Abstract
Clinical correlations between bacterial infections and rejection suggest a hypothesis that innate immune stimulation by bacterial infections results in the production of inflammatory cytokine that facilitate bystander T cell activation, increased alloreactivity and inhibition of tolerance induction. Previous studies demonstrated that IFNs produced during an infection with a model bacterium, Listeria monocytogenes, prevented the induction of transplantation tolerance in mice with anti-CD154 and donor-specific transfusion (DST) (1). We investigated the impact of two clinically relevant bacterial infections at the time of transplantation on the ability of anti-CD154 and DST to induce skin allograft acceptance in mice. Staphylococcus aureus (SA) infection prevented skin allograft acceptance whereas maximally tolerated doses of Pseudomonas aeruginosa infection had no effect. SA induced an acute production of IL-6, which was necessary and sufficient for the prevention of skin allograft acceptance VSports最新版本. Furthermore, a single pulse of methylprednisolone modulated IL-6 production during SA infection and facilitated skin allograft acceptance in SA-infected recipients. Taken together, our results suggest that bacterial infections elicit specific pro-inflammatory cytokines signatures that can serve as barriers to tolerance induction, and that inhibiting the production of or neutralizing these inflammatory cytokines can synergize with co-stimulatory blockade-based therapies to facilitate the development of transplantation tolerance.
Keywords: bacterial infection, donor-specific alloreactivity, donor-specific transfusion, immunosuppression, transplant immunology and tolerance, transplant rejection, immune regulation
Introduction
In the absence of immunosuppression, transplantation across histocompatibility barriers inevitably results in acute allograft rejection. Over the past several decades, pharmacologic immunosuppressive regimens have improved significantly from the rudimentary regimen of steroids and azathioprine to the more sophisticated multi-drug regimens involving anti-proliferative agents, calcineurin inhibitors, and a variety of biologics aimed at suppressing T cell reactivity (2). These current regimens result in global immunosuppression leaving the recipient vulnerable to severe infections and virus-mediated cancers (3) VSports注册入口. Furthermore, while these immunosuppressive agents have dramatically improved rates of acute graft rejection, they have proven less efficacious in fostering long-term graft survival and chronic rejection remains an important barrier to the goal of long-term graft survival.
These observations have led the field to postulate that many of the shortcomings associated with current immunosuppressive strategies can only be overcome by developing transplantation tolerance, characterized by extended suppression of pathologic allograft-specific immune responses in the absence of maintenance immunosuppression, while leaving the remainder of the immune system competent to fight infections and malignancies. Several strategies for tolerance induction have been investigated, the most promising currently being the induction of peripheral operational tolerance using costimulation-targeting therapies that block the engagement of costimulatory receptors on T cells to ligands on APCs that is essential for optimal T cell stimulation (4). Targeting of the CD40-CD154 pathway with a blocking anti-CD154 monoclonal antibody has resulted in successful tolerance to cardiac allografts in mice, and in long-term survival of kidney allografts in non-human primates (5) V体育官网入口. Mechanistically, this therapy is thought to be dependent on induction of alloreactive T cell anergy, deletion, and regulation (6, 7).
Identifying potential barriers to the induction of such tolerance as well as events that may reverse established allograft tolerance could prove to have profound implications in clinical practice. Clinical data support a correlation between viral infections and acute rejection of established allografts via a variety of mechanisms including bystander activation, memory and heterologous immunity, as well as molecular mimicry (8–11). In addition to viruses, bacteria that express ligands capable of activating innate immunity may also be capable of enhancing the adaptive immune response to allografts, thereby preventing tolerance. In line with this possibility are the observations that graft survival is affected by the type of organ or tissue that is transplanted. Sterile organs such as heart, liver, and kidney tend to have an overall half-life superior to organs such as skin, lung, and intestine that serve a barrier function with inherent exposure to the environment and resident microbes VSports在线直播. These observations have led us to hypothesize that local microbes contribute to the enhanced allogenicity observed after transplantation of the latter set of organs. Indeed, reports in the clinical literature reveal an association between various types of bacterial infections and poor graft survival from both acute and chronic rejection (12).
We have previously reported as proof-of-principle that the model bacterium, Listeria monocytogenes, can both prevent the induction of and reverse established allograft tolerance in murine models utilizing co-stimulation blockade (1, 13). We have extended these observations with this investigation to address an important but under-investigated area in transplantation immunology, namely, whether clinically significant nosocomial bacterial infections can prevent and reverse allograft tolerance V体育2025版. Staphylococcus aureus (SA) and Pseudomonas aerugenosa (PA) are prevalent bacterial infections in the initial months following transplantation (8–10, 14). Staphylococcus aureus is a gram-positive commensal bacterium of human skin and mucous membranes, colonizing the nostrils of up to 50 percent of patients tested (15, 16). Upon breach of normal skin and mucosal barriers, SA becomes an important human pathogen and is a leading cause of bloodstream infection, skin and soft-tissue infection, and lower respiratory infection (15, 17). Pseudomonas is a gram-negative rod bacteria that has become an important cause of infection, especially in patients with compromised host defense mechanisms (18). Our investigations reveal that infection with SA, but not PA, at the time of transplantation, prohibits successful skin allograft acceptance by inducing inflammation and IL-6 production, which allows for CD154-independent bystander activation of alloreactive T cells and precipitates acute allograft rejection.
Materials and Methods
"VSports注册入口" Mice
Female C57BL/6 (B6, H-2b), BALB/c (B/c, H-2d), C3H/HeJ (H-2k) inbred mice, age 8–9 weeks, were purchased from either The Jackson Laboratory (Bar Harbor, ME), The Division of Cancer Treatment at the National Cancer Institute (Frederick, MD), or Charles River (Wilmington, MA). RAG2−/− mice on the C57BL/6 background were purchased from Taconic (Hudson, NY). TCRβδ−/− mice, CD8−/− mice, IL-6−/− mice, and mAct-OVA transgenic mice, all on the C57BL/6 background, were purchased from the Jackson Laboratory. MyD88−/− mice on the BALB/c and C57BL/6 backgrounds were provided by Dr. S. Akira (Osaka University, Osaka, Japan) (19). OTI transgenic mice on a C57BL/6 (CD90. 1 congenic) background whose T cells recognize the OVA257–264 peptide in the context of H-2Kb were a gift from Dr. A. Sperling (University of Chicago, Chicago, IL). OTII transgenic mice on C57BL/6 background (CD45. 1 congenic) whose T cells recognize the OVA323–339 peptide presented by I-Ab were a gift from Dr. Y. X. Fu (University of Chicago, Chicago, IL). TCR75 transgenic mice on a C57BL/6 background whose T cells recognize the H-2Kd-derived peptide (Kd54–68) presented on I-Ab were a kind gift form Dr. P. Bucy (University of Alabama, Birmingham, AL) VSports. Animals were kept in a biohazard facility and used in agreement with the University of Chicago Institutional Animal Care and Use Committee according to the National Institutes of Health guidelines for animal use.
Skin Transplantation
Anti-CD154 mAbs were generated from the anti-CD154 (MR1) hybridoma, which was a generous gift from Dr. K. Bishop (University of Michigan, Ann Arbor, MI). After growth in protein-free hybridoma medium (Invitrogen), anti-CD154 mAbs were purified from hybridoma supernatants using 45% ammonium sulfate precipitation and then dialyzed in PBS for 72 hours VSports app下载. Donor grafts were prepared by harvesting full thickness dorsal flank skin approximately 1 cm in diameter from either wild type BALB/c, MyD88−/− BALB/c, or mAct-OVA transgenic C57BL/6 mice where indicated. Skin grafts were then transplanted onto the dorsal flank of recipient mice that were either wild type C57BL/6 or RAG2−/−, TCRβδ−/−, CD8−/−, MyD88−/−, or IL-6−/− mice all on the C57BL/6 background where indicated. Recipient animals were then treated with anti-CD154 (1mg/dose i. v. on day 0, and i. p. on days 7 and 14 post-transplantation) in combination with DST (107 donor splenocytes on the day of transplantation). Skin grafts were followed visually for rejection, as defined by evidence of graft necrosis or contraction. For IL-6 neutralization studies, purified anti-IL-6 antibody (MP5-20F3, Rat IgG1) was purchased from BioXcell (West Lebanon, NH) and injected post-transplantation (500 μg i. v. , day 0; 250 μg i. p. , days 1, 3, 5, 7, 10, and 14). For IL-17 neutralization studies, purified anti-IL-17 antibody (clone 50104, Rat IgG2a) was purchased from R&D Systems (Minneapolis, MN) and injected post-transplantation (100 μg/mouse i. v. day 0, i. p days 2, 3, 4, 7, 9, 11 and 14). For pharmacologic immunosuppression studies, mice were treated with either methylprednisolone (20 mg/kg, day 0, i. p), cyclosporin (20 mg/kg, day 0, i. p. ), or sirolimus (0. 3 mg/kg, day 0, i. p) purchased from the University of Chicago Hospitals Pharmacy.
V体育官网 - Bacterial Preparations and Mouse Infections
SA strain EsxB:erm, a generous gift from Dr. D. Missiakas (University of Chicago, Chicago, IL) (20), was grown at 37°C overnight in tryptic soy broth (BD Biosciences) containing 10μg/ml erythromycin. PA strain PA01, a generous gift from Dr. J. Alverdy (University of Chicago, Chicago, IL), was grown in tryptic soy broth overnight at 37°C. After overnight growth, SA and LM cultures were diluted 100-fold in fresh media, and incubated at 37°C for 5 hours. LM strain, LM-OVA, was grown in brain-heart infusion broth (BD Biosciences) at 37°C overnight. Bacteria were grown to log phase cultures and titers were quantified by OD600. Bacteria were collected by centrifugation, washed twice, and resuspended in PBS. Mice receiving live infections were injected i.p. on the day of transplantation with SA (2×108 cfu/200μl), LM (2×105 cfu/200μl), or PA (2×107 cfu/200μl). Heat killed SA was generated by incubating cultured SA at 60°C for 3 hours. Heat killed SA particles were then washed twice with PBS. Mice receiving heat killed SA were injected with 2×109 cfu/200 μl i.p. on the day of transplantation. Bacterial doses were chosen as the highest sub-lethal dose acceptable after performing an LD50 analysis for each strain.
T cell Proliferation Assays (VSports)
In vivo CD4+ and CD8+ T cell proliferation was analyzed after adoptive transfer into recipients of skin transplantation. Splenocytes from OTI and OTII mice were processed into single cell suspensions and enriched for CD8+ and CD4+ T cells, respectively, by negative magnetic separation (Miltenyi Biotec). They were then labeled with carboxyfluorescein succinimidyl ester (CFSE) and adoptively transferred into mice on the day of transplantation (2–5×105 OTI and OTII cells, i.v.). Five days after transplantation splenocytes were harvested, processed into single cell suspensions, and stained for flow cytometry with the following antibodies in 1%BSA/0.02% sodium azide (FACS buffer) for 1 hour at 4°C: 1) OTI staining: PE-labeled anti-TCRVα2 (B20.1, rat IgG2a) was purchased from eBioscience (San Diego, CA). Biotinylated TCRVβ5.1/5.2 (MR9-4, mouse IgG1), PerCP-labeled anti-CD90.1 (OX-7, mouse IgG1), PECy7-labeled anti-CD8 (53–6.7, rat IgG2a), APC-labeled anti-GR1 (RB6-8C5, rat IgG2b), APC-labeled anti-CD19 (1D3, rat IgG2a), APC-labeled anti-CD11b (M1/70, rat IgG2b), APC-labeled anti-CD11c (HL3, hamster IgG1) were purchased from BD Biosciences (Franklin Lakes, NJ). 2) OTII staining: PE-labeled anti-TCRVα2 (B20.1, rat IgG2a), biotinylated anti-CD45.1 (A20, mouse IgG2a) were purchased from eBioscience (San Diego, CA). PECy7-labeled anti-CD4, APC-labeled anti-GR1 (RB6-8C5, rat IgG2b), APC-labeled anti-CD19 (1D3, rat IgG2a), APC-labeled anti-CD11b (M1/70, rat IgG2b), APC-labeled anti-CD11c (HL3, hamster IgG1) were purchased from BD Biosciences (Franklin Lakes, NJ). Secondary staining was performed with APCCy7-labeled streptavidin, purchased from BD Biosciences (Franklin Lakes, NJ). After all staining, cells were washed twice with FACS buffer. Samples were run on the LSR-II (BD Biosciences, Franklin Lakes, NJ) and analyzed for evidence of proliferation as indicated by dilution of CFSE intensity using FlowJo cytometry analysis software (Tree Star, Ashland, OR).
Serum IL-6 Measurements
After C57BL/6 were injected with SA (2×108), LM (2×105), PA (2×105), or IL-6-expressing or control plasmids, blood was collected by retro-orbital puncture at the indicated time points and serum was separated by centrifugation. Serum samples were stored at -20°C for subsequent analysis. Initial serum cytokine screening after SA infection was conducted by Mouse cytokine 20-Plex panel (Invitrogen, Carlesbad, CA). The concentrations of IL-6 were subsequently measured by ELISA (eBioscience, San Diego, CA).
Hydrodynamic IL-6 Gene Delivery
Mouse IL-6 expressing and empty control plasmids were purchased from Invivogen (San Diego, CA). Stock plasmids were isolated and prepared using EndoFree Plasmid Mega Kit (Qiagen, Valencia, CA). Plasmid DNA concentration was determined by spectrometer and confirmed in by agarose gel electrophoresis. Plasmid DNA (5 μg/mouse) was diluted in 1.9ml sterile nonpyrogenic 0.9% sodium chloride solution (Hospira Inc., Lake Forest, IL), and injected through tail vein within 5–6 seconds using a 3ml syringe with 27G1/2 needle (BD Bioscience, Franklin Lakes, NJ) two days prior to transplantation.
"VSports" IFNγ and IL-17 ELISPOT Assays
The IFN γ and IL-17 ELISPOT assays were conducted according to the instructions of the manufacturer (BD Biosciences, Franklin Lakes, NJ). Briefly, ELISPOT plates (Millipore, Billerica, MA) were coated with purified anti-IFN γ capture antibody or purified anti-IL-17 capture antibody (BD Biosciences, Franklin Lakes, NJ) overnight at 4°C and then blocked with 10%FBS/PBS. Responder splenocytes (106/well, in triplicate) harvested from transplanted mice were co-cultured with γ-irradiated (3000 rads) splenocytes (5×105/well) that were either syngeneic (C57B/L6), allogeneic (BALB/c), third-party (C3H), or syngeneic splenocytes pulsed overnight with heat killed SA (108 cfu/107 splenocytes). Responder cells were also stimulated with or without anti-CD3 (2C11). Cell cultures were incubated for 12 hours at 37°C, 5% CO2. Biotinylated anti-IFNγ or anti-IL-17 detection antibodies (BD Biosciences, Franklin Lakes, NJ) were added, followed by HRP-conjugated anti-biotin (BD Biosciences, Franklin Lakes, NJ). Plates were developed using 3-amino-9-ethylcarbazole (AEC) substrate. The numbers of spots per well were enumerated using the ImmunoSpot Analyzer (CTL Analyzers LLC, Cleveland).
Statistics
The two-tailed Student’s t test or the Tukey test for multiple comparisons was performed to determine statistical differences between groups.
Results
V体育官网 - SA infection at the time of transplantation prevents the induction of skin allograft acceptance with anti-CD154/DST
We have previously shown that a model intracellular organism, LM, can prevent the induction of skin allograft acceptance (1); however, we were interested in extending those observations and exploring the impact of two clinically important bacterial species, the gram-positive commensal and pathogen, SA, as well as the gram-negative opportunistic pathogen, Pseudomonas aerugninosa (PA). Using a model of skin allograft acceptance induced by anti-CD154/DST, we tested the impact of SA and PA infections on the ability of anti-CD154/DST treatment to induce skin allograft acceptance (Figure 1A). In the absence of anti-CD154/DST treatment, allogeneic skin grafts were rejected within 12 days. Upon treatment with anti-CD154/DST, long-term skin allograft survival was achieved with a median survival of 68 days. Infection with a maximally tolerated dose of PA had no effect on allograft acceptance and the median graft survival was >60 days. Despite administration of anti-CD154/DST, SA infection at the time of transplantation precipitated an acute rejection in 75 percent of recipients of allogeneic skin grafts, with a median graft survival of 15 days. Importantly, SA infection had no effect on survival of syngeneic skin grafts, which persisted long-term indicating that SA did not induce non-specific inflammation that prevented healing of skin grafts, but rather, SA was augmenting allo-specific immune responses.
Figure 1.
(A) SA infection at the time of transplantation prevents tolerance induction. Allogeneic skin transplantation was performed by grafting full-thickness trunk skin from BALB/c donors onto the flank of C57BL/6 recipients. Transplant recipients were either left untreated (N=29) or treated (N=22) with anti-CD154 (1mg/mouse, day 0 i.v., day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0, i.v.,) in the absence or presence of infection with PA (2×107 cfu, i.p.; N=4), LM (2×105 cfu, i.p.; N=4), or SA (2×108 cfu, i.p.; N=46) on the day of transplantation. Recipients of syngeneic grafts (N=11) were untreated, but received the same dose of SA as the anti-CD154/DST treated recipients. (p<0.0001 for untreated versus anti-CD154/DST groups, p<0.0001 for anti-CD154/DST versus anti-CD154/DST +SA groups, p<0.0001 for anti-CD154/DST +SA versus Syn +SA groups). (B) SA-mediated rejection is associated with innate recognition and requires MyD88-signaling. Allogeneic skin transplantation was performed using full thickness trunk skin from Balb/c MyD88−/− grafts transplanted onto C57BL/6 MyD88−/− or wild type C57BL/6 recipients. Recipients were either left untreated (N=5) or given anti-CD154 (1mg/mouse, day 0 i.v., day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0 i.v.), then injected with heat killed SA (HKSA) on the day of transplantation (2×109 cfu, i.p.; N=8 (MyD88−/−) or 6 (WT)). (p<0.05 for WT +anti-CD154/DST +HKSA versus MyD88−/− +anti-CD154/DST +HKSA; p<0.001 for MyD88−/− +anti-CD154/DST +HKSA versus MyD88−/− No Rx). (C) SA-mediated prevention of tolerance is mediated by the adaptive immune system and requires T cells. Allogeneic skin transplantation was performed using full thickness trunk skin from Balb/c with recipients that were either wild type C57BL/6 (N=46), or RAG−/− (N=3), TCRβδ−/− (N=4), or CD8−/− (N=3) on the C57BL/6 background. All groups were treated with anti-CD154 (1mg/mouse, day 0 i.v., day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0 i.v.), then infected with SA on the day of transplantation (2×108 cfu, i.p.). (p<0.05 for wild type versus RAG−/−, p<0.05 for wild type versus TCRβδ−/−, p<0.05 for wild type versus CD8−/−).
Rejection associated with SA infection is dependent upon innate immune recognition and signaling
The primary host immune response against SA involves innate immune recognition and activation. SA components can be recognized by TLR2 and TLR4, and these TLRs together with downstream MyD88-signaling have been reported to be critically important to host-defense against SA infection (21–24). We tested whether these early innate events contribute to SA-mediated prevention of graft acceptance by using MyD88−/− donor-recipient pairs. Because MyD88−/− mice are exquisitely sensitive to SA infection (23, 25) heat-killed bacteria were administered in lieu of live infection to avoid mortality. Similar to live SA infection, heat-killed SA were capable of preventing graft acceptance resulting in acute skin allograft rejection in wild type animals (Figure 1B). Acute skin allograft rejection with normal kinetics was observed in MyD88−/− mice in the absence of anti-CD154/DST treatment; however, SA was unable to precipitate acute rejection in anti-CD154/DST-treated MyD88−/− recipients (Figure 1B). These data indicate that innate recognition of SA and events downstream of MyD88-signalling are necessary for the ability of SA to prevent graft acceptance.
Rejection associated with SA infection is a classical, T cell-mediated response
Allograft rejection is considered a T cell-dependent phenomenon that can be shaped by stimuli that active innate immunity (26). A role of T cells, especially Th17 cells, in controlling acute SA infection has recently been revealed (27, 28). We tested whether the adaptive immune system was required for SA-mediated prevention of skin allograft acceptance or whether innate immunity was sufficient to mediate rejection. Using RAG−/− recipients, we observed that an innate immune activation alone was not sufficient for SA to mediate rejection and that lymphocytes were required. Using TCRβδ−/−, which lack both conventional αβ T cells as well as Γδ T cells, we also observed that T cells were required for SA-induced prevention of tolerance. Specifically, we demonstrate by using CD8−/− recipients, that CD8+ T cells were necessary for SA-mediated allograft rejection (Figure 1C). We could not specifically address the question of whether CD4+ T cells were required in this model of SA-mediated prevention of skin allograft tolerance, as tolerance could not be established in their absence (29). These data led us to conclude that, although innate immune responses elicited by SA are important for host defense against the bacterium, they were, in and of themselves, not sufficient to induce rejection in anti-CD154/DST-treated recipients. Rather, our data suggest that SA infection over-rides the effects of anti-CD154/DST to facilitate a T cell-dependent rejection process.
SA promotes the CD154-independent expansion of alloreactive CD4+ and CD8+ T cells
We sought to visualize the direct effects of infection on the fate of alloreactive T cell populations by using an adoptive transfer system in which OVA-specific CD4+ OTII and CD8+ OTI TCR transgenic cells were CFSE labeled and transferred into C57BL/6 recipients of mOVA.B6 skin grafts. Animals were sacrificed 5 days after transfer and transplantation for flow cytometric analysis of the transgenic populations within the spleen (Figure 2). We observed that in the absence of transplantation, OTII and OTI T cells remained largely undivided either in the absence or presence of SA. Upon transplantation and in the absence of anti-CD154/DST treatment, OTII and OTI T cells proliferated vigorously. This proliferation was largely suppressed with anti-CD154/DST treatment. In contrast, in the setting of concomitant transplantation and SA infection, proliferation of both CD4+ (OTII) and CD8+ (OTI) alloreactive T cells was observed in anti-CD154/DST-treated recipients.
Figure 2.
SA promotes the CD154-independent proliferation of alloreactive CD4+ and CD8+ T cells. On the day of transplantation with mAct-OVA transgenic C57BL/6 donor grafts, mice were adoptively transferred CFSE labeled congenically marked OTI (5×105) and OTII (5×105) T cells. Transplanted mice were either left untreated (N=5), treated with anti-CD154 (1mg/mouse, day 0 i.v., day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0 i.v.) in the absence (N=5) or presence SA (2×108 cfu, i.p.; (N=6)). Untransplanted mice with (N=4) or without SA (N=4) were used as controls. Five days after transplantation, spleen (shown) and lymph node (not shown) cells were harvested and analyzed by flow cytometry for evidence of proliferation marked by CFSE dilution. The data are displayed as representative histograms (A) and composite analysis of all groups (B). (for both OTI and OTII analysis, p value n.s. for naïve versus +SA, p<0.0001 for naïve versus untreated, p<0.0001 for untreated versus anti-CD154/DST, p<0.001 for anti-CD154/DST versus anti-CD154/DST +SA).
SA-induced IL-6 is necessary and sufficient for prevention of skin allograft acceptance
We hypothesized that proinflammatory cytokines induced downstream of MyD88 following SA infection may impact T cell alloreactivity and confer resistance to anti-CD154/DST treatment. Experiments using a mouse cytokine 20-Plex panel (Invitrogen) quantified the levels of 20 cytokines and chemokines in the serum 1–24 hours after SA infection. IL-6 was observed to be a cytokine that was strongly induced early following SA infection, and moderately increased in LM-infected animals, but not following PA infection (Figure 3A). To test whether IL-6 was important for the ability of SA to prevent graft acceptance, we used IL-6−/− recipients as well as anti-IL-6 neutralization in wild-type recipients treated with anti-CD154/DST. We observed that infection with SA did not result in the rejection of skin allografts in the absence of IL-6, and conclude that IL-6 is critical for the ability of SA to prevent skin allograft acceptance (Figure 3B). Normal rejection kinetics was observed in IL-6−/− recipients that were not treated with anti-CD154/DST. In addition, LM infection, with mediates IFNs-dependent allograft rejection (13), was able to precipitate acute rejection in IL-6−/− recipient animals treated with anti-CD154/DST (Figure 3B).
Figure 3.
SA-mediated prevention of tolerance requires IL-6 induction. A) In vivo production of IL-6 from C57BL/6 mice infected with SA (2×108 cfu, i.p.), LM (2×105), or PA (2×107) was measured in serum samples at the designated time points by ELISA (N=2–3). B) Allogeneic skin transplantation was performed using Balb/c full thickness trunk skin transplanted onto IL-6−/− or wild type C57BL/6 recipients treated (N=3) with or without anti-IL6 mAb (500ug i.v. day 0; 250ug i.p. days 1, 3, 5, 7, 10, and 14 (N=3)). Long-term graft survival was induced with anti-CD154 (1mg/mouse, day 0 i.v. day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0 i.v.), then infected with either LM (2×105 cfu, i.p. (N=3)), or SA (2×108 cfu, i.p. (N=6)). (p<0.01 for IL-6−/− No Rx versus both IL-6−/− +anti-CD154/DST +SA and WT +anti-CD154/DST +anti-IL-6 +SA; p<0.01 for WT +anti-CD154/DST +SA versus both IL-6−/− +anti-CD154/DST +SA and WT +anti-CD154/DST +anti-IL-6 +SA; p<0.01 for IL-6−/− +anti-CD154/DST +LM versus both IL-6−/− +anti-CD154/DST +SA and WT +anti-CD154/DST+anti-IL-6 +SA).
We next sought to determine whether IL-6 alone, in the absence of infection with SA, was sufficient to prevent graft acceptance. To this end, we used the system of hydrodynamic gene delivery (30–33) to achieve levels of systemic IL-6 comparable to those seen after SA infection (Figure 4A). The delivery of the IL-6 expressing plasmid, but not the empty control plasmid, was able to prevent induction of allograft acceptance (Figure 4B). From these data, we infer that IL-6 produced during inflammation or upon bacterial infection is sufficient to promote activation of alloreactive T cells and the acute rejection of the allograft in anti-CD154/DST treated recipients.
Figure 4.
IL-6 alone is sufficient to prevent tolerance induction. A) Exogenous IL-6 was delivered by hydrodynamic gene delivery. In vivo production of IL-6 by C57BL/6 mice infected with SA (2×108 cfu, i.p.) or an IL-6 expressing plasmid (pORF-IL6, 5ug/1.9ml i.v.) versus empty vector control (pORF-mcs, 5ug/1.9ml i.v.) was measured by ELISA from serum samples at the indicated time points (N=4–6). (p<0.0001 for pORF-mcs versus both pORF-IL6 and +SA; p value n.s. for pORF-IL6 versus +SA) B) Allogeneic skin transplantation was performed in C57BL/6 recipients treated with anti-CD154 (1mg/mouse, day 0 i.v., day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0 i.v. (N=4)). Mice were either treated with SA infection (2×108 cfu i.p.) on the day of transplantation (N=47) or hydrodynamic delivery of IL-6 expressing plasmid (N=11) versus empty vector control (N=4) two days prior to transplantation. (p<0.05 for anti-CD154/DST +pORF-IL6 versus anti-CD154/DST +pORF-mcs; p value n.s. for anti-CD154/DST +pORF-IL6 versus anti-CD154/DST +SA).
VSports app下载 - SA and IL-6 promote the differentiation of an alloreactive Th1, but not alloreactive Th17, responses
SA-induced IL-6 has the potential to influence the activity of alloreactive T cells in a number of ways. IL-6 is known to promote the proliferation of Th1 effector cells, thereby leading to their escape from regulation (34). In addition, IL-6 has recently been described to be a pivotal cytokine in driving the differentiation of Th17 effector cells at the expense of peripheral Treg induction (35). Because this model of costimulation blockade-mediated allograft acceptance with anti-CD154/DST is thought to be dependent upon a balance of effector and regulatory T cells (6), we characterized the impact of IL-6 on this equilibrium. To this end, we used a 24-hour IFNγ or IL-17 ELISPOT assay with recipient splenocytes as responders and irradiated splenocytes that were syngeneic, allogeneic or syngeneic pulsed with heat-killed SA as stimulators. In the absence of anti-CD154/DST treatment, we observed that the donor-stimulated response was primarily associated with the presence of IFNγ-producing cells. These cells were largely absent in the setting of graft acceptance induced by anti-CD154/DST. In the setting of a SA infection or exogenous IL-6 treatment, there was a partial restoration of this primed IFNγ-producing population of alloreactive cells (Figure 5A). Following SA infection, there was a detectable population of primed, IL-17-producing anti-SA cells, however there were no detectable IL-17-producing alloreactive cells upon SA- or IL-6-mediated rejection (Figure 5B). These observations suggested that IL-6 enhanced conventional Th1 alloreactive responses but had minimal affect in skewing to a Th17 response in the setting of allograft rejection. This conclusion was further substantiated by the inability of neutralizing anti-IL-17A mAbs to restore graft acceptance to recipients treated with anti-CD154/DST and infected with HKSA (Figure 5C). This dose of anti-17A mAb had previously been shown to prevent CpG-mediated cardiac allograft rejection in IL-6-KO mice treated with anti-CD154/DST (36). HKSA was used to ameliorate potential defects in clearing SA in the absence of IL-17, and has been shown to be able to prevent graft acceptance in anti-CD154/DST-treated recipients (Fig 1B).
Figure 5.
SA and IL-6 promote the differentiation of Th1, but not Th17, alloreactive cells. Splenocytes were harvested from mice 3–4 weeks after transplantation and stimulated with irradiated syngeneic (C57BL/6), allogenic (BALB/c), or SA pulsed syngeneic splenocytes in a 12 hour co-culture. The frequency of (A) primed IFNγ-producing or (B) IL-17-producing cells was analyzed by ELISPOT. (p<0.001 for syngeneic versus alloreactive IFNγ-production in unmodified rejection (N=18), SA rejection (N=19), IL-6 rejection (N=6), IL-6−/− unmodified rejection (N=9), and p>0.05 for tolerant (N=4); p<0.001 for syngeneic versus SA-reactive IFNγ-production in SA rejection; p<0.001 for syngeneic versus alloreactive IL-17-production in unmodified rejection; p<0.001 for syngeneic versus SA-reactive IL-17-production in SA rejection). (C) Inability of anti-IL17 mAbs (clone 50104, 100 μg/mouse i.v. day 0, i.p days 2, 3, 4, 7, 9, 11 and 14; (N=6)). to abrogate the ability of heat-killed SA (HKSA), administered on the day of transplantation, to induce skin allograft rejection in anti-CD154/DST-treated recipients (N=6).
Treatment with conventional immunosuppression blunts IL-6 production following SA infection allowing for the establishment of graft acceptance despite infection
Steroids have been a cornerstone of immunosuppressive regimens and are known to potently suppress the production of a number of proinflammatory cytokines (2, 37). We hypothesize that inclusion of low-dose steroids to tolerance inducing regimens may modulate the production of inflammatory cytokines after SA infection, thereby allowing for the establishment of allograft acceptance. Consistent with previous reports, we showed that in a naïve animal, a single treatment on the day of transplantation with methylprednisolone (20 mg/kg, i.p), but not the T cell directed therapies of cyclosporine (20 mg/kg, i.p.) or sirolimus (0.3 mg/kg, i.p), blunted the induction of IL-6 following SA infection (Figure 6A). Importantly, upon addition of steroids, we observed that SA infection at the time of transplantation no longer precipitated graft rejection and skin allografts were maintained long-term (Figure 6B).
Figure 6.
Treatment with steroids blunts SA-induced IL-6 production and facilitates allograft acceptance in SA-infected anti-CD154/DST-treated recipients. A) In vivo production of IL-6 from C57BL/6 mice infected with SA (2×108 cfu, i.p.) without or with immunosuppression, either methylprednisolone (20 mg/kg day 0, i.p.), cyclosporine (20 mg/kg day 0, i.p.), or sirolimus (0.3 mg/kg day 0, i.p.) (N=3). IL-6 concentrations in the serum samples were analyzed by ELISA. B) Allogeneic skin transplantation was performed using full thickness trunk skin from Balb/c donors grafted onto C57BL/6 recipients treated with or without methylprednisolone (Steroid; N=4), with or without anti-CD154 (1mg/mouse, day 0 i.v., day 7 and 14 i.p.) and DST (107 donor splenocytes, day 0 i.v.) in the absence (N=4) or presence SA (2×108 cfu, i.p.). (p<0.0001 for Steroid +anti-CD154/DST +SA (N=5) versus anti-CD154/DST +SA (N=47)).
Discussion
Our current study extends previous observations with the intracellular Listeria bacterium and demonstrates that infections with the clinically-relevant bacterial species, SA, can prevent the induction of allograft acceptance. While innate stimulation and MyD88-signaling is necessary for SA infection to prevent anti-CD154/DST-mediated graft acceptance, allograft rejection remained T cell-dependent. We also demonstrated that SA-mediated allograft rejection in anti-CD154/DST-treated recipients was associated with the activation of alloreactive CD4+ and CD8+ T cells.
SA infection results in an early production of IL-6, which peaks within 24 hours of infection, and IL-6 was necessary and sufficient to prevent skin allograft acceptance in this model utilizing costimulation blockade. This observation is consistent with previously published descriptions of the numerous effects that IL-6 imparts upon T cells, including stimulating the proliferation and differentiation of both CD4+CD8− and CD4−CD8+ thymocytes (38–40), as well as promoting the proliferation of both naïve and memory T cells in the periphery (40–44). These activities are due in part to the co-stimulatory capacity of IL-6, which is able to bypass the requirement for IL-2 as well as the need for APCs and accessory molecule interaction (41), including CD40-CD154 as described in this study. In addition, IL-6 has the ability to effect differentiation programs depending on the context in which T cells are being activated. IL-6 has been shown to have the ability to dramatically enhance the proliferation of Th1 cells, rendering them resistant to suppression (34). It has also become clear that IL-6 plays a pivotal role in the dichotomous differentiation programs of Th17 and induced Tregs (35, 45, 46). Under situations in which TGFβ is present, IL-6 drives the development of Th17 cells at the expense of Treg cells (35). This production of IL-6 is consistent with the increased frequencies of Th17-producing, as well as IFNγ-producing, SA-reactive T cells following SA infection. However, we were unable to detect increased frequencies of donor-reactive Th17 cells after SA- or IL-6-mediated rejection, rather, the alloreactive response appeared to be dominated by Th1 cells. This observation, by itself, does not exclude the possibility of these anti-SA Th17 cells contributing to allograft injury either through bystander damage or cross-reactivity, or that a Th17 alloreactive response can only be detected locally in the allograft, as we described for the abrogation of tolerance by CpG (36). However, IL-17 neutralization of SA-infected recipients treated with anti-CD154/DST did not affect graft rejection kinetics (Figure 5C), consistent with the conclusion that even if alloreactive and SA-specific Th17 responses had been induced by SA, they were not necessary for SA-induced graft rejection in anti-CD154/DST-treated recipients.
An unexpected observation from our studies comes from comparing the effects of various microbes on transplantation tolerance and induction of acute rejection. The extracellular bacterium, SA, like the intracellular bacteria LM (1), can prevent tolerance induction, however, the mechanism by which they do so are quite distinct. Listeria prevents tolerance induction through the MyD88-independent production of IFN-s while SA does so by inducing IL-6. Equally important, we demonstrate that another extracellular bacterial species, PA, which generally induces more immunomodulatory cytokines, such as IL-10 (47–49), did not alter alloreactivity nor impact allograft acceptance in anti-CD154/DST-treated recipients. From these observations, it is clear that although these three species of bacteria possess many conserved molecular patterns that can activate pattern-recognition receptors, the innate immune responses to specific bacteria are different, which in turn, had unique effects on alloreactivity and skin allograft acceptance. The differences in innate immune responses elicited by different bacterial species are likely to be multifactorial. Firstly, there are important differences between host responses to intracellular versus extracellular pathogens, whereby intracellular pathogens tend to evoke a Th1-dominated response whereas extracellular pathogens tended to elicit Th17 responses (50–52). Secondly, bacteria have evolved sophisticated survival strategies to evade, subvert, or usurp host immune responses (53–58). Third, the site of infection may also affect the nature of the innate immune response. As such, it may be that the composite milieu post-infection is critical for directing bystander T cell responses. Fourth, some bacterial strains, including SA and Streptococcus pyogenes, secrete up to 20 toxins with superantigen properties. Superantigens bind to Class II and the Vs subunit of the TCR and cause the polyclonal activation and subsequent depletion of up to 30% of the T cells, and it is possible that these events can also contribute to alterations in the alloreactive response. Thus controlling infections and infection-induced inflammation at the time of transplantation may be critical for successful induction of long-term allograft acceptance.
One of the earliest modalities of pharmacologic immunosuppression that allowed transplantation to emerge as a viable treatment for end stage organ disease was steroid therapy. Steroids, through complex mechanisms of action, are potent at blunting inflammatory responses and remain a major component of immunosuppressive regimens in transplantation and autoimmunity. Often, patients are treated with high doses of steroids at the time of transplantation and although these are gradually tapered, a low dose of maintenance steroid therapy in concert with T cell suppression is typically required to prevent allograft rejection (2, 59). Our observations that steroids can blunt IL-6 production and facilitate graft acceptance in SA-infected recipients, without significantly compromising protective immunity to SA support a hypothesis that control of inflammatory cytokine production by steroids may be critical for the long-term control of alloreactivity and for permitting superior allograft survival. These observations may also explain why attempts at steroid minimization or withdrawal have been met with limited success and with higher rates of acute rejection (59), and theorize that maintenance steroid therapy may prevent acute rejection by blunting of the pro-inflammatory response during infections that may otherwise be sufficient to stimulate alloreactivity causing graft rejection. We also speculate that the continued use of steroids may be the reason why it has been more difficult to establish an association between bacterial infections and allograft rejection in the clinical population.
A major goal in transplantation immunology is to induce donor-specific tolerance that will result in the extended suppression of allograft-specific immune responses while leaving the remainder of the immune system fully competent. Thus the identification of potential barriers to tolerance as well as events that may reverse established allograft tolerance is critically necessary. The interaction between the immune response to infections and the immune response to foreign tissues triggering rejection in organ transplant recipients is a topic of great importance with implications on both graft and patient survival. The findings described here provide insight into the mechanisms by which bacterial infections, and specifically SA, precipitate rejection. We demonstrate that IL-6, produced as a result of SA infection, facilitates CD154-independent T cell activation. These observations are in contrast to the intracellular Listeria infections, which prevent tolerance induction through IFN-s, and to Pseudomonas infections, which have no impact on tolerance induction. Our study raise a cautionary note that minimization/avoidance of steroids may not be an optimum long-term immunosuppressive therapy in transplant recipients. Collectively, this study provide new insights into host-bacteria interactions, further our understanding of how select types of bacterial species can serve as barriers to transplantation tolerance by their ability to trigger the production of pro-inflammatory cytokines and underscore a novel approach of inhibiting the production of or neutralizing inflammatory cytokines that can synergize with co-stimulatory molecule-based therapies to facilitate the development of transplantation tolerance.
Acknowledgments (VSports在线直播)
This work was supported in part by grants, NIAID RO1 AI071080 to MLA, and ROTRF 979162997 and NIAID R01 AI072630 to ASC, and an Illinois Transplant Society Grant to EBA and ASC. We also thank the Frank W. Fitch Monoclonal Antibody facility of the University of Chicago Cancer Center for their assistance in the production of anti-mouse CD16/32. The facility is supported by NCI Cancer Center Support Grant #5P30CA014599-35.
"VSports app下载" Abbreviations
- DST
donor-specific transfusion
- SA
Staphylococcus aureus
- MR1
anti-CD154
- CFSE
carboxyfluorescein succinimidyl ester
- PA
Pseudomonas aerugninosa
Footnotes (VSports注册入口)
Disclosure Section: The authors of this manuscript have no conflicts of interest to disclose as described by the American Journal of Transplantation.
References
- 1.Wang T, Chen L, Ahmed E, Ma L, Yin D, Zhou P, et al. Prevention of allograft tolerance by bacterial infection with Listeria monocytogenes. J Immunol. 2008;180(9):5991–5999. doi: 10.4049/jimmunol.180.9.5991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Allison AC. Immunosuppressive drugs: the first 50 years and a glance forward. Immunopharmacology. 2000;47(2–3):63–83. doi: 10.1016/s0162-3109(00)00186-7. [DOI] [PubMed] [Google Scholar]
- 3.Fehr T, Sykes M. Tolerance induction in clinical transplantation. Transpl Immunol. 2004;13(2):117–130. doi: 10.1016/j.trim.2004.05.009. [DOI] [PubMed] [Google Scholar]
- 4.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–548. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
- 5.Yamada And A, Sayegh MH. The CD154-CD40 costimulatory pathway in transplantation. Transplantation. 2002;73(1 Suppl):S36–39. doi: 10.1097/00007890-200201151-00012. [DOI] [PubMed] [Google Scholar]
- 6.Quezada SA, Jarvinen LZ, Lind EF, Noelle RJ. CD40/CD154 interactions at the interface of tolerance and immunity. Annu Rev Immunol. 2004;22:307–328. doi: 10.1146/annurev.immunol.22.012703.104533. [DOI (VSports注册入口)] [PubMed] [Google Scholar]
- 7.Monk NJ, Hargreaves RE, Marsh JE, Farrar CA, Sacks SH, Millrain M, et al. Fc-dependent depletion of activated T cells occurs through CD40L-specific antibody rather than costimulation blockade. Nat Med. 2003;9(10):1275–1280. doi: 10.1038/nm931. [DOI] [PubMed] [Google Scholar]
- 8.Reinke P, Fietze E, Ode-Hakim S, Prosch S, Lippert J, Ewert R, et al. Late-acute renal allograft rejection and symptomless cytomegalovirus infection. Lancet. 1994;344(8939–8940):1737–1738. doi: 10.1016/s0140-6736(94)92887-8. [DOI] [PubMed] [Google Scholar]
- 9.McLaughlin K, Wu C, Fick G, Muirhead N, Hollomby D, Jevnikar A. Cytomegalovirus seromismatching increases the risk of acute renal allograft rejection. Transplantation. 2002;74(6):813–816. doi: 10.1097/00007890-200209270-00014. [V体育平台登录 - DOI] [PubMed] [Google Scholar]
- 10.Sageda S, Nordal KP, Hartmann A, Sund S, Scott H, Degre M, et al. The impact of cytomegalovirus infection and disease on rejection episodes in renal allograft recipients. Am J Transplant. 2002;2(9):850–856. doi: 10.1034/j.1600-6143.2002.20907.x. ["V体育平台登录" DOI] [PubMed] [Google Scholar]
- 11.Vilchez RA, Dauber J, McCurry K, Iacono A, Kusne S. Parainfluenza virus infection in adult lung transplant recipients: an emergent clinical syndrome with implications on allograft function. Am J Transplant. 2003;3(2):116–120. doi: 10.1034/j.1600-6143.2003.00024.x. ["VSports" DOI] [PubMed] [Google Scholar]
- 12.Ahmed EB, Chong AS, Alegre M. The Role of Bacterial Infections in Allograft Rejection. Expert Reviews in Clinical Immunology. 2008;4(2):281–293. doi: 10.1586/1744666X.4.2.281. [DOI] [PubMed] [Google Scholar]
- 13.Wang T, Ahmed EB, Chen L, Xu J, Tao J, Wang CR, et al. Infection with the intracellular bacterium, Listeria monocytogenes, overrides established tolerance in a mouse cardiac allograft model. Am J Transplant. 2010;10(7):1524–1533. doi: 10.1111/j.1600-6143.2010.03066.x. ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fishman JA, Rubin RH. Infection in organ-transplant recipients. N Engl J Med. 1998;338(24):1741–1751. doi: 10.1056/NEJM199806113382407. [V体育ios版 - DOI] [PubMed] [Google Scholar]
- 15.Lowy FD. Staphylococcus aureus infections. N Engl J Med. 1998;339(8):520–532. doi: 10.1056/NEJM199808203390806. ["V体育ios版" DOI] [PubMed] [Google Scholar]
- 16.von Eiff C, Becker K, Machka K, Stammer H, Peters G. Nasal carriage as a source of Staphylococcus aureus bacteremia. Study Group. N Engl J Med. 2001;344(1):11–16. doi: 10.1056/NEJM200101043440102. [DOI (V体育2025版)] [PubMed] [Google Scholar]
- 17.Diekema DJ, Pfaller MA, Schmitz FJ, Smayevsky J, Bell J, Jones RN, et al. Survey of infections due to Staphylococcus species: frequency of occurrence and antimicrobial susceptibility of isolates collected in the United States, Canada, Latin America, Europe, and the Western Pacific region for the SENTRY Antimicrobial Surveillance Program, 1997–1999. Clin Infect Dis. 2001;32 (Suppl 2):S114–132. doi: 10.1086/320184. [DOI] [PubMed] [Google Scholar]
- 18.Nampoory MR, Johny KV, Costandy JN, Nair MP, Said T, Homoud H, et al. Infection related renal impairment: a major cause of acute allograft dysfunction. Exp Clin Transplant. 2003;1(1):60–64. [PubMed] [Google Scholar]
- 19.Adachi O, Kawai T, Takeda K, Matsumoto M, Tsutsui H, Sakagami M, et al. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity. 1998;9(1):143–150. doi: 10.1016/s1074-7613(00)80596-8. [DOI] [PubMed] [Google Scholar]
- 20.Bae T, Banger AK, Wallace A, Glass EM, Aslund F, Schneewind O, et al. Staphylococcus aureus virulence genes identified by bursa aurealis mutagenesis and nematode killing. Proc Natl Acad Sci U S A. 2004;101(33):12312–12317. doi: 10.1073/pnas.0404728101. ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lien E, Sellati TJ, Yoshimura A, Flo TH, Rawadi G, Finberg RW, et al. Toll-like receptor 2 functions as a pattern recognition receptor for diverse bacterial products. J Biol Chem. 1999;274(47):33419–33425. doi: 10.1074/jbc.274.47.33419. [DOI] [PubMed] [Google Scholar]
- 22.Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J Biol Chem. 1999;274(25):17406–17409. doi: 10.1074/jbc.274.25.17406. [DOI] [PubMed] [Google Scholar]
- 23.Takeuchi O, Hoshino K, Akira S. Cutting edge: TLR2-deficient and MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection. J Immunol. 2000;165(10):5392–5396. doi: 10.4049/jimmunol.165.10.5392. [DOI] [PubMed] [Google Scholar]
- 24.Yoshimura A, Lien E, Ingalls RR, Tuomanen E, Dziarski R, Golenbock D. Cutting edge: recognition of Gram-positive bacterial cell wall components by the innate immune system occurs via Toll-like receptor 2. J Immunol. 1999;163(1):1–5. [PubMed] [Google Scholar]
- 25.Miller LS, O'Connell RM, Gutierrez MA, Pietras EM, Shahangian A, Gross CE, et al. MyD88 mediates neutrophil recruitment initiated by IL-1R but not TLR2 activation in immunity against Staphylococcus aureus. Immunity. 2006;24(1):79–91. doi: 10.1016/j.immuni.2005.11.011. [DOI] [PubMed] [Google Scholar]
- 26.Issa F, Schiopu A, Wood KJ. Role of T cells in graft rejection and transplantation tolerance. Expert Rev Clin Immunol. 6(1):155–169. doi: 10.1586/eci.09.64. [DOI] [PubMed] [Google Scholar]
- 27.Lin L, Ibrahim AS, Xu X, Farber JM, Avanesian V, Baquir B, et al. Th1-Th17 cells mediate protective adaptive immunity against Staphylococcus aureus and Candida albicans infection in mice. PLoS Pathog. 2009;5(12):e1000703. doi: 10.1371/journal.ppat.1000703. ["V体育2025版" DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ma CS, Chew GY, Simpson N, Priyadarshi A, Wong M, Grimbacher B, et al. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J Exp Med. 2008;205(7):1551–1557. doi: 10.1084/jem.20080218. ["VSports注册入口" DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Markees TG, Phillips NE, Gordon EJ, Noelle RJ, Shultz LD, Mordes JP, et al. Long-term survival of skin allografts induced by donor splenocytes and anti-CD154 antibody in thymectomized mice requires CD4(+) T cells, interferon-gamma, and CTLA4. J Clin Invest. 1998;101(11):2446–2455. doi: 10.1172/JCI2703. [V体育ios版 - DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Herweijer H, Wolff JA. Progress and prospects: naked DNA gene transfer and therapy. Gene Ther. 2003;10(6):453–458. doi: 10.1038/sj.gt.3301983. [DOI] [PubMed] [Google Scholar]
- 31.Knapp JE, Liu D. Hydrodynamic delivery of DNA. Methods Mol Biol. 2004;245:245–250. doi: 10.1385/1-59259-649-5:245. [DOI] [PubMed] [Google Scholar]
- 32.Pan H, Hong F, Radaeva S, Gao B. Hydrodynamic gene delivery of interleukin-22 protects the mouse liver from concanavalin A-, carbon tetrachloride-, and Fas ligand-induced injury via activation of STAT3. Cell Mol Immunol. 2004;1(1):43–49. [PubMed] [Google Scholar]
- 33.Yazawa H, Murakami T, Li HM, Back T, Kurosaka K, Suzuki Y, et al. Hydrodynamics-based gene delivery of naked DNA encoding fetal liver kinase-1 gene effectively suppresses the growth of pre-existing tumors. Cancer Gene Ther. 2006;13(11):993–1001. doi: 10.1038/sj.cgt.7700970. [DOI] [PubMed] [Google Scholar]
- 34.Pasare C, Medzhitov R. Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science. 2003;299(5609):1033–1036. doi: 10.1126/science.1078231. [DOI] [PubMed] [Google Scholar]
- 35.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- 36.Chen L, Ahmed EB, Wang T, Wang Y, Orchando J, Chong AS, Alegre M. TLR Signals Promote IL-6/IL-17-Dependent Transplant Rejection. Journal of Immunology. 2009;182(10) doi: 10.4049/jimmunol.0803842. Article in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lund T, Fosby B, Korsgren O, Scholz H, Foss A. Glucocorticoids reduce pro-inflammatory cytokines and tissue factor in vitro and improve function of transplanted human islets in vivo. Transpl Int. 2008;21(7):669–678. doi: 10.1111/j.1432-2277.2008.00664.x. [DOI] [PubMed] [Google Scholar]
- 38.Okada M, Kitahara M, Kishimoto S, Matsuda T, Hirano T, Kishimoto T. IL-6/BSF-2 functions as a killer helper factor in the in vitro induction of cytotoxic T cells. J Immunol. 1988;141(5):1543–1549. ["VSports" PubMed] [Google Scholar]
- 39.Takai Y, Wong GG, Clark SC, Burakoff SJ, Herrmann SH. B cell stimulatory factor-2 is involved in the differentiation of cytotoxic T lymphocytes. J Immunol. 1988;140(2):508–512. [PubMed] [Google Scholar]
- 40.Uyttenhove C, Coulie PG, Van Snick J. T cell growth and differentiation induced by interleukin-HP1/IL-6, the murine hybridoma/plasmacytoma growth factor. J Exp Med. 1988;167(4):1417–1427. doi: 10.1084/jem.167.4.1417. [DOI (VSports app下载)] [PMC free article] [PubMed] [Google Scholar]
- 41.Lotz M, Jirik F, Kabouridis P, Tsoukas C, Hirano T, Kishimoto T, et al. B cell stimulating factor 2/interleukin 6 is a costimulant for human thymocytes and T lymphocytes. J Exp Med. 1988;167(3):1253–1258. doi: 10.1084/jem.167.3.1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Garman RD, Jacobs KA, Clark SC, Raulet DH. B-cell-stimulatory factor 2 (beta 2 interferon) functions as a second signal for interleukin 2 production by mature murine T cells. Proc Natl Acad Sci U S A. 1987;84(21):7629–7633. doi: 10.1073/pnas.84.21.7629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Longhi MP, Wright K, Lauder SN, Nowell MA, Jones GW, Godkin AJ, et al. Interleukin-6 is crucial for recall of influenza-specific memory CD4 T cells. PLoS Pathog. 2008;4(2):e1000006. doi: 10.1371/journal.ppat.1000006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Rochman I, Paul WE, Ben-Sasson SZ. IL-6 increases primed cell expansion and survival. J Immunol. 2005;174(8):4761–4767. doi: 10.4049/jimmunol.174.8.4761. [DOI] [PubMed] [Google Scholar]
- 45.Mangan PR, Harrington LE, O'Quinn DB, Helms WS, Bullard DC, Elson CO, et al. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441(7090):231–234. doi: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
- 46.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24(2):179–189. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
- 47.Grohmann U, Van Snick J, Campanile F, Silla S, Giampietri A, Vacca C, et al. IL-9 protects mice from Gram-negative bacterial shock: suppression of TNF-alpha, IL-12, and IFN-gamma, and induction of IL-10. J Immunol. 2000;164(8):4197–4203. doi: 10.4049/jimmunol.164.8.4197. ["V体育官网入口" DOI] [PubMed] [Google Scholar]
- 48.Kobayashi M, Tsuda Y, Yoshida T, Takeuchi D, Utsunomiya T, Takahashi H, et al. Bacterial sepsis and chemokines. Curr Drug Targets. 2006;7(1):119–134. doi: 10.2174/138945006775270169. ["VSports最新版本" DOI] [PubMed] [Google Scholar]
- 49.Varma TK, Durham M, Murphey ED, Cui W, Huang Z, Lin CY, et al. Endotoxin priming improves clearance of Pseudomonas aeruginosa in wild-type and interleukin-10 knockout mice. Infect Immun. 2005;73(11):7340–7347. doi: 10.1128/IAI.73.11.7340-7347.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bettelli E, Korn T, Kuchroo VK. Th17: the third member of the effector T cell trilogy. Curr Opin Immunol. 2007;19(6):652–657. doi: 10.1016/j.coi.2007.07.020. [DOI (VSports注册入口)] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen Z, O'Shea JJ. Th17 cells: a new fate for differentiating helper T cells. Immunol Res. 2008;41(2):87–102. doi: 10.1007/s12026-007-8014-9. [DOI] [PubMed] [Google Scholar]
- 52.Takatori H, Kanno Y, Chen Z, O'Shea JJ. New complexities in helper T cell fate determination and the implications for autoimmune diseases. Mod Rheumatol. 2008;18(6):533–541. doi: 10.1007/s10165-008-0099-z. [V体育安卓版 - DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fedtke I, Gotz F, Peschel A. Bacterial evasion of innate host defenses--the Staphylococcus aureus lesson. Int J Med Microbiol. 2004;294(2–3):189–194. doi: 10.1016/j.ijmm.2004.06.016. [DOI] [PubMed] [Google Scholar]
- 54.Woolard MD, Frelinger JA. Outsmarting the host: bacteria modulating the immune response. Immunol Res. 2008;41(3):188–202. doi: 10.1007/s12026-008-8021-5. [DOI] [PubMed] [Google Scholar]
- 55.Rooijakkers SH, van Strijp JA. Bacterial complement evasion. Mol Immunol. 2007;44(1–3):23–32. doi: 10.1016/j.molimm.2006.06.011. [DOI] [PubMed] [Google Scholar]
- 56.Hornef MW, Normark S, Henriques-Normark B, Rhen M. Bacterial evasion of innate defense at epithelial linings. Chem Immunol Allergy. 2005;86:72–98. doi: 10.1159/000086652. [DOI] [PubMed] [Google Scholar]
- 57.Pieters J. Evasion of host cell defense mechanisms by pathogenic bacteria. Curr Opin Immunol. 2001;13(1):37–44. doi: 10.1016/s0952-7915(00)00179-5. [DOI] [PubMed] [Google Scholar]
- 58.Kaufmann SH. Immunity to intracellular bacteria. Annu Rev Immunol. 1993;11:129–163. doi: 10.1146/annurev.iy.11.040193.001021. ["VSports" DOI] [PubMed] [Google Scholar]
- 59.Pascual J, Zamora J, Galeano C, Royuela A, Quereda C. Steroid avoidance or withdrawal for kidney transplant recipients. Cochrane Database Syst Rev. 2009;(1):CD005632. doi: 10.1002/14651858.CD005632.pub2. [DOI] [PubMed] [Google Scholar]