Moreover, the specificity of the HCV C effect was established by incubating PBMC with the recombinant core protein of the hepatitis B virus, HBcAg (2.5 µg/ml). The response of the cells was investigated at both transcriptional and peptide level, by real time quantitative RT-PCR and ELISA, respectively. The results of these experiments are shown on Figs. 4.20. and 4.21. Upon incubation with HCV C, HNP transcription rose markedly (up to five-fold increase) in PBMC from both diseased and healthy individuals. The experiment was repeated several times on PBMC from healthy controls and twice in PBMC from chronic C patients, with similar results. An enhancement of transcriptional level was detected also in PBMC from 52 Relative HNP transcriptional level acute C patients (data not shown). The increase in the transcription level was a fast response, since it become evident already after 30 min incubation with HCV C, as detected in preliminary time-course experiments. The same studies indicated that the maximal transcriptional induction was reached after 3-4 h of incubation (Fig. 4.20). 6 PBMC from chronic C patient 5 PBMC from healthy control 4 3 2 1 0 HCV C CsA HBcAg Relative HNP transcriptional level in HCV C, CsA and HBcAg–treated PBMC from chronic C patient and healthy control. The relative transcriptional level of HNP genes is defined as the ratio of the transcriptional level observed in the stimulated cells normalized to that of the untreated cells (negative control). Relative HNP level Fig. 4.20. CsA+ HCV C 2,5 PBMC from chronic C patient 2,0 PBMC from healthy control 1,5 1,0 0,5 0,0 HCV C Fig. 4.21. CsA + HCV C CsA HBcAg Relative HNP concentration in peptide extracts from HCV C, CsA and HBcAgtreated PBMC from chronic C patient and healthy control. The HNPs level was detected in PBMC peptide extracts by ELISA test. The relative HNP level was obtained normalizing values observed in treated cells to those of the untreated cells (negative control). We could notice that the induction of HNP expression was generally greater in cells from HCV patients, especially in acute stage, rather than in those from healthy individuals. Nonetheless, since cells from both diseased and healthy individuals could respond to HCV C induction, it is conceivable that this type of reaction is initiated by innate immune 53 mechanisms and is not exclusive of individuals who have experienced HCV infection and mounted an adaptive immune response to it. The induction of α-defensins expression was clearly inhibited upon CsA treatment, prior to HCV C addition; while CsA alone kept the transcriptional levels close to those observed in the untreated sample. These data suggest that the α−defensin synthesis induced by HCV C and, presumably, by hepatitis C virus infection, may be mediated by the NFAT pathway. Moreover, the up-regulation of the HNP genes seems a selective response to HCV C, as no variation in α-defensins mRNA was observed in PBMC upon incubation with HBcAg, a protein that shares with HCV C both the structural function and immunomodulatory properties (Manigold et al., 2003; Hyodo et al., 2004). Overall, the results obtained by qRT-PCR were confirmed by ELISA at the peptide level (Fig. 4.21), even if the inducing effect of HCV C was less evident and a slight increase in peptide level was discerned also upon treatment with the recombinant HBcAg. The presence of the three major isoforms of HNPs, in the analysed samples, was confirmed by MALDI-ToF spectrometry. Effect of phorbol 12-myristate 13-acetate, ionomycin and phytohemagglutinin The potential involvement of NFAT in transcriptional control of HNP genes was further investigated by evaluating the expression of both HNP and IL-2 genes upon treating PBMC with combinations of HCV C, CsA and canonical activators of NFAT cascade, such as phorbol 12-myristate 13-acetate (PMA) and ionomycin (phorbol esters activate protein kinase C, while ionophores, such as ionomycin, trigger intracellular calcium release, hence concurring to activation of the NFAT pathway). The effect of the T cell mitogen, phytohemagglutinin (PHA), was also studied: this lectin triggers T-cell activation and IL-2 production by binding non-specifically to the cell surface receptor complex; the combination of PHA and PMA results in a greatly increased IL-2 production (Manger et al., 1986; Liu et al., 1992). IL-2 gene was chosen as positive control, for it is classically known to be transcriptionally regulated via NFAT. Relative transcriptional levels of HNP and IL-2 genes were assessed by real time qRTPCR and were calculated dividing the level detected in treated cells by that observed in untreated (negative control) cells: hence, values greater than 1 indicate an increased transcription, while values equal to or minor than 1 indicate no variation or decreased transcription, in relation to untreated cells. Transcriptional level of IL-2 gene was studied using LUX primer-detection and a relative quantitation method. Data are reported on Fig. 4.22. 54 1000 IL-2 100 10 PH A A A+ PH sA PM M +P M A+ io n) +C A+ io n sA +C C+ P V (H CV C HC (P M A+ io n) V HC A+ io n V HC 0,1 PM C+ Cs A 1 C Relative mRNA level HNP Fig. 4.22. Relative transcriptional level of HNP and IL-2 genes in PBMC from acute C patient, following incubation with HCV C, CsA, PMA, ionomycin (ion) and PHA, as indicated on the x axis. Dotted boxes highlight data obtained combining NFAT inducers with the inhibitor (CsA). Both genes showed a similar trend of transcription. HCV C alone induced not only HNP, but also IL-2; by adding CsA, transcriptional level decreased. NFAT inducing agents, PMA and ionomycin, up-regulated the expression of both genes and the addition of CsA made the level lower again: the effect of this treatment on IL-2 may appear obvious, since several other works have already demonstrated it; yet, in our case it was a sort of cue reassuring us that the conditions used in our in vitro system were correct. The combination of HCV C and canonical activators resulted in enhanced transcription, even if at level not so high as with PMA and ionomycin alone; pre-treatment with CsA again inhibited the inducing effect. PHA, alone or combined with PMA, determined the transcriptional activation of IL-2 and HNP. On the whole, HNP and IL-2 transcription seem to vary accordingly, suggesting that a common mechanism, possibly NFAT-mediated, might control both genes. However, addition of HCV C to PMA and ionomycin did not result in a further enhance of transcription, which rather decreased when compared to PBMC treated only with PMA and ionomycin: this might be due to a perturbing effect of the combination of more drugs, possibly even a feed-back mechanism of control; on the other hand, HCV C might trigger a transcriptional cascade, only partly overlapping with that induced by PMA and ionomycin. In agreement with this latter hypothesis, we found that a general inducer of T cells activation and proliferation, such as PHA, can trigger HNP transcription. These observations induce us to speculate that rather a complex mechanism, probably not only NFAT-mediated, might control HNP genes. Another interesting feature is that IL-2 gene is much more strongly modulated in comparison to HNP. IL-2 transcriptional level was found extremely low in unstimulated cells, as it could be detected only in a very few naive PBMC samples (from both healthy and diseased individuals) and, mainly, in those obtained from acute C and B hepatitis patients. The transcriptional level was greatly enhanced, and thus easily detected, after appropriate cell treatment. This finding corresponds to data already available in the scientific literature: IL-2, in fact, seems to be produced at appreciable levels only in particular physiological conditions and especially in acute inflammation states (Hartel et al., 1999). 55 Selectivity of HCV C induction In order to understand the cellular response to HCV C treatment, the effect on the expression of LL37 and IL-15 genes was investigated, at the transcriptional level, and compared to the modulation already observed for HNP and IL-2. The expression of these genes was monitored by real-time RT-PCR in HCV-treated PBMC from both healthy individuals and acute C patients. The transcriptional levels observed in treated PBMC were normalized to those found in untreated PBMC. Data are shown in Fig. 4.23. Relative mRNA level 8 7 6 5 4 3 2 1 0 HNP Fig. 4.23. IL-2 IL-15 LL37 Relative transcriptional levels of HNP, IL-2, IL-15 and LL37 genes in HCV Ctreated PBMC. The greatest modulation was observed for the two cytokine genes; to a minor extent, also HNP transcription was enhanced, whereas no appreciable variation was detected in LL37 expression. These data, deriving from in vitro experiments, seem to agree with what was already assessed when studying the expression profile in PBMC freshly isolated from HCV patients: transcription of HNP and IL-15 was found significantly higher, while non-relevant variation was noticed for LL37. 4.8. HNP expression profile in HCV patients under antiviral therapy HNP expression was studied in PBMC from chronic C patients undergoing a pharmacological treatment based on interferon-α (INF-α), an antiviral drug that inhibits viral replication and stimulates the immune response of the host. The profile of expression was monitored over a period of time ranging from 2 to 9 months after treatment beginning. Data obtained by real time qRT-PCR are reported in Fig. 4.24. With the exception of patient 2, we can notice a general trend toward a clear drop of α-defensin transcription, especially in the first two-three months of therapy. Moreover, the expression levels tend to stabilize in the range of values observed for the healthy controls. 56 H N P m R N A le v e l 75 p1 p2 60 p3 p4 45 p5 p6 30 15 Healthy range 0 -1 0 1 2 3 4 5 6 7 8 9 10 months Fig. 4.24. HNP transcriptional profile in six chronic HCV patients (p1→ →p6) undergoing therapy. HNP mRNA level (HNP/GAPDH) 150 120 90 60 30 0 before therapy Fig. 4.25. 2 m onths after therapy HNP transcriptional modulation in an HCC patient under INFα/ribavirin-combined therapy. A similar result was found in a patient with HCC, undergoing a more severe therapy, based on INF-α combined with ribavirin (Fig. 4.25). The HNP transcriptional level was measured before and after treatment. These profiles of expression strengthen the hypothesis that α-defensins are somehow associated to HCV pathogenesis, and that their expression may be stimulated just by the presence of the virus. In an attempt to explain such a trend, we can therefore assume that INFα, by preventing HCV replication, might indirectly contribute to limit α-defensins expression. In perspective, it would be worth evaluating the expression profile of HNP genes in relation to the type of response that the patients develop as a consequence of the therapy. 57 5 CONCLUSIONS The studies undertaken in the present thesis allow us to draw some relevant conclusions. i) A significant increase of HNP expression in PBMC from HCV-infected individuals was demonstrated. This was verified by multiple approaches based on real time qRT-PCR, ELISA and biological activity analyses. The HNP over-expression might be either a direct or indirect effect of the presence of the virus: in the former case, over-expression could be mediated by HCV core protein, which was found to stimulate HNP genes transcription in vitro. In the latter, the phenomenon could be cytokine-mediated, for instance IL-15mediated, as we found this cytokine being over-expressed in HCV-patients, as well as being able to enhance HNP genes transcription in vitro. Possibly, both types of mechanisms are involved in this up-regulation: a direct effect of the virus is particularly likely during the stages of high viraemia, such as during acute infection; conversely, in the phase of chronicization, cytokines and other immune factors may contribute to maintain high HNP levels. ii) The elevated levels of α-defensins, their correlation with liver fibrosis progression, and the pattern of expression in patients under therapy, all suggest the involvement of these antimicrobial peptides in HCV pathogenesis. It would be interesting to know the kind of role played by HNPs in this context. HNPs might be protective and thus have a positive function for the host organism, giving that they are antimicrobial and antiviral, though a direct activity against HCV has not been documented yet. At the same time, HNPs might be even disadvantageous for the host: at physiologic concentrations, in fact, α-defensins promote tissue healing by triggering fibroblast proliferation and deposition of extracellular matrix (Murphy et al., 1993; Oono et al., 2002). Therefore, it is possible that, at pathological concentrations, that is when over-expressed, as in this case, α-defensins might increase fibrogenesis and enhance substitution of functional hepatic tissue with fibrotic non-functional tissue. Whatever the function of α-defensins in HCV pathogenesis, their involvement raises particular interest from a clinical point of view: firstly, it represents a further little step in understanding the pathogenesis of this infection, which is still rather obscure, under several aspects; secondly, it opens up perspectives for new, complementary diagnostic tools and possible therapeutic targets, in a disease for which no plainly successful therapy has been found, yet. iii) Our data support the participation of NFAT to the transcriptional regulation of HNP genes. NFAT is a major factor controlling several immune-related genes, including genes encoding AMPs; however, its involvement in the control of α-defensin genes is a novel aspect, since, so far, the regulation of their expression has been mainly studied in relation to cell differentiation and maturation processes. The position of the putative NFAT core site in the promoter of HNP genes is peculiar, in that it is overlapping with an Ets-like regulatory element, which was demonstrated to be essential for α-defensin expression during myeloid blood cell differentiation (Ma et al., 1998). However, adjacent and/or overlapping Ets and NFκB/NFAT binding sites have been identified in the promoters of several inducible lymphoid genes, suggesting that interaction between members of these families of transcription factors might be an evolutionary conserved mechanism for regulating immune-related genes (Bassuk et al., 1997). We can therefore speculate that such a cooperative mechanism might operate also for HNP genes. It is noteworthy that in the promoter of the gene encoding the chemokine CCL5/RANTES there is a potential NFAT site: interestingly, also this gene is over58 expressed in chronic hepatitis C, in such a way that positively correlates with the severity of hepatic inflammation (Apolinario et al., 2002; Melchiorsen et al., 2003). The overexpression of CCL5 was detected in hepatic cells, which represent the main target of the virus; it is plausible that, if HCV promotes HNP transcription via NFAT, we should expect a more considerable effect on the dysregulation of these genes, just at the hepatic level, that is in infiltrating T-lymphocytes, as well as hepatocytes. iv) Our study confirms the immunomodulatory properties of the HCV core protein: in this specific case, HCV C was shown to function - and possibly trigger NFAT pathway - even exogenously with respect to the affected cells. This is of particular concern, as this viral protein may be released in the blood stream of infected patients (Kanto et al., 1995; Sabile et al., 1999); consequently, by taking advantage of this strategic location, HCV C could modulate gene expression even in nondirectly infected-cells, and also in tissues and organs which are distant from the primary site of infection. 59 6 ACKNOWLEDGMENTS I wish to thank my tutor, Prof. Donatella Barra, whose extraordinary human and professional qualities I could appreciate during the years I have spent in the Department of Biochemical Sciences, University of Rome La Sapienza. I am grateful to Prof. Maurizio Simmaco, for his constant and contagious enthusiasm and for giving me the opportunity to work in a brand new laboratory, in a stimulating environment. I thank Dott. Maria Luisa Mangoni, for the technical and moral support, and for her friendly and precious collaboration. I thank the Coordinator Prof. Paolo Sarti and the teaching body of the Ph.D course, for their contribution to my scientific formation. I thank Prof. Antonio Aceti and his collaborators, especially Dott. Barbara Zechini, for contributing to the realization of this study. A big thank goes to all my lab mates, for their help and for having created a nice and enjoyable atmosphere, which has been so important to me, particularly during my “rainy days”. Finally, I thank my family, my parents and Peppe, for their encouragement and sweet patience: to them I dedicate this thesis. 60 7 REFERENCES Agawa Y, Lee S, Ono S, Aoyagi H, Ohno M , Taniguchi T, Anzai K, and Kirino Y (1991) Interaction with phospholipids bilayers, ion channel formation, and antimicrobial activity of basic amphipatic α-helical model peptides of various chain length. J. Biol. Chem. 266: 20218-22 Agerberth B, Charo G, Werr J, Olsson B, Idali F, Lindbom L, Kiessling R, Jörnvall H, Wigzell H, and Gudmundsson GH (2000) The human antimicrobial and chemotactic peptides LL-37 and α-defensins are expressed by specific lymphocyte and monocyte populations. Blood 96: 3086-3093 Agerberth B, Gunne H, Odelberg J, Kogner P, Boman HG, and Gudmunsson GH (1995) FALL39, a putative human peptide antibiotic, is cysteine-free and expressed in bone marrow and testis. Proc. Natl. Acad. Sci. USA 92: 195-199 Agerberth B, Lee JY, Bergman T, Carlquist M, Boman HG, Mutt V, and Jornvall H (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 Apolinario A, Majano PL, Alvarez-Perez E, Saez A, Lozano C, Vargas J, and Garcia-Monzon (2002) Increased expression of T cell chemokines and their receptors in chronic hepatitis C: relationship with the histological activity of liver disease. Am. J. Gastroenterol. 97: 2861-2870 Bals R and Wilson JM (2003) Cathelicidins- a family of multifunctional antimicrobial peptides. Cell. Mol. Life Sci. 60: 711-720 Bassuk AG, Anadappa RT, and Leiden JM (1997) Physical interactions between Ets and NFκB/NFAT proteins play an important role in the cooperative activation of the human immunodeficiency virus enhancer in T cells. J. Virol. 71: 3563-3573 Beals CR, Sheridan CM, Turck CW, Gradner P, and Crabtree GR (1997) Nuclear export of NFATc enhanced by glycogen synthase kinase-3. Science 275: 1930-1933 Bergqvist A and Rice CM (2001) Transcriptional activation of the interleukin-2 promoter by hepatitis C virus core protein. J. Virol. 75: 772-781 Boman HG (1995) Antimicrobial peptides and their role in innate immunity. Annu. Rev. Immunol. 13: 61-92 Boman HG (1996) Peptide antibiotics: holy or heretic grails of innate immunity? Scand. J. Immunol. 43: 475-482 Boman HG (1998) Gene-encoded peptide antibiotics and the concept of innate immunity: an update review. Scand. J. Immunol. 48: 12-25 Boman HG (2003) Antibacterial peptides: basic facts and emerging concepts. J. Intern. Med. 254: 197-215 Boman HG and Hultmark D (1987) Cell free immunity of insects. Annu. Rev. Microbiol. 41: 103126 Brokaert WF, Terras FR, Cammue BP, and Osborn RW (1995) Plant defensins: novel antimicrobial peptides as components of the host defence system. Plant Physiol. 108: 1353-1358 Bukh J, Purcell RH, and Miller RH (1994) Sequence analysis of the core gene of 14 hepatitis C virus genotypes. Proc. Natl. Acad. Sci. USA 91: 8239-8243 61 Bulet P, Dimarcq JL, Hetru C, Lagueux M, Charlet M, Hegy G, VanDorsselaer A, and Hoffmann JA (1993) A novel inducible antibacterial peptide of Drosophila carries an Oglycosylated substitution. J. Biol. Chem. 268: 14893-14897 Bustin S A (2002) Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems. J. Endocrinol. 29: 23-39 Casteels P, Ampe C, Jacobs F, Vaeck M, and Tempst P (1989) Apidaecins: antibacterial peptides from honeybee. EMBO J. 8: 2387-2391 Cerny A and Chisari FV (1999) Pathogenesis of chronic hepatitis C: immunological features of hepatic injury and viral persistence. Hepathology 30: 595-601 Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, and Houghton M (1989) Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science 244: 359-362 Christensen B, Fink J, Merrifield RB, and Mauzerall D (1988) Channel forming properties of cecropins and related model compounds incorporated into planar lipid membranes. Proc. Natl. Acad. Sci. USA 85: 5072-5076 Cole AM, and Lehrer RI (2002) Minidefensins: antimicrobial peptides with activity against HIV-1. Curr. Pharm. Des. 9: 1463-1473 Cooper S, Erickson AL, Adams EJ, Kansopon J, Weiner AJ, Chien DY, Houghton M, Parham P, and Walker CM (1999) Analysis of a successful immune response against hepatitis C virus. Immunity 10: 439-449 Corbett Research Group www.corbettresearch.com (2004) Real time amplification on the Rotor-Gene. Crabtree GR, and Clipstone, NA (1994) Signal transmission between the plasma membrane and the nucleus of T lymphocytes. Annu. Rev. Biochem. 63: 1045-1083 Daher KA, Selsted ME, and Lehrer RI (1986) Direct inactivation of viruses by human granulocytes defensins. J. Virol. 60: 1068-1074 Diamond G, Zasloff M, Eck H, Bressaur M, Maloy WL, and Bevins CL (1991) Tracheal antimicrobial peptide, a novel cysteine-rich peptide from mammalian tracheal mucosa: peptide isolation and cloning of a cDNA. Proc. Natl. Acad. Sci. USA 88: 3952-3956 Eisen-Vandervelde AL, Waggoner SN, Yao ZQ, Cale EM, Hahn CS, and Hahn YS (2004) Hepatitis C virus core selectively suppresses interleukin-12 synthesis in human macrophages by interfering with AP-1 activation. J. Biol. Chem. 279: 43479-43486 Fang XM, Shu Q, Chen QX, Book M, Sahl HG, Hoeft A, and Stiber F (2003) Differential expression of α- and β-defensins in human peripheral blood. Eur. J. Clin. Invest. 33: 82-87 Fearon DT, and Locksley RM (1996) The instructive role of innate immunity in the acquired immune response. Science 272: 50-54 Frank RW, Gennaro R, Schneider K, Przybylski M, and Romeo D (1990) Amino acid sequence of 2 proline-rich bactenecins-antimicrobial peptides of bovine neutrophils. J. Biol. Chem. 265: 18871-18874 Fried MW, Shiffman ML, Reddy R, Smith C, Marinos G, Goncales Jr FL, et al. (2002) Peg interferon alfa-2a plus ribavirin for chronic hepatitis C virus infection. N. Engl. J. Med. 347: 975-982 62 Gallo RL and Nizet V (2003) Endogenous production of antimicrobial peptides in innate immunity and human disease. Curr. Aller. Asth. Rep. 3: 402-409 Ganz T, Selsted ME, Szlarek D, Harwig SS, Daher K et al (1985) Defensins: natural peptide antibiotics of human neutrophils. J. Clin. Invest. 76: 1427-1435 (1985) Goldman MJ, Anderson GM, Stolzenberg ED, Kari UP, Zasloff M, and Wilson JM (1997) Human beta-defensin-1 is a salt sensitive antibiotic in lung that is inactivated in cystic fibrosis. Cell 88: 553-560 Gough M, Hancock REW, and Kelly NM (1996) Anti endotoxic potential of cationic peptides antimicrobials. Infect. Immun. 64: 4922-4927 Grakoui A, Wychowski C, Lin C, Feinstone SM, and Rice CM (1993)Expression and identification of hepatitis C virus polyprotein cleavage products. J. Virol. 67:1385-1395 Hancock REW (1997) Peptide antibiotics. Lancet 349: 418-422 Hancock REW and Lehrer R (1998) Cationic peptides: a new source of antibiotics. Trends Biotechnol. 16: 82-87 Hancock REW, Falla T and Brown MM (1995) Cationic bactericidal peptides. Adv. Microbial Physiol. 37: 135-75 Härtel C, Bein G, Kirchner H, and Klüter H (1999) A human whole-blood assay for analysis of Tcell function by quantification of cytokine mRNA. Scand. J. Immunol. 49: 649-654 Hill CP, Yee J, Selsted M, and Eisenberg D (1991) Crystal structure of defensin HNP-3, an amphiphilic dimmer: mechanism of membrane permeabilization. Science 251: 1481-1485 Hoffmann JA and Hetru C (1992) Insect defensins: inducible antibacterial peptides. Immunol. Today 13: 411-415 Horsley V, and Pavlath GK (2002) NFAT: ubiquitous regulator of cell differentiation and adaptation. J. Cell. Biol. 156: 771-774 Houghton M, Weiner AJ, Han J, Kuo G, Choo QL (1991) Molecular biology of the hepatitis C viruses: implications for diagnosis, development and control of viral disease. Hepathology 14: 381-388 Hultmark D, Engstrom A, Andersson K, Steiner H, Bennich H, Boman HG (1983) Insect immunity. Attacins, a family of antibacterial proteins from Hyalophora cecropia. EMBO J. 2: 571-6. Hyodo N, Nakamura I, and Imawari M (2004) Hepatitis B core antigen stimulates interleukin-10 secretion by both T cells and monocytes from peripheral blood of patients with chronic hepatitis B virus infection. Clin. Exp. Immunol. 135: 462-466 Ihi T, Nakazato M, Mukae H, and Matsukura S (1997) Elevated concentrations of human neutrophil peptides in plasma, blood, and body fluids from patients with infections. Clin. Infect. Disease 25: 1134-1140 Jin DY, Wang HL, Zhou Y, Chun A, V Kibler K, Hou YD, Kung H, and Jeang KT (2000) Hepatitis C virus core protein-induced loss of LZIP function correlates with cellular transformation. EMBO J. 19 (4): 729-740 63 Kagan BL, Selsted ME, Ganz T, and Lehrer RI (1990) Antimicrobial defensin peptides form voltage dependent ion-permeable channels in lipid bilayer membranes. Proc. Natl. Acad. Sci. USA 87: 210-214 Kakumu S, Okumura A, Ishikawa T, Yano M, Enomoto A, Nishimura H, Yoshioka K, and Yoshikai Y (1997) Serum levels of IL-10, IL-15 and soluble tumour necrosis factor-alpha (TNF-alpha) receptors in type C chronic liver disease. Clin. Exp. Immunol.109:458-463 Kanto T, Hayashi N, Takehara T, Hagiwara H, Mita E, Naito M, Kasahara A, Fusamoto H, and Kamada T (1995) Density analysis of hepatitis C virus particle population in the circulation of infected hosts: implications for virus neutralization and persistence. J. Hepatol. 4: 440-448 Kuo G, ChooQ-L, Alter HJ, Gitnik GL, Redeker AG, Purcell RH, Miyamura T, Dienstag JL, Alter MJ, Stevens CE, Tegmeier G, Bonino F, Colombo M, Lee WS, Kuo C, Berger K, Shuster JR, Overby LR, Bradley DW, and Houghton M (1989) An assay for circulating antibodies to a major etiologic virus of human non-A, non-B hepatitis. Science 244: 362-364 Large MK, Kittlesen DJ, and Hahn YS (1999) Suppression of host immune response by the core protein of hepatitis C virus: possible implications for hepatitis C virus persistence. J. Immunol. 162: 931-938 Lehrer, RI, Selsted ME, Szklarek D, and Fleischmann J (1983) Antibacterial activity of microbicidal cationic proteins 1 and 2, natural peptide antibiotics of rabbit lung macrophages. Infect. Immun. 42: 10-14 Lehrer RI (2004) Primate defensins. Nature Rev. Microbiol. 2: 727-738 Lichtestein A, Ganz T, Sested ME, and Lehrer RI (1986) In vitro tumor cells cytolysis mediated by peptide defensins of human and rabbit granulocytes. Blood 68: 1407-1410 Liu J, Sakane T, and Tsunematsu T (1992)The effects of FK-506 and cyclosporin A on the proliferation of PHA-stimulated T cells in response to IL-2, IL-4 or IL-6. Int. Arch. Allergy Immunol. 98 :293-8 Liu L, Roberts A, and Ganz T (2003) By IL-1 signaling, monocytes-derived cells dramatically enhance the epidermal antimicrobial response to lipopolysaccharide. J. Immunol. 170: 575-580 Liu L, Zhao C, Heng HHQ, and Ganz T (1997) The human β-defensin-1 and α-defensins are encoded by adjacent genes: two peptide families with differing disulfide topology share a common ancestry. Genomics 43: 316-320 Ma Y, Su Q, and Tempst P (1998) Differentiation stimulated activity binds an ETS-like, essential regulatory element in the human promieleocytic defensin promoter. J. Biol. Chem. 273: 8727 Mackewicz CE, Yuan J, Tran P, Diaz L, Mack E, Selsted ME, and Levy JA. (2003) α-Defensins can have anti-HIV activity but are not CD8 cell anti-HIV factors. AIDS 17: F23-F32 Mallow EB, Harris A, Salzman N, Russell JP, DeBerardinis R, Ruchelli E, and Bevins CL (1996) Human enteric defensins. J. Biol. Chem. 271: 4038-4045 Manger B, Hardy KJ, Weiss A, and Stobo JD (1986) Differential effect of cyclosporin A on activation signaling in human T cell lines. Clin. Invest. 77: 1501-6. Manigold T, Bocker U, Chen J, Gundt J, Traber P, V. Singer M, and Rossol S (2003) Hepatitis B core antigen is a potent inductor of interleukin-18 in peripheral blood mononuclear cells of healthy controls and patients with hepatitis B infection. J. Med. Virol. 71:31-40 64 Marusawa H, Hijikata M, Chiba T, and Shimotohno K (1999) Hepatitis C virus core protein inhibits Fas- and tunor necrosis factor alpha-mediated apoptosis via NF-κB activation. J. Virol. 73:4713-4720 Matsuzaki K, Harada M, Handa T, Funakoshi S, Fujii N, Yajima H, and Miyajima K (1989) Magainin1-induced leakage of entrapped calcein out of negatively charged lipid vesicles. Biochim. Biophys. Acta 981: 260-266 Mattila PS, Ullman KS, Fiering S, Emmel EA, McCutcheon M, Crabtree GR, and Herzenberg LA (1990) The actions of cyclosporin A and FK506 suggest a novel step in the activation of T lymphocytes EMBO J. 9: 4425-33 Mc Michael AJ and Phillips RE (1997) Escape of human immunodeficiency virus from immune control. Ann. Rev. Immunol. 15: 271-296 Medzhitov R, and Janeway CA Jr (1997) Innate immunity: the virtues of a nonclonal system of recognition. Cell 91: 295-298 Melchiorsen J, Sørensen LN, and Paludan S (2003) Expression and function of chemokines during viral infections: from molecular mechanisms to in vivo function. J. Leuk. Biol. 74: 331-343 Moore KS, Bevins CL, Brasseur MM, Tomassini N, Turner K, Eck H, and Zasloff M (1991) Antimicrobial peptides in the stomach of Xenopus laevis. J. Biol. Chem. 266: 19851-19857 Morikawa N, Hagiwara K, and Nakajima T (1992) Brevinin -1 and -2, unique antimicrobial peptides from the skin of the frog Rana brevipoda porsa. Biochem. Biophys. Res. Commun. 189: 184-190 Morrison G, Kilanowski F, Davidson D, and Dorin J (2002) Characterization of the mouse beta defensin1, Defb1, mutant mouse model. Infect. Immun.70: 3053-3060 Murphy CJ, Foster BA, Mannis MJ, Selsted ME, and Reid TW (1993) Defensins are mitogenic for epithelial cells and fibroblasts. J. Cell. Physiol. 155: 408-413 Nakashima H, Yamamoto N, Masuda M, and Fujii N (1993)Defensins inhibit HIV replication in vitro. AIDS 7: 1129 Nizet V, OhtakeT, Lauth X, Trowbridge J, Rudisill J, Dorschner RA, Pestonjamasp V, Piraino J, Huttner K, and Gallo RL. (2001) Innate antimicrobial peptides protects the skin from invasive bacterial infection. Nature 414: 454-457 Northrop JP, Ullman KS, and Crabtree GR (1993) Characterization of the nuclear and cytoplasmic components of the lymphoid-specific nuclear factor of activated T cells (NF-AT) complex. J. Biol. Chem. 268, 2917-23 Obata-Onai A, Hashimoto S, Onai N, Kurachi M, Nagai S, Shizuno K, Nagahata T, and Mathushima K (2002) Comprehensive gene expression analysis of human NK cells and CD8(+) T lymphocytes. Int. Immunol. 14: 1085-1098 Oono T, Shirafuji Y, HuhWK, Akiyama H, and Iwatsuki K (2002) Effects of human neutrophil peptide-1 on the expression of interstitial collagenase and type I collagen in human dermal fibroblasts. Arch. Dermatol. Res. 294: 185-189 Ouelette AJ and Selsted ME (1996) Paneth cell defensins: endogenous components of intestinal host defense. FASEB J. 10: 1280-1289 Pavio N (2003) The hepatitis C virus persistence: how to evade the immune system? J. Biosci. 28: 287-304. 65 Purcel RH (1994) Hepatitis viruses: changing patterns of human disease. Proc. Natl. Acad. Sci. USA 91, 2401-2406 Pütsep K, Carlsson G, Boman HG, and Anderson M (2002) Deficiency of antibacterial peptides in patients with morbus Kostmann: an observation study. Lancet 360:1144-1149 Rao A, Luo C, and Hogan PG (1997) Transcription factors of the NFAT family- Regulation and function. Annu. Rev. Immunol. 15: 707-747 Ray RB, Lagging LM, Meyer K, and Ray R (1996) Hepatitis C virus core protein cooperates with ras and transforms primary rat embryo fibroblasts to tumorigenic phenotype. J. Virol. 70: 44384443 Romagnani S (1992) Induction of TH1 and TH2 responses: a key role for the natural immune response? Immunol. Today 13: 379-381 Sabile A, Perlemuter G, Bono F, Kohara K, Demaugre F, Kohara M, Matsuura Y, Miyamura T, Brechot C, and Barba G (1999) Hepatitis C virus core protein binds to apolipoprotein AII and its secretion is modulated by fibrates. Hepathology 30: 1064-1076 Salzman NH, Ghosh D, Huttner K, Paterson Y, and Bevins CL. (2003) Protection against enteric salmonellosis in transgenic mice expressing a human intestinal defensin. Nature 422: 522-526 Schutte BC, Mitros J, Bartlett JA, Walters JD, Jia HP, Welsh MJ, Csavant T, and McCray PB Jr (2002) Discovery of five conserved β-defensin gene clusters using a computational search strategy. Proc. Natl. Acad. Sci. USA 99: 2129-2133 Selsted ME, Brown DM, De Lange RJ, and Leherer RI (1983) Primary structures of MCP-1 and MCP-2, natural peptide antibiotics of rabbit lung macrophages. J. Biol. Chem. 258: 1448514489 Selsted ME, Harwig SS, Ganz T, Schilling JW, and Lehrer RI (1985) Primary structure of three human neutrophil defensins. J. Clin. Invest. 76: 1436-1439 Selsted ME, Novotny MJ, Morris WL, Tang YQ, Smith W, and Cullor JS (1992) Indolicin, a novel bactericidal tridecapeptide amide from neutrophils. J. Biol. Chem. 267: 4292-4295 Selsted ME, Tang YQ, Morris WL; McGuire PA, Novotny MJ, Smith W, Henschen AH, and Cullor JS (1993) Purification, primary structure and antibacterial activities of β-defensins, a new family of antimicrobial peptides from bovine neutrophils. J. Biol. Chem. 268: 6641-6648 Shai Y (2002) Mode of action of membrane-active antimicrobial peptides. Biopolymers 66: 236-248 Shaw JP, Utz PJ, Durand DB, Toole JJ, Emmel EA, and Crabtree GR (1988) Identification of a putative regulator of early T cell activation genes. Science 241: 202-5 Simmaco M, Barra D, Chiarini F, Noviello L, Melchiorri P, Kreil G, and Richter K (1991) A family of bombinin-related peptides from the skin of Bombina variegata. Eur. J. Biochem. 199: 217-222 Simmaco M, Mangoni ML, Boman A, Barra D, Boman HG (1998) Experimental infections of Rana esculenta with Aeromonas hydrophila: a molecular mechanism for the control of the normal flora. Scand. J. Immunol. 48: 357-363. Simmaco M, Mignogna G, Barra D, and Bossa F (1994) Antimicrobial peptides from skin secretion of Rana esculenta. Molecular cloning of cDNA encoding esculentin and isolation of new active peptides. J. Biol. Chem. 269: 11956-11961 66 Simmaco M, Mignogna G, Canofeni S, Miele R, Mangoni ML, and Barra D (1996) Temporins, antimicrobial peptides from the European red frog Rana temporaria. Eur. J. Biochem. 242: 788792 Tang YQ, Yuan J, Ösapay G, Ösapay K, Tran D, Miller C, Ouellette AJ, and Selsted ME (1999) A cyclic antimicrobial peptide produced in primate leukocytes by the ligation of two truncated α-defensins. Science 286: 498-502 Timmerman LA, Clipstone NA, Ho SN, Northrop JP, and Crabtree GR (1996) Rapid shuttling of NF-AT in discrimination of Ca2+ signals and immunosuppression. Nature 383, 837-40 Trabattoni D, Caputo SL, Maffeis G, Vichi F, Biasin M, Pierotti P, Fasano F, Saresella M, Franchini M, Ferrante P, Mazzotta F, Clerici M. (2004) Human α-defensin in HIV-exposed but uninfected individuals. J. Acquir. Immune. Defic. Syndr. 35: 455-463 Tsuchinara K, Hijikata M, Fukuda K, Kukori T, Yamamoto N, and Shimotohno K (1999) Hepatitis C virus core protein regulates cell growth and signal transduction pathway transmitting growth stimuli. Virology 258:100-107 Tsutsumi-Ishii Y, Hasebe T, and Nagaoka I (2000) Role of CCAAT/Enhancer- binding protein site in transcription of human neutrophil peptide- 1 and -3 defensin genes. J Immunol. 164: 32643273 van den Berg RH, Faber-Krol MC, van Wetering S, Hiemstra PS, and Daha MR (1998) Inhibition of activation of classical pathway of complement by human neutrophil defensins. Blood 92: 3898-3903 Waldmann TA and Tagaya Y (1999)The multifaceted regulation of interleukin-15 expression and the role of this cytokine in NK-cell differentiation and host response to intracellular pathogens. Annu. Rev. Immunol. 17: 19-49 Wang W, Cole AM, Hong T, Waring AJ, Lehrer RI (2003) Retrocyclin, an antiretroviral θdefensin, is a lectin. J. Immunol. 170: 4708-4716 Wang X, Zhang Z, Louboutin J, Moser C, Weiner DJ, and Wilson JM (2003) Airway epithelia regulate expression of human β-defensin 2 through Toll-like receptor 2. FASEB J. 17: 17271729 Woodrow MA, Rayter S, Downward J, and Cantrell DA (1993) p21ras function is important for T cell antigen receptor and protein kinase C regulation of nuclear factor of activated T cells. J. Immunol. 150: 3853-61 Yang D, Biragyn A, Kwak LW, and Oppenheim JJ (2002) Mammalian defensins in immunity: more than just microbicidal. Trends Immunol. 23: 291-296. Yang D, Chen Q, Chertov O, and Oppenheim JJ (2000) Human neutrophil defensins selectively chemoattract naive T and immature dendritic cells. J. Leuk. Biol. 68: 9-14 Yang D, Chertov O, Bykovskaia SN , Chen Q, Buffo MJ, Slogan J, Anderson M, Shröder JM, Wang JM, Howard OMZ, and Oppenheim JJ (1999) β-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6. Science 286:525-527 Zanetti M, Gennaro R, and Romeo D (1995) Cathelicidins: a novel protein family with a common proregion and a variable C-terminal antimicrobial domain. FEBS Lett. 374: 1-5 Zasloff M (1987) Magainins, a class of antimicrobial peptides from Xenopus skin: isolation, characterization of two active forms, and partial cDNA sequence of precursor. Proc. Natl. Acad. Sci. USA 84: 5449-5453 67 Zhang L, Yu W, He T, Yu J, Caffrey RE, Dalmasso EA, Fu S, Pham T, Mei J, Ho JJ, Zhang W, Lopez P, and Ho DD (2002) Contribution of human α-defensin-1,-2, and -3 to the anti-HIV-1 activity of CD8 antiviral factor. Science 298: 995-1000 Zhu Q, Bateman A, Singh A, and Solomon S (1989) Isolation and biological activity of corticostatic peptides (anti-ACTH). Endocr. Res. 15: 129-149 68 8 PUBLICATIONS Mangoni ML, Fiocco D, Mignogna G, Barra D, and Simmaco M. (2003) Functional characterization of the 1-18 fragment of esculentin -1b, an antimicrobial peptide from Rana esculenta. Peptides, 24, 1771-1777 Simmaco M, Mangoni ML, Miele R, Borro M, Fiocco D, and Barra D. (2003) Defence peptides in the amphibian immune system. Bacterial, Plant & Animal Toxins, 155-167. Carzaniga R, Fiocco D, Bowyer P, and O’Connell RJ. (2002) Localization of melanin in conidia of Alternaria alternata using phage display antibodies. Mol. Plant-Microbe Interactions, 15, 216-224 Miele R, Borro M, Fiocco D, Barra D, and Simmaco M. (2000) Sequence of a gene from Bombina orientalis coding for the antimicrobial peptide BLP-7. Peptides, 21, 16811686 69