1-s2.0-S1097276511007179-main

download 1-s2.0-S1097276511007179-main

of 14

Transcript of 1-s2.0-S1097276511007179-main

  • 7/27/2019 1-s2.0-S1097276511007179-main

    1/14

    Molecular Cell

    Article

    Granzyme B-Dependent Proteolysis Acts as a Switchto Enhance the Proinflammatory Activity of IL-1a

    Inna S. Afonina,1 Graham A. Tynan,2 Susan E. Logue,1 Sean P. Cullen,1 Michael Bots,3 Alexander U. Luthi,1

    Emer P. Reeves,4 Noel G. McElvaney,4 Jan P. Medema,3 Ed C. Lavelle,2 and Seamus J. Martin1,*1Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland2Adjuvant Research Group, School of Biochemistry and Immunology, Trinity College, Dublin 2, Ireland3LEXOR (Lab of Experimental Oncology and Radiobiology), Center for Experimental Molecular Medicine,

    Academic Medical Center (AMC), Amsterdam, The Netherlands4Respiratory Research Division, Department of Medicine, Education and Research Centre, Royal College of Surgeons in Ireland,

    Beaumont Hospital, Dublin 9, Ireland

    *Correspondence: [email protected]

    DOI 10.1016/j.molcel.2011.07.037

    SUMMARY

    Granzyme B is a cytotoxic lymphocyte-derived pro-

    tease that plays a central role in promoting apoptosis

    of virus-infected target cells, through direct proteol-

    ysis and activation of constituents of the cell death

    machinery. However, previous studies have also

    implicated granzymes A and B in the production of

    proinflammatory cytokines, via a mechanism that

    remains undefined. Here we show that IL-1a is a sub-

    strate for granzyme B and that proteolysis potently

    enhanced the biological activity of this cytokine

    in vitro as well as in vivo. Consistent with this, com-

    pared with full-length IL-1a, granzyme B-processed

    IL-1a exhibited more potent activity as an immu-

    noadjuvant in vivo. Furthermore, proteolysis of

    IL-1a within the same region, by proteases such as

    calpain and elastase, was also found to enhance its

    biological potency. Thus, IL-1a processing by

    multiple immune-related proteases, including gran-

    zyme B, acts as a switch to enhance the proinflam-

    matory properties of this cytokine.

    INTRODUCTION

    Granzyme (Gzm) B is a protease typically found in the cytotoxicgranules of Natural Killer (NK) and cytotoxic T cells (CTLs) and,

    upon delivery into the cytoplasm of virus-infected target cells,

    promotes apoptosis to limit virus replication and dissemination

    (reviewed in Afonina et al., 2010). Gzm B promotes apoptosis

    through proteolysis of the BH3-only protein Bid, as well as via

    proteolytic processing and activation of caspase-3 (Afonina

    et al., 2010). However, accumulating evidence suggests that

    granzymes A and B may also influence the production of proin-

    flammatory cytokines, although the molecular targets of these

    proteases in inflammatory contexts remain to be defined (Sower

    et al., 1996a,b; Metkaret al., 2008; Froelich et al., 2009). Interest-

    ingly, a number of noncytotoxic cell types, such as mast cells

    and basophils, have also been found to secrete Gzm B upon

    activation (Tschopp et al., 2006; Strik et al., 2007). Furthermore,Gzm B is found at elevated levels in serum from rheumatoid

    arthritis (RA) patients and in other inflammatory conditions asso-

    ciated with elevated levels of IL-1 (Lauw et al., 2000; Tak et al.,

    1999; Kim et al., 2007). For these reasons, we explored whether

    Gzm B may contribute to the processing and activation of IL-1.

    IL-1 is a cytokine with diverse biological activities and is

    produced in the early stages of infection or sterile injury, where

    it plays an importantrole in instigating immune responses(Dinar-

    ello, 1996; Kono et al., 2010). Although IL-1 is produced in two

    different forms (IL-1a and IL-1b), encoded by distinct genes,

    these cytokines bind to the same receptor and share similar bio-

    logical activities (Dinarello, 1996). Despite their relatedness in

    structure and biological activity, IL-1a and IL-1b are posttransla-

    tionally processed in a very different manner. IL-1a is expressed

    as a $31 kDa polypeptide, and in this form is capable of binding

    to the IL-1 receptor and is biologically active (Mosley et al.,

    1987a,b). IL-1b is also expressed as a $31 kDa polypeptide,

    but in marked contrast to IL-1a, full-length IL-1b is incapable of

    binding to the IL-1 receptor and, consequently, exhibits little bio-

    logical activity (Mosley et al., 1987a,b; Hazudaet al., 1991). IL-1b

    acquires biological activity upon limited internal processing by

    the aspartic acid-specific protease caspase-1 (Thornberry

    et al., 1992). Caspase-1-dependent processing of IL-1b at

    Asp116 unlocks the biological activity of this cytokine. Gzm A

    has also been found to cleave IL-1b in vitro, however the func-

    tional consequences of this event remain unclear (Irmler et al.,

    1995).IL-1a is typically released from cells as a result of injury or

    death, and circulating IL-1a levels are persistently elevated in

    inflammatory conditions such as RA. Although full-length IL-1a

    displays biological activity, this cytokine is susceptible to

    proteolysis by certain intracellular proteases. For example, the

    calcium-activated protease, calpain-1, promotes restricted

    proteolysis of IL-1a to produce a $17 kDa protein, although

    this reportedly has little effect on the biological activity of this

    cytokine (Kobayashi et al., 1990; Carruth et al., 1991). Therefore,

    it remains unclear what role calpain-mediated processing of

    IL-1a serves. As a full-length protein, IL-1a is typically cell-asso-

    ciated, and a portion of this cell-associated form may be present

    Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc. 265

    mailto:[email protected]://dx.doi.org/10.1016/j.molcel.2011.07.037http://dx.doi.org/10.1016/j.molcel.2011.07.037mailto:[email protected]
  • 7/27/2019 1-s2.0-S1097276511007179-main

    2/14

    Figure 1. IL-1a Is a Substrate for Granzyme B

    (A) 35S-labeled IL-1a, IL-1b, and Bid were incubated at 37C for 2 hr, either alone or in the presence of recombinant caspase-1, -4, -5 (20 nM), caspase-3, -7, or

    granzyme B (200 nM), followed by analysis by SDS-PAGE/fluorography.

    (B)35

    S-labeled IL-1a, IL-1b, and Bid were incubated for 2 hr at 37C with the indicated concentrations of granzyme B and analyzed as in (A).

    (C) Recombinant purified full-length IL-1a was incubated at 37C for 2 hr with the indicated concentrations of granzyme B and analyzed by SDS-PAGE with

    Coomassie staining. IL-1a cleavage fragments were excised from the gel and analyzed by MALDI-TOF mass spectrometry. Mass spectra of cleavage fragments

    are indicated, with coverage of the two cleavage fragments underlined in blue and red, respectively.

    (D) Schematic representation of IL-1a indicating nuclear localization signal and granzyme B and calpain-1 cleavage sites. A sequence alignment of the putative

    granzyme B cleavage site in IL-1a from a number of mammals is shown to the right. The P1 Asp residue is indicated.

    (E) 35S-labeled IL-1aWT and IL-1aD103A mutant were incubated at 37C for 2 hr with the indicated concentrations of granzyme B, followed by analysis by

    SDS-PAGE/fluorography.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    266 Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc.

  • 7/27/2019 1-s2.0-S1097276511007179-main

    3/14

    at the plasma membrane (Kurt-Jones et al., 1985). Proteolytic

    processing of IL-1a may remove a nuclear localization signal or

    permit shedding of membrane-associated IL-1a and increase

    thelikelihood of secretion (Watanabe and Kobayashi, 1994; Mat-

    sushima et al., 1986). Thus, proteolysis of IL-1a may convert thiscytokine from a cell-associated form to a soluble molecule that

    could exert more systemic effects. An additional possibility is

    that proteolysis of IL-1a may modulate the biological potency

    of this cytokine, although this issue has not been explored in

    any detail.

    Here we report that human IL-1a, but not IL-1b, is a substrate

    for the CTL/NK cell protease, Gzm B, and that processing of this

    cytokine by the latter protease enhanced its biological activity

    several-fold. A similar increase in IL-1a activity was also seen

    upon processing of IL-1a by other proteases, such as calpain-1,

    elastase, and mast cell chymase. Thus, Gzm B and other prote-

    ases that are produced during inflammatory responses may

    promote inflammation, at least in part, through conversion of

    pro-IL-1a to its mature form.

    RESULTS

    IL-1a Is a Substrate for Granzyme B

    Because recent studies have suggested that CTL/NK granzymes

    may have extracellular proinflammatory roles, we set out to

    explore whether Gzm B was capable of processing any of the

    major proinflammatory cytokines. To address this issue, we

    screened a panel of in vitro transcribed and translated members

    of the interleukin family (IL-1 to IL-17) for susceptibility to prote-

    olysis by Gzm B, as well as a range of other aspartic acid-

    specific proteases as controls (data not shown). All proteases

    were confirmed to be active by virtue of their ability to cleave

    synthetic tetrapeptide substrates (data not shown) or control

    proteins such as Bid (Figure 1A). The outcome of this screen

    identified full-length IL-1a as a substrate for Gzm B but not for

    any of the other proteases tested. As Figure 1A shows, whereas

    caspases-1, -3, -4, -5, -and -7 failed to cleave IL-1a, Gzm B

    produced a major cleavage product at $17 kDa. In contrast,

    Gzm B did not cleave IL-1b, whereas caspase-1 did so, as ex-

    pected (Figure 1A). To confirm the specificity of IL-1a proteolysis

    by Gzm B, we titrated this protease over a range of concentra-

    tions from 12.5 to 100 nM. IL-1b failed to be cleaved by Gzm B

    at any of these concentrations, whereas proteolysis of IL-1a,

    as well as theknown Gzm B substrate, Bid, wasreadily observed

    (Figure1B).Importantly, because human andmurineGzmB have

    been shown to have divergent substrate preferences (Cullenet al., 2007), we also tested whether murine IL-1awasa substrate

    for Gzm B and found thatboth human and mouse Gzm B cleaved

    murine IL-1a within the nanomolar range (Figures S1A and S1B).

    IL-1a Is Cleaved by Granzyme B at Asp103

    To map the Gzm B cleavage site within IL-1a, we expressed and

    purified the full-length cytokine from E. coli and incubated re-

    combinant IL-1a with Gzm B to promote proteolysis. Cleavage

    products were then analyzed by mass spectrometry, which sug-

    gested that proteolysis occurred between amino acids 80 and

    112 (Figure 1C). Because Gzm B has an almost absolute prefer-

    ence for proteolysis after Asp residues and typically cleaves

    within IXXD/S motifs, this suggested that IL-1a was cleaved afterAsp103, within the motif IAND103, which is conserved among

    mammals (Figure 1D). To confirm this as the site of proteolysis,

    we generated a point mutation Asp > Ala103 in IL-1a and tested

    the susceptibility of this mutant to proteolysis by Gzm B. As

    Figure 1E shows, this mutant fully resisted proteolysis, as did a

    deletion mutant of IL-1a lacking the N-terminal 103 amino acids

    (Figure 1F), confirming that Gzm B cleaves IL-1a at Asp103.

    Granzyme B-Mediated Proteolysis of IL-1a Potentiates

    Biological Activity

    Previous studies have shown that calpain-1 can process IL-1a at

    Phe118 (Kobayashi et al., 1990), however the significance of this

    cleavage event for IL-1a bioactivity has not been determined.

    Indeed, it is widely assumed that proteolytic processing of

    IL-1a within its N-terminal region has little effect on the biological

    activity of this cytokine. For these reasons, we wondered

    whether Gzm B-mediated processing of IL-1a could impact

    upon its biological activity. To explore this issue, we measured

    the ability of IL-1a to stimulate production of IL-6 and IL-8 from

    HeLa cells and primary HUVECs, as both cell types are known

    to respond to this cytokine (Bertelsen and Sanfridson, 2007;

    Rhim et al., 2008).

    To mimic the Gzm B cleavage product of IL-1a, we generated

    a truncated form of this cytokine, missing the N-terminal 103

    amino acids (IL-1a104271). Using this form of IL-1a we confirmed

    that both HeLa cells and primary HUVECs were highly respon-

    sive to this cytokine (Figure 2A), with both cell types producing

    IL-6 and IL-8 in response to IL-1a104271. HUVECs also pro-

    duced GM-CSF under the same conditions (Figure 2A). These

    observations confirmed that truncation of IL-1a after Asp103

    produced a biologically active cytokine. However, this type of

    artificial truncation does not necessarily faithfully reproduce

    the natural cleaved form of full-length IL-1a, as proteolysis of a

    protein does not necessarily disassociate the resulting cleavage

    products.

    To compare the effects of Gzm B-mediated proteolysis of

    IL-1a with the full-length cytokine, we incubated bacterially ex-

    pressed IL-1a precursor in the presence or absence of Gzm B,

    followed by assessment of biological activity as above. Surpris-

    ingly, proteolytic processing of IL-1a by Gzm B increased the

    activity of this cytokine several-fold (Figure 2B). Similar observa-tions were also made in time course experiments, once again

    indicating that Gzm B-mediated proteolysis dramatically

    increased the bioactivity of IL-1a (Figure 2C).

    Importantly, we ruled out the possibility that co-purifying

    microbial contaminants in the Gzm B preparations contributed

    to the effects seen. First, we expressed and purified a catalyti-

    cally inactive Gzm B mutant (Gzm BSA) in the same yeast system

    that was used to generate wild-type Gzm B (Figures S2AS2C).

    (F) Recombinant IL-1a1-271

    and IL-1a104-271

    were incubated for 2 hr at 37C with the indicated concentrations of granzyme B, then analyzed by SDS-PAGE and

    Coomassie staining. All data shown are representative of at least three independent experiments.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc. 267

  • 7/27/2019 1-s2.0-S1097276511007179-main

    4/14

    Figure 2. Granzyme B-Dependent Proteolysis of IL-1a Enhances Cytokine Bioactivity

    (A) HeLa(top panels) or HUVEC cells (lower panels) were incubated with 1 nM of recombinant IL-1a104-271

    or annexin V as a control protein. Culture supernatants

    were collected at the indicated time-points, with IL-6, IL-8, and GM-CSF concentrations then determined by ELISA.

    (B) HeLa or HUVEC cells were incubated with the indicated concentrations of full-length or granzyme B-cleaved IL-1a for 8 hr, and concentrations of IL-6, IL-8,

    and GM-CSF in the supernatants were determined by ELISA.

    (C) HeLa cells were left untreated or incubated with 2 nM of full-length or granzyme B-cleaved IL-1a. Culture supernatants were collected at the indicated time-

    points, with IL-6 and IL-8 concentrations subsequently determined by ELISA. All results are representative of at least three independent experiments. Error bars

    represent the mean the SEM of triplicate experiments.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    268 Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc.

  • 7/27/2019 1-s2.0-S1097276511007179-main

    5/14

    This mutant failed to cleave IL-1a and also failed to enhance the

    activity of IL-1a on HeLa or HUVECs (Figures S2C and S2D).

    Second, neither wild-type nor mutant forms of Gzm B inducedcytokines when added directly to HeLa cells in the absence of

    IL-1a (Figure S2E).Third, whereas HeLa cells arevery responsive

    to IL-1a, these cells fail to produce IL-6 or IL-8 in response to

    a wide panel of microbial components (i.e., pathogen-associ-

    ated molecular patterns), such as lipopolysaccharide, which

    would represent possible contaminants (Figure S2F).

    Granzyme B Does Not Directly Influence Inflammatory

    Cytokine Production by HeLa or HUVECs

    To further rule out the possibility that residual Gzm B activity

    within the cleaved IL-1a preparations was responsible for the

    increased proinflammatory effects seen, we neutralized this

    Figure 3. Granzyme B Activity Does Not

    Directly Promote Inflammatory Cytokine

    Production

    (A) Recombinant IL-1a was incubated with gran-

    zyme B (200 nM) for 3 hr at 37C. Residual gran-

    zyme B activity was then measured after furtherincubation for 30 min either alone or in the pres-

    ence of the granzyme B inhibitor PI-9 (1 mM).

    Granzyme B activity was measured by monitoring

    hydrolysis of the synthetic granzyme B substrate

    IETD-AFC by fluorometry.

    (B)35S-labeled Bid was incubated, either alone or

    withthe indicated concentrations of active or PI-9-

    treated granzyme B for 2 hr at 37C. Reactions

    were analyzed by SDS-PAGE followed by fluo-

    rography.

    (C) HeLa cells were incubated for 8 hr with the

    indicated concentrations of full-length or gran-

    zymeB-cleaved IL-1a, where residual granzyme B

    activity after proteolysis of IL-1a was inhibited by

    addition of saturating amounts of PI-9. IL-6 and

    IL-8 levels in culture supernatants were deter-

    mined by ELISA.

    (D) HeLa cells were incubated for 8 hr with the

    indicated concentrations of IL-1aFL

    or IL-1a104-271

    .

    IL-6 and IL-8 levels were determined by ELISA.

    (E) HeLa cells were incubated for 8 hr with the

    indicated concentrations of IL-1aFL

    , granzyme

    B-treated IL-1aFL

    , IL-1a104-271

    , or granzyme

    B-treated IL-1a104-271

    , as indicated. IL-6 and IL-8

    levels were determined by ELISA. Results are

    representative of at least three independent ex-

    periments. Error barsrepresentthe mean SEMof

    determinations from three independent experi-

    ments.

    granzyme with the serpin protease inhib-

    itor PI-9, confirming that no residual

    protease activity remained after the incu-

    bation period with IL-1a (Figures 3A and

    3B). Addition of PI-9 after Gzm B-medi-

    ated proteolysis of IL-1a had no effect

    on the increased potency of the Gzm

    B-treated IL-1a preparations (Figure 3C),

    demonstrating that Gzm B was not

    directly acting upon the cells used to assay IL-1a activity.

    Similar observations were also made with HUVECs (data not

    shown).We also directly comparedthe potency of full-length IL-1awith

    the artificially-truncated IL-1a104-271 protein (which mimics the

    Gzm B-cleaved form of this cytokine) and once again observed

    considerable differences in activity between these forms of

    IL-1a (Figure 3D). Furthermore, incubation of truncated

    IL-1a104-271 with Gzm B did not have any effect on the activity

    of this cytokine, as expected (Figure 3E). Similarly, incubation

    of the Gzm B-noncleavable form of IL-1a (IL-1aD103A) with this

    granzyme also failed to enhance the activity of this protein

    (data not shown). Collectively, these data provide strong

    evidence that proteolysis of IL-1a by Gzm B enhances the

    biological activity of this protein up to 10-fold.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc. 269

  • 7/27/2019 1-s2.0-S1097276511007179-main

    6/14

    Figure 4. Proteolysis of IL-1a by Calpain and Other Proteases Also Enhances Cytokine Bioactivity

    (A)35

    S-labeled IL-1aWT

    andIL-1aD103A

    wereincubatedfor 2 hrat 37C withthe indicatedconcentrations of granzymeB or calpain-1. Reactions wereanalyzed by

    SDS-PAGE/fluorography.

    (B) HeLa or HUVEC cells were incubated with the indicated concentrations of full-length IL-1a, or granzyme B- or calpain-1-cleaved IL-1a for 8 hr. IL-6 and IL-8

    levels were determined by ELISA.

    (C)35

    S-labeled in vitro transcribed/translated IL-1a andIL-1b were incubatedfor 2 hr at37C with theindicated concentrationsof elastase, or with granzymeB or

    calpain-1 (both at 200 nM) as controls. Reactions were then analyzed as in (A).

    (D) HeLacells wereincubated for 8 hr withthe indicated concentrations of full-length IL-1a, orIL-1a cleaved witheither granzymeB or elastase, as indicated. IL-6

    and IL-8 levels were then determined by ELISA.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    270 Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc.

  • 7/27/2019 1-s2.0-S1097276511007179-main

    7/14

  • 7/27/2019 1-s2.0-S1097276511007179-main

    8/14

    Figure 5. IL-1a Is Cleaved, either Extracellularly or Intracellularly, by Endogenous Granzyme B

    (A) YT cells were either left untreated or were permeabilized via 2 cycles of freeze-thaw (Fr-Th), or addition of 10 mg/ml of streptolysin O (SLO) for 90 min.

    Recombinant IL-1a was added to the supernatants, which were further incubated for 2 hr at 37C and analyzed for IL-1a processing by immunoblotting.

    (B) YT cells wereincubated for 18 hr withHeLa target cells at the indicated effector:target ratios in the presenceof recombinant IL-1a. Culture supernatants were

    analyzed by immunoblot for IL-1a processing. Serum-derived transferrin served as a loading control.

    (C) HeLacells wereincubated withrecombinantextracellularIL-1a for the indicated times in the presenceor absence of YT cells, as indicated. Supernatantswere

    then analyzed for IL-1a processing by immunoblot.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    272 Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc.

  • 7/27/2019 1-s2.0-S1097276511007179-main

    9/14

    IL-1a Cleaving Activity in Inflammatory Patients

    To explore whether IL-1a also underwent processing during

    inflammation, we obtained bronchoalveolar lavage fluids

    (BALF) from patients with persistent inflammatory lung condi-

    tions (cystic fibrosis, chronic obstructive pulmonary disease,bronchiectasis) as well as healthy donors. Previous studies

    have shown that such fluids contain high levels of inflammatory

    proteases, particularly elastase, due to invasion of the lungs by

    neutrophils and other cells of the innate immune system (McEl-

    vaney et al., 1991; Reeves et al., 2010). As Figure 5H illustrates,

    IL-1a underwent rapid proteolysis to fragments very similar to

    the Gzm B/elastase/calpain-cleaved forms of this cytokine in

    BALF from several cystic fibrosis and bronchiectasis patients,

    whereas little processing was seen in controls. Thus, proteases

    capable of processing IL-1a to its mature form are secreted into

    the extracellular space under inflammatory conditions in vivo.

    Granzyme B-Cleaved IL-1a Exhibits Enhanced Activity

    In Vivo

    We next explored whether the Gzm B-cleaved form of IL-1a also

    exhibited greater bioactivity in vivo. To address this question, we

    compared the adjuvant activity of full-length versus Gzm

    B-cleaved IL-1a in an antigen-driven model, using ovalbumin

    (OVA). BALB/c mice were immunized with OVA, either alone,

    or in combination with full-length or Gzm B-cleaved IL-1a,

    followed by boosting with the same combinations two weeks

    later. One week after boosting, OVA-specific antibodies were

    measured in the serum as well as OVA-specific T cell responses

    in the peritoneal lavage and spleens of these mice. As Figures 6A

    and S3 show, both forms of IL-1a significantly boosted antibody

    responses to OVA, with the cleaved form of this cytokine exhib-

    iting considerably greater potency in this regard. Furthermore,

    OVA-specific IL-4, IL-5, and IL-10 production from peritoneal

    lavage cell preparations was also significantly greater in mice

    that received the Gzm B-cleaved form of IL-1a (Figure 6B). Poly-

    clonal stimulation with anti-CD3 also produced more robust

    cytokine responses from mice that had received cleaved IL-1a

    (Figure 6C). A similar trend toward more robust OVA-specific

    responses was also observed in splenocytes from the same

    mice, but this was less statistically significant than in peritoneal

    exudates (Figure 6D). Collectively, these data lend further

    support to our in vitro observations that Gzm B-dependent

    proteolysis of IL-1a acts to enhance the proinflammatory effects

    of this cytokine.

    To again rule out the possibility that a contaminant within the

    Gzm B preparations could contribute to the enhanced IL-1aactivity seen in vivo, we used the catalytically inactive mutant

    form of this protease (Gzm BSA) (Figures S2AS2C) to explore

    whether this mutant could enhance IL-1a activity in vivo. As Fig-

    ure 7A shows, preincubation with Gzm BSA failed to potentiate

    IL-1a activity under conditions where incubation with wild-type

    Gzm B clearly did so.We also asked whether the IL-1a mutant (D103A) incapable

    of undergoing processing by Gzm B (Figure 1E) possessed

    reduced activity in vivo, when compared to wild-type IL-1a.

    As Figure 7B illustrates, IL-1a-dependent enhancement of OVA-

    specific antibody responses were consistently less pronounced

    with IL-1aD103A than with wild-type IL-1a, suggesting that endog-

    enous Gzm B contributes to the potency of IL-1a in vivo.

    Finally, we also explored IL-1a-dependent enhancement

    of OVA-specific antibody responses in wild-type versus

    gzmB/mice. Although it is importantto note that several prote-

    ases can process IL-1a to a more active form, as we have shown

    here, we were nonetheless interested to explore whether the

    absence of Gzm B had a significant impact on IL-1a activity

    in vivo. As Figures 7C and 7D show, we found that IL-1a-depen-dent enhancement of OVA-specific antibodies was greater in

    wild-typeanimals than that seen ingzmB/ mice. Note however

    that baseline antibody responses to OVA alone were higher in

    gzmB/ animals for reasons that are unclear. Nonetheless,

    the enhancement effect of IL-1a on OVA-specific responses

    was significantly higher in wild-type animals than in knockouts,

    again suggesting that endogenous Gzm B contributes to IL-1a

    processing in vivo.

    Collectively, these data reveal a hitherto unappreciated

    consequence of IL-1a processing to the mature $17 kDa cyto-

    kine. The N-terminal IL-1a propeptide appears to function as a

    sensor for multiple proinflammatory proteases, proteolysis of

    which switches the activity of this cytokine from a basal to a

    hyperactive state.

    DISCUSSION

    Here we report that IL-1a is a substrate for the CTL/NK protease

    Gzm B and that proteolysis of this cytokine by the latter protease

    very significantly increased its biological potency. These data

    suggest that Gzm B may enhance inflammatory responses in

    certain situations through proteolytic processing of IL-1a. We

    have further shown that proteolysis of IL-1a by calpain-1, elas-

    tase, and mast cell chymase, at sites close to but distinct from

    the Gzm B processing site, also potentiated the activity of this

    cytokine. This suggests that proteolysis of IL-1a within its N

    terminus, irrespective of the protease involved, acts as a switchto increase the biological potency of this cytokine. Thus,contrary

    (D) YT cells were incubated for 6 hr with HeLa target cells at an E:T ratio of 10:1 in the presence of recombinant IL-1a and PI-9, as indicated. Supernatants were

    then analyzed for IL-1a processing by immunoblot. Serum-derived transferrin served as a loading control.

    (E) YT cells were incubated for 6 hr with HeLa target cells at an E:T ratio of 10:1 in the presence of either recombinant IL-1aWT or IL-1aD103Amutant, as indicated.

    Supernatants were then analyzed for IL-1a processing by immunoblot. Serum-derived transferrin served as a loading control.

    (F andG) HeLa cellsweretransfected with IL-1a expression plasmid.24 hr latercells were exposed to YT cells(F) or NK-92 cells(G) cellsat an E:Tratio of 20:1 or

    5:1, respectively. NK cells were removed 35 hr later and HeLa cells were further incubated for the total of 24 hr, after which cell lysates were generated and

    immunoblotted for the indicated proteins. Cell death was assessed by annexin V/propidium iodide binding, measured by flow cytometry. All data shown are

    representative of at least three independent experiments.

    (H) Recombinant purified full-length IL-1a was incubated at 37C for 20 min with 10 ml of BALF samples from control patients or patients with cystic fibrosis,

    bronchiectasis, or chronic obstructive pulmonary disease (COPD). IL-1a processing was analyzed by immunoblot. As controls, full-length IL-1a and IL-1a104-271

    were expressed in HEK293T cells and were included to facilitate size comparison of cleavage products.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc. 273

  • 7/27/2019 1-s2.0-S1097276511007179-main

    10/14

    to current thinking, limited proteolysis of IL-1a increases biolog-

    ical activity and serves as a further level of regulation on this

    cytokine.

    Proteolysis of IL-1a may induce a conformational change in

    this protein that increases its affinity for the IL-1 receptor

    complex, thereby increasing biological potency. Alternatively,

    Figure 6. Granzyme B-Cleaved IL-1a Exhibits Enhanced Activity In Vivo

    BALB/c mice(five miceper group) wereimmunized either withovalbumin (OVA) alone(200mg), OVA in combination withfull-lengthIL-1a (5 mg permouse), orOVA

    in combination with granzyme B-cleaved IL-1a (5 mg per mouse). All mice were boosted with the same combinations on day 14. Peripheral blood, spleens, and

    peritoneal lavages were collected on day 21.

    (A) OVA-specific total IgG, IgG1, IgG2a, and IgG2b in plasma samples were determined by ELISA.

    (BD) Cells from peritoneal lavages (B and C) and spleens (D) were restimulated for 3 days with indicated concentrations of OVA, anti-CD3, or anti-CD3/PMA, as

    indicated. IL-4,IL-5, IL-10, and IFN-g productionwas determined by ELISA. All measurementswere taken in triplicate. Error barsrepresent the mean SEMfrom

    each group of five mice. Significance levels, *** = p < .01, ** = p < .05, * = p < .1, by Students t test.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    274 Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc.

  • 7/27/2019 1-s2.0-S1097276511007179-main

    11/14

    Figure 7. Granzyme B Contributes to IL-1a Processing In Vivo

    (A and B) BALB/c mice (fiveanimals per group) were immunized either with ovalbumin (OVA) alone (200mg) or OVA in combination with the full-length IL-1a, Gzm

    BWT

    -treated IL-1a, Gzm BSA

    -treated IL-1a, or noncleavable mutant IL-1aD103A

    (5 mg per mouse). On day 21, peripheral bloods were collected and OVA-specific

    total IgG, IgG1, IgG2a, and IgG2b antibodies in plasma samples were determined by ELISA.

    (C)WT orGranzymeB/C57BL/6 mice(five per group) wereimmunized either withOVA alone (200mg)or with OVA in combinationwithfull-lengthIL-1a (5 mg per

    mouse). On day 21, peripheral bloods were collected and OVA-specific total IgG, IgG1, IgG2a, and IgG2b in plasma samples were determined by ELISA.

    (D) Fold increasein antibody production in WT versus GranzymeB/ micetreated with OVA plus IL-1a, compared with OVA alone. The analysis shown is based

    on the data presented in (C). Error bars represent the mean SEM from each group of five mice.

    Significance levels, *** = p < .01, ** = p < .05, * = p < .1, by Students t test.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc. 275

  • 7/27/2019 1-s2.0-S1097276511007179-main

    12/14

    the IL-1a N terminus may partly occlude the receptor binding

    domain, removal of which now permits a more stable interaction

    with the IL-1R complex. This possibility is consistent with a

    previous study that utilized sensitivity to proteinase K-mediated

    proteolysis to explore whether IL-1a underwent a conformationalchange as a consequence of removal of its N terminus (Hazuda

    et al., 1991). Although the latter study did not explore the func-

    tional effects of IL-1a proteolysis in any detail, it did suggest

    that proteolysis of this cytokine initiated a profound conforma-

    tional change in the molecule from a proteinase K-sensitive to

    a proteinase K-insensitive state (Hazuda et al., 1991). This

    change is most likely reflected in an altered conformation that

    has increased IL-1 receptor affinity.

    There are a number of scenarios where Gzm B may encounter

    IL-1a. First, in a classical CTL/NK killing reaction, where the

    target cell expresses IL-1a, this cytokine may become pro-

    cessed within the target cell and exacerbate proinflammatory

    responses if released from dying cells prior to clearance of the

    latter by phagocytes. In this context, it is relevant to note thatBid was substantially more sensitive to Gzm B-mediated prote-

    olysis than IL-1a. Thus, although the concentration of Gzm B

    achieved in a CTL/NK killing reaction is unknown, at low nano-

    molar concentrations of Gzm B robust proteolysis of Bid and

    apoptosis might occur without significant proteolysis of IL-1a,

    with proteolysis of the latter becoming more significant upon

    exposure to higher granzyme concentrations.

    Second, there arenow a numberof studies suggesting that NK

    cells and CTLs, as well as other cell types, may actively secrete

    granzymes into the extracellular space (Spaeny-Dekking et al.,

    1998; Prakash et al., 2009). In this scenario, where injury or infec-

    tion has resulted in necrosis and release of IL-1a, Gz m B

    released from CTL/NK cellsor indeed one of the growing

    number of noncytotoxic cell types that have been found to

    express this granzymecould process this cytokine and

    enhance its potency. In this context it is noteworthy to mention

    that several studies have detected circulating Gzm B at elevated

    levels in rheumatoid arthritis patients and in patients with melioi-

    dosis (Spaeny-Dekking et al., 1998; Tak et al., 1999; Lauw et al.,

    2000). Furthermore, Gzm B expression and secretion has been

    repeatedly detected in noncytotoxic, proinflammatory immune

    cells such as macrophages, plasmacytoid dendritic cells, acti-

    vated mast cells, basophils, B lymphocytes, macrophages, ker-

    atinocytes, platelets, human articular chondrocytes, and breast

    carcinoma cells (reviewed in Afonina et al., 2010), further sug-

    gesting that this granzyme may have alternative, noncytotoxic

    functions. Proteases such as elastase and mast cell chymaseare abundantly released via degranulation of neutrophils and

    mast cells during inflammatory reactions, providing a ready

    means by which these proteases may encounter IL-1a released

    through cell damage. Indeed, several recent studies have shown

    that IL-1a is a major initiator of inflammatory responses to

    necrotic cells (Chen et al., 2007; Kono and Rock, 2008; Kono

    et al., 2010).

    A recent study found that whereas IL-1a was readily released

    from cells undergoing necrosis, this cytokine was retained within

    the nucleus of cells undergoing apoptosis (Cohen et al., 2010).

    Retention of IL-1a within the nucleus appears to be facilitated

    by a nuclear localization signal within its N terminus, the region

    that is separated from the molecule through Gzm B-dependent

    proteolysis. Thus, in addition to increasing biological potency

    through enhancing IL-1a binding to its receptor, proteolysis of

    IL-1a by proteases such as Gzm B may also facilitate its release

    by overriding the nuclear retention mechanisms that appear tooperate during some forms of cell death.

    In summary, in addition to acting as a direct instigator of cell

    death upon delivery into target cells, Gzm B may also serve in

    a hitherto unsuspected capacity as an amplifier of inflammatory

    responses through restricted proteolysis of IL-1a. We have also

    shown that proteolysis of IL-1a by several other proteases, each

    of which cleave this cytokine at distinct sites within its N

    terminus, enhanced the biological potency of this cytokine.

    IL-1a processing to the mature $17 kDa protein acts as a switch

    to enhance the proinflammatory properties of this cytokine, from

    a basal level of activity to a hyperactive one. Thus, in common

    with IL-1b, the biological activity of IL-1a is also regulated

    through restricted proteolysis.

    EXPERIMENTAL PROCEDURES

    Reagents

    Recombinant caspase-1, -3, -4, -5, and -7 were expressed and purified from

    bacteria as described previously (Luthi et al., 2009). Recombinant polyhisti-

    dine-tagged human PI-9 was expressed and purified from bacteria according

    to standard procedures. Recombinant wild-type Gzm B (both human and

    murine) and the catalytically inactive mutant Gzm BSA were expressed and

    purified from Pichia pastoris yeast as described previously (Cullen et al.,

    2007). cDNA clones encoding full-length human and murine IL-1a were

    obtained from OriGene and subcloned into pET45b. Recombinant IL-1a and

    derivatives were expressed in E. coli and purified as described in Supple-

    mental Experimental Procedures. Human calpain-1 and human mast cell

    chymase were purchased from Calbiochem, human elastase was purchased

    from SERVA. Polyclonal anti-IL-1a antibody was from AbD Serotec, goat

    anti-rabbit HRPO secondary antibody was from Jackson ImmunoResearch.

    Ac-IETD-AFC was purchased from Bachem (UK). Lipopolysaccharides

    (LPS), mannan, lipoteichoic acid (LTA), and muramyl dipeptide (MDP) were

    purchased from Sigma (Ireland) Ltd. Pam3CSK4, flagellin, and CpG were

    purchased fromInvivogen (France).Unlessotherwisestated,all other reagents

    were purchased from Sigma (Ireland) Ltd.

    Cell Culture

    HeLa cells were cultured in RPMI media supplemented with 5% fetal calf

    serum, and YT cells were cultured in RPMI supplemented with 10% serum

    and HUVEC (PromoCell, Germany)in endothelial cellgrowthmedia withadded

    growth supplement (PromoCell, Germany). Cells were cultured at 37C in

    humidified atmosphere with 5% CO2.

    Cell-Killing Assays

    HeLa cells were transiently transfected with IL-1a expression plasmid using

    GeneJuice transfection reagent (Merck, Ireland). For NK-mediated killing,

    HeLa cells were incubated with YT or NK-92 cells at 20:1 and 5:1 effector-

    to-target ratios, respectively. NK cells were removed from the target cell

    monolayer after 35 hr, and HeLa cells were further incubated for a total of

    24 hr. Celllysateswere generated at 23 107 cells/mland analyzed by western

    blotting.

    Animals

    BALB/c mice were purchased from Harlan (UK). Animal experiments were in

    accordance with the regulations of the Trinity College Dublin ethics committee

    and the Irish Department of Health. Granzyme B null mice, on the C57BL/6

    background, were bred at Academic Medical Center (AMC), Amsterdam,

    The Netherlands.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    276 Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc.

  • 7/27/2019 1-s2.0-S1097276511007179-main

    13/14

    Determination of Cytokine and Antibody Concentrations

    Cytokines were determined by ELISA as described previously (Luthi et al.,

    2009). Paired antibodies for mouse IL-4, IL-5, IL-10, and IFN-g were

    purchased fromBD Pharmingen(UK); human IL-6,IL-8, and GM-CSF DuoSets

    were purchased from R&D Systems (UK). Serum immunoglobulin titers were

    determined as described previously (Lavelle et al., 2001).

    IL-1a Bioactivity Assay

    HeLa cells were plated at a density of 4 3 104 cells per well, and primary

    HUVECs were platedat 23 104

    cellsper well on24-wellplates. 24hr latercells

    were incubated with the indicated concentrations of recombinant IL-1a, or

    recombinant annexin V (as a control protein), and culture supernatants were

    collected at the specified time-points for quantitative cytokine analysis by

    ELISA.

    Bronchoalveolar Lavage Fluid Analysis

    Bronchoalveolar lavage fluid samples from control individuals or patients

    with cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD), or

    bronchiectasis were collected as described previously (Reeves et al., 2010).

    Informed patient consent was obtained and ethical approval was granted by

    the Beaumont Hospital Institutional Review Board.

    SUPPLEMENTAL INFORMATION

    Supplemental Information includes three figures and Supplemental

    Experimental Procedures and can be found with this article online at

    doi:10.1016/j.molcel.2011.07.037.

    ACKNOWLEDGMENTS

    Work in the Martin laboratory is supported by SRC and PI grants from Science

    Foundation Ireland (07/SRC/B1144 and 08/IN.1/B2031) and The Wellcome

    Trust UK (082749). The Lavelle laboratory is supportedby Science Foundation

    Ireland grant 07/SRC/B1144. I.S.A. was a Health Research Board of Ireland

    scholar. S.J.M. is a Science Foundation Ireland Principal Investigator. The

    authors have no conflicting financial interests to declare.

    Received: September 1, 2010

    Revised: June 17, 2011

    Accepted: July 15, 2011

    Published: October 20, 2011

    REFERENCES

    Afonina, I.S., Cullen, S.P., and Martin, S.J. (2010). Cytotoxicand non-cytotoxic

    roles of the CTL/NK protease granzyme B. Immunol. Rev. 235, 105116.

    Bertelsen, M., and Sanfridson, A. (2007). TAB1 modulates IL-1alpha mediated

    cytokine secretion but is dispensable for TAK1 activation. Cell. Signal. 19,

    646657.

    Black, R.A., Kronheim, S.R., Cantrell, M., Deeley, M.C., March, C.J., Prickett,

    K.S., Wignall, J., Conlon, P.J., Cosman, D., Hopp, T.P., et al. (1988).Generation of biologically active interleukin-1 beta by proteolytic cleavage of

    the inactive precursor. J. Biol. Chem. 263, 94379442.

    Carruth, L.M., Demczuk, S., and Mizel, S.B. (1991). Involvement of a calpain-

    like protease in the processing of the murine interleukin 1 alpha precursor.

    J. Biol. Chem. 266, 1216212167.

    Chen, C.J., Kono,H., Golenbock, D.,Reed, G.,Akira,S., and Rock, K.L.(2007).

    Identification of a key pathway required for the sterile inflammatory response

    triggered by dying cells. Nat. Med. 13, 851856.

    Cohen, I.,Rider,P., Carmi, Y., Braiman, A.,Dotan, S.,White, M.R., Voronov,E.,

    Martin, M.U., Dinarello, C.A., and Apte, R.N. (2010). Differential release of

    chromatin-bound IL-1alpha discriminates between necrotic and apoptotic

    cell death by the ability to induce sterile inflammation. Proc. Natl. Acad. Sci.

    USA107, 25742579.

    Cullen, S.P., Adrain, C., Luthi, A.U., Duriez, P.J., and Martin, S.J. (2007).

    Human and murine granzyme B exhibit divergent substrate preferences.

    J. Cell Biol. 176, 435444.

    Dinarello, C.A. (1996). Biologic basis for interleukin-1 in disease. Blood 87,

    20952147.

    Froelich, C.J., Pardo, J., and Simon, M.M. (2009). Granule-associated serine

    proteases: granzymes might not just be killer proteases. Trends Immunol.

    30, 117123.

    Hazuda, D.J., Strickler, J., Simon, P., and Young, P.R. (1991). Structure-func-

    tion mapping of interleukin 1 precursors. Cleavage leads to a conformational

    change in the mature protein. J. Biol. Chem. 266, 70817086.

    Hernandez-Pigeon, H., Jean, C., Charruyer, A., Haure, M.J., Baudouin, C.,

    Charveron, M., Quillet-Mary, A., and Laurent, G. (2007). UVA induces gran-

    zymeB in human keratinocytes through MIF: implication in extracellular matrix

    remodeling. J. Biol. Chem. 282, 81578164.

    Irmler, M., Hertig, S., MacDonald, H.R., Sadoul, R., Becherer, J.D., Proudfoot,

    A., Solari, R., and Tschopp, J. (1995). Granzyme A is an interleukin 1 beta-con-

    verting enzyme. J. Exp. Med. 181, 19171922.

    Kagi, D., Ledermann, B., Burki, K., Seiler, P., Odermatt, B., Olsen, K.J.,

    Podack, E.R., Zinkernagel,R.M.,and Hengartner, H. (1994). Cytotoxicitymedi-

    ated by T cells and natural killer cells is greatly impaired in perforin-deficient

    mice. Nature 369, 3137.

    Kim, W.J., Kim, H.,Suk,K., andLee,W.H. (2007). Macrophages expressgran-

    zyme B in the lesion areas of atherosclerosis and rheumatoid arthritis.

    Immunol. Lett. 111, 5765.

    Kobayashi, Y., Yamamoto, K., Saido, T., Kawasaki, H., Oppenheim, J.J., and

    Matsushima, K. (1990). Identification of calcium-activated neutral protease as

    a processing enzyme of human interleukin 1 alpha. Proc. Natl. Acad. Sci. USA

    87, 55485552.

    Kono, H., and Rock, K.L. (2008). How dying cells alert the immune system to

    danger. Nat. Rev. Immunol. 8, 279289.

    Kono, H., Karmarkar, D., Iwakura, Y., and Rock, K.L. (2010). Identification of

    the cellular sensor that stimulates the inflammatory response to sterile cell

    death. J. Immunol. 184, 44704478.

    Kurt-Jones, E.A., Beller, D.I., Mizel, S.B., and Unanue, E.R. (1985).Identification of a membrane-associated interleukin 1 in macrophages. Proc.

    Natl. Acad. Sci. USA 82, 12041208.

    Lauw, F.N., Simpson, A.J., Hack, C.E., Prins, J.M., Wolbink, A.M.,

    van Deventer, S.J., Chaowagul, W., White, N.J., and van Der Poll, T. (2000).

    Soluble granzymes are released during human endotoxemia and in patients

    with severe infection due to gram-negative bacteria. J. Infect. Dis. 182,

    206213.

    Lavelle, E.C., Grant, G., Pusztai, A., Pfuller, U., and OHagan, D.T. (2001). The

    identification of plant lectins with mucosal adjuvant activity. Immunology 102,

    7786.

    Lowin, B., Beermann, F., Schmidt, A., and Tschopp, J. (1994). A null mutation

    in theperforingene impairscytolytic T lymphocyte-and naturalkillercell-medi-

    ated cytotoxicity. Proc. Natl. Acad. Sci. USA 91, 1157111575.

    Luthi, A.U., Cullen, S.P., McNeela, E.A., Duriez, P.J., Afonina, I.S., Sheridan,

    C., Brumatti, G., Taylor, R.C., Kersse, K., Vandenabeele, P., et al. (2009).Suppression of interleukin-33 bioactivity through proteolysis by apoptotic

    caspases. Immunity 31, 8498.

    Matsushima, K., Taguchi, M., Kovacs, E.J., Young, H.A., and Oppenheim, J.J.

    (1986). Intracellularlocalization of human monocyteassociated interleukin 1 (IL

    1) activityand release of biologically active IL 1 from monocytes by trypsin and

    plasmin. J. Immunol. 136, 28832891.

    McElvaney, N.G., Hubbard, R.C., Birrer, P., Chernick, M.S., Caplan, D.B.,

    Frank, M.M., and Crystal, R.G. (1991). Aerosol alpha 1-antitrypsin treatment

    for cystic fibrosis. Lancet 337, 392394.

    Metkar, S.S., Menaa, C., Pardo, J., Wang, B., Wallich, R., Freudenberg, M.,

    Kim, S., Raja, S.M., Shi, L., Simon, M.M., and Froelich, C.J. (2008). Human

    and mouse granzyme A induce a proinflammatory cytokine response.

    Immunity 29, 720733.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity

    Molecular Cell 44, 265278, October 21, 2011 2011 Elsevier Inc. 277

    http://dx.doi.org/doi:10.1016/j.molcel.2011.07.037http://dx.doi.org/doi:10.1016/j.molcel.2011.07.037
  • 7/27/2019 1-s2.0-S1097276511007179-main

    14/14

    Mizutani, H., Schechter, N., Lazarus, G., Black, R.A., and Kupper, T.S. (1991).

    Rapid and specific conversion of precursor interleukin 1 beta (IL-1 beta) to an

    active IL-1 species by human mast cell chymase. J. Exp. Med. 174, 821825.

    Mosley, B., Urdal, D.L., Prickett, K.S., Larsen, A., Cosman, D., Conlon, P.J.,

    Gillis, S., and Dower, S.K. (1987a). The interleukin-1 receptor binds the human

    interleukin-1 alpha precursor but not the interleukin-1 beta precursor. J. Biol.

    Chem. 262, 29412944.

    Mosley, B., Dower, S.K., Gillis, S., and Cosman, D. (1987b). Determination of

    the minimum polypeptide lengths of the functionally active sites of human

    interleukins 1 alpha and 1 beta. Proc. Natl. Acad. Sci. USA 84, 45724576.

    Prakash, M.D., Bird, C.H., and Bird, P.I. (2009). Active and zymogen forms of

    granzyme B are constitutively released from cytotoxic lymphocytes in the

    absence of target cell engagement. Immunol. Cell Biol. 87, 249254.

    Reeves, E.P., Williamson, M., Byrne, B., Bergin, D.A., Smith, S.G., Greally, P.,

    OKennedy, R., ONeill,S.J., and McElvaney,N.G. (2010). IL-8dictatesglycos-

    aminoglycan binding and stability of IL-18 in cystic fibrosis. J. Immunol. 184,

    16421652.

    Rhim, J.H., Kim, S.A., Lee,J.E., Kim, D.J., Chung, H.K., Shin, K.J., and Chung,

    J. (2008). Cancer cell-derived IL-1alpha induces IL-8 release in endothelial

    cells. J. Cancer Res. Clin. Oncol. 134, 4550.

    Sedelies, K.A., Sayers, T.J., Edwards, K.M., Chen, W., Pellicci, D.G., Godfrey,

    D.I., and Trapani, J.A. (2004). Discordant regulation of granzyme H and gran-

    zyme B expression in human lymphocytes. J. Biol. Chem. 279, 2658126587.

    Sower, L.E., Froelich, C.J., Allegretto, N., Rose, P.M., Hanna, W.D., and

    Klimpel, G.R. (1996a). Extracellular activities of human granzyme A.

    Monocyte activation by granzyme A versus alpha-thrombin. J. Immunol.

    156, 25852590.

    Sower, L.E., Klimpel, G.R., Hanna, W., and Froelich, C.J. (1996b). Extracellular

    activities of human granzymes. I. Granzyme A induces IL6 and IL8 production

    in fibroblast and epithelial cell lines. Cell. Immunol. 171, 159163.

    Spaeny-Dekking, E.H., Hanna, W.L., Wolbink, A.M., Wever, P.C., Kummer,

    J.A., Swaak, A.J., Middeldorp, J.M., Huisman, H.G., Froelich, C.J., and

    Hack, C.E. (1998). Extracellular granzymes A and B in humans: detection of

    native species during CTL responses in vitro and in vivo. J. Immunol. 160,

    36103616.

    Strik, M.C., de Koning, P.J.,Kleijmeer, M.J., Bladergroen,B.A., Wolbink, A.M.,

    Griffith, J.M., Wouters, D., Fukuoka, Y., Schwartz, L.B., Hack, C.E., et al.

    (2007). Human mastcellsproduce and releasethe cytotoxic lymphocyte asso-

    ciated protease granzyme B upon activation. Mol. Immunol. 44, 34623472.

    Tak, P.P., Spaeny-Dekking, L., Kraan, M.C., Breedveld, F.C., Froelich, C.J.,

    and Hack, C.E. (1999). The levels of soluble granzyme A and B are elevated

    in plasma and synovial fluid of patients with rheumatoid arthritis (RA). Clin.

    Exp. Immunol. 116, 366370.

    Thornberry, N.A., Bull, H.G., Calaycay, J.R., Chapman, K.T., Howard, A.D.,

    Kostura, M.J., Miller, D.K., Molineaux, S.M., Weidner, J.R., Aunins, J., et al.

    (1992). A novel heterodimeric cysteine protease is required for interleukin-1

    beta processing in monocytes. Nature 356, 768774.

    Tschopp, C.M., Spiegl, N., Didichenko, S., Lutmann, W., Julius, P., Virchow,

    J.C., Hack, C.E., and Dahinden, C.A. (2006). Granzyme B, a novel mediator

    of allergic inflammation: its induction and release in blood basophils and

    human asthma. Blood 108, 22902299.

    Watanabe,N., and Kobayashi, Y. (1994). Selective releaseof a processed form

    of interleukin 1 alpha. Cytokine 6, 597601.

    Molecular Cell

    Proteolysis of IL-1a Potentiates Bioactivity