Inducible expression of an antimicrobial peptide of the ... - CiteSeerX

2 downloads 0 Views 353KB Size Report
cathelicidin family of antimicrobial peptides and is stored in the cytoplasmic granules of bovine neu- trophils. We investigated the expression of this gene in ...
Inducible expression of an antimicrobial peptide of the innate immunity in polymorphonuclear leukocytes Linda Tomasinsig,*,†, Marco Scocchi,† Carla Di Loreto,‡ Daria Artico,‡ and Margherita Zanetti*,† Dipartimento di *Scienze e Tecnologie Biomediche and ‡Ricerche Mediche e Morfologiche, Universita` di Udine, Italy; and †Laboratorio Nazionale CIB, AREA Science Park, Trieste, Italy

Abstract: Epithelia- and leukocyte-associated antimicrobial peptides provide immediate protection against microbial infections by rapidly inactivating potential pathogens. Bac5 is a member of the cathelicidin family of antimicrobial peptides and is stored in the cytoplasmic granules of bovine neutrophils. We investigated the expression of this gene in airway and intestine, and although the gene was not found to be locally expressed in these tissues, a strong Bac5 induction signal was detected by in situ hybridization in neutrophils infiltrating infected lung, consistent with expression of this gene in activated neutrophils. The Bac5 gene was also induced in bovine peripheral neutrophils stimulated with Escherichia coli or purified lipopolysaccharide (LPS) but not in other blood cells and in resting neutrophils. The levels of Bac5 mRNA increased at 12–24 h post-stimulation, and a dosedependent increase in Bac5 expression was determined in the presence of increasing amounts of LPS. A metabolically labeled product with a molecular weight compatible with that of proBac5 was immunoprecipitated from cell-free media of stimulated neutrophils, suggesting that the newly synthesized polypeptide is released extracellularly. Collectively, these results provide the first evidence that fully differentiated neutrophils are capable of de novo synthesis and secretion of a granule-associated antimicrobial peptide. J. Leukoc. Biol. 72: 1003–1010; 2002. Key Words: Bac5 䡠 neutrophil 䡠 gene expression

INTRODUCTION The antimicrobial peptides of innate immunity are a front-line, host-defence response to microbial invasion [1– 4]. In mammals, these peptides are released by epithelial cells at the interface with the external environment and by activated leukocytes recruited at sites of inflammation. Their broad and rapid microbicidal effect on release may be critically important to clear tissues from pathogens and to prevent the onset of infection [5–7]. A variety of mammalian antimicrobial peptides belong to the cathelicidin protein family [8 –11]. Bac5 was the first reported

member of this family [8, 12]. Its sequence was deduced from bovine bone marrow (BM) cells mRNAs [12], and early studies of this molecule contributed substantially to elucidate the biosynthesis and maturation process of cathelicidins in myeloid-derived cells [13, 14]. Bac5 is actively synthesized during myelopoiesis but not in mature resting neutrophils [13] and is stored in the cytoplasmic neutrophil granules as a propeptide (proBac5), in which the C-terminal antimicrobial domain is joined to a conserved, cathelin-like propiece [13]. The antimicrobial domain displays a proline-rich cationic sequence of 43 amino acid residues and is liberated from the propeptide coincident with neutrophil degranulation [14, 15]. It is remarkable that whereas the propiece is fairly conserved in all cathelicidin members, the peptide regions are highly heterogeneous and in addition to proline-rich peptides, may include ␣-helical, tryptophan-rich, and disulfide-bridged peptides displaying distinct spectra of antimicrobial activity [8 –11]. All known cathelicidins were originally identified in myeloid-derived cells. However, expression studies of the human LL-37 [16] have indicated that this gene is also expressed in mucosal epithelia and in other nonmyeloid cells [5, 17–19]. In addition, the human LL-37 [16] and the murine cathelinrelated antimicrobial peptide (CRAMP) [20] cathelicidin genes are up-regulated in skin in response to inflammatory and infectious stimuli [6, 21]. These studies thus suggest a more widespread pattern of cathelicidin expression than previously recognized. To gain a better insight into the regulated expression of the Bac5 gene, we have extended here the analysis of the expression of this gene to normal and inflamed nonmyeloid tissues that are known to be sites of antimicrobial peptide synthesis. We find that the expression of the Bac5 gene is induced in neutrophils accumulated at inflammation sites in infected lung tissue, and we analyze the expression of this gene in primary cultures of peripheral neutrophils in response to bacteria and lipopolysaccharide (LPS), providing evidence for de novo synthesis of Bac5 in mature neutrophils.

Correspondence: Margherita Zanetti, Dept. Biomedical Sciences and Technology, University of Udine, P.le Kolbe 4, I-33100 Udine, Italy. E-mail: [email protected] Received June 28, 2002; revised August 3, 2002; accepted August 5, 2002.

Journal of Leukocyte Biology Volume 72, November 2002 1003

MATERIALS AND METHODS Tissue samples Specimens from tracheal, bronchial, and lung tissue and from jejunum, duodenum, ileum, and rectum were obtained from freshly killed, 12- to 24-monthold healthy cows. Samples from a minimum of two different animals were analyzed. Lung tissue from four animals with pneumonia was kindly provided by Dr. M. Galeotti from the Dip. Produzione Animale, Faculty of Veterinary Sciences, University of Udine (Italy). Samples were dissected, washed in phosphate-buffered saline (PBS), pH 7.4, fixed in PBS containing 4% paraformaldehyde, and embedded in paraffin wax. Samples from the same animals were set apart for RNA extraction, placed in RNase-free denaturing solution (25 mM sodium citrate, pH 7.0, 4 M guanidinium thiocyanate, 0.5% lauroylsarcosine, 0.1 M 2-mercaptoethanol), and immediately frozen in dry ice.

Cell isolation and stimulation BM cells were isolated from bovine ribs as described [13]. Polymorphonuclear leukocytes (PMN) and mononuclear cells (MNC) were purified from whole blood of freshly killed cows. After differential centrifugation, PMN were isolated by hypotonic lysis of the supernatant, whereas the MNC fraction was obtained from the interface buffy coat after lysis with isotonic NH4Cl of the contaminating erythrocytes [13]. As assessed by Wright staining, PMN preparations contained ⬎97% granulocytes (92% neutrophils and 5% other granulocytes, mainly eosinophils) and ⬍2% MNC. Promyelocytes and myelocytes were absent in all the neutrophil preparations used, and metamyelocytes and band cells ranged from 0 to 1.4% of the total cells. Less than 5% of granulocytes was present in the MNC preparations. Cell viability of both cell fractions exceeded 98% as determined by the trypan blue dye exclusion after overnight culture. PMN and MNC were plated at 5 ⫻ 106 cells/ml into 10-cm tissueculture Petri dishes and maintained in RPMI 1640 supplemented with 100 units/ml penicillin (Gibco-BRL, Grand Island, NY), 100 ␮g/ml streptomycin (Gibco-BRL), and 2 mM L-glutamine (Gibco-BRL) in the presence of 5% heat-inactivated fetal bovine serum (FBS; Sigma Chemical Co., St. Louis, MO) at 37°C in a humidified atmosphere with 5% CO2. Cells were stimulated by addition of serum-opsonized, heat-inactivated Escherichia coli to the culture medium at a 10:1 ratio bacteria-to-neutrophils or by purified LPS from E. coli serotype 026:B6 (Sigma Chemical Co.) at 10, 100, and 1000 ng/ml. When indicated, polymyxin B (PB) at 1 ␮g/ml was preincubated with LPS (100 ng/ml) for 30 min at room temperature before addition to the cell culture.

Reverse transcriptase-polymerase chain reaction (RT-PCR) and Southern blot analysis Total RNA was isolated from homogenized tissue and cell samples as described [22]. RT was performed by using 1 ␮g DNase-treated, total RNA primed with oligo(dT) adaptor primer (Amersham Biosciences, Little Chalfont, UK) in a final volume of 20 ␮l containing 0.5 mM dNTPs (Amersham Biosciences), 10 mM dithiothreitol, 40 units RNaseOUT inhibitor (GibcoBRL), and 200 units SUPERSCRIPT II RT (Gibco-BRL). The reaction was carried out at 42°C for 2 h. PCR amplification was performed in a total volume of 25 ␮l including 0.6 units Amplitaq Gold DNA polymerase (Perkin Elmer, Foster City, CA), 2.5 mM MgCl2, 0.2 mM dNTPs, 0.5 ␮M each sense and antisense primer pairs, and 5 ␮l 1:10 diluted RT sample. After 10 min at 94°C, 25 or 30 cycles were performed with the following program: 30 s at 94°C, 40 s at the specific annealing temperature, and 1 min at 72°C. PCR products were analyzed in 1.5% agarose gel, and their sizes were compared with a 100-bp DNA ladder (New England Biolabs, Beverly, MA). Primer sequences were designed to specifically amplify the transcript regions of bovine cathelicidins, interleukin-8 (IL-8), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). For some cathelicidins, a sense primer (5⬘-GTGGAATTCAGGGTGAAGGAGAC-3⬘) based on the conserved proregion of bovine cathelicidins and sequence-specific antisense primers were used as follows: Bac5: 5⬘CTCAGGGCAAAAGAGAT-3⬘; Bac7: 5⬘-AGTGCTAACCTTGATGTT-3⬘; indolicidin: 5⬘-TCTGAACAAATCAGACACTTA-3⬘; BMAP-27: 5⬘-CGGAATTCACCCCAAATGGAGTA-3⬘; and BMAP-28: 5⬘-AATTGGGCCATACTTCTTCC-3⬘. For the cyclic dodecapeptide amplification, the sense and antisense primers were, respectively: 5⬘-GATGATGAAGACCCAGACAG-3⬘ and 5⬘GACGAATTCGAAAACCCTTAGGACTC-3⬘; for BMAP-34: 5⬘-ACCGAATTCAGCTACAGGGAGGCCGT-3⬘ (sense) and 5⬘-ACCTGATCCTAAGGACT-

1004

Journal of Leukocyte Biology Volume 72, November 2002

TT-3⬘ (antisense); for IL-8: 5⬘-GTGTGAAGCTGCAGTTCTG-3⬘ (sense) and 5⬘-TTCTGCACCCACTTTTCC-3⬘ (antisense); and for GAPDH: 5⬘-CATGCCATCACTGCCACCC-3⬘ (sense) and 5⬘-ACCTGGTCCTCAGTGTAGC-3⬘ (antisense). Bovine ␤-defensin (BBD)-1S and BBD-2A primer sequences were designed to amplify a 163-bp-long region that is highly conserved among ␤-defensins as reported [23]. The identity of amplified products was confirmed by DNA sequencing with the BigDye Terminator kit (PE Applied Biosystems, Foster City, CA). For Southern analysis, RT-PCR amplified fragments run on a 1.5% agarose gel were blotted onto GeneScreen Plus nylon membrane (NEN Life Sciences, Boston, MA). Filters were prehybridized 1 h at 50°C in 0.5 M NaH2PO4, pH 7.2, containing 5% sodium dodecyl sulfate (SDS), 1 mM EDTA, and 100 ␮g/ml denatured salmon sperm DNA. The hybridization was carried out overnight at 50°C using a [32P] end-labeled probe corresponding to nt 429– 449 of Bac5 cDNA [12]. The membranes were washed twice for 5 min with 0.1% SDS in 2 ⫻ saline sodium citrate (SSC; 30 mM tri-sodium citrate, pH 7.0, 0.3 M NaCl) and twice with 0.1% SDS in 0.1 ⫻ SSC at 50°C for 15 min and were then rinsed in 2 ⫻ SSC and exposed to X-ray film (Kodak).

Northern blot analysis Total RNA (7 ␮g) was size-separated on a 1% denaturing agarose gel, denaturated by incubation in 8 mM NaOH for 20 min, and then transferred to a GeneScreen Plus nylon membrane (NEN Life Sciences) in 3 M NaCl, 8 mM NaOH. The filter was prehybridized in ULTRAhyb solution (Ambion, Austin, TX) for 30 min at 42°C and then hybridized for 6 –24 h at the same temperature with a [32P]-labeled fragment corresponding to 3⬘ ends Bac5 cDNA coding for the antimicrobial domain [12], to IL-8, and to ␤-actin cDNAs. After hybridization, the membranes were washed twice for 5 min with 0.1% SDS in 2 ⫻ SSC and twice for 15 min with 0.1% SDS in 0.1 ⫻ SSC at 42°C, rinsed in 2 ⫻ SSC, and then exposed to X-ray film (Kodak).

Preparation of RNA probes and in situ hybridization Digoxigenin (DIG)-labeled RNA probes were based on the 3⬘ end of Bac5 cDNA [12] coding for the antimicrobial domain, cloned in pBluescript-II SK. After linearization of template DNA with the appropriate restriction enzyme, in vitro transcription was performed by using T7 or T3 RNA polymerase according to the instructions of the DIG RNA labeling kit (Boehringer Mannheim, Mannheim, Germany). Lung tissue (7-␮m-thick sections) was mounted on slides and deparaffinized at 60°C for 15 min. Following dehydration, sections were treated with 1 ␮g/ml proteinase K and fixed in 4% paraformaldehyde in PBS. Acetylation was performed by incubating slides twice for 5 min with 0.25% acetic anhydride in 0.1 M triethanolamine, pH 8.0. Sections were prehybridized for 90 min at 50°C in 20 mM Tris-HCl, pH 7.5, containing 1⫻ Denhardt’s solution, 300 mM NaCl, 100 mM dithiothreitol, 1 mM EDTA, 0.5 mg/ml salmon sperm DNA, 0.5 mg/ml polyadenylic acid (Boehringer Mannheim), and 50% formamide. Hybridization was performed overnight at 50°C in the hybridization solution plus 200 ng/ml DIG-labeled antisense or sense Bac5 probe. Slides were then washed three times for 20 min at 55°C with 50% formamide in 2 ⫻ SSC containing 0.1% Tween-20 (SSCT), 20 min at 55°C with 2 ⫻ SSCT, and if necessary, 20 min at 60°C in 0.2 ⫻ SSCT. Detection of the DIG-labeled probe was carried out by incubating sections with alkaline phosphatase-conjugated anti-DIG antibodies (Boehringer Mannheim) diluted 1:200 in PBS containing 0.1% Tween-20 and 10% FBS. Slides were rinsed four times in PBS with 0.1% Tween-20 for 10 min and incubated in a chromogen solution of nitro-blue tetrazolium/5-bromo4-cloro-3-indolyl-phosphate plus 1 mM Levamisole. Nuclear Fast Red (BDH Chemicals, Poole, UK) was used as counterstain. Control of specificity was achieved by hybridizing with the sense riboprobe and by treating sections with 20 ␮g/ml RNase A at 37°C for 1 h before hybridization with the antisense probe. Hematoxylin/eosin and Giemsa-staining reagents were from Sigma Chemical Co.

Metabolic labeling, immunoprecipitation, and Western blotting Peripheral blood neutrophils (2⫻107 cells/ml in 3-cm tissue-culture dishes) were maintained in RPMI 1640 in the presence of 5% heat-inactivated FBS and stimulated by addition of heat-inactivated E. coli opsonized with autologous serum at a 1:10 cells-to-bacteria ratio. The cell-free media were discarded

http://www.jleukbio.org

Fig. 1. (A) RT-PCR analysis of airway and intestine samples from healthy cows. (B) RT-PCR analysis of normal (Control) and infected cow lung. RT products were amplified using Bac5-, BBD-, or GAPDH-specific primer pairs, as indicated. The size of the amplified fragments is indicated on the left.

after 6 h and replaced with Cys-free RPMI 1640 containing 5% FBS, 125 ␮Ci/ml [35S]Cys (Amersham Biosciences), E. coli at 1:10 cell-to-bacteria ratio, and 1 mM phenylmethylsulfonyl fluoride. Incubations at 37°C were prolonged for further 6, 16, and 22 h. At each time point, cell-free media were collected, and cells were washed twice with ice-cold PBS. Cell pellets were solubilized at 4 ⫻ 107 cell equivalents/ml in lysis buffer (20 mM Tris-HCl, pH 8.2, 1.5% SDS) as described [13]. Bac5 was immunoprecipitated from cell extracts and cell-free media using affinity-purified Bac5 antibodies and protein A-Sepharose [13]. Protein recovered by immunoprecipitation was analyzed by a 15% polyacrylamide gel followed by Western blotting using Bac5-specific antibodies or fluorography [13]. Exposure times at ⫺80°C ranged from 15 to 21 days.

Bac5 was also found absent in airway and intestinal tissues by in situ hybridization and immunohistochemistry (not shown), further supporting that the gene is not constitutively expressed in these tissues. To find out if the Bac5 gene is induced under inflammatory conditions, lung tissue from cows with clinical and histopathological signs of acute pulmonary infection versus noninflamed lung tissue from healthy control animals was analyzed by RT-PCR (Fig. 1B) and by in situ hybridization (Fig. 2). A Bac5 product of the predicted size was amplified from lung apical tissue with marked histopathological signs of inflammation (Fig. 1B). Analysis of this tissue by in situ hybridization, using an antisense riboprobe corresponding to the peptide-coding region of Bac5, revealed a strong and diffuse Bac5-positive signal (Fig. 2, A and C, representative sample), whereas sections of the same tissue hybridized with a sense riboprobe (Fig. 2, B and D) or treated with RNase (not shown) resulted in a low, nonspecific background. Morphological examination of the signal-positive cells clearly localized Bac5 mRNA to neutrophils at sites of acute inflammation. Massive neutrophilic infiltration was observed in these areas by hematoxylin/eosin staining of adjacent tissue sections (Fig. 2E). Cell counts in hematoxylin/ eosin and Giemsa-stained sections adjacent to those used for in situ hybridization indicated over 94% morphologically mature

Sequencing and analysis of 5⬘ flanking region of the gene The sequence of the Bac5 gene was obtained from the genomic clone ␭Cl1. This was identified by screening of a ␭DASH II bovine genomic library with the conserved 5⬘ cDNA of the bovine cathelicidins [24]. Phage DNA was directly sequenced on both strands, and the 5⬘ flanking region of the gene was analyzed for the presence of transcription factors binding sites using the MatInspector software [25].

RESULTS Expression of the Bac5 gene in healthy and infected bovine tissue To determine if the Bac5 gene is expressed in mucosal epithelia, a collection of bovine tissues from the respiratory and the digestive tracts was analyzed by a semiquantitative RTPCR using Bac5-specific primer pairs, and bovine BM RNA was used as a positive control. As shown in a representative PCR (Fig. 1A), a Bac5 product of the expected size was amplified from BM mRNA, whereas intestinal, tracheal, bronchial, and lung tissues were consistently negative. Amplification of the housekeeping gene GAPDH served as a positive control of RT reaction. In addition, as a quality control for RNA integrity, the same tissues were also PCR-analyzed for the presence of BBD mRNA using primer pairs from regions that are highly conserved among myeloid and epithelial members of this antimicrobial peptide family [23]. BBD gene expression was also detected in the airways, in ileum, and rectum (Fig. 1A), at variance with Bac5 and consistent with published results indicating expression of ␤-defensins in these sites [26].

Fig. 2. In situ hybridization of inflamed bovine pulmonary tissue. Lung sections hybridized with antisense (A, C, F) and sense (B, D) Bac5 riboprobes. (E) Hematoxylin/eosin-stained serial section. Original magnification: A, B, E, F, 20⫻; C, D, 100⫻.

Tomasinsig et al. De novo synthesis of Bac5 in neutrophils

1005

Fig. 3. (A) Northern blot analysis of RNA from bovine PMN of a single adult animal. Peripheral blood neutrophils were cultured for up to 20 h in RPMI supplemented with 5% FBS in the absence (⫺) or the presence (⫹) of E. coli or 100 ng/ml E. coli LPS. Filters were sequentially hybridized under high-stringency conditions with Bac5 and IL-8 cDNA probes. Hybridization with ␤-actin probe served as a control of the amount and quality of the RNA present in each lane. (B) Induction of Bac5 mRNA in peripheral neutrophils after 20 h incubation with increasing amounts of E. coli LPS in the absence (⫺) or the presence (⫹) of 1 ␮g/ml PB.

neutrophils (e.g., 472 out of 486 neutrophilic cells counted), and the vast majority of the neutrophilic cells in these areas appeared to be Bac5 transcript-positive by the in situ analysis. All other cell types were Bac5-negative in the samples analyzed (see also bronchial tissue in Fig. 2F), indicating that the Bac5 gene is not locally expressed in this epithelium in response to infectious stimuli.

Induction of the Bac5 gene in bacteria- and LPSstimulated neutrophils The presence of Bac5 mRNA in lung-infiltrating neutrophils prompted us to investigate the potential of fully differentiated neutrophils to express this gene. Specifically, we evaluated the ability of heat-killed E. coli or of purified LPS, a major component of the gram-negative bacterial membrane, to induce the Bac5 gene in primary cultures of bovine peripheral neutrophils. Cells were incubated with each stimulus for various lengths of time in the presence of 5% FBS, and expression of Bac5 mRNA was investigated using RT-PCR and Northern blotting. The expression of the gene encoding IL-8, a chemotactic cytokine that is released by PMN in response to microbial challenge [27, 28], and of the housekeeping genes GAPDH and ␤-actin was monitored as positive controls, respectively, for PMN stimulation and RNA integrity and relative amounts. As shown in the Northern blot in Figure 3A, the Bac5 gene was not expressed at detectable levels in unstimulated neutrophils but was markedly induced after 20 h incubation of the cells with E. coli or LPS. The expression levels increased in a dose-dependent manner in the presence of increasing amounts of LPS and were found decreased when LPS was added in combination with polymyxin B (Fig. 3B), a molecule which is known to bind LPS and neutralize its effects. The IL-8 gene was also strongly induced after a 20-h incubation of neutrophils with E. coli or E. coli LPS (Fig. 3A), in good agreement with published results indicating expression of this gene in stimulated neutrophils [27, 28]. Unlike Bac5, a basal expression of the IL-8 gene was also detected in the absence of added stimuli (Fig. 3A). Peripheral blood MNC were examined for the presence of Bac5 mRNA as compared with neutrophils from the same 1006

Journal of Leukocyte Biology Volume 72, November 2002

blood samples by RT-PCR and Southern blot analysis of the amplified products (Fig. 4). This analysis failed to reveal a Bac5-corresponding transcript in freshly isolated MNC and after incubation for up to 20 h in the presence or absence of E. coli or of E. coli LPS. These results thus suggest that the Bac5 gene is not expressed in circulating lymphocytes and monocytes, also in the presence of infectious stimuli. A time-course for the induction of the Bac5 gene in neutrophils was determined using RT-PCR (Fig. 5). The identity of the amplified product as Bac5 in these experiments was confirmed by Southern blotting (Fig. 5A, top row) and sequencing of the PCR product. The expression of this gene was found to be substantially increased at 12, 16, and 20 h after addition of heat-killed E. coli (Fig. 5A) or LPS (Fig. 5B) and remained high at 24 h in at least five separate experiments with blood samples from different animals. Conversely, Bac5 mRNA was barely detected in neutrophils incubated in the absence of stimuli for up to 20 h. In addition to Bac5, the expression of the other bovine cathelicidin genes, i.e., Bac7, cyclic dodecapeptide, indolicidin, BMAP-27, BMAP-28, and BMAP-34 [9], was

Fig. 4. RT-PCR from total RNA isolated from bovine PMN and MNC and Southern analysis of amplified Bac5 products (top row). Cells were purified from a single adult animal and incubated for 20 h in RPMI supplemented with 5% FBS in the presence (⫹) or the absence (⫺) of heat-killed, opsonized E. coli or 100 ng/ml E. coli LPS. Bac5- or GAPDH-specific primer pairs were used for PCR amplification. Southern blot analysis was carried out using a Bac5-specific probe.

http://www.jleukbio.org

Fig. 5. RT-PCR from total RNA isolated from bovine PMN cultured for the indicated time lengths in RPMI supplemented with 5% FBS, in the absence (⫺) or the presence (⫹) of heat-killed, opsonized E. coli (A) or 100 ng/ml E. coli LPS (B). Primer pairs specific for Bac5, IL-8, or GAPDH cDNAs were used for PCR amplification. Southern analysis of amplified products (A, top row) was carried out using a Bac5-specific probe.

also analyzed using RT-PCR. Importantly, no products were amplified using primers based on the cDNA sequences of these cathelicidins in neutrophils stimulated for 20 h with E. coli or LPS and in unstimulated neutrophils (not shown), suggesting that among the bovine members of this family, Bac5 is selectively induced under these conditions. To obtain evidence for Bac5 expression at the protein level, peripheral neutrophils were incubated for 6 h in the presence or absence of E. coli and then metabolically labeled for 6, 16, and 22 h with [35S]Cys (four Cys residues are present in the cathelin-like, N-terminal region of proBac5). As exposure of neutrophils to bacteria is known to cause release of the granule contents in the extracellular medium, cell extracts and cellfree media were separately immunoprecipitated using Bac5specific antibodies. Products recovered by immunoprecipitation were analyzed by SDS-polyacrylamide gel electrophoresis, followed by Western blotting or by fluorography to reveal the unlabeled and the metabolically labeled (newly synthesized) proBac5. Western blotting of neutrophil lysates (Fig. 6A, lane

Fig. 6. (A) Western blot of products recovered by immunoprecipitation from cell-free media of neutrophils incubated for 6 h in the absence (a, d) or presence (b, e) of E. coli and metabolically labeled with [35S]Cys for 6 h (a, b) and 16 h (d, e). Western blotting of SDS-lysed neutrophils (c). Bac5-specific antibodies were used for immunoprecipitation and Western analyses. The apparent molecular weight of the heavy and light immunoglobulin G chains and of proBac5 is indicated on the left. (B) Fluorographic analysis of Bac5related products recovered by immunoprecipitation from cell-free media of neutrophils incubated for 6 h in the absence (a⬘, d⬘) or presence (b⬘, e⬘) of E. coli and biosynthetically labeled for 6 h (a⬘, b⬘) and 16 h (d⬘, e⬘).

c) served here as a control for mol wt of proBac5 (approximately 16.5 kDa). Western analysis of the immunoprecipitates allowed detection of a molecule with mol wt compatible with that of proBac5 in the cell-free media of neutrophils incubated with E. coli and labeled with [35S]Cys for 6 h (Fig. 6A, lane b), 16 h (Fig. 6A, lane e), and 22 h (not shown). Conversely, proBac5 was absent or weakly detected at corresponding incubation times in the media of unstimulated neutrophils (Fig. 6A, lanes a and d). Fluorographic analysis of the same immunoprecipitates revealed a radiolabeled proBac5 in cell-free media of neutrophils stimulated with E. coli and labeled with [35S]Cys for 16 h (Fig. 6B, lane e⬘) and 22 h (not shown) but not in those of stimulated and 6-h-labeled cells (Fig. 6B, lane b⬘). No labeled molecules were detected at corresponding times in the incubation media of unstimulated cells (Fig. 6B, lanes a⬘ and d⬘). The electrophoretic mobility of the labeled product (Fig. 6B, lane e⬘) corresponded to that of proBac5 in two experiments, and in two other experiments (not shown), radiolabeled molecules of approximately 10 and 11 kDa were detected, which may correspond to proteolytic proBac5 fragments. Western analysis of immunoprecipitates recovered from cell extracts revealed the presence of proBac5 in stimulated and unstimulated neutrophils, and decreased amounts were detected in stimulated versus unstimulated cells (results not shown), consistent with granule depletion upon neutrophil activation. Fluorographic analysis of the same immunoprecipitates failed to detect any labeled Bac5-related product inside [35S]Cys-labeled, unstimulated neutrophils or in neutrophils stimulated and [35S]Cys-labeled for up to 22 h (not shown), suggesting continued secretion of the newly synthesized proBac5. As a preliminary step toward studies aimed to understand the regulatory mechanisms that govern the production of Bac5 in response to bacteria and bacterial components, the 5⬘ flanking region of the Bac5 gene was sequenced, and the sequence was analyzed for the presence of putative consensus sites for binding to transcription factors. Several putative binding sites for factors that are known to be involved in the regulation of granulocyte genes were deduced, including stimulating protein-1 (SP-1), activator protein-1 (AP-1), myeloid zinc finger-1 (MZF-1), cellular equivalent to avian myoblastosis virus onco-

Tomasinsig et al. De novo synthesis of Bac5 in neutrophils

1007

Fig. 7. Nucleotide sequence of the 5⬘ flanking region of the Bac5 gene (uppercase). The coding sequence is in lowercase, and putative transcription factor binding sites are underlined.

gene-v-myb (c-Myb), cellular equivalent to avian leukemia virus E26 (Ets-2), nuclear factor-␬B (NF-␬B), nuclear factorinterleukin-6 (NF-IL-6) (Fig. 7).

DISCUSSION The peptide-based host defence of mammals counts on epithelial and myeloid components that exert rapid and direct microbicidal effects on release [1, 2, 6, 11]. The epithelial antimicrobial peptides are constitutively produced and/or transcriptionally induced [6, 21, 29, 30] in epithelial cells in response to inflammatory and infectious stimuli. Those associated with the neutrophil granules are synthesized in the early stages of neutrophil maturation, and their expression is downregulated with terminal cell differentiation [13, 31, 32]. The antimicrobial peptides belonging to the cathelicidin family [8 –11] are neutrophil-associated molecules, and some, i.e., LL-37 [16] and CRAMP [20], are also found in nonmyeloid tissues, particularly skin and other specialized epithelia [6, 17–21]. Bac5 is a cathelicidin family member, which is stored in fairly large amounts (2 ␮g/106 cells) in the bovine neutrophil granules [14] and is processed to a mature, proline-rich, antimicrobial peptide in activated neutrophils [14]. We have investigated here the expression of this gene in epithelial tissues such as airways and intestine, which are common sites of antimicrobial peptide synthesis. Results obtained using PCR, in situ hybridization, and immunohistochemistry concur to demonstrate that unlike the human LL-37, the Bac5 gene is not expressed in these sites and is not induced in lung epithelium in response to infectious stimuli. Several other cathelicidins, however, are present in the bovine neutrophils in addition to Bac5 [9], and we cannot rule out that some of these have a different pattern of expression. An interesting result of this analysis is the detection of a Bac5-corresponding mRNA in the neutrophils massively recruited at inflammation sites in lung 1008

Journal of Leukocyte Biology Volume 72, November 2002

tissue. This result highlights the potential for de novo synthesis of Bac5 in fully differentiated neutrophils. To date, no other antimicrobial peptide genes have been reported to be induced in mature neutrophils of adult mammals. All previous work considered expression of the granule-associated antimicrobial peptides to be restricted to specific stages of cell differentiation [13, 31, 32], despite the known ability of neutrophils to synthesize cytosolic components [33] and a recent published report on the induction of the granule-associated enzyme proteinase 3 in cytokine-primed neutrophils [34]. Consistent with the in situ results suggesting induction of Bac5 in activated neutrophils, we demonstrate that E. coli and purified E. coli LPS trigger expression of this gene in primary cultures of peripheral neutrophils at the mRNA and protein levels. The kinetics of Bac5 induction in stimulated neutrophils suggests that this is a relatively late response (16 –20 h post-stimulation), and the same is suggested by the detection of a radiolabeled product with a molecular weight compatible with that of proBac5 at correspondingly late times in the cell-free media of activated neutrophils. No labeled Bac5related products are recognized in cell extracts of stimulated neutrophils. This result suggests that the sorting machinery for storage in the cytoplasmic granules is not active. The newly synthesized polypeptide is thus redirected to the constitutive secretory pathway and accumulates in the extracellular medium. The long cell incubation times in these experiments provide a likely explanation for the presence of lower molecular weight (10 –11 kDa) proteolytic fragments in the incubation media of stimulated cells. Whether the newly synthesized propeptide is processed to the mature antimicrobial peptide, however, remains open to future studies. To obtain a preliminary insight into possible regulatory mechanisms for gene induction in the mature neutrophils, we have inspected the 5⬘ flanking region of the Bac5 gene and found several putative consensus sequences for binding to transcription factors involved in granulocyte gene regulation. Particularly, consensus sites for NF-␬B and NF-IL-6 are present in this sequence and appear ubiquitous in cathelicidin gene promoters [34 –36]. Their engagement in LPS-mediated induction of antimicrobial genes is conserved and widespread, as evidenced by studies on the transcriptional regulation of insect [3], amphibian [37], and mammalian antimicrobial genes [4]. In mammals, NF-␬B appears to be essential and sufficient for full LPS responsiveness of the human ␤-defensin-2 gene in RAW264.7 cells [38], and NF-IL-6 and NF-␬B are required for LPS induction of the bovine tracheal antimicrobial peptide and human ␤-defensin-2 genes in epithelial cells [39, 40]. It is interesting that LPS has also been shown to up-regulate the porcine cathelicidin genes PR-39 and protegrin in immature BM cells [36], and although the mechanism has not been completely described, a role for NF-IL-6 and NF-␬B together with other transcription factors has been proposed in this case [36]. The kinetics of induction of the two genes differs markedly from that observed for Bac5. The levels of PR-39 and protegrin mRNAs in marrow cells increase rapidly, peak at 6 h, and are decreased 12 h after LPS stimulation [36], whereas we find here that the level of Bac5 mRNA in the mature neutrophils increases at approximately 16 h and is maintained high at 24 h after LPS stimulation. This distinct kinetics suggests that http://www.jleukbio.org

somewhat different mechanisms govern the cathelicidin induction in response to LPS in immature and mature neutrophils, likely involving different modules/combinations of transcription factors. Dissecting the pathways of induction at the molecular level will lead to a better understanding of these mechanisms. A late boost of Bac5 expression in activated neutrophils could be consistent with the need to maintain a significant level of antimicrobial defense at sites of infection many hours after degranulation, although it is difficult to explain why this applies to only Bac5 out of all the bovine cathelicidins. An alternative explanation is the involvement of the newly synthesized Bac5 in other inflammatory responses. For instance, a signaling role would not require large amounts of the peptide but would require that the peptide be present in the inflammatory environment at the right time. The delayed Bac5 expression would seem to indicate that its presence is required for events whose timing is not coincident with degranulation. In this respect, it is interesting to note that the storage peptide (proBac5) may be largely depleted at the time of de novo synthesis, as suggested by Western analysis of neutrophil extracts after 22 h stimulation with E. coli (results not shown). The proposed hypothesis of a signaling role finds its rationale in the ability of other cathelicidin family members to mediate various host responses. For example, LL-37 is chemotactic for neutrophils, monocytes, and lymphocytes through interaction with formyl peptide-like receptor-1 [41], and the proline-rich PR-39 interacts with SH3 domains of cytoplasmic proteins and modulates expression of proteoglycans as part of wound repair in mammalian cells [42, 43]. Preliminary results (L. Tomasinsig, unpublished data) indicate that Bac5 peptide is also capable of translocating across the plasma membrane of mammalian cells without causing membrane damage, and this further supports the potential for this peptide to interact with intracellular targets and possibly trigger cellular responses.

ACKNOWLEDGMENTS This work was supported by the Italian Ministry for University and Research (MURST) P.R.I.N. Cofin. 2000; CNR Agenzia 2000; Commissariato di Governo della Regione FVG; and Regione Friuli Venezia Giulia. We are grateful to Prof. M. Galeotti from the Dip. Produzione Animale, Faculty of Veterinary Sciences, University of Udine, for help with histopathological examination of the animal tissues and to Dr. A. Tossi and R. Gennaro, Dept. of Biochemistry of the University of Trieste, for critically reading the manuscript.

REFERENCES 1. Lehrer, R. I., Ganz, T. (1999) Antimicrobial peptides in mammalian and insect host defence. Curr. Opin. Immunol. 11, 23–27. 2. Ganz, T. (2002) Antimicrobial polypeptides in host defense of the respiratory tract. J. Clin. Investig. 109, 693– 697. 3. Hoffmann, J. A., Reichhart, J. M. (2002) Drosophila innate immunity: an evolutionary perspective. Nat. Immunol. 3, 121–126. 4. Zasloff, M. (2002) Antimicrobial peptides of multicellular organisms. Nature 415, 389 –395.

5. Bals, R., Wang, X., Zasloff, M., Wilson, J. M. (1998) The peptide antibiotic LL-37/hCAP-18 is expressed in epithelia of the human lung where it has broad antimicrobial activity at the airway surface. Proc. Natl. Acad. Sci. USA 95, 9541–9546. 6. Dorschner, R. A., Pestonjamasp, V. K., Tamakuwala, S., Ohtake, T., Rudisill, J., Nizet, V., Agerberth, B., Gudmundsson, G. H., Gallo, R. L. (2001) Cutaneous injury induces the release of cathelicidin anti-microbial peptides active against group A Streptococcus. J. Investig. Dermatol. 117, 91–97. 7. Nizet, V., Ohtake, T., Lauth, X., Trowbridge, J., Rudisill, J., Dorschner, R. A., Pestonjamasp, V., Piraino, J., Huttner, K., Gallo, R. L. (2001) Innate antimicrobial peptide protects the skin from invasive bacterial infection. Nature 414, 454 – 457. 8. Zanetti, M., Gennaro, R., Romeo, D. (1995) Cathelicidins: a novel protein family with a common proregion and a variable C-terminal antimicrobial domain. FEBS Lett. 374, 1–5. 9. Gennaro R., Zanetti, M. (2000) Structural features and biological activities of the cathelicidin-derived antimicrobial peptides. Biopolymers 55, 31– 49. 10. Zanetti, M., Gennaro, R., Scocchi, M., Skerlavaj, B. (2000) Structure and biology of cathelicidins. Adv. Exp. Med. Biol. 479, 203–218. 11. Lehrer, R. I., Ganz, T. (2002) Cathelicidins: a family of endogenous antimicrobial peptides. Curr. Opin. Hematol. 9, 18 –22. 12. Zanetti, M., Del Sal, G., Storici, P., Schneider, C., Romeo, D. (1993) The cDNA of the neutrophil antibiotic Bac5 predicts a pro-sequence homologous to a cysteine proteinase inhibitor that is common to other neutrophil antibiotics. J. Biol. Chem. 268, 522–526. 13. Zanetti, M., Litteri, L., Gennaro, R., Horstmann, H., Romeo, D. (1990) Bactenecins, defense polypeptides of bovine neutrophils, are generated from precursor molecules stored in the large granules. J. Cell Biol. 111, 1363–1371. 14. Zanetti, M., Litteri, L., Griffiths, G., Gennaro, R., Romeo, D. (1991) Stimulus-induced maturation of probactenecins, precursors of neutrophil antimicrobial polypeptides. J. Immunol. 146, 4295– 4300. 15. Scocchi, M., Skerlavaj, B., Romeo, D., Gennaro, R. (1992) Proteolytic cleavage by neutrophil elastase converts inactive storage proforms to antibacterial bactenecins. Eur. J. Biochem. 15, 589 –595. 16. Agerberth, B., Gunne, H., Odeberg, J., Kogner, P., Boman, H. G., Gudmundsson, G. H. (1995) FALL-39, a putative human peptide antibiotic, is cysteine-free and expressed in bone marrow and testis. Proc. Natl. Acad. Sci. USA 92, 195–199. 17. Frohm Nilsson, M., Sandstedt, B., Sorensen, O., Weber, G., Borregaard, N., Stahle-Backdahl, M. (1999) The human cationic antimicrobial protein (hCAP18), a peptide antibiotic, is widely expressed in human squamous epithelia and colocalizes with interleukin-6. Infect. Immun. 67, 2561– 2566. 18. Malm, J., Sorensen, O., Persson, T., Frohm-Nilsson, M., Johansson, B., Bjartell, A., Lilja, H., Stahle-Backdahl, M., Borregaard, N., Egesten, A. (2000) The human cationic antimicrobial protein (hCAP-18) is expressed in the epithelium of human epididymis, is present in seminal plasma at high concentrations, and is attached to spermatozoa. Infect. Immun. 68, 4297– 4302. 19. Hase, K., Eckmann, L., Leopard, J. D., Varki, N., Kagnoff, M. F. (2002) Cell differentiation is a key determinant of cathelicidin LL-37/human cationic antimicrobial protein 18 expression by human colon epithelium. Infect. Immun. 70, 953–963. 20. Gallo, R. L., Kim, K., Bernfield, M., Kozak, C., Zanetti, M,. Merluzzi, L., Gennaro, R. (1997) Identification of CRAMP, a cathelin related antimicrobial peptide expressed in the embryonic and adult mouse. J. Biol. Chem. 272, 13088 –13093. 21. Frohm, M., Agerberth, B., Ahangari, G., Stahle-Backdahl, M., Liden, S., Wigzell, H., Gudmundsson, G. H. (1997) The expression of the gene coding for the antibacterial peptide LL-37 is induced in human keratinocytes during inflammatory disorders. J. Biol. Chem. 272, 15258 –15263. 22. Chomczynski, P., Sacchi, N. (1987) Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal. Biochem. 162, 156 –159. 23. Ryan, L. K., Rhodes, J., Bhat, M., Diamond, G. (1998) Expression of beta-defensin genes in bovine alveolar macrophages. Infect. Immun. 66, 878 – 881. 24. Scocchi, M., Wang, S., Zanetti, M. (1997) Structural organization of the bovine cathelicidin gene family and identification of a novel member. FEBS Lett. 417, 311–315. 25. Quandt, K., Frech, K., Karas, H., Wingender, E., Werner, T. (1995) MatInd and MatInspector: new fast and versatile tools for detection of consensus matches in nucleotide sequence data. Nucleic Acids Res. 23, 4878 – 4884.

Tomasinsig et al. De novo synthesis of Bac5 in neutrophils

1009

26. Yount, N. Y., Yuan, J., Tarver, A., Castro, T., Diamond, G., Tran, P. A., Levy, J. N., McCullough, C., Cullor, J. S., Bevins, C. L., Selsted, M. E. (1999) Cloning and expression of bovine neutrophil beta-defensins. Biosynthetic profile during neutrophilic maturation and localization of mature peptide to novel cytoplasmic dense granules. J. Biol. Chem. 274, 26249–26258. 27. Bazzoni, F., Cassatella, M. A., Rossi, F., Ceska, M., Dewald, B., Baggiolini, M. (1991) Phagocytosing neutrophils produce and release high amounts of the neutrophil-activating peptide 1/interleukin 8. J. Exp. Med. 173, 771–774. 28. Caswell, J. L., Middleton, D. M., Sorden, S. D., Gordon, J. R. (1998) Expression of the neutrophil chemoattractant interleukin-8 in the lesions of bovine pneumonic pasteurellosis. Vet. Pathol. 35, 124 –131. 29. Schonwetter, B. S., Stolzenberg, E. D., Zasloff, M. A. (1995) Epithelial antibiotics induced at sites of inflammation. Science 267, 1645–1648. 30. Tarver, A. P., Clark, D. P., Diamond, G., Russell, J. P., ErdjumentBromage, H., Tempst, P., Cohen, K. S., Jones, D. E., Sweeney, R. W., Wines, M., Hwang, S., Bevins, C. L. (1998) Enteric beta-defensin: molecular cloning and characterization of a gene with inducible intestinal epithelial cell expression associated with Cryptosporidium parvum infection. Infect. Immun. 66, 1045–1056. 31. Sorensen, O., Arnljots, K., Cowland, J. B., Bainton, D. F., Borregaard, N. (1997) The human antibacterial cathelicidin, hCAP-18, is synthesized in myelocytes and metamyelocytes and localized to specific granules in neutrophils. Blood 90, 2796 –2803. 32. Nagaoka, I., Hirata, M., Sugimoto, K., Tsutsumi-Ishii. Y., Someya, A., Saionji, K., Igari, J. (1998) Evaluation of the expression of human CAP18 gene during neutrophil maturation in the bone marrow. J. Leukoc. Biol. 64, 845– 852. 33. Cassatella, M. A. (1999) Neutrophil-derived proteins: selling cytokines by the pound. Adv. Immunol. 73, 369 –509. 34. Zhou, Z., Richard, C., Menard, H. A. (2000) De novo synthesis of proteinase 3 by cytokine primed circulating human polymorphonuclear neutrophils and mononuclear cells. J. Rheumatol. 27, 2406 –2411.

1010

Journal of Leukocyte Biology Volume 72, November 2002

35. Gudmundsson, G. H., Agerberth, B., Odeberg, J., Bergman, T., Olsson, B., Salcedo, R. (1996) The human gene FALL39 and processing of the cathelin precursor to the antibacterial peptide LL-37 in granulocytes. Eur. J. Biochem. 238, 325–332. 36. Wu, H., Zhang, G., Minton, J. E., Ross, C. R., Blecha, F. (2000) Regulation of cathelicidin gene expression: induction by lipopolysaccharide, interleukin-6, retinoic acid, and Salmonella enterica serovar typhimurium infection. Infect. Immun. 68, 5552–5558. 37. Miele, R., Bjorklund, G., Barra, D., Simmaco, M., Engstrom, Y. (2001) Involvement of Rel factors in the expression of antimicrobial peptide genes in amphibia. Eur. J. Biochem. 268, 443– 449. 38. Tsutsumi-Ishii, Y., Nagaoka, I. (2002) NF-kappa B-mediated transcriptional regulation of human beta-defensin-2 gene following lipopolysaccharide stimulation. J. Leukoc. Biol. 71, 154 –162. 39. Becker, M. N., Diamond, G., Verghese, M. W., Randell, S. H. (2000) CD14-dependent lipopolysaccharide-induced beta-defensin-2 expression in human tracheobronchial epithelium. J. Biol. Chem. 275, 29731–29736. 40. Diamond, G., Kaiser, V., Rhodes, J., Russell, J. P., Bevins, C. L. (2000) Transcriptional regulation of beta-defensin gene expression in tracheal epithelial cells. Infect. Immun. 68, 113–119. 41. Yang, D., Chertov, O., Oppenheim, J. J. (2001) Participation of mammalian defensins and cathelicidins in anti-microbial immunity: receptors and activities of human defensins and cathelicidin (LL-37). J. Leukoc. Biol. 69, 691– 697. 42. Gallo, R. L., Ono, M., Povsic, T., Page, C., Eriksson, E., Klagsbrun, M., Bernfield, M. (1994) Syndecans, cell surface heparan sulfate proteoglycans, are induced by a proline-rich antimicrobial peptide from wounds. Proc. Natl. Acad. Sci. USA 91, 11035–11039. 43. Chan, Y. R., Gallo, R. L. (1998) PR-39, a syndecan-inducing antimicrobial peptide, binds and affects p130Cas. J. Biol. Chem. 273, 28978 – 28985.

http://www.jleukbio.org