Inhibition of Fas/Fas ligand signaling improves septic survival ...

6 downloads 0 Views 318KB Size Report
and Alfred Ayala*,1. *Surgical Research, Department of Surgery, Brown University School of Medicine and Rhode Island Hospital, ... (FasL) pathway plays a central role in immune- .... tory-Tek II eight-chamber slides (Nalge NUNC, Naperville, IL), fixed with ..... extensive cell death of hepatocytes, corresponding with a high.
Inhibition of Fas/Fas ligand signaling improves septic survival: differential effects on macrophage apoptotic and functional capacity Chun-Shiang Chung,* Grace Y. Song,* Joanne Lomas,* H. Hank Simms,† Irshad H. Chaudry,‡ and Alfred Ayala*,1 *Surgical Research, Department of Surgery, Brown University School of Medicine and Rhode Island Hospital, Providence; †Department of Surgery, North Shore University Hospital, Manhasset, New York; and ‡Department of Surgery, University of Alabama at Birmingham School of Medicine

Abstract: Signaling through the Fas/Fas ligand (FasL) pathway plays a central role in immunecell response and function; however, under certain pathological conditions such as sepsis, it may contribute to the animal’s or patient’s morbidity and mortality. To determine the contribution of FasL to mortality, we conducted survival studies by blocking Fas/FasL with Fas receptor fusion protein (FasFP) in vivo. C3H/HeN mice received FasFP or the saline vehicle (veh) immediately (0 h) or delayed (12 h), after sepsis induced by cecal ligation and puncture (CLP). Subsequently, we examined the effect of FasFP treatment (12 h post-CLP) on macrophage apoptosis and functional capacities. Peritoneal and splenic macrophages and Kupffer cells from sham-veh-, CLPveh-, sham-FasFP-, or CLP-FasFP-treated mice were harvested 24 h after CLP and stimulated with lipopolysaccharide (LPS) for 24 h. The results indicate that only delayed (12 h) but not 0 h administration of FasFP demonstrated a significant increase in survival. The ability of all macrophage populations to release interleukin (IL)-6 was significantly depressed, and IL-10 release was augmented after CLP. FasFP treatment attenuated the increased IL-10 release in Kupffer cells. However, althogh enhanced susceptibility to LPS-induced apoptosis could be suppressed in CLP mouse Kupffer cells by FasFP, FasFP did not change the peritoneal or splenic macrophage response. Furthermore, FasFP attenuated the elevated plasma levels of liver enzymes after sepsis. These data indicate that in vivo inhibition of Fas/FasL signaling has tissue-specific effects on the induction of macrophage apoptosis, functional changes, and liver damage, which may contribute to the host’s ability to ward off a septic challenge. J. Leukoc. Biol. 74: 344 –351; 2003. Key Words: cecal ligation and puncture 䡠 mice 䡠 peritoneal macrophage 䡠 splenic macrophage 344



Kupffer cells

Journal of Leukocyte Biology Volume 74, September 2003

INTRODUCTION Despite the advent of a number of modern therapeutic approaches, including aggressive operative intervention, specific antibiotic treatment, nutritional support, and antiendotoxin or antiproinflammatory cytokine therapies, sepsis and subsequent multiple organ dysfunction continue to be the leading cause for mortality of patients in the intensive care unit [1]. Yet, information on the pathogenesis of such a complex condition remains incomplete. A current hypothesis is that the exaggerated proinflammatory mediators released in the course of sepsis appear to contribute to the initiation of cell and organ dysfunction [2]. However, recent clinical trials directed at anticytokine-based therapeutic strategies have been unsuccessful [3]. The reason for this is at least in part a result of our lack of an adequate understanding of this complex, pathophysiological process. As substantial resources continued to be devoted to the treatment of septic patients, it remains critical to better understand the precise mechanisms that underlie the pathophysiology of sepsis if we are to develop more appropriate therapies. Studies have suggested that cytokines such as tumor necrosis factor (TNF), interleukin (IL)-1, or IL-6 may be involved in the initiation of organ dysfunction associated with sepsis and multiple organ failure [4]. Activated macrophages are thought to be one of the major producers of cytokines and growth factors, which not only regulate innate- and acquired-immune responses but also play a key role in the clearance and killing of invading pathogens during septic insult. We have previously demonstrated that macrophages isolated from septic mice show a marked loss of important immune functional capacity (i.e., decreased inducible TNF, IL-1, and IL-6 production), which is associated with a significant increase in macrophage apoptosis when subsequently challenged with lipopolysaccharide (LPS) [5–7]. However, the mechanism responsible for these changes as well as the interactions between cytokine production and apoptotic processes in macrophages are poorly understood.

1

Correspondence: Surgical Research, Aldrich 2, Rhode Island Hospital, 593 Eddy St., Providence, RI 02903. E-mail: [email protected] Received August 20, 2002; accepted August 20, 2002; doi: 10.1189/ jlb.0102006.

http://www.jleukbio.org

Apoptotic cell death plays a critical role in normal physiologic processes and pathologic conditions [8]. Studies have demonstrated that sepsis induces an increase in apoptosis in lymphocytes, monocytes, and epithelial cells in patients and experimental animals [9, 10]. It has been proposed that loss of immune (T and B cells and monocytes) and nonimmune (epithelial, cardiac myocytes, hepatocytes, and vascular endothelial) cells through the apoptotic process may contribute to the immunosuppression seen in sepsis, which in turn, may lead to the subsequent multiple organ dysfunction [2, 11, 12]. Furthermore, studies in animal models of sepsis show that prevention of lymphocyte apoptosis by administration of caspase inhibitors or through overexpression of Bcl-2, an antiapoptotic protein, improves survival during sepsis [13]. However, the mechanisms by which apoptosis is regulated during sepsis as well as its contribution to organ dysfunction remain to be established. The Fas/Fas ligand (FasL) system has been identified as a major pathway involved in the induction of apoptotic cell death in many cell types, including lymphocytes and other leukocytes [14 –16]. Fas is widely expressed in many cell types, and FasL expression is restricted and usually requires activation [17, 18]. The expression of Fas and FasL has been reported in monocytes and macrophages in low but detectable levels [19]. In a previous study, we demonstrated that FasL deficiency improves the ability of mice to survive septic challenge [20]. However, the role of the Fas/FasL system in the regulation of macrophage apoptosis during sepsis has not been established. In the present study, we attempted to determine whether inhibition of the activation of Fas by FasL through the administration of Fas fusion protein (FasFP) in septic mice could improve survival as well as alter the apoptotic response and functional capacity of various macrophage populations in response to LPS stimulation.

MATERIALS AND METHODS Animals Male inbred C3H/HeNCrlBR (C3H/HeN endotoxin-sensitive strain, Charles River Laboratories, Wilmington, MA) mice, 8 –10 weeks of age and weighing 20 –25 g, were used in all experiments. The studies described here were performed according to the National Institutes of Health Guide for the Care and Use of Laboratory Animals and were approved by the Brown University and Rhode Island Hospital Committee on Animal Use and Care (Providence, RI).

Model of polymicrobial sepsis Polymicrobial sepsis was induced in mice using the method described in our previous study [5]. In brief, mice were lightly anesthetized with Metofane (Methoxyflurane, Pitman-Moore, Mundelein, IL), and the abdomen was shaved and scrubbed with betadine. A midline incision (1.5–2.0 cm) was made below the diaphragm to expose the cecum, which was isolated, ligated, and punctured twice with a 22-gauge needle and gently compressed to extrude a small amount of cecal contents through the punctured holes. The cecum was returned to the abdomen, and the incision was closed in layers with a 6-0 Ethilon suture (Ethicon, Somerville, NJ). The animals were then resuscitated with 1.0 mL lactated Ringer’s solution by subcutaneous (s.c.) injection. For sham controls, the animals were subjected to the same surgical procedure, i.e., laparotomy and cecal isolation, but the cecum was neither ligated nor punctured.

Reagent FasFP (kindly provided by Dr. Carl Edwards, Amgen, Thousand Oaks, CA) is a chimeric fusion protein consisting of the extracellular domain of murine Fas

covalently linked to the Fc region and hinge protein of a human immunoglobulin G (IgG) [21]. For the survival study, mice were subjected to cecal ligation and puncture (CLP), received a single dose of 200 ␮g/kg FasFP or saline at 0 h or 12 h after surgery, and were monitored for mortality over a 10-day period. In all subsequent in vivo studies, animals subjected to CLP or sham procedures were randomized into four groups (sham-saline vehicle, sham-FasFP, CLP-saline vehicle, and CLP-FasFP), receiving FasFP (200 ␮g/kg body weight) or vehicle s.c. 12 h postsurgery. It should be noted that in prior studies, we have not seen vasoactive effects of this agent on mean arterial pressure [22].

Cell preparations Animals were killed 24 h after CLP or sham CLP operations by Metofane overdose, and splenic macrophages were obtained as described previously [23]. The spleens were removed and gently glass ground, and erythrocytes were lysed using hypotonic buffers; isolated splenocytes were then washed, counted, plated at 1 ⫻ 107 cells per mL in plastic tissue-culture plates, and incubated at 37°C for 2 h. The adherent splenic macrophages were then washed with fresh Dulbecco’s modified Eagle’s medium (DMEM; Gibco-BRL, Grand Island, NY). Peritoneal macrophage monolayers were established by harvesting peritoneal exudate cells [24] via lavage of the peritoneal cavity with 2 ⫻ 5 mL cold DMEM. The cells were centrifuged (800 g at 4°C for 15 min), resuspended in fresh DMEM at 2 ⫻ 106 cells per mL, plated onto plastic tissue-culture plates, and incubated at 37°C for 2 h. The nonadherent cells were removed by washing three times with fresh DMEM. These protocols provided adherent cells that were greater than 95% positive by nonspecific esterase staining and exhibited typical macrophage morphology. Kupffer cells were harvested as described previously [25]. In brief, the liver was retrograde perfused with 25 mL ice-cold Hanks’ balanced salt solution (HBSS; Gibco-BRL) through the portal vein and immediately followed by perfusion with 10 mL prewarmed 0.05% collagenase IV (Sigma Chemical Co., St. Louis, MO) in HBSS. The liver was then removed, minced finely in a Petri dish containing 5 mL warm collagenase, and incubated at 37°C for 20 min. The cell suspension was passed through a steel screen, centrifuged, resuspended in DMEM, layered onto 16% Metrizamide (Accurate Chemical, Westbury, NJ) in HBSS, and centrifuged at 3000 g, 4°C for 45 min. The cells from the interface were collected, washed, resuspended in DMEM medium containing 10% fetal bovine serum (FBS) at 4 ⫻ 106 cells per mL, plated onto plastic tissue-culture plates, and incubated at 37°C overnight. The nonadherent cells were removed by washing three times with fresh DMEM. Adherent macrophage monolayers were stimulated with 10 ␮g LPS per mL DMEM medium supplemented with 10% FBS for 24 h (37°C, 5% CO2; 95% humidity). At the end of the incubation period, cells and the cultured supernatants were collected for analysis.

Analysis of apoptosis and Fas expression Apoptosis of macrophages was determined by DNA fragmentation, morphological changes, and caspase-3 activity. The percent of apoptotic-positive cells was detected by the DNA-binding agent propidium iodide (PI; Sigma Chemical Co.) according to the methods of Telford et al. [26]. Macrophages were collected after culture, resuspended in 0.5 mL PI-staining reagent (0.1% Nonidet P-40, 0.1% sodium citrate, and 50 ␮g PI per mL), and immediately analyzed using a fluorescein-activated cell sorter flow cytometer (Becton Dickinson, San Jose, CA). To assess apoptosis by morphology, macrophages were cultured in Laboratory-Tek II eight-chamber slides (Nalge NUNC, Naperville, IL), fixed with ethanol, stained with Giemsa stain (Gibco-BRL), and counted. Apoptotic cells were characterized by the presence of nuclear condensation and loss of cytoplasm. Two hundred cells were counted per slide, three slides per group. Fas expression in Kupffer cells was analyzed by immunofluorescent staining. Cells were cultured in Laboratory-Tek II slides (Nalge NUNC), fixed with 2% paraformaldehyde (Sigma Chemical Co.), and blocked in phosphatebuffered saline (PBS) containing 2% normal goat serum (Pierce, Rockford, IL) for 30 min at room temperature. Phycoerythrin (PE)-labeled anti-Fas or hamster IgG in PBS with 2% goat serum was added to cells and incubated overnight at 4°C. Cells were washed and mounted in Vectashield (Vector Laboratories, Burlingame, CA) and viewed by fluorescent microscopy.

Chung et al. Blocking Fas/FasL reduces septic mortality and apoptosis

345

Fig. 1. Percent survival of mice following sepsis induced by CLP. Administration of FasFP to animals immediately (0 h) after CLP had no effect on survival compared with vehicle (veh)-treated mice. However, FasFP treatment at the beginning of the hypodynamic stage (12 h) of sepsis significantly increased survival compared with vehicle and 0 h-treated mice. *, P ⬍ 0.05, versus vehicle-treated mice; Fisher Exact test; n ⫽ 19 –20 mice/group.

Caspase-3 activity assays

RESULTS

Caspase activity was determined by fluorogenic assay according to Fukuzuka et al. [27] with minor modifications. LPS-stimulated macrophages were collected and lysed with lysis buffer {50 mM HEPES, 0.1% 3-[(3-cholamidopropyl) dimethylammonio]-1-propansulfonate (CHAPS), 1 mM dithiothreitol (DTT), 0.1 mM EDTA, 0.4% Triton X-100, pH 7.4} at 4°C for 15 min. The cell lysate was collected after centrifugation at 13,000 g for 10 min and incubated with the fluorescent caspase-3 substrate, Ac-DEVD-AFC (N-acetyl-Asp-GluVal-Asp-7-amino-4-trifluoromethyl coumarin; Enzyme Systems Products, Livermore, CA), at 100 ␮M in the assay buffer (50 mM HEPES, 1% sucrose, 0.1% CHAPS, 10 mM DTT, pH 7.4). The fluorescence of the cleaved substrates was followed spectrofluorometrically (excitation of 400 nm and emission of 505 nm) with a Bio-Tek FL500 microplate fluorescence reader (Winooski, VT). The protein content of the cell lysate was determined with a Bio-Rad (Hercules, CA) protein assay reagent, and the data are presented as picomole-cleaved AFC per mg protein per min, which is calculated from a standard curve plot with free AFC.

Delayed FasFP treatment improves survival to polymicrobial sepsis As our previous study demonstrated that FasL gene deficiency had protected mice from mortality in a CLP model of sepsis, we determined whether in vivo administration of FasFP, as a block of the Fas signaling pathway, had a similar effect on survival rate. Our results indicate that delayed treatment (12 h postCLP) with FasFP resulted in a significant increase in survival following CLP when compared with the vehicle-treated control CLP mice (Fig. 1). However, this survival advantage was absent in mice receiving FasFP at 0 h, as the treated group showed a comparable mortality rate to the control mice (Fig. 1).

Determination of macrophage cytokine production by enzyme-linked immunosorbent assay (ELISA) IL-1␤, IL-6, IL-10, and IL-12 produced from macrophages upon LPS stimulation were measured in culture supernatants by the “sandwich ELISA” technique as described previously [28] with monoclonal antibody pairs and the appropriate cytokine standards obtained from R&D Systems (Minneapolis, MN; IL-1␤) and BD-Pharmingen (San Diego, CA; IL-6, IL-10, and IL-12), respectively.

Plasma levels of liver enzymes and soluble FasL Blood was collected in a syringe containing 2 units heparin, transferred to a microtube, and centrifuged immediately at 10,000 g for 10 min at 4°C. Plasma samples were stored at –70°C until analyzed. Plasma levels of glutamatepyruvate-transaminase (GPT) and lactate dehydrogenase (LDH) were determined by using a Sigma assay kit (Sigma Chemical Co.), according to the manufacturer’s instruction. Soluble FasL concentration in plasma was determined with a commercially available coated ELISA kit (R&D Systems).

Presentation of data and statistical analysis The data are presented as mean ⫾ SE of n ⫽ 5–7 animals for each group. Differences were considered to be significant if P ⬍ 0.05, as determined using the one-way ANOVA and Tukey’s test. The survival data were compared using the Fisher exact test and were considered significant at P ⬍ 0.05.

346

Journal of Leukocyte Biology Volume 74, September 2003

Fig. 2. Apoptosis in LPS-stimulated peritoneal macrophages from four groups of mice. Cells were harvested 24 h after surgery and stimulated with 10 ␮g LPS per mL for another 24 h. PI cell-cycle analysis was used to determine the frequency of apoptosis. Peritoneal macrophages from vehicle-treated CLP mice show an increase in apoptosis in response to stimulation. Cells from FasFPtreated (12 h) CLP mice did not show changes in their apoptotic frequency. *, P ⬍ 0.05, versus sham. One-way ANOVA and Tukey’s test. Mean ⫾ SE; n ⫽ 5–7 mice/group.

http://www.jleukbio.org

Fig. 3. Giemsa staining of representative LPS-stimulated peritoneal macrophages (⫻400 original magnification). Cells from Sham (A) and CLP (B) were harvested 24 h after surgery and stimulated with 10 ␮g LPS per mL for another 24 h. Cells at arrows show feature characteristics of apoptosis, i.e., nuclear condensation, pyknosis, and shrinking cytoplasm.

Effects of FasFP treatment on inducible macrophage apoptosis in late polymicrobial sepsis are tissue-specific To examine the effects of FasFP treatment on apoptosis, macrophage cultures from different sources, such as the peritoneal cavity (peritoneal macrophages), spleen (splenic macrophages), and liver (Kupffer cells) of the four experimental groups of mice were established and stimulated with LPS. The results in Figure 2 show a significant increase in apoptosis, as determined by PI staining, in LPS-stimulated peritoneal macrophages derived from septic mice 24 h after CLP. The in vivo administration of FasFP did not alter the percentage of inducible peritoneal macrophage apoptosis. These results were confirmed by morphological analysis. Figure 3 shows the typical Giemsa staining of peritoneal macrophages obtained from a sham or a septic mouse following in vitro LPS stimulation in which the CLP mouse macrophage population exhibits an increase in the number of cells with condensed/pyknotic nuclei (typifying apoptosis). Repeated assessment of cells from different animals indicated the percentage of apoptosis for shamvehicle, sham-FasFP, CLP-vehicle, and CLP-FasFP was 9.3 ⫾ 0.6%, 9.2 ⫾ 0.3%, 45.6 ⫾ 3.4%*, and 47.3 ⫾ 3.2%*, respectively (*P⬍0.05 vs. sham; n⫽4/group). Alternatively, although LPS-stimulated Kupffer cell apoptosis (PI staining) was also significantly increased in vehicle-treated septic mice, treatment with FasFP significantly decreased Kupffer cell apoptosis (P⬍0.05 vs. CLP; Fig. 4). The results were con-

firmed by morphological analysis for which typical samples are shown in Figure 5. Multiple sampling of different animals indicated the % apoptosis for sham-vehicle, sham-FasFP, CLP-vehicle, and CLP-FasFP was 28.6 ⫾ 1.2%, 24.2 ⫾ 0.7%, 66.2 ⫾ 1.9%*, and 43.8 ⫾ 0.8%#, respectively (*P⬍0.05 vs. sham; #P⬍0.05 vs. CLP-vehicle; n⫽3/group). To examine whether Fas expression is altered on Kupffer cells, PE-labeled anti-Fas antibody was used. The result showed that there was no change in Fas expression on Kupffer cells between groups (data not shown). For splenic macrophages, we have consistently observed that there were no changes in the extent of apoptosis induced by LPS in cells derived from either group (data not shown).

Effects of FasFP treatment on caspase-3 activity in LPS-stimulated Kupffer cells after sepsis In a previous study [7], we demonstrated that caspases-3 activity is significantly increased in LPS-stimulated peritoneal macrophages of septic mice that were associated with increased apoptosis in these cells. Here, we determined whether the induction of caspase-3 activity in Kupffer cells from CLP mice was affected by FasFP treatment. As shown in Figure 6, caspase-3 activity in LPS-stimulated Kupffer cells from septic mice is up-regulated significantly when compared with cells from sham mice. Consistent with the morphological data/apoptosis rates, FasFP treatment decreased caspase-3 activity in these cells.

Fig. 4. Apoptosis in LPS-stimulated Kupffer cells from four groups of mice. Cells were harvested 24 h after surgery and stimulated with 10 ␮g LPS per mL for another 24 h. PI cell-cycle analysis was used to determine the frequency of apoptosis. Kupffer cells from vehicle-treated CLP mice show an increase in apoptosis in response to stimulation. FasFPtreated (12 h) CLP mice show a decrease in their apoptotic frequency. *, P ⬍ 0.05, versus sham; #, P ⬍ 0.05, versus CLP. One-way ANOVA and Tukey’s test. Mean ⫾ SE; n ⫽ 5–7 mice/group.

Chung et al. Blocking Fas/FasL reduces septic mortality and apoptosis

347

Effects of FasFP treatment on cytokine release from different macrophages in response to LPS stimulation after sepsis Figure 7A illustrates that the capacity of peritoneal macrophages to release proinflammatory cytokines (IL-1␤, IL-6, and IL-12) in response to LPS stimulation is significantly depressed, and release of the anti-inflammatory cytokine IL-10 is increased in peritoneal macrophages from septic mice when compared with vehicle and FasFP-treated sham animals. Treatment of septic mice with FasFP did not alter the capacity of these cells to release cytokines in response to LPS. The release of IL-6, IL-12, and IL-10 (IL-1␤ was not detectable) in LPS-stimulated splenic macrophages (Fig. 7B) was similar to those results obtained with peritoneal macrophages in which a depression in IL-6 and IL-12 release and an enhancement in IL-10 release were observed after CLP. FasFP treatment had no effect on cytokine release from splenic macrophages from septic mice. Kupffer cell IL-6 (IL-1␤ and IL-12 were not detectable) release upon LPS stimulation was significantly depressed in

Fig. 5. Giemsa staining of representative LPS-stimulated Kupffer cells (⫻400 original magnification). Cells from Sham (A), CLP (B), and CLP-FasFP (12 h; C) were harvested 24 h after surgery and stimulated with 10 ␮g LPS per mL for another 24 h. Cells at arrows show feature characteristics of apoptosis, i.e., nuclear condensation, pyknosis, and shrinking cytoplasm.

Fig. 6. Caspase-3 activity in LPS-stimulated Kupffer cells from four groups of mice. Enzyme activity was measured by the cleavage of the caspase-3 substrate Ac-DEVD-AFC. Increased caspase-3 activity was observed in Kupffer cells after sepsis. FasFP treatment (12 h) significantly decreased caspase-3 activity in Kupffer cells. *, P ⬍ 0.05, versus sham; #, P ⬍ 0.05, versus CLP. One-way ANOVA and Tukey’s test. Mean ⫾ SE; n ⫽ 5–7 mice/group.

348

Journal of Leukocyte Biology Volume 74, September 2003

Fig. 7. Cytokine release in LPS-stimulated peritoneal macrophages (A), splenic macrophages (B), and Kupffer cells (C). Cells were harvested 24 h after surgery and stimulated with 10 ␮g LPS per mL for another 24 h. Cytokine release in cultured supernatants was determined by ELISA. Although a marked decrease in IL-1␤ (only detected in peritoneal macrophages), IL-6, and IL-12 levels in vehicle-treated CLP mice compared with sham and shamvehicle groups, IL-10 release is increased in all three macrophages from septic mice. FasFP treatment (12 h) only attenuated IL-10 but not the proinflammatory cytokine release in Kupffer cells. *, P ⬍ 0.05, versus sham; #, P ⬍ 0.05, versus CLP. One-way ANOVA and Tukey’s test. Mean ⫾ SE; n ⫽ 5–7 mice/group. ND, Not detectable.

http://www.jleukbio.org

septic mice (Fig. 7C). Like peritoneal and splenic macrophages, treatment of CLP animals with FasFP did not restore IL-6 release. However, although Kupffer cell IL-10 release increased in vehicle-treated animals after sepsis, the administration of FasFP significantly attenuated the rise of IL-10 release.

Effects of FasFP treatment on plasma levels of liver enzymes, GPT and LDH, and soluble FasL after sepsis To investigate whether the changes in Kupffer cell apoptosis or function might be associated with the onset of liver damage, we determined the plasma levels of GPT and LDH in these mice. GPT (Fig. 8A) and LDH (Fig. 8B) were increased significantly at 24 h after the onset of sepsis; however, these enzyme levels were significantly attenuated in FasFP-treated animals. To determine whether FasFP treatment affects soluble FasL in circulation, the plasma levels of soluble FasL were measured. We observed very low concentrations (ranging from 18 to 27 pg/mL), which did not vary significantly between groups (data not shown).

DISCUSSION The results presented here demonstrate that blockade of the Fas/FasL system 12 h after CLP, during the end of the hyperdynamic phase but before the onset of the hypodynamic or immunosuppressive phase, provides a survival benefit in septic mice. Alternatively, when FasFP was given immediately after

Fig. 8. Plasma levels of GPT (A) and LDH (B) at 24 h after CLP. Both enzymes were significantly increased in vehicle-treated CLP mice; treatment of FasFP (12 h) significantly attenuated the production of these enzymes. *, P ⬍ 0.05, versus sham; #, P ⬍ 0.05, versus CLP. One-way ANOVA and Tukey’s test. Mean ⫾ SE; n ⫽ 5–7 mice/group.

CLP, there was no effect on survival compared with vehicletreated controls. Prior studies from our laboratory indicated that a deficiency of the gene encoding FasL provides significant protection from the development of immune dysfunction and mortality [20]. However, the survival advantage only becomes apparent over time (⬎6 days post-CLP), although improvement in immune function can be seen at 24 h post-CLP. This agrees with the concept that increased apoptosis in the immune system is not evident until 12 h post-CLP and beyond. Thus, the ability of delayed FasFP administration to inhibit a more chronic process, such as the development of immune dysfunction and late apoptosis, is consistent with the observations made in the FasL gene-deficient animals. This may also reflect that FasL is a somewhat minor contributor to the proinflammatory process during the early period following sepsis [22]. Therefore, neither FasFP nor FasL gene deficiency would exhibit its most marked effects at this stage. Recently, we have reported that treatment of animals with FasFP at 12 h post-CLP significantly restored the hepatic and intestinal blood flow that was depressed in septic mice [22]. Although we postulate here that the protection of FasFP in the septic mouse may be an indirect result of the maintenance of organ function, the precise mechanism remains to be established thoroughly. The role of the Fas/FasL system in the regulation of lymphocyte apoptosis is well described [29 –31]; however, less is known about its effect on specific macrophage populations. In vivo studies have demonstrated that various agents, such as LPS, TNF, IL-1, IL-10, IL-4, nitric oxide, and FasL [19, 32, 33], can regulate macrophage apoptosis. We have previously observed that macrophages isolated from septic mice that are not subjected to further stimulation or culture exhibited variable degrees of apoptosis, with an increase in peritoneal macrophage basal apoptosis, a small but significant decline in Kupffer cell apoptosis, and no change in splenic macrophage apoptosis [6]. In contrast, when cells from septic mice are stimulated in vitro with LPS, peritoneal macrophages and Kupffer cells exhibit a consistent increase beyond their own base level of apoptosis compared with sham animal cells [6]. It is interesting that the apoptotic frequency in splenic macrophages from septic mice remains unchanged in the presence or absence of LPS [5, 6]. Here, we have continued to expand our understanding of this divergence and the changes in susceptibility to the onset of apoptosis in macrophages by inhibiting the Fas/FasL system during sepsis. The results indicate that blockade of Fas/FasL signaling at 12 h after sepsis has a differential effect on the apoptotic frequency in macrophages harvested from various tissue sites. FasFP treatment attenuated the elevation in Kupffer cell apoptosis, which was seen in vehicle-treated CLP mice. However, the increase in peritoneal macrophage apoptosis was not affected by FasFP treatment, and similarly, it had no effect on splenic macrophage apoptosis. Macrophages express Fas and FasL constitutively; however, studies demonstrate that treatment with LPS did not alter the expression of either protein [19, 34]. Consistent with these findings, our results show that there was no difference in Fas expression on Kupffer cells taken from sham or CLP mice (data not shown). It has been proposed that monocyte FasL is not used in the autocrine induction of apoptosis but may be in-

Chung et al. Blocking Fas/FasL reduces septic mortality and apoptosis

349

volved in effector (paracrine/endocrine) functions when released from the cell surface [29]. It is possible that in peritoneal and liver macrophages, the Fas/FasL system is not the central apoptotic pathway activated during sepsis but may be serving as a regulatory mechanism. This may in part explain why blocking Fas activation did not decrease peritoneal macrophage apoptosis. It appears that other factors besides Fas and FasL are induced during LPS stimulation, which may contribute to the sensitivity of Kupffer cells to apoptosis. Additionally, it has been suggested that soluble FasL in circulation may also play some role in a paracrine regulation of apoptosis [35]. However, soluble FasL concentration in plasma was nearly undetectable in our animals regardless of treatment. Thus, the mechanism by which FasFP mediates its divergent effects on apoptosis and function of macrophages from various tissue sites remains to be further elucidated. In keeping with our previous studies [5, 36], macrophages from mice subjected to polymicrobial sepsis exhibited a depression of proinflammatory cytokine release in response to LPS. This macrophage hyporesponsiveness is closely associated with enhanced macrophage apoptosis, particularly in peritoneal macrophages and Kupffer cells. In addition, anti-inflammatory cytokine IL-10 release is significantly increased in septic macrophages following LPS stimulation. In vivo administration of FasFP at 12 h post-CLP had no effect on the decline in proinflammatory cytokine release from each macrophage population tested here. Similarly, it did not affect the release of IL-10 from peritoneal and splenic macrophages, but it did significantly attenuate IL-10 release from Kupffer cells. The liver is one potential target organ of Fas-mediated cell death, as injection of an agonistic anti-Fas antibody induces extensive cell death of hepatocytes, corresponding with a high level of liver enzymes and lethality in mice [37]. We have demonstrated that the changes in apoptosis and IL-10 release of Kupffer cells were associated with a reduction in liver damage (attenuated plasma levels of liver enzymes) subsequent to septic insult when FasFP was administered in vivo. Therefore, protecting Kupffer cells from apoptosis with FasFP treatment may directly or indirectly provide a benefit for the liver by preventing bystander cellular damage and dysfunction, thereby improving survival to septic challenge. Alternatively, attenuating IL-10 production by FasFP treatment may play some role in the decrease in inducible Kupffer cell apoptosis seen during sepsis, as studies have shown that the T helper cell type 2 cytokines IL-10 and IL-4 can enhance monocyte apoptosis [38]. In summary, our data indicate that in vivo blockade of the Fas/FasL pathway by the administration of FasFP 12 h after the onset of sepsis improves survival and has a divergent effect on macrophage apoptosis and function, which is dependent on the tissue of origin. FasFP treatment attenuates liver damage, suppresses Kupffer cell apoptosis, and blocks the increased IL-10 release seen in response to LPS stimulation in septic mice. However, FasFP neither restores function nor decreases apoptosis in peritoneal macrophages. Although the precise mechanism is not known, we speculate that blocking Fasmediated apoptosis may preserve specific/key organ and immune functions, contributing to the host’s defense against 350

Journal of Leukocyte Biology Volume 74, September 2003

septic challenge and thus, may provide a novel target for therapeutic intervention.

ACKNOWLEDGMENTS This investigation was supported by National Institutes of Health Grant R01-GM53209. We thank Sara Spangenberger, Ginny Hovanesian, and Paul Monfils at the Core Laboratories Facilities at Rhode Island Hospital for their assistance with the flow cytometry and histologic examination.

REFERENCES 1. Goris, R. J. A. (1993) Sepsis and multiple organ failure: the result of whole body inflammation. In Host Defense Dysfunction in Trauma, Shock and Sepsis E. Faist, J. Meakins, F. W. Schildberg, Berlin-Heidelberg, Springer-Verlag, 161–170. 2. Bone, R. C. (1996) Sir Issac Newton, sepsis, SIRS and CARS. Crit. Care Med. 24, 1125–1128. 3. Abraham, E. (2000) Cytokine neutralization: an overview. In Immune Response in the Critically Ill J. C. Marshall, J. Cohen, Berlin-Heidelberg, Springer-Verlag, 385–392. 4. Barriere, S. L., Lowry, S. F. (1995) An overview of mortality risk prediction in sepsis. Crit. Care Med. 23, 376 –393. 5. Ayala, A., Perrin, M. M., Kisala, J. M., Ertel, W., Chaudry, I. H. (1992) Polymicrobial sepsis selectively activates peritoneal but not alveolar macrophage to release inflammatory mediators (IL-1, IL-6 and TNF). Circ. Shock 36, 191–199. 6. Ayala, A., Urbanich, M. A., Herdon, C. D., Chaudry, I. H. (1996) Is sepsis-induced apoptosis associated with macrophage dysfunction? J. Trauma 40, 568 –574. 7. Chung, C. S., Song, G. Y., Moldawer, L. L., Chaudry, I. H., Ayala, A. (2000) Neither Fas ligand nor endotoxin is responsible for inducible phagocyte apoptosis during sepsis/peritonitis. J. Surg. Res. 91, 147–153. 8. Nagata, S., Golstein, P. (1995) The Fas death factor. Science 267, 1449 –1456. 9. Ayala, A., Herdon, C. D., Lehman, D. L., Ayala, C. A., Chaudry, I. H. (1996) Differential induction of apoptosis in lymphoid tissues during sepsis: variation in onset, frequency, and the nature of the mediators. Blood 87, 4261– 4275. 10. Hotchkiss, R. S., Swanson, P. E., Freeman, B. D., Tinsley, K. W., Cobb, J. P., Matuschak, G. M., Buchman, T. G., Karl, I. E. (1999) Apoptotic cell death in patients with sepsis, shock and multiple organ dysfunction. Crit. Care Med. 27, 1230 –1251. 11. Ayala, A., Chung, C. S., Chaudry, I. H. (1998) Lymphocyte apoptosis in sepsis. Sepsis 2, 55–71. 12. Meldrum, D. R. (1998) Tumor necrosis factor in the heart. Am. J. Physiol. 274, R577–R595. 13. Hotchkiss, R. S., Tinsley, K. W., Swanson, P. E., Chang, K. C., Cobb, J. P., Buchman, T. G., Korsmeyer, S. J., Karl, I. E. (1999) Prevention of lymphocyte cell death in sepsis improves survival in mice. Proc. Natl. Acad. Sci. USA 96, 14541–14546. 14. Pinkoski, M. J., Brunner, T., Green, D. R., Lin, T. (2000) Fas and Fas ligand in gut and liver. Am. J. Physiol. Gastrointest. Liver Physiol. 278, G354 –G366. 15. French, L. E., Hahne, M., Viard, I., Radlgruber, G., Zanone, R., Becker, K., Muller, C., Tschopp, J. (1996) Fas and Fas ligand in embryos and adult mice: ligand expression in several immune-privileged tissues and coexpression in adult tissues characterized by apoptotic cell turnover. J. Cell Biol. 133, 335–343. 16. Squier, M. K., Sehnert, A. J., Cohen, J. J. (1995) Apotosis in leukocytes. J. Leukoc. Biol. 57, 2–10. 17. Miyawaki, T., Uehara, T., Nibu, R., Tsuji, T., Yachie, A., Yonehara, S., Taniguchi, N. (1992) Differential expression of apoptosis-related Fas antigen on lymphocyte subpopulations in human peripheral blood. J. Immunol. 149, 3753–3758. 18. Suda, T., Takahashi, T., Golstein, P., Nagata, S. (1993) Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 75, 1169 –1178. 19. Kiener, P. A., Davis, P. M., Starling, G. C., Mehlin, C., Klebanoff, S. J., Ledbetter, J. A., Liles, W. C. (1997) Differential induction of apoptosis by

http://www.jleukbio.org

20. 21.

22. 23.

24. 25.

26. 27.

28.

Fas-Fas ligand interactions in human monocytes and macrophages. J. Exp. Med. 185, 1511–1516. Chung, C. S., Xu, Y. X., Wang, W., Chaudry, I. H., Ayala, A. (1998) Is Fas ligand or endotoxin responsible for mucosal lymphocyte apoptosis in sepsis? Arch. Surg. 133, 1213–1220. Mountz, J. D., Zhou, T., Su, X., Cheng, J., Pierson, M., Bluethmann, H., Edwards, C. K. (1996) Autoimmune disease results from multiple interactive defects in apoptosis induction molecules and signaling pathways. Behring Inst. Mitt. 97, 200 –219. Chung, C. S., Yang, S. L., Song, G. Y., Lomas, J., Wang, P., Simms, H. H., Chaudry, I. H., Ayala, A. (2001) Inhibition of Fas signaling prevents hepatic injury and improves organ blood flow. Surgery 130, 339 –345. Ayala, A., Perrin, M. M., Wang, P., Ertel, W., Chaudry, I. H. (1991) Hemorrhage induces enhanced Kupffer cell cytotoxicity while decreasing peritoneal or splenic macrophage capacity: involvement of cell-associated TNF and reactive nitrogen. J. Immunol. 147, 4147– 4154. Ayala, A., Perrin, M. M., Chaudry, I. H. (1990) Defective macrophage antigen presentation following haemorrhage is associated with the loss of MHC class II (Ia) antigens. Immunology 70, 33–39. Ayala, A., Perrin, M. M., Ertel, W., Chaudry, I. H. (1992) Differential effects of haemorrhage on Kupffer cells: decreased antigen presentation despite increased inflammatory cytokine (IL-1, IL-6 and TNF) release. Cytokine 4, 66 –75. Telford, W. G., King, L. E., Fraker, P. J. (1992) Comparative evaluation of several DNA binding dyes in the detection of apoptosis-asssociated chromatin degradation by flow cytometry. Cytometry 13, 137–143. Fukuzuka, K., Rosenberg, J. J., Gaines, M. D., Edwards, C. K., ClareSalzler, M., MacKay, S. L. D., Moldawer, L. L., Copeland, E. M., Mozingo, D. W. (1999) Caspase-3 dependent organ apoptosis early after burn injury. Ann. Surg. 229, 851– 858. Ayala, A., Deol, Z. K., Lehman, D. L., Herdon, C. D., Chaudry, I. H. (1994) Polymicrobial sepsis but not low dose endotoxin infusion causes decreased splenocyte IL-2/IFN-gamma release while increasing IL-4/ IL-10 production. J. Surg. Res. 56, 579 –585.

29. Tanaka, A., Suda, T., Takahashi, T., Nagata, S. (1995) Expression of the functional soluble form of human Fas ligand in activated lymphocytes. EMBO J. 14, 1129 –1135. 30. Schattner, E., Friedman, S. M. (1996) Fas expression and apoptosis in human B cells. Immunol. Res. 15, 246 –257. 31. Suda, T., Okazaki, T., Naito, Y., Yokota, T., Arai, N., Ozaki, S., Nakao, K., Nagata, S. (1995) Expression of the Fas ligand in cells of T cell lineage. J. Immunol. 154, 3806 –3813. 32. Managan, D. F., Welch, G. R., Wahl, S. M. (1991) Lipopolysaccharide, tumor necrosis factor-␣ and IL-1␤ prevent programmed cell death (apoptosis) in human peripheral blood monocytes. J. Immunol. 146, 1541– 1546. 33. Sarih, M., Souvannavong, V., Adam, A. (1993) Nitric oxide synthase induces macrophage death by apoptosis. Biochem. Biophys. Res. Commun. 191, 503–508. 34. Um, H. D., Orenstein, J. M., Wahl, S. M. (1996) Fas mediates apoptosis in human monocytes by a reactive oxygen intermediate dependent pathway. J. Immunol. 156, 3469 –3477. 35. Liles, W. C., Kiener, P. A., Ledbetter, J. A., Aruffo, A., Klebanoff, S. J. (1996) Differential expression of Fas (CD95) and Fas ligand on normal human phogocytes: implications for the regulation of apoptosis in neutrophils. J. Exp. Med. 184, 429 – 440. 36. Ayala, A., Perrin, M. M., Wang, P., Chaudry, I. H. (1992) Sepsis induces an early increased spontaneous release of the hepatocellular stimulatory factor (IL-6) by Kupffer cells in both endotoxin tolerant or intolerant mice. J. Surg. Res. 52, 635– 641. 37. Rodriguez, I., Matsuura, K., Ody, C., Nagata, S., Vassalli, P. (1996) Systemic injection of a tripeptide inhibits the intracellular activation of CPP32-like proteases in vivo and fully protects mice against Fas-mediated fulminant liver destruction and death. J. Exp. Med. 184, 2067–2072. 38. Estaquier, J., Ameisen, J. C. (1997) A role for T-helper type-1 and type-2 cytokines in the regulation of human monocyte apoptosis. Blood 90, 1618 –1625.

Chung et al. Blocking Fas/FasL reduces septic mortality and apoptosis

351