+ All Categories
Home > Documents > Granzyme B Is Critical for T Cell Receptor-Induced Cell Death of Type 2 Helper T Cells

Granzyme B Is Critical for T Cell Receptor-Induced Cell Death of Type 2 Helper T Cells

Date post: 28-Apr-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
11
Immunity 25, 237–247, August 2006 ª2006 Elsevier Inc. DOI 10.1016/j.immuni.2006.06.011 Granzyme B Is Critical for T Cell Receptor-Induced Cell Death of Type 2 Helper T Cells Satish Devadas, 1,4 Jyoti Das, 1,4 Catherine Liu, 1 Liying Zhang, 1 Arthur I. Roberts, 1 Zui Pan, 2 Paul A. Moore, 3,5 Gobardhan Das, 1 and Yufang Shi 1, * 1 Department of Molecular Genetics, Microbiology, and Immunology 2 Department of Physiology and Biophysics Robert Wood Johnson Medical School University of Medicine and Dentistry of New Jersey 661 Hoes Lane Piscataway, New Jersey 08854 3 Human Genome Sciences, Inc. Rockville, Maryland 20850 Summary Although CD95L is required for T cell receptor (TCR)- induced cell death (TCR-ICD) in T helper 1 cells, the molecular mechanisms mediating TCR-ICD in Th2 cells are unknown. We found that death receptors were not involved in TCR-ICD of Th2 cells because blocking their cognate ligands had no effect on apo- ptosis of activated Th2 cells. Furthermore, we showed that caspases were not actively involved in TCR-ICD of Th2 cells. However, inhibition of granzyme B (GrB) ac- tivity abolished TCR-ICD in Th2 cells but not Th1 cells. Likewise, Th2 cells derived from GrB-deficient mice were resistant to TCR-ICD, and GrB deficiency or inhi- bition of GrB activity consequently enhanced the pro- duction of Th2 cytokines. GrB-deficient mice exhibited increased susceptibility to allergen-induced asthma. Thus, GrB plays a critical role in the TCR-ICD of Th2 cells. Introduction Mature T cells are at rest until they recognize antigen and can survive for several months in rodents and years in humans (Sprent and Tough, 1994). Upon T cell recep- tor (TCR) stimulation, T cells become activated and initi- ate specific immune responses. These activation signals are often robust: with an appropriate costimulus, contin- uous TCR stimulation can cause cell numbers to in- crease by a factor of 10 9 to 10 11 in just 60 days (Levine et al., 1997). Unchecked T cell expansion after TCR acti- vation is detrimental. Thus, effective control mecha- nisms have evolved alongside this extraordinary po- tential for T cell expansion. Activated T cells require substantial amounts of cytokines, especially interleu- kin-2 (IL-2) and IL-7, for sustained survival. Once the ac- tivation signals diminish, either by depletion of antigen or by increased inhibitory signals, cytokine production drops off, resulting in death or clonal contraction of T cells by cytokine deprivation (Akbar and Salmon, 1997; Janssen et al., 2000; Vella et al., 1998). In addition, pre- viously activated T cells undergo apoptosis after en- gagement of their TCR, helping to maintain peripheral tolerance and cellular homeostasis (Lenardo, 1991; Mercep et al., 1988; Shi et al., 1989). The discovery of the T helper 1 (Th1) and Th2 dichot- omy was a landmark in the understanding of the mech- anisms regulating immune responses during infections, autoimmunity, and tumorigenesis (Mosmann et al., 1986; Murphy et al., 2000). It is well established that the cytokine milieu plays a critical role in helper T cell dif- ferentiation (Paul and Seder, 1994). Modulation of the availability of key cytokines such as IL-4, interferon-g, (IFN-g), IL-12, and IL-10 either by antibody neutralization or through exogenous supplementation leads to distinct patterns of helper T cell differentiation (Mosmann and Sad, 1996; Zhang et al., 2003). CD4 + helper T cells differ- entiate into two phenotypically distinct subpopulations: Th1 cells, which are promoted by IL-12 and IFN-g in the absence of IL-4, and Th2 cells, which are favored by IL-4 and IL-10 in the absence of IL-12 and IFN-g (Constant and Bottomly, 1997; Mosmann et al., 1986; Paul and Seder, 1994). The granzyme family of serine proteases are pivotal mediators of apoptosis utilized by cytotoxic T lympho- cytes and natural killer cells against targets such as virus-infected cells and tumor cells (Lieberman, 2003). The most demonstrated function of granzymes is their induction of target cell apoptosis after entry via a still- uncharacterized perforin-assisted mechanism (Lieber- man, 2003; Raja et al., 2003). Interestingly, granzymes can also act upon the cells harboring them; it has been found that CD16 ligation-induced apoptosis of NK cells results from the release of granzyme B (GrB) from cyto- plasmic granules (Ida et al., 2003). GrB has also been re- ported in the cytosol of CD2-activated cytotoxic T cells (Bidere et al., 2002). In addition, granzymes may be in- volved in glucocorticoid-induced apoptosis in B leuke- mia cells (Yamada et al., 2003). The presence of GrB in CD4 + T cells was reported by several studies soon after its discovery (Fruth et al., 1987) and was believed to be associated with their cytotoxic activity. The function of GrB in CD4 + helper T cells has been largely ignored, however, and remains a mystery, even as much prog- ress has been made in elucidating the function of gran- zymes in NK cells and cytotoxic T cells. Th1 and Th2 cells differ not only in their cytokine- secretion profiles, but also in their tendencies to un- dergo TCR-induced cell death (TCR-ICD) and in their ex- pression of CD95L (Oberg et al., 1997; Ramsdell et al., 1994). We have reported that CD95L and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) are differentially expressed in Th1 and Th2 cells after TCR activation; TRAIL is expressed mainly by Th2 cells, and CD95L is detected preferentially in Th1 cells (Zhang et al., 2003). Blockade of TRAIL, CD95L, or tumor necro- sis factor-a (TNF-a) did not markedly reduce death in Th2 cells. Hence, TCR-ICD in Th2 cells occurs through an unidentified mechanism. Here we showed that Th2 cells, but not Th1 cells, treated with a GrB inhibitor or derived from GrB-deficient mice were resistant to TCR-ICD. *Correspondence: [email protected] 4 These authors contributed equally to this work. 5 Present address: Celera Genomics Rockville, 45 West Gude Drive, Rockville, Maryland 20850.
Transcript

Immunity 25, 237–247, August 2006 ª2006 Elsevier Inc. DOI 10.1016/j.immuni.2006.06.011

Granzyme B Is Criticalfor T Cell Receptor-InducedCell Death of Type 2 Helper T Cells

Satish Devadas,1,4 Jyoti Das,1,4 Catherine Liu,1

Liying Zhang,1 Arthur I. Roberts,1 Zui Pan,2

Paul A. Moore,3,5 Gobardhan Das,1 and Yufang Shi1,*1Department of Molecular Genetics, Microbiology,

and Immunology2Department of Physiology and BiophysicsRobert Wood Johnson Medical SchoolUniversity of Medicine and Dentistry of New Jersey661 Hoes LanePiscataway, New Jersey 088543Human Genome Sciences, Inc.Rockville, Maryland 20850

Summary

Although CD95L is required for T cell receptor (TCR)-induced cell death (TCR-ICD) in T helper 1 cells, the

molecular mechanisms mediating TCR-ICD in Th2

cells are unknown. We found that death receptorswere not involved in TCR-ICD of Th2 cells because

blocking their cognate ligands had no effect on apo-ptosis of activated Th2 cells. Furthermore, we showed

that caspases were not actively involved in TCR-ICD ofTh2 cells. However, inhibition of granzyme B (GrB) ac-

tivity abolished TCR-ICD in Th2 cells but not Th1 cells.Likewise, Th2 cells derived from GrB-deficient mice

were resistant to TCR-ICD, and GrB deficiency or inhi-bition of GrB activity consequently enhanced the pro-

duction of Th2 cytokines. GrB-deficient mice exhibitedincreased susceptibility to allergen-induced asthma.

Thus, GrB plays a critical role in the TCR-ICD of Th2cells.

Introduction

Mature T cells are at rest until they recognize antigenand can survive for several months in rodents and yearsin humans (Sprent and Tough, 1994). Upon T cell recep-tor (TCR) stimulation, T cells become activated and initi-ate specific immune responses. These activation signalsare often robust: with an appropriate costimulus, contin-uous TCR stimulation can cause cell numbers to in-crease by a factor of 109 to 1011 in just 60 days (Levineet al., 1997). Unchecked T cell expansion after TCR acti-vation is detrimental. Thus, effective control mecha-nisms have evolved alongside this extraordinary po-tential for T cell expansion. Activated T cells requiresubstantial amounts of cytokines, especially interleu-kin-2 (IL-2) and IL-7, for sustained survival. Once the ac-tivation signals diminish, either by depletion of antigenor by increased inhibitory signals, cytokine productiondrops off, resulting in death or clonal contraction of Tcells by cytokine deprivation (Akbar and Salmon, 1997;Janssen et al., 2000; Vella et al., 1998). In addition, pre-

*Correspondence: [email protected] These authors contributed equally to this work.5 Present address: Celera Genomics Rockville, 45 West Gude Drive,

Rockville, Maryland 20850.

viously activated T cells undergo apoptosis after en-gagement of their TCR, helping to maintain peripheraltolerance and cellular homeostasis (Lenardo, 1991;Mercep et al., 1988; Shi et al., 1989).

The discovery of the T helper 1 (Th1) and Th2 dichot-omy was a landmark in the understanding of the mech-anisms regulating immune responses during infections,autoimmunity, and tumorigenesis (Mosmann et al.,1986; Murphy et al., 2000). It is well established thatthe cytokine milieu plays a critical role in helper T cell dif-ferentiation (Paul and Seder, 1994). Modulation of theavailability of key cytokines such as IL-4, interferon-g,(IFN-g), IL-12, and IL-10 either by antibody neutralizationor through exogenous supplementation leads to distinctpatterns of helper T cell differentiation (Mosmann andSad, 1996; Zhang et al., 2003). CD4+ helper T cells differ-entiate into two phenotypically distinct subpopulations:Th1 cells, which are promoted by IL-12 and IFN-g in theabsence of IL-4, and Th2 cells, which are favored by IL-4and IL-10 in the absence of IL-12 and IFN-g (Constantand Bottomly, 1997; Mosmann et al., 1986; Paul andSeder, 1994).

The granzyme family of serine proteases are pivotalmediators of apoptosis utilized by cytotoxic T lympho-cytes and natural killer cells against targets such asvirus-infected cells and tumor cells (Lieberman, 2003).The most demonstrated function of granzymes is theirinduction of target cell apoptosis after entry via a still-uncharacterized perforin-assisted mechanism (Lieber-man, 2003; Raja et al., 2003). Interestingly, granzymescan also act upon the cells harboring them; it has beenfound that CD16 ligation-induced apoptosis of NK cellsresults from the release of granzyme B (GrB) from cyto-plasmic granules (Ida et al., 2003). GrB has also been re-ported in the cytosol of CD2-activated cytotoxic T cells(Bidere et al., 2002). In addition, granzymes may be in-volved in glucocorticoid-induced apoptosis in B leuke-mia cells (Yamada et al., 2003). The presence of GrB inCD4+ T cells was reported by several studies soon afterits discovery (Fruth et al., 1987) and was believed to beassociated with their cytotoxic activity. The function ofGrB in CD4+ helper T cells has been largely ignored,however, and remains a mystery, even as much prog-ress has been made in elucidating the function of gran-zymes in NK cells and cytotoxic T cells.

Th1 and Th2 cells differ not only in their cytokine-secretion profiles, but also in their tendencies to un-dergo TCR-induced cell death (TCR-ICD) and in their ex-pression of CD95L (Oberg et al., 1997; Ramsdell et al.,1994). We have reported that CD95L and tumor necrosisfactor (TNF)-related apoptosis-inducing ligand (TRAIL)are differentially expressed in Th1 and Th2 cells afterTCR activation; TRAIL is expressed mainly by Th2 cells,and CD95L is detected preferentially in Th1 cells (Zhanget al., 2003). Blockade of TRAIL, CD95L, or tumor necro-sis factor-a (TNF-a) did not markedly reduce death in Th2cells. Hence, TCR-ICD in Th2 cells occurs through anunidentified mechanism. Here we showed that Th2 cells,but not Th1 cells, treated with a GrB inhibitor or derivedfrom GrB-deficient mice were resistant to TCR-ICD.

Immunity238

Figure 1. Kinetics of TCR-ICD in Th1 and Th2 Cells

Helper T cell subsets differentiated from BALB/c splenocytes were cultured on anti-CD3-coated plastic to initiate TCR-ICD as described in Ex-

perimental Procedures. Apoptosis was determined by flow cytometric analysis of cells costained with annexin V-FITC and propidium iodide (PI).

Cells positive for Annexin V and negative for PI are early apoptotic, as shown 8 hr postactivation (A). Anti-CD3-activated Th1 cells or Th2 cells

were analyzed at the indicated time points (B). Viability was determined by analysis of cell membrane permeability to PI and plotted against FSC

at 8 hr (C); values for multiple time points are shown (D). Values are means 6 SE from four experiments. TCR-ICD at each time point is significantly

different as determined by repeated measures ANOVA, p < 0.01.

We found that CD4+ T cells from OVA-immunized miceproduced more IL-4, IL-5, and IL-10 when treated witha GrB inhibitor. Similarly, GrB-deficient mice producedmore of these Th2 cytokines. Most importantly, GrB-deficient mice were found to be highly susceptible toallergen-induced asthma. We conclude that granzyme Bactivation is required for the TCR-ICD of Th2 cells, andthus likely plays a role in modulating the Th2 response.

Results

TCR-ICD Is More Rapid in Th1 Cells than in Th2 CellsStimulation of T cell hybridomas (Mercep et al., 1988; Shiet al., 1989) or restimulation of previously activated pri-mary T cells (Lenardo, 1991) usually leads to apoptosis.Previous studies revealed that Th2 cells are more resis-tant than Th1 cells to TCR-induced cell death (TCR-ICD)(Zhang et al., 1997), but the differences in their kineticsof apoptosis have not been formally determined. Wegenerated Th1 and Th2 cells from murine splenocytesex vivo and then restimulated them with immobilizedCD3 antibody to initiate TCR-ICD. Cell death was moni-tored by flow cytometry to detect Annexin V binding (ameasure of early apoptosis) and changes in light-scattercharacteristics, as well as subsequent membrane per-meability to propidium iodide (PI) (Figures 1A and 1C).Th1 cells died more quickly and more extensively thanTh2 cells, as detected by either technique (Figures 1B

and 1D). Furthermore, DNA fragmentation as indicatedby emergence of a hypodiploid PI-stained DNA peak oc-curred more quickly in Th1 cells at all time points from8 to 18 hr (data not shown). These results demonstratethat both Th1 and Th2 cells undergo apoptosis after re-stimulation; Th1 cell death, however, takes place morequickly and more profoundly. The difference in kineticsof TCR-ICD in Th1 and Th2 cells suggest a likely diver-gence in their mechanisms of apoptosis.

TCR-ICD in Th2 Cells Does Not Require TNF Family

Members or Major CaspasesTo determine whether ligands of the TNF superfamilyare involved in TCR-ICD of helper T cell subsets, weadded specific blocking agents to anti-CD3-restimu-lated Th1 or Th2 cells and observed the effect on apo-ptosis by DNA content analysis (Figure 2A). As ex-pected, the TCR-ICD of Th1 cells was inhibited byantibody against CD95L (MFL3) and by recombinant hu-man TR6 (DcR3), which binds to both CD95L and LIGHT(Migone et al., 2002). None of these agents, however, af-fected TCR-ICD in Th2 cells, corroborating our earlier re-port that apoptosis in Th1 cells relies mostly on CD95L(Zhang et al., 2003). In contrast, soluble TRAIL receptor,DR5, had no effect on either helper T cell subset. Thus,TRAIL is not involved in the TCR-ICD of Th2 cells, al-though it is expressed by these cells. As demonstratedearlier, TRAIL as expressed by Th2 cells can kill Th1

Granzyme B and Th2 Apoptosis239

Figure 2. Differential Caspase Activation Profiles and Responses to Apoptosis Inhibitors in Th1 and Th2 Cells

Differentiated Th1 cells or Th2 cells were plated on immobilized anti-CD3 to initiate TCR-ICD in the presence of the indicated inhibitors used at

optimal concentration: TR6 (100 ng/mL), DR5 (10 mg/mL), osteoprotegerin (OPG, 250 ng/mL), CD95L antibody (MFL3, 100 ng/mL), or z-VAD (5–

250 mM). The percentage of cells in late apoptosis at 16 hr was revealed by hypodiploid DNA content in permeabilized cells stained with PI and

analyzed by flow cytometry. Net increases in percentage of apoptotic cells above spontaneous levels were calculated and are presented as

means 6 SE of four experiments. Differences between Th1 and Th2 cells were found to be significant by Student’s t test (A and B): activation

of caspase-8 was measured by flow cytometric detection of a caspase 8-specific fluorogenic synthetic substrate. The difference between

Th1 and Th2 is significant by repeated measures ANOVA, p < 0.01 (C). General caspase activation was measured with a pan-caspase substrate,

zVAD-FITC. Dead cells were excluded by PI permeability (D).

cells, but not Th2 cells themselves (Zhang et al., 2003).Addition of osteoprotegerin (OPG), which blocks RANKLand TRAIL, also affected neither cell type (Figure 2A).Involvement of TNF-a was similarly ruled out by antibodyneutralization. Therefore, although CD95L is critical forthe TCR-ICD of Th1 cells, none of the known TNF familymembers is implicated in the TCR-ICD in Th2 cells, sug-gesting that Th2 cells employ a distinct mechanism ofTCR-ICD compared to Th1 cells.

To further dissect the mechanisms of TCR-ICD, cas-pase utilization in each helper T cell subset was ana-lyzed by adding z-VAD, which blocks the activity of sev-eral major caspases, especially caspases 1, 3, 8, and 9(Komoriya et al., 2000). We found that z-VAD signifi-cantly inhibited TCR-ICD in Th1 cells, but not Th2 cells,at all concentrations tested (Figure 2B); z-VAD at 100 mMor less was shown to be nontoxic during the 24 hr cultureas determined by cell-viability analysis and prolifera-tion (data not shown). Because TCR-ICD in Th1 cells isinhibited by z-VAD as well as by antibody againstCD95L, Th1 apoptosis must depend on the CD95-trig-gered caspase pathway. Therefore, we examined acti-vation kinetics of initiator caspase 8 by using relativelyspecific fluorogenic substrates. Caspase 8 was acti-vated markedly in Th1 cells, but weakly in Th2 cells(Figure 2C). General caspase activation, assayed via

the relatively broad substrate, z-VAD-FITC, was alsodramatically higher in Th1 cells compared to Th2 cells(Figure 2D). Therefore, although TCR-ICD of Th2 cellsexhibits the typical characteristics of TCR-ICD, it is notmediated by known ligands of the TNF superfamily,nor does it involve the major caspases. Because wehave reported that the TCR-ICD of Th2 cells does notoccur through fratricide (Zhang et al., 2003), it is likely tobe an internal process.

Role of Granzyme B in TCR-ICD of Th2 Cells

Granzymes can act upon the cells containing them, ashas been demonstrated in CD16-stimulated NK cells(Ida et al., 2003) and glucocorticoid-treated B leukemiacells (Yamada et al., 2003). Thus, we hypothesize thatgranzymes may be involved in Th2 cell death. One ofthese, granzyme B (GrB), is a serine protease that wasidentified in the granules of cytotoxic T cells and NKcells. To determine its possible role in Th2 cell TCR-ICD, we added a GrB inhibitor, z-AAD-CMK, to culturesof restimulated Th2 cells and examined its effect on ap-optosis. GrB inhibitor prevented apoptosis in a dose-dependent manner, whereas Th1 cell cultures wereunaffected (Figure 3A). A control peptide, z-FA-CMK,had no effect on either cell type. Furthermore, blockadeof cathepsin C (dipeptidyl peptidase I, DPP-I), whose

Immunity240

Figure 3. GrB Inhibition or GrB Deficiency Abolishes Caspase Activity and DNA Fragmentation in Th2 Cells

Cultures of differentiated Th1 cells or Th2 cells from BALB/c mice were restimulated with plastic bound anti-CD3 in the presence of graded con-

centrations of GrB inhibitor (z-AAD-CMK) or a negative control (z-FA-CMK), or a DPP-I inhibitor (Gly-Phe-CHN2) to prevent activation of cathep-

sin C, and the percentage of apoptotic cells was determined by hypodiploid DNA content after 16 hr (A and B). Differentiated Th1 cells or Th2 cells

from Gzmb2/2/DPGK-neo or 129/SvJ were restimulated with anti-CD3 for 8 hr, and caspase 3 activation was measured by flow cytometric de-

tection of caspase-specific fluorogenic synthetic substrates (C). The percentage of apoptotic cells after 16 hr with and without restimulation is

shown by hypodiploid DNA content (D).

activity is required for GrB activation (Meade et al.,2006), also reduced TCR-ICD in Th2 cells specifically(Figure 3B). All apoptotic characteristics, including An-nexin V binding, caspase activation, and loss of genomicDNA, were inhibited in Th2 cells by inhibition of GrB orcathepsin C (data not shown); Th1 cells were unaffected.

To further verify the role of granzyme B in TCR-ICD ofTh2 cells, we employed mice deficient in granzyme B(Revell et al., 2005). Th1 and Th2 cells from Gzmb2/2/DPGK-neo mice and control 129/SvJ mice were acti-vated with plate bound anti-CD3 and assessed for theactivation of caspases 3 and 8, and for TCR-ICD bythe appearance of a hypodiploid DNA peak (Figure 3D).TCR-induced activation of caspase 3 (Figure 3C) andcaspase 8 (not shown) was observed in Th1 cells derivedfrom both control and GzmB2/2 mice. In contrast, Th2cells derived from control mice had a moderate increasein caspase 3 activation, while Th2 cells from GzmB2/2

mice showed no activation of caspase 3 (Figure 3C) orother caspases, as detected by z-VAD-FITC (notshown), indicating that what little caspase activationdoes occur in Th2 cells is GrB dependent. Further, whileGzmB2/2 Th1 cells underwent normal TCR-ICD, Th2cells derived from these mice were resistant to TCR-ICD. As a control, Th2 cells similarly derived from wild-type 129/SvJ mice underwent normal TCR-ICD thatwas sensitive to DPP-I inhibitor (data not shown). Similarresults were obtained with GzmB2/2/+GK-neo mice,which are deficient in granzymes C and F, as well as

GrB (data not shown). Taken together, these resultsdemonstrate that the TCR-ICD of Th2 cells, but notTh1 cells, is likely to depend on a GrB-mediated apopto-sis pathway.

Differential Activity of GrB in Helper T Cell Subsets

Since we have shown that GrB inhibitors block TCR-ICDin Th2 cells, but not Th1 cells, and Th2 cells derived fromGrB knockout mice are resistant to TCR-ICD, we exam-ined GrB mRNA levels in Th1 and Th2 cells by Northernblotting analysis and real-time PCR. Surprisingly, bothresting and activated Th1 cells, as well as activatedTh2 cells, were found to express high levels of GrBmRNA. GrB mRNA was low in resting Th2 cells, but in-creased significantly upon activation (Figure 4A). Thisresult was verified by real-time PCR analysis. First,equal loading was determined by b-actin mRNA detec-tion, and then the Th1 or Th2 character of our cell prep-arations was confirmed by examining IFN-g and IL-4, re-spectively (Figure 4B). Finally, real-time PCR analysis ofGrB mRNA was found in the same unexpected patternas seen in our Northern blot (Figure 4B). Thus, the GrBmRNA expression pattern detected by both Northernanalysis and real-time PCR does not explain why GrB in-hibitors block TCR-ICD in Th2 cells but not Th1 cells.

GrB is sequestered in the cytoplasmic granules ofcells that harbor this enzyme (Catalfamo and Henkart,2003; Raja et al., 2003). Since we have shown that Th2cells are not killed by cell-cell contact (Zhang et al.,

Granzyme B and Th2 Apoptosis241

Figure 4. Differential Expression of Serine Protease Inhibitors and GrB in Th1 Cells and Th2 Cells

Differentiated helper T cell subsets were restimulated with immobilized anti-CD3 for 6 hr, and total mRNA was prepared and analyzed for GrB,

SPI-6, SPI-2A, IFN-g, FasL, GAPDH, and b-Actin by Northern blotting analysis (A) and real-time PCR (B). GAPDH or b-actin served as endogenous

controls to normalize total RNA. The presence of mRNA for IFN-g and IL-4, respectively, was used to verify that Th1 and Th2 cells were properly

differentiated. The results are representative of three experiments.

2003), it is likely that the effect of GrB on TCR-ICD is ex-erted by intracellular release of the enzyme. Therefore,we examined GrB degranulation in both Th1 cells andTh2 cells before and after stimulation with CD3 antibody.Because all granules possess LAMP-1 (lysosome-asso-ciated membrane protein-1, CD107a) (Alter et al., 2004;Peters et al., 1991), we used confocal microscopy to ex-amine for the colocalization of GrB (anti-GrB-PE) andLAMP-1 (anti-LAMP-1-FITC). As shown in Figure 5A,GrB and LAMP-1 colocalize in resting Th1 cells and

Th2 cells, shown in yellow in the overlaid images.Upon activation, GrB and LAMP-1 remain colocalizedin Th1 cells even after the cells have died, as indicatedby fragmented nuclei. In activated Th2 cells, separationof red and green fluorescence was seen. Furthermore,GrB appears in the nuclei of activated Th2 cells (Fig-ure 5A), demonstrating that GrB is being released fromthe granules. Therefore, activation of Th2 cells resultsin the intracellular degranulation of GrB. Interestingly,adding a cathepsin C (DPP-I) inhibitor (Gly-Phe-CHN2)

Figure 5. Differential GrB Degranulation in Th1 Cells and Th2 Cells

Th1 and Th2 cells were reactivated with anti-CD3 for 12 hr, permeabilized, and stained with anti-LAMP-1-FITC and anti-GrB-PE, then washed

and counterstained with Hoechst 33342 nuclear stain. The cells were then visualized by laser-scanning confocal microscopy. Images are as

follows: top left, Hoechst 33342 nuclear stain; top right, LAMP-1; bottom left, GrB-FITC; bottom right, their overlay. Degranulation of GrB is

visualized as the diffusion of the normal punctate distribution of red fluorescence into the rest of the cell including the nucleus, which can

be observed to a great extent only in activated Th2 cells. In the overlaid images, GrB degranulation is shown by the disassociation of GrB

(red) from LAMP-1 (green) (A). Addition of a DPP-I inhibitor to Th2 cell cultures prevented TCR-induced GrB degranulation (B). Th2 cells derived

from Gzmb2/2 and control mice were activated and stained to show the specificity of GrB staining. There is no GrB staining in cells derived from

Gzmb2/2 mice (C).

Immunity242

Figure 6. Effect of Granzyme B on Th1-Th2 Cytokine Production

Gzmb2/2 and 129/SvJ mice were immunized with 10 mg OVA in ALUM, and cultures of CD4+ splenocytes were assayed for IFN-g, IL-4, IL-5, and

IL-13; IFN-g was not detectable. Differences in cytokine levels between control and Gzmb2/2 mice were analyzed (A). BALB/c mice were immu-

nized by tail base injection of 10 mg OVA in incomplete Freund’s adjuvant. After 7 days, splenocytes were stimulated (7.5 3 106 cells/mL) with OVA

(100 mg/mL) in the presence or absence of z-VAD or GrB inhibitor (each at 100 mM). Supernatant medium was collected after 48 hr and assayed for

IFN-g and IL-4. (B) Th1 cells or Th2 cells differentiated from BALB/c mouse splenocytes were reactivated with anti-CD3 in the presence of the

indicated inhibitors at optimal concentrations as in Figure 2. Supernatants were collected after 48 hr and analyzed for the indicated cytokines via

a multiplexed assay (C and D). Values represent means 6 SE of three experiments. Comparisons by Student’s t test are indicated.

blocked TCR-induced degranulation in Th2 cells(Figure 5B). The specificity of the GrB antibody was as-certained by comparing staining of Th2 cells derivedfrom 129/SvJ and GzmB2/2 mice. Upon activation withanti-CD3, the red staining (anti-GrB-PE) was observedonly in cells derived from 129/SvJ mice, not those fromGzmB2/2 mice (Figure 5C). Similarly, GrB staining wasnot observed in activated Th1 or resting Th1 and Th2cells from GzmB2/2 mice (data not shown). These dataprovide direct evidence for TCR-induced degranulationof GrB uniquely in Th2 cells.

Recent studies have revealed that cells possessinggranzymes generally also have inhibitors or mecha-nisms to inactivate them in order to prevent autodes-truction (Bots et al., 2005). Hence, we compared the ex-pression of certain serine protease inhibitors in Th1 cellsand Th2 cells. Serine protease inhibitor (SPI)-6, whichspecifically inhibits granzyme B activity, was found to in-crease after TCR activation only in Th1 cells (Figure 4A).Interestingly, SPI-2A, which inhibits both caspases 3and 9, was found to increase only in activated Th2 cells.Thus, while Th1 cells express some GrB, it remains con-fined to granules; in case of minor leakage, GrB activ-ity would be blocked by SPI-6. Real-time PCR analysisalso demonstrated this divergent pattern of SPI-6 andSPI-2A expression among activated Th1 and Th2 cells(data not shown). Thus, the preferential expression ofSPI-6 by Th1 cells may provide another defense mecha-nism against GrB-induced death in these cells. How-ever, we currently have no direct evidence to supportsuch a role for SPI-6.

Modulation of GrB Activity Favors the Development

of Th2-Type Immune ResponsesOur results show that the TCR-ICD of Th1 cells is medi-ated by CD95L, while GrB mediates Th2 cell death. Ourprevious studies have shown that CD4+ T cells derivedfrom lpr mice produce more Th1 cytokines under unbi-ased conditions, indicating that Th1 cell expansion is fa-vored by the absence of CD95L (Zhang et al., 2003). Toexamine the role of GrB in influencing the type of im-mune response in vivo, GzmB2/2 mice and control129/SvJ mice were immunized with OVA in alum (see Ex-perimental Procedures). CD4+ cells were isolated 5 dayspost-boost and restimulated with OVA in vitro for 48 hr,and then supernatants were assayed for cytokine levels.We found that IL-4, IL-5, and IL-13 were significantlyhigher in Gzmb2/2 mice (Figure 6A), while IFN-g wasundetectable. Thus, GrB deficiency favors the develop-ment of Th2-type immunity in response to specificantigen challenge, suggesting that GrB may play an im-portant role in modulating Th2-type immune responsesin vivo.

We then asked whether Th subset-specific manipula-tion of TCR-ICD could bias an immune response to anti-gen. To test this possibility, we used specific inhibitorsof TCR-ICD during the generation of OVA-specifichelper T cell subsets and examined the resultant im-mune response. Splenocytes isolated from normalBALB/c mice 7 days after immunization with OVA in in-complete Freund’s adjuvant were stimulated with OVAin vitro under unbiased growth conditions. To these cul-tures, we added either the caspase inhibitor, z-VAD,

Granzyme B and Th2 Apoptosis243

which rescues Th1 cells from apoptosis, or a GrB inhib-itor, which protects Th2 cells. Cells cultured without in-hibitors served as controls. These expanded cultureswere subsequently restimulated with OVA and cytokineproduction measured in the supernatant by multiplexedcytokine assay (BioPlex System, Bio-Rad). We founda striking divergence in the Th1 and Th2 signature cyto-kines: while both types of cytokines were produced bycontrol cultures, inhibition of caspase activity by z-VAD resulted in significantly increased IFN-g productionand less IL-4; conversely, inhibition of GrB caused a dra-matic drop in IFN-g and significantly increased IL-4(Figure 6B). A similar boost in IL-4 levels resulted whenDPP-I inhibitor was used to block cathepsin C activity.Thus, inhibition of caspase activity resulted in a Th1-skewed response, while GrB inhibition caused an exag-gerated Th2-type response. This result not only con-firms the selective protective effect of these inhibitorson Th1 and Th2 cells, but also shows that we wereable to manipulate this antigen-specific secondary im-mune response toward the Th1 or Th2 type by addingspecific protease inhibitors that favor the survival ofone type of helper T cell over another.

To verify the influence of TCR-ICD on the expansion ofhelper T cell subsets, anti-CD3-stimulated differenti-ated Th1 and Th2 cell cultures were rescued fromTCR-ICD, and changes in cytokine expression were ex-amined. Rescue with FasL antibody caused IFN-g levelsto double in Th1 cell cultures (Figure 6C); the same oc-curred with the addition of z-VAD (not shown). Con-versely, inhibiting the activation of GrB by blockade ofcathepsin C (DPP-I) activation resulted in significantincreases in IL-4, IL-5, and IL-10 (Figure 6C). These re-sults clearly demonstrate that specifically blocking thedeath of Th1 or Th2 cells after TCR activation results inprolonged cytokine production, providing further sup-port for the notion that TCR-ICD can regulate the typeof immune response.

Gzmb2/2 Mice Develop More SevereAllergen-Induced Asthma

To further evaluate the possible role of GrB in the gener-ation of Th2 responses in a pathological setting in vivo,we employed the mouse model of OVA-induced allergiclung inflammation. It is well established that the devel-opment of asthmatic airway inflammation (AAI) in thismodel is solely dependent on the Th2-type immuneresponse. Therefore, we performed AAI experimentswith Gzmb2/2 mice and wild-type controls. Mice weresensitized with i.p. injections of OVA (100 mg) plus alumi-num hydroxide (Alum, 1 mg) as adjuvant on days 0 and 7.On days 14–17, mice were challenged daily by intranasaladministration of OVA (50 ml of 0.5 mg/mL in sterile PBS)or PBS alone. 24 hr after the last challenge, mice wereeuthanized and the bronchio-alveolar lavage (BAL) fluidwas obtained by intratracheal infusion of PBS. Cells inthe BAL fluid were analyzed after H&E staining and leu-kocyte differentials were enumerated. Airway inflamma-tion is characterized by infiltration of leukocytes into theareas surrounding the airways and some of the alveolarspace and by eosinophilia in the BAL fluid. Inflammatorymediators in cell-free BAL fluid were determined by mul-tiplexed assay for IL-4, IL-5, and IFN-g. Histologicalchanges in the lung are shown in Figure 7A. The severity

of leukocyte infiltration was also quantified by two inde-pendent observers in a double-blind fashion accordingto the method established by Herrick (Herrick et al.,2000); scores are presented in Figure 7B. Wild-typemice show significant inflammatory cell infiltration afterOVA sensitization and challenge. Such infiltration ismore dramatic in Gzmb2/2 mice. Increased lung inflam-mation was also manifest in the cellularity of the BAL, asindicated by the fact that total cell numbers and eosino-phil counts were significantly higher in Gzmb2/2 micethan in wild-type controls (Figure 7C). The level of IL-4and IL-5 in the BAL fluid of Gzmb2/2 mice was severalfold higher than in wild-type mice (w30 pg/mL in theasthmatic BAL of wild-type mice; Figure 7D; Takedaet al., 2005). IFN-g was undetectable in the BAL of eithergroup. Therefore, deficiency in GrB leads to a more po-tent Th2-type immune response in vivo, even in a patho-logical setting.

Discussion

Naive CD4+ T cells differentiate into Th1 or Th2 cells de-pending on the cytokine environment and display differ-ent immune functions and cytokine profiles upon stimu-lation (Mosmann and Sad, 1996; Murphy, 1998).Although much is known about the transcription factorsthat regulate the production of several cytokines by Th1and Th2 cells, the exact mechanisms controlling the es-tablishment and persistence of a Th1- or Th2-type im-mune response are not well defined. We have previouslyshown that CD95L and TRAIL are induced after restimu-lation of Th1 and Th2 cells, respectively, and that bothCD95L and TRAIL are able to kill Th1 cells (Zhanget al., 2003). In the current study, we show that theTCR-ICD of Th1 cells depends on the CD95-CD95L in-teraction, while apoptosis of Th2 cells requires GrB ac-tivation. Significantly, we were able to manipulate theimmune response in vivo toward a Th1 or a Th2 bias inan asthma model by specifically rescuing one or theother helper T cell subset from TCR-ICD, an interventionthat may have significant implications for treating dis-eases perpetuated by a dysfunctional Th1-Th2 balance.

Our conclusion that GrB is important in the TCR-ICDof Th2 cells is supported by several lines of evidence.First, inhibitors of GrB or DPP-I prevented TCR-ICDonly in Th2 cells, not in Th1 cells. Second, Th2 cells de-rived from Gzmb2/2 mice were resistant to TCR-ICD. Fi-nally, our confocal microscopy detection of the colocal-ization of GrB and LAMP-1 provides direct evidence ofTCR-induced degranulation of GrB in Th2 cells. InTCR-restimulated Th1 cells, in contrast, GrB still colo-calized with LAMP-1 even after nuclear fragmentationbecame apparent. Therefore, GrB degranulation is criti-cal for the TCR-ICD of Th2 cells.

In experiments with anti-CD3-induced cell death, Th2cells were not protected by CD95L antibody or the cas-pase inhibitor, z-VAD, as were Th1 cells, suggesting dif-ferent pathways of TCR-ICD in Th1 and Th2 cells. Thisimplies the possibility of altering the Th1/Th2 balancein an immune response by manipulation of the differentpathways of TCR-ICD in Th1 and Th2 cells. The bestevidence is that in the absence of exogenous cytokines,inhibition of apoptosis by anti-CD95L and soluble DR5leads to an increase in IFN-g-producing cells (Zhang

Immunity244

Figure 7. Granzyme B-Deficient Mice Exhibit More Severe Allergen-Induced Asthma

GzmB2/2/DPGK-neo mice or 129/SvJ controls were sensitized by two i.p. injections of OVA plus ALUM on days 0 and 7. On days 14 to 17, mice

were challenged daily by intranasal injection of OVA (0.5 mg/mL in 50 ml sterile PBS) or PBS alone. Mice were euthanized on day 18, and bronchio-

alveolar lavage (BAL) fluid was obtained by intratracheal infusion of PBS. Lung tissue was fixed for histological sections and stained with H&E.

Arrows indicate infiltrated leukocytes (A). Microscopic examination was performed by two individuals and relative inflammatory index scored in

double-blinded fashion (B). Cells in BAL fluid were analyzed for total numbers of leukocytes and eosinophils per lung (C). Levels of IL-4 and IL-5 in

the BAL fluid were measured by multiplexed ELISA (D).

et al., 2003). Our studies revealed that z-VAD has no ef-fect on TCR-ICD and IL-4 production in differentiatedTh2 cells. This is also shown by Sehra et al. (2005) thatz-VAD has no effect on IL-4 production by committedTh2 cells. However, the increase in TCR-induced IL-4production by naive T cells (Sehra et al., 2005) couldbe due to a different role of caspases at different stagesof T cell differentiation. Our conclusion of the role ofTCR-ICD in the Th1/Th2 balance is also supported byour finding that Gzmb2/2 mice exhibit a dramatic in-crease in Th2-type immune response and are highly sus-ceptible to allergen-induced asthma. Therefore, GrBparticipates in the regulation of Th2 responses.

By investigating the mechanism of CD95-mediatedapoptosis, we found that CD95L antibody, TR6, andz-VAD were each able to block death in Th1 cells, whileTh2 cells were little affected. We conclude that Th1cell death is triggered by the CD95-CD95L interactionand mediated via caspase 8, which initiates the activa-tion cascade of caspases 3 and 9, finally leading to celldeath. The TCR-ICD of Th2 cells, which occurs at

a slower rate than for Th1 cells, was unrelated to theTNF-family proteins, since neither anti-CD95L, TR6,DR5, OPG, nor anti-TNF affected Th2 cell TCR-ICD. Inaddition, our previous studies have shown that reacti-vated Th2 cells can kill Th1 cells but not themselves,while Th1 cells cannot kill Th2 cells. Instead, Th2 cellsundergo apoptosis by death receptor-independentmechanisms (Zhang et al., 2003). Since apoptotic Th2cells exhibit the typical early and late signs of TCR-ICD, their apoptosis machinery must be intact—thoughless vigorously activated—leading to lower levels ofcaspase activation than in Th1 cells. The resistance ofTh2 cells to TCR-ICD may be attributable to the pres-ence of FLIP (FLICE-like Inhibitory Protein), a caspase 8homolog that inhibits caspase 8 activation. In fact, wehave reported much higher amount of FLIP protein inTh2 cells compared to Th1 cells (Zhang et al., 2003), andFLIP has been reported to downregulate caspase activa-tion mediated by other receptors. Furthermore, we haveshown that the intracellular caspase inhibitor, SPI-2A,is highly expressed after TCR-induced activation of

Granzyme B and Th2 Apoptosis245

Th2 cells, but not Th1 cells. Therefore, besides the lackof FasL expression, it is likely the increased expressionof FLIP and SPI-2A that confers upon Th2 cells theirresistance to TCR-ICD.

Although some reports have shown that the TCR-ICDof both Th1 and Th2 cells depends on the CD95-CD95Linteraction (Watanabe et al., 1997), recent reports are in-creasingly showing strong evidence that Th1 and Th2cells use different mechanisms to execute TCR-ICD(Ramsdell et al., 1994; Varadhachary et al., 1997; Yateset al., 2000; Zhang et al., 1997, 2003). The role of theCD95-CD95L interaction in TCR-ICD has been reportedto be more important in Th1 cells than in Th2 cells(Ramsdell et al., 1994; Zhang et al., 2001, 2003). Addi-tional evidence from other groups suggests that thepresence of CD95/Fas-associated phosphatase (FAP)potently unlinks CD95L signaling in Th2 cells (Varad-hachary et al., 1997; Zhang et al., 1997). Surprisingly,very little has been reported about the mechanism ofTCR-ICD in Th2 cells. The data presented here regardingthe critical role of GrB in the Th2 cell death suggesta novel mechanism regulating the balance betweenTh1 and Th2 cells.

We also report that activated Th1 cells express theGrB inhibitor, SPI-6, while Th2 cells instead expresshigh levels of intracellular SPI-2A, an endogenous inhib-itor of caspases 3 and 9. This reciprocal expression ofSPI-6 and SPI-2A shows that Th2 cells have no protec-tion against GrB-mediated death but are protectedfrom the activity of several caspases; the opposite istrue for Th1 cells. Thus, it is not surprising that Th2 celldeath is characterized by low caspase activity and islargely unaffected by caspase inhibitors, while Th1death involves marked increases in caspase activityand is unaffected by granzyme B inhibitors. Therefore,the absence of SPI-6 in Th2 cells renders them suscep-tible to death by TCR-activated granzymes.

The conversion of pro-granzyme B to active granzymeB is mediated by cathepsin C. We have shown that exog-enous inhibition of cathepsin C protects Th2 cells fromTCR-ICD. The role of GrB was confirmed in vivo by dem-onstrating that Gzmb2/2 mice have a Th2-skewed im-mune response. We also showed that the immune re-sponse in OVA can be biased toward the Th1 or a Th2type by rescue of the corresponding helper T cell withspecific inhibitors of apoptosis. Moreover, in the patho-logical setting of OVA-induced asthma, a Th2-driven dis-ease, significantly greater disease severity occurred inGzmb2/2 mice compared to normal mice. Therefore,GrB plays a critical role in regulating Th2 response.This seems discordant with the recent report suggestinga critical role of NKT cells in asthma (Meyer et al., 2006).We recently reexamined the role of CD4+ T cells via ge-netically modified mice and showed that CD4+ T cellsare required for OVA-induced asthmatic airway inflam-mation in several strains of mice tested (data not shown).

We have shown that GrB is the pivotal mediator of Th2cell apoptosis. GrB can independently activate multiplecaspases, including caspases 1, 2, 3, 6, 7, 9, and 10 (Lie-berman, 2003), and some caspase substrates, includingPARP and NuMA (Beresford et al., 1999). GrB has alsobeen shown to activate t-BID and cleave ICAD indepen-dent of caspases (Barry et al., 2000; Thomas et al., 2001).While it is uncertain whether the GrB-mediated Th2 cell

apoptosis depends on the caspases or acts directly oncellular substrates, we have shown that GrB activity iselevated in Th2 cells and that inhibition of GrB rescuesTh2 cells from apoptosis, implicating its central role inTh2 cell death. Further investigation of the mechanismsby which GrB regulates TCR-ICD in Th2 cells will allowus to better understand the mechanisms that modulateTh2 response in normal and disease states.

Experimental Procedures

Reagents and Mice

Antibodies against murine CD95L (MFL3), CD95 (JO2), and FITC-

conjugated AnnexinV were from BD Biosciences/Pharmingen (San

Diego, CA). Antibodies against IFN-g, IL-4, IL-12, and GrB (16G6)

were purchased from eBiosciences (San Diego, CA). TNF-a, TRAIL,

IFN-g, and anti-TNF-a were from R&D Systems (Minneapolis, MN).

DR5 was a kind gift of Dr. Youhai Chen, University of Pennsylvania.

Caspases 1, 2, 3, 6, 8, and 9 were assayed by kits from Intergen, Inc.

(Purchase, NY). The GrB inhibitor, z-AAD-CMK, and its control,

z-FA-CMK, were from Calbiochem (San Diego, CA). Caspase inhib-

itor z-VAD was from Enzyme Systems (Livermore, CA). TR6 recombi-

nant protein was made by Human Genome Sciences (Rockville, MD).

The DPP-I inhibitor, Gly-Phe-CHN2 (MP Biochemicals, Aurora, OH),

was used to inhibit cathepsin C activity.

Female BALB/c mice at 5–8 weeks old were obtained from

National Cancer Institute (Frederick, MD), and Gzmb2/2 mice

(GzmB2/2/DPGK-neo) and wild-type controls (129/SvJ) (Revell

et al., 2005) were generously provided by Dr. Timothy Ley, Washing-

ton University Medical School. Mice were maintained in the Robert

Wood Johnson Medical School Vivarium. Animals were matched

for age and gender in each experiment. All animal experiments

were approved by the institutional Animal Care and Use Committee.

Differentiation of Helper T Cell Subsets

CD4+ T cells were purified from splenocytyes by negative selection

by means of immunoaffinity columns (mouse CD4+ T cell subset iso-

lation kit, R&D Systems) and differentiated under Th1- or Th2-gener-

ating conditions. CD4+ lymphocytes at 106/mL were activated with

anti-CD3 (1 mg/mL) bound to plastic and soluble anti-CD28 (2 mg/

mL). IL-12 (10 ng/mL) and anti-IL-4 (10 mg/mL) were included in

Th1 cultures, while IL-4 (5 ng/mL), anti-IL-12, and anti-IFN-g (each

at 10 mg/mL) were supplied for Th2 cultures. After 24 hr, IL-2 was

added to all cultures. Cells were divided at a 1:4 ratio after 3 days

and allowed to rest under the above cytokine conditions in the

absence of anti-CD3 and anti-CD28 for another 2 days. These cells

(1 3 106/mL) were then restimulated with plate bound anti-CD3

(1 mg/mL) to initiate TCR-ICD. All cell cultures were maintained

in RPMI 1640 medium supplemented with 2 mM L-glutamine,

50 mM 2-mercaptoethanol, 10% heat-inactivated FBS, and 10 mM

Gentamycin.

Northern Blot Analysis

Total RNA was isolated from differentiated helper T cell subsets via

affinity columns (Qiagen, Chatsworth, CA), according to the manu-

facturer’s protocol. RNA samples were fractionated on 1% aga-

rose/2.2 M formaldehyde denaturing gel and transferred onto a Ny-

tran membrane (Schleicher & Schuell, Inc., Keene, NH). DNA probes

were labeled with 32P-dCTP by random priming (Roche, Indianapo-

lis, IN) according to manufacturer’s instructions. Prehybridization

and hybridization were carried out at 42�C in a solution containing

5 3 SSC (10 3 SSC is 1.5 M NaCl, 0.15 M sodium citrate), 2.5 mM

EDTA, 0.1% SDS, 5 3 Denhardt’s solution, 2 mM sodium pyrophos-

phate, 50 mM sodium phosphate, and 50% formamide. Membranes

were washed with 0.2 3 SSC, 0.1% SDS at 56�C for 1 hr, and hybrid-

ization signals were detected by autoradiography.

Real-Time PCR

Total RNA was isolated with an RNeasy Mini Kit (Qiagen). Genomic

DNA was removed with RNase-free DNase prior to cDNA synthesis.

First-strand cDNA synthesis was performed for each RNA sample

with Sensiscript RT Kit (Qiagen). Reverse-transcribed cDNA was de-

termined by real-time PCR with SYBR Green Master Mix (Applied

Immunity246

Biosystems, Foster City, CA). The primers were: GrB: 50-ACTCTTG

ACGCTGGGACCTA, 50-AGTGGGGCTTGACTTCATGT; b-actin: 50-

GTGGGCCGCCCTAGGCACCA, 50-CTCTTTGATGTCACGCACGATT

TC; FasL: 50-TGGGTAGACAGCAGTGCCAC, 50-GCCCACAAGATG

GACAGGG; IFN-g: 50-TCAAGTGGCATAGATGTGGAAGAA, 50-TG

GCTCTGCAGGATTTTCATG; IL-4: 50-ACAGGAGAAGGGACGCCAT,

50-GAAGCCCTACAGACGAGCTCA. b-Actin gene was used as an en-

dogenous control to normalize for the amount of total RNA in each

sample. All values are expressed as relative fold increase or de-

crease compared to a control.

Confocal Microscopy

For subcellular localization of LAMP-1 (CD107a) and GrB, the cells

were fixed and permeabilized with a kit from BD Biosciences and

were then stained with FITC-labeled LAMP-1 and PE-labeled anti-

GrB (eBiosciences) for 20 min, counterstained with Hoechst 33342

nuclear stain, then washed and mounted on slides. The cells were

then visualized with a Zeiss LSM510 META laser scanning confocal

microscope with a 633 water immersion objective (NA 1.2). Sepa-

rate filter sets for FITC (488 nm excitation, a 505–550 nm band

pass emission), PE (543 nm excitation, 560 nm long pass emission),

and DAPI (351 nm excitation, 385–470 nm emission) were used for

image acquisition.

Blocking of Apoptosis

Differentiated helper T cells (106 cells/mL) were analyzed for apopto-

sis after activation in the presence or absence of the following

reagents: z-VAD (5–250 mM), z-AAD-CMK (a GrB inhibitor, 5–250

mM), anti-CD95L (MFL3, 50–1000 ng/mL), osteoprotegerin (OPG,

50–1000 ng/mL, R&D), DR5 (5–20 mg /mL), DMSO (0.1%–5%), TR6

(50–1000 ng/mL), or the DPP-I inhibitor, Gly-Phe-CHN2 (5–250

mM), each added 15–20 min prior to stimulation. Apoptosis was an-

alyzed by Annexin V binding and propidium iodide (PI) permeability.

PBS-washed cells were resuspended in 5 ml cold binding buffer con-

taining FITC-conjugated AnnexinV. The cell suspension was diluted

to 250 ml in a staining buffer (PBS with 2% FCS and 0.1% sodium

azide) with PI (25 mg/mL) and analyzed by flow cytometry. Viable

cells are defined as those negative for both PI and Annexin V, while

early apoptotic cells are Annexin V+PI2. Double-positive cells are

late apoptotic or necrotic. Only early apoptotic cells are included

in the data analysis. To determine total DNA content, cells were

resuspended in DNA staining buffer consisting of 0.5% saponin,

50 mg/mL PI, and 0.1 mg/mL RNase A in PBS and analyzed by flow

cytometry for the percentage of cells in the hypodiploid region.

Caspase Measurement

Activated caspases were detected with fluorogenic caspase sub-

strates (Intergen, Purchase, NY) according to the manufacturer’s

protocol. In brief, cells (1 3 106/mL) were stimulated with anti-

CD3, and 5 ml of the diluted caspase substrate was added. The cells

were then incubated at 37�C for 1 hr, washed and resuspended in

PBS with 5 mg/mL PI, and analyzed by flow cytometry. The increase

in caspase+ PI2 population of the stimulated cells compared to un-

stimulated cells was calculated and plotted for each of the cas-

pases.

Immunization and Cytokine Measurement

BALB/c mice were immunized by tail-base injection of 10 mg OVA

(Grade V; Sigma) in 50 ml saline mixed with 50 ml of incomplete

Freund’s adjuvant. After 7 days, spleens were harvested, and

CD4+ cells were purified and stimulated (7.5 3 106 cells/mL) with

OVA (100 mg/mL) in the presence or absence of z-VAD or GrB inhib-

itor (each at 100 mM). Supernatant was collected at 48 hr and as-

sayed for IFN-g and IL-4 by ELISA. Gzmb2/2 or 129/SvJ wild-type

mice were immunized i.p. with 10 mg OVA mixed with 2 mg ALUM

in 200 ml PBS on days 0 and 7, and cell cultures were similarly pre-

pared and analyzed.

Induction of Allergic Airway Inflammation

8-week-old female Gzmb2/2 and wild-type mice (H2b) were sensi-

tized by i.p. injections of 100 mg of OVA plus 1 mg ALUM in 0.2 ml

PBS on days 0 and 7. On days 14–17, mice were challenged daily in-

tranasally with OVA (0.5 mg/mL in 50 ml PBS) or PBS alone. Mice

were euthanized 24 hr later, and BAL fluid was obtained by intratra-

cheal infusion of 2 ml PBS, as described (Das et al., 2001). Cells in the

BAL fluid were analyzed on slides after H&E staining. Leukocyte dif-

ferentials were acquired on a CELL-DYN 3700 Hematology Analyzer.

Inflammatory mediators were determined in cell-free BAL fluid by

multiplexed cytokine assay for IL-4, IL-5, and IFN-g (BioPlex, Bio-

Rad, Hercules, CA).

Statistical Analysis

Differences in treatment groups were assessed by paired two-tail

Student’s t test or by analysis of variance (ANOVA) for repeated

measures, as indicated. The significance levels are indicated as fol-

lows: *p < 0.05; **p < 0.01; ***p < 0.001.

Acknowledgments

We thank Dr. Timothy Ley for the GrB-deficient mice, Dr. Chris

Bleackley for advice on GrB studies, Dr. Lianjun Zhang for statistical

analysis, Dr. Xi Yang for advice on immunization, Dr. Sidney Pestka

for discussions, and Ms. Julie Friedman for histology. This work was

supported in part by USPHS grants (AI43384, AI057596, and

AI50222) and the National Space Biomedical Research Institute

(IIH00405), which is supported by the National Aeronautics and

Space Administration through the Cooperative Agreement NCC

9-58.

Received: October 21, 2005

Revised: April 18, 2006

Accepted: June 13, 2006

Published online: August 10, 2006

References

Akbar, A.N., and Salmon, M. (1997). Cellular environments and

apoptosis: tissue microenvironments control activated T-cell death.

Immunol. Today 18, 72–76.

Alter, G., Malenfant, J.M., and Altfeld, M. (2004). CD107a as a func-

tional marker for the identification of natural killer cell activity. J. Im-

munol. Methods 294, 15–22.

Barry, M., Heibein, J.A., Pinkoski, M.J., Lee, S.F., Moyer, R.W.,

Green, D.R., and Bleackley, R.C. (2000). Granzyme B short-circuits

the need for caspase 8 activity during granule-mediated cytotoxic

T-lymphocyte killing by directly cleaving Bid. Mol. Cell. Biol. 20,

3781–3794.

Beresford, P.J., Xia, Z., Greenberg, A.H., and Lieberman, J. (1999).

Granzyme A loading induces rapid cytolysis and a novel form of

DNA damage independently of caspase activation. Immunity 10,

585–594.

Bidere, N., Briet, M., Durrbach, A., Dumont, C., Feldmann, J., Char-

pentier, B., de Saint-Basile, G., and Senik, A. (2002). Selective inhi-

bition of dipeptidyl peptidase I, not caspases, prevents the partial

processing of procaspase-3 in CD3-activated human CD8(+)

T lymphocytes. J. Biol. Chem. 277, 32339–32347.

Bots, M., Kolfschoten, I.G., Bres, S.A., Rademaker, M.T., de Roo,

G.M., Kruse, M., Franken, K.L., Hahne, M., Froelich, C.J., Melief,

C.J., et al. (2005). SPI-CI and SPI-6 cooperate in the protection

from effector cell-mediated cytotoxicity. Blood 105, 1153–1161.

Catalfamo, M., and Henkart, P.A. (2003). Perforin and the granule

exocytosis cytotoxicity pathway. Curr. Opin. Immunol. 15, 522–527.

Constant, S.L., and Bottomly, K. (1997). Induction of Th1 and Th2

CD4+ T cell responses: the alternative approaches. Annu. Rev.

Immunol. 15, 297–322.

Das, J., Chen, C.H., Yang, L., Cohn, L., Ray, P., and Ray, A. (2001). A

critical role for NF-kappa B in GATA3 expression and TH2 differen-

tiation in allergic airway inflammation. Nat. Immunol. 2, 45–50.

Fruth, U., Sinigaglia, F., Schlesier, M., Kilgus, J., Kramer, M.D., and

Simon, M.M. (1987). A novel serine proteinase (HuTSP) isolated

from a cloned human CD8+ cytolytic T cell line is expressed and se-

creted by activated CD4+ and CD8+ lymphocytes. Eur. J. Immunol.

17, 1625–1633.

Herrick, C.A., MacLeod, H., Glusac, E., Tigelaar, R.E., and Bottomly,

K. (2000). Th2 responses induced by epicutaneous or inhalational

Granzyme B and Th2 Apoptosis247

protein exposure are differentially dependent on IL-4. J. Clin. Invest.

105, 765–775.

Ida, H., Nakashima, T., Kedersha, N.L., Yamasaki, S., Huang, M.,

Izumi, Y., Miyashita, T., Origuchi, T., Kawakami, A., Migita, K.,

et al. (2003). Granzyme B leakage-induced cell death: a new type

of activation-induced natural killer cell death. Eur. J. Immunol. 33,

3284–3292.

Janssen, O., Sanzenbacher, R., and Kabelitz, D. (2000). Regulation

of activation-induced cell death of mature T-lymphocyte popula-

tions. Cell Tissue Res. 301, 85–99.

Komoriya, A., Packard, B.Z., Brown, M.J., Wu, M.L., and Henkart,

P.A. (2000). Assessment of caspase activities in intact apoptotic thy-

mocytes using cell-permeable fluorogenic caspase substrates.

J. Exp. Med. 191, 1819–1828.

Lenardo, M.J. (1991). Interleukin-2 programs mouse alpha beta

T lymphocytes for apoptosis. Nature 353, 858–861.

Levine, B.L., Bernstein, W.B., Connors, M., Craighead, N., Lindsten,

T., Thompson, C.B., and June, C.H. (1997). Effects of CD28 costimu-

lation on long-term proliferation of CD4+ T cells in the absence of ex-

ogenous feeder cells. J. Immunol. 159, 5921–5930.

Lieberman, J. (2003). The ABCs of granule-mediated cytotoxicity:

new weapons in the arsenal. Nat. Rev. Immunol. 3, 361–370.

Meade, J.L., de Wynter, E.A., Brett, P., Malik Sharif, S., Woods, C.G.,

Markham, A.F., and Cook, G.P. (2006). A family with Papillon-Lefevre

Syndrome reveals a requirement for Cathepsin C in Granzyme B ac-

tivation and NK cell cytolytic activity. Blood 107, 3665–3668.

Mercep, M., Bluestone, J.A., Noguchi, P.D., and Ashwell, J.D. (1988).

Inhibition of transformed T cell growth in vitro by monoclonal anti-

bodies directed against distinct activating molecules. J. Immunol.

140, 324–335.

Meyer, E.H., Goya, S., Akbari, O., Berry, G.J., Savage, P.B., Kronen-

berg, M., Nakayama, T., DeKruyff, R.H., and Umetsu, D.T. (2006).

Glycolipid activation of invariant T cell receptor+ NK T cells is suffi-

cient to induce airway hyperreactivity independent of conventional

CD4+ T cells. Proc. Natl. Acad. Sci. USA 103, 2782–2787.

Migone, T.S., Zhang, J., Luo, X., Zhuang, L., Chen, C., Hu, B., Hong,

J.S., Perry, J.W., Chen, S.F., Zhou, J.X., et al. (2002). TL1A is a TNF-

like ligand for DR3 and TR6/DcR3 and functions as a T cell costimu-

lator. Immunity 16, 479–492.

Mosmann, T.R., and Sad, S. (1996). The expanding universe of T-cell

subsets: Th1, Th2 and more. Immunol. Today 17, 138–146.

Mosmann, T.R., Cherwinski, H., Bond, M.W., Giedlin, M.A., and Coff-

man, R.L. (1986). Two types of murine helper T cell clone. I. Definition

according to profiles of lymphokine activities and secreted proteins.

J. Immunol. 136, 2348–2357.

Murphy, K.M. (1998). T lymphocyte differentiation in the periphery.

Curr. Opin. Immunol. 10, 226–232.

Murphy, K.M., Ouyang, W., Farrar, J.D., Yang, J., Ranganath, S., As-

nagli, H., Afkarian, M., and Murphy, T.L. (2000). Signaling and tran-

scription in T helper development. Annu. Rev. Immunol. 18, 451–494.

Oberg, H.H., Lengl-Janssen, B., Kabelitz, D., and Janssen, O. (1997).

Activation-induced T cell death: resistance or susceptibility corre-

late with cell surface fas ligand expression and T helper phenotype.

Cell. Immunol. 181, 93–100.

Paul, W.E., and Seder, R.A. (1994). Lymphocyte responses and cyto-

kines. Cell 76, 241–251.

Peters, P.J., Borst, J., Oorschot, V., Fukuda, M., Krahenbuhl, O.,

Tschopp, J., Slot, J.W., and Geuze, H.J. (1991). Cytotoxic T lympho-

cyte granules are secretory lysosomes, containing both perforin and

granzymes. J. Exp. Med. 173, 1099–1109.

Raja, S.M., Metkar, S.S., and Froelich, C.J. (2003). Cytotoxic granule-

mediated apoptosis: unraveling the complex mechanism. Curr.

Opin. Immunol. 15, 528–532.

Ramsdell, F., Seaman, M.S., Miller, R.E., Picha, K.S., Kennedy, M.K.,

and Lynch, D.H. (1994). Differential ability of Th1 and Th2 T cells to

express Fas ligand and to undergo activation-induced cell death.

Int. Immunol. 6, 1545–1553.

Revell, P.A., Grossman, W.J., Thomas, D.A., Cao, X., Behl, R., Rat-

ner, J.A., Lu, Z.H., and Ley, T.J. (2005). Granzyme B and the down-

stream granzymes C and/or F are important for cytotoxic lympho-

cyte functions. J. Immunol. 174, 2124–2131.

Sehra, S., Patel, D., Kusam, S., Wang, Z.Y., Chang, C.H., and Dent,

A.L. (2005). A role for caspases in controlling IL-4 expression in

T cells. J. Immunol. 174, 3440–3446.

Shi, Y.F., Sahai, B.M., and Green, D.R. (1989). Cyclosporin A inhibits

activation-induced cell death in T-cell hybridomas and thymocytes.

Nature 339, 625–626.

Sprent, J., and Tough, D.F. (1994). Lymphocyte life-span and mem-

ory. Science 265, 1395–1400.

Takeda, K., Miyahara, N., Kodama, T., Taube, C., Balhorn, A., Da-

khama, A., Kitamura, K., Hirano, A., Tanimoto, M., and Gelfand,

E.W. (2005). S-carboxymethylcysteine normalises airway respon-

siveness in sensitised and challenged mice. Eur. Respir. J. 26,

577–585.

Thomas, D.A., Scorrano, L., Putcha, G.V., Korsmeyer, S.J., and Ley,

T.J. (2001). Granzyme B can cause mitochondrial depolarization and

cell death in the absence of BID, BAX, and BAK. Proc. Natl. Acad.

Sci. USA 98, 14985–14990.

Varadhachary, A.S., Perdow, S.N., Hu, C., Ramanarayanan, M., and

Salgame, P. (1997). Differential ability of T cell subsets to undergo

activation-induced cell death. Proc. Natl. Acad. Sci. USA 94, 5778–

5783.

Vella, A.T., Dow, S., Potter, T.A., Kappler, J., and Marrack, P. (1998).

Cytokine-induced survival of activated T cells in vitro and in vivo.

Proc. Natl. Acad. Sci. USA 95, 3810–3815.

Watanabe, N., Arase, H., Kurasawa, K., Iwamoto, I., Kayagaki, N.,

Yagita, H., Okumura, K., Miyatake, S., and Saito, T. (1997). Th1 and

Th2 subsets equally undergo Fas-dependent and -independent ac-

tivation-induced cell death. Eur. J. Immunol. 27, 1858–1864.

Yamada, M., Hirasawa, A., Shiojima, S., and Tsujimoto, G. (2003).

Granzyme A mediates glucocorticoid-induced apoptosis in leuke-

mia cells. FASEB J. 17, 1712–1714.

Yates, A., Bergmann, C., Van Hemmen, J.L., Stark, J., and Callard, R.

(2000). Cytokine-modulated regulation of helper T cell populations.

J. Theor. Biol. 206, 539–560.

Zhang, X., Brunner, T., Carter, L., Dutton, R.W., Rogers, P., Bradley,

L., Sato, T., Reed, J.C., Green, D., and Swain, S.L. (1997). Unequal

death in T helper cell (Th)1 and Th2 effectors: Th1, but not Th2, effec-

tors undergo rapid Fas/FasL-mediated apoptosis. J. Exp. Med. 185,

1837–1849.

Zhang, J., Bardos, T., Mikecz, K., Finnegan, A., and Glant, T.T.

(2001). Impaired Fas signaling pathway is involved in defective

T cell apoptosis in autoimmune murine arthritis. J. Immunol. 166,

4981–4986.

Zhang, X.R., Zhang, L.Y., Devadas, S., Li, L., Keegan, A.D., and Shi,

Y.F. (2003). Reciprocal expression of TRAIL and CD95L in Th1 and

Th2 cells: role of apoptosis in T helper subset differentiation. Cell

Death Differ. 10, 216–224.


Recommended