干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 535165|回复: 259
go

Fas Ligand Enhances Hematopoietic Cell Engraftment Through Abrogation of Alloimm [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:53 |只看该作者 |倒序浏览 |打印
作者:Michal Pearl-Yafea, Esma S. Yolcub, Jerry Steina, Ofer Kaplanc, Isaac Yaniva, Haval Shirwanb, Nadir Askenasya作者单位:aFrankel Laboratory, Center for Stem Cell Research, Department of Pediatric Hematology-Oncology, Schneider Children ( _& ~6 R" q9 V% i0 j+ D1 S3 ^  r
                  1 C3 H8 ~+ J& E( C
                  # o: Y3 k! F3 R- Z! n+ x/ E( _
         
. u) E/ c/ j& i3 R                        
5 p4 P( b5 F( P! h. Q3 W            # u' H4 Y6 a6 @7 j- f; X
            
4 O# C# h) g1 p            
' _. m* J6 Y1 J6 H  t, c) q            
9 K% ~6 H. X- ]$ n; f- i/ b6 R                      & M  b2 z' M, N0 m1 j3 }# c
        - {) j9 _4 |# b  }5 f8 l2 b
        
2 M  e9 M( H- E9 [- y8 m        # w$ T/ S" H+ C# i" V' s& q
          【摘要】" C9 y, Q: ]% c0 @
      Early after transplantation, donor lineage-negative bone marrow cells (lin¨C BMC) constitutively upregulated their expression of Fas ligand (FasL), suggesting an involvement of the Fas/FasL axis in engraftment. Following the observation of impaired engraftment in the presence of a dysfunctional Fas/FasL axis in FasL-defective (gld) donors or Fas-defective (lpr) recipients, we expressed a noncleavable FasL chimeric protein on the surface of donor lin¨C BMC. Despite a short life span of the protein in vivo, expression of FasL on the surface of all the donor lin¨C BMC improved the efficiency of engraftment twofold. The FasL-coated donor cells efficiently blunted the host alloimmune responses in primary recipients and retained their hematopoietic reconstituting potential in secondary transplants. Surprisingly, FasL protein improved the efficiency of engraftment in syngeneic transplants. The deficient engraftment in lpr recipients was not reversed in chimeric mice with Fas¨C stroma and Fas  BMC, demonstrating that the host marrow stroma was also a target of donor cell FasL. Hematopoietic stem and progenitor cells are insensitive to Fas-mediated apoptosis and thus can exploit the constitutive expression of FasL to exert potent veto activities in the early stages of engraftment. Manipulation of the donor cells using ectopic FasL protein accentuated the immunogenic and nonimmunogenic interactions between the donor cells and the host, alleviating the requirement for a megadose of transplanted cells to achieve a potent veto effect.
% O9 R0 u* D5 N- u% s
0 V" ]" }- K# |2 E3 dDisclosure of potential conflicts of interest is found at the end of this article. ) Y8 C% E& |% s# c' s9 b
          【关键词】 Adult stem cells Apoptosis Hematopoietic stem cell transplantation Fas8 A5 A# }% ~: Y' c. d+ U$ l
                  INTRODUCTION/ F1 S) g- L; L/ @# ^

2 [4 E, G& Q9 PActivation-induced cell death (AICD) is crucial to the maintenance of immune homeostasis. One of the major regulatory mechanisms by which AICD occurs is the Fas receptor, the trimerization of which signals apoptosis and inhibits cell proliferation . Recent evidence from our laboratory indicates that these beneficial effects achieved by FasL-mediated AICD may be mediated by the differential sensitivities of T regulatory and T effector cells to activation of this apoptotic pathway (A. Kaminitz, M.P.-Y, E.S.Y., J.S., O.K., I.Y., H.S., N.A., unpublished data).
$ L" K/ y  y! u# `) Y0 q, I4 E% I: E+ ~" q# ?/ W
Research on the role of the Fas/FasL axis in the context of hematopoietic stem and progenitor cell (HSPC) transplants has focused on the Fas-mediated activities of immune cells, either in support of HSPC engraftment or graft versus host disease . These data suggest that physiological FasL expression by donor cells plays a role in engraftment through interaction with the Fas  cells in the host.& Q; d' H& y; b3 Q0 I8 d$ l

' z! x7 g! m9 ]4 \+ D; ^9 EWe recently found that the Fas receptor and its ligand are coexpressed by murine HSPC in the early phases of homing to the bone marrow, suggesting that this molecular interaction plays a role in early engraftment (submitted manuscript). As opposed to reports that the Fas receptor suppresses donor cell activity .
0 w1 g% a& |* L: M) A7 j2 d  f5 B+ p( D# g' M
In this study we characterized the functional involvement of FasL in HSPC engraftment. The upregulation of Fas and FasL expression on bone marrow-homed donor HSPC indicated that this molecular interaction is relevant to the very early stages of engraftment. Therefore, we used a FasL protein that has a very short lifetime in vivo . The protein aggregates spontaneously to deliver potent apoptotic signals through trimerization of the Fas receptor and also may be adhered to the surface of cells via biotinylation.- H; m- |3 @  ^) n5 b  q, ~- Y  D
8 {/ V$ S2 I3 n/ c
MATERIALS AND METHODS
- x/ ]1 u1 u3 R
8 E* m; J% X- V$ L$ u' xAnimal Preparation and Transplantation
" X' B1 a1 s0 q* [6 L$ E8 w6 |1 N0 y& Q+ `$ E/ q1 c* w! e
Mice used in this study were C57Bl/6J (B6, H2Kb, CD45.2), B6.SJL-Ptprca Pepcb/BoyJ (H2Kb, CD45.1), B6.MRL-Fas-lpr/J (lpr, H2Kb, CD45.2), B6Smn.C3-Fasl-gld/J (gld, H2Kb), BALB/c (H2kd), B10.BR-H2k-H2-T18a/SgSnJJrep (C57Bl/BR, H2Kk), and C57BL/6-TgN(ACTbEGFP)1Osb (GFP, H2kb), purchased from Jackson Laboratory (Bar Harbor, ME, http://www.jax.org). The mice were housed in a barrier facility. All of the procedures were approved by the Institutional Animal Care Committee. Recipients were conditioned using total body irradiation delivered by an x-ray irradiator (Rad Source 2000; Rad Source Technologies Inc., Alpharetta, GA, http://www.radsource.com) at a rate of 106 rad/minute. Irradiation was routinely performed 18¨C24 hours before transplantation. In all experiments, sublethal irradiation and lethal irradiation were administered at 850 rad and 950 rad total body irradiation (TBI), respectively. Donor cells were suspended in 0.2 ml of phosphate-buffered saline (PBS) and infused into the lateral tail vein. In secondary transplants, half of the cellular content of one femur was transplanted into irradiated recipients.
! r2 p& g. W" K; w" Z8 P" O  ], E- I  z& q# T0 ?+ V! \& C
Cell Isolation, Characterization, and Staining
: e# p8 L. _) I+ r2 p
# @& A; n# k: t7 A. ]4 [4 TWhole BMC were harvested from femurs and tibia of donors, and low-density cells were collected as previously described . For immunomagnetic separation of lineage-negative (lin¨C) BMC, the cells were incubated for 45 minutes at 4¡ãC with saturating amounts of biotinylated anti-mouse monoclonal antibodies (mAb) specific for CD5, B220, TER-119, Mac-1, Gr-1, and NK1.1. All antibodies were obtained from hybridoma cell cultures, except Ter-119 and NK1.1 (eBioscience, Santiago, CA, http://www.ebioscience.com). mAb-coated cells were washed twice with PBS containing 2% fetal calf serum and were incubated with sheep-anti-rat IgG conjugated to M-450 magnetic beads at a ratio of four beads per cell (Dynal Inc., Lake Success, NY, www.invitrogen.com). The lineage-positive (lin ) cells conjugated to beads were precipitated by exposure to a magnetic field, and supernatant containing lin¨C BMC was collected. The efficiency of the lin¨C cell separation procedure was reassessed by flow cytometry using a cocktail of primary labeled mAb against the lineage markers listed above. The average yield of this procedure was 4%¨C5% lin¨C BMC, and the viability was >95% as determined by the trypan blue and propidium iodide exclusion. To achieve a higher degree of purity (>95%), the immunomagnetic separation was repeated in some cases.2 t/ v6 S* }. n1 I, |

. K+ }! {; `8 e  N! @' I( n8 FFlow Cytometry6 w0 A$ ^0 y; V. D6 N

. `) J4 D* R' m4 s$ K* z! \; YDonor chimerism was determined from the percentage of donor and host peripheral blood lymphocytes. Blood was collected in heparinized serum vials in 200 µl of M199 and centrifuged over 1.5 ml of lymphocyte separation medium (Cedar Lane, Hornby, ON, Canada, http://www.cedarlanelabs.com). Before analysis, red blood cells (RBC) were lyzed using ammonium chloride solution for 4 minutes at room temperature, and the process was arrested by addition of excess cold solution. The nucleated cells were incubated for 45 minutes at 4¡ãC with phycoerythrin-anti-H2Kb (clone CTKb; Caltag Laboratories, Burlingame, CA, http://www.caltag.com) and fluorescein isothiocyanate-anti-H2Kd mAb (clone SF-1.1; BD Pharmingen, San Diego, http://www.bdbiosciences.com/index_us.shtml). Measurements were performed with a Vantage SE flow cytometer (Becton, Dickinson and Company, Franklin Lakes, NJ, http://www.bd.com). Positive staining was determined on a log scale and normalized with control cells stained with isotype control mAb. Cell death and apoptosis were determined in cells incubated with 5 mg/ml 7-aminoactinomycin-D and Annexin-V (IQ-Products, Groningen, The Netherlands, http://www.iqproducts.nl). The Fas receptor was identified with a primary labeled antibody (eBioscience), and FasL was stained with a biotinylated MFL4 mAb (BD Pharmingen) and fluorochrome-labeled streptavidin (eBioscience).* Z# e  |2 b, j$ T. W' n( s: z

5 {& {! D" y; g* Y( ?$ C8 A; ?3 l. X, FAdsorption of FasL Protein on the Surface of Cells) ]/ g4 i: l9 ]6 R2 v, F
0 W, _( [/ e* _
Nucleated BMC and splenocytes harvested under aseptic conditions were suspended in 5 µM freshly prepared EZ-Link Sulfo-NHS-LC-Biotin (Pierce, Rockford, IL, http://www.piercenet.com) in PBS for 30 minutes at room temperature . After washing twice with PBS, the cells were incubated with streptavidin-FasL chimeric protein (100 ng of protein per 1 x 106 cells in PBS). After two washes, the efficiency of adsorption was evaluated by flow cytometry using anti-streptavidin and anti-FasL antibodies.
! @* w' Q8 X$ o, ]
4 ~) Y& z% b: }, o" R) T( ?9 zApoptotic Challenge Using FasL Protein
7 z; x1 |: v5 m0 [6 {% ?$ C3 P
1 q% m0 S! {/ Z% TA20 murine lymphoblastoma cells and splenocytes served as controls for the apoptotic activity of the chimeric protein. Bone marrow cells were incubated (5 x 106 cells per milliliter) for 24 hours in -minimal essential medium culture medium supplemented with StemPro Nutrient Supplement (Stem Cell Technologies, Vancouver, BC, Canada, http://www.stemcell.com), 2 mM L-glutamine, 50 µM 2¦Â-mercaptoethanol (ME), 10 ng/ml stem cell factor, and 100 ng/ml thrombopoietin. All factors were purchased from Peprotech (Rocky Hill, NJ, http://www.peprotech.com). The cells were challenged by addition of 75¨C250 ng/ml streptavidin-FasL chimeric protein for 18¨C24 hours. Apoptosis and death were monitored by flow cytometry.
9 r0 E8 |" L. M- X" j7 [7 k9 `2 e7 @5 `8 }$ R/ c0 q
Mixed Lymphocyte Reaction* J3 L3 D% ~- V+ K  G
6 M; L) y0 M& X
Splenocytes were labeled with 2.5 µM of the intracellular dye 5-(and-6-)-carboxyfluorescein diacetate succinimidyl ester (CFSE; Molecular Probes, Carlsbad, CA, http://probes.invitrogen.com), and 50 x 106 cells were plated on Petri dishes for 45 minutes to enrich for lymphocytes. After 45 minutes, nonadherent cells were collected, washed, and incubated with irradiated (2,000 rad) stimulator splenocytes in Dulbecco's modified Eagle's medium supplemented with 2 mM L-glutamine, 1 mM sodium pyruvate, 13.6 µM folic acid, 270 µM L-asparagine, 548 µM L-arginine, 10 mM HEPES, 50 µM 2-ME, 100 mg/ml streptomycin, 100 U/ml penicillin, 5% heat-inactivated fetal bovine serum, and 1% heat-inactivated mouse serum. All of the ingredients were purchased from Biological Industries (Kibbutz Beit Haemek, Israel, http://www.bioind.com) and Sigma-Aldrich (St. Louis, http://www.sigmaaldrich.com). Controls included irradiated syngeneic and third party cells. Cultures were harvested after a 5 days incubation period, and the dilution of CFSE was analyzed by flow cytometry by gating on the live lymphocytes. Data were analyzed using CellQuest software (Becton Dickinson). All mixed lymphocyte reaction (MLR) assays were performed in triplicates and are representative of a minimum of three animals per group.
# L; j* t4 H) \0 r  \& l
  L5 l0 H( r  ]  j$ Z( G1 a1 w: L- s+ Z# IOrgan Harvesting
4 ^$ S; H) i- i: y2 z) P2 b. Z
  r1 Y2 T9 {& C  }# ISpleen, lung, and liver were harvested after intracardiac perfusion of 30 ml of cold PBS containing 100 units of heparin. The tissues were sectioned into pieces and processed; the lung was digested in 380 U/ml collagenase type V (Sigma) for 60 minutes at 37¡ãC, and the liver was digested in 1,500 U/ml collagenase for 20 minutes at 37¡ãC. All tissues, including spleen, were filtered over a 40-µm mesh, and cell suspensions were washed twice with PBS.
" G) Z  O! n* p0 e! L6 o
  V& O  V! s4 ?! h  W! F# ILong-Term Culture of Bone Marrow Stroma* V+ k, @$ R. s8 D6 o' ?9 u

) y* D) A6 C. v4 ]. mB6.lpr mice irradiated with 850 rad TBI were transplanted with 107 whole BMC from wild-type syngeneic mice (CD45.1CD45.2). After 3 weeks, donor chimerism was measured in peripheral blood, and the BMC were harvested from transplanted and naïve mice (control). Following RBC lysis, the cells were plated at a density of 106 cells per cm2 in murine mesenchymal medium (Stem Cell Technologies) supplemented with 100 U/ml penicillin and 100 µg/ml streptomycin. The medium was replaced twice per week to eliminate the nonadherent cells, and the culture was maintained until attaining confluency (approximately 3 weeks). The adherent cells were then detached by trypsinization and stained with rat anti-mouse mAb against CD11b, CD45.1, and CD45.2 (eBioscience). The donor (CD45.1) and host (CD45.2) origin of the cultured stromal cells was determined in flow cytometry by gating out the CD11C  cells.7 g$ n' M2 _3 @

/ H4 T3 z/ n* J- rStatistical Analysis
1 H3 {, ]8 G" k8 x9 Y0 g
! m8 T8 F, n# u4 FData are presented as means ¡À SD for each experimental protocol. Results in each experimental group were evaluated for reproducibility by linear regression of duplicate measurements. Differences between the experimental protocols were estimated with a post hoc Scheffe t test, and significance was considered at p
/ {# x5 r# r( Y3 M- b; {7 x- Z2 A! j
RESULTS
- ]/ ?# ^' W1 P0 K# h  C
) c8 j- {# d9 p. c* _& NThe Fas Receptor and Ligand Play a Role in Hematopoietic Cell Engraftment: R6 N' C! j! K! Q6 V
2 R1 Y* V+ w* {7 s. S- p
Two days after transplantation of allogeneic lin¨C BMC into irradiated hosts (H2KdH2Kb), the bone marrow-homed cells were harvested and analyzed for the expression of Fas and FasL. Whereas the residual BMC of the host displayed a moderate upregulation in the expression of these molecules, the donor cells showed a remarkable upregulation of Fas and FasL within hours after homing to the bone marrow (Fig. 1A). Approximately 20% of the transplanted cells displayed lineage markers during the first 2 days after transplantation, and approximately 75% of these cells displayed the Fas receptor and ligand. Fas and FasL expression was also pronounced in the lin¨C subset of bone marrow-homed donor cells. The constitutive joint expression of the Fas receptor and ligand was likely the result of the skewed cytokine environment in the irradiated bone marrow and upregulation of these molecules by cycling bone marrow cells . To determine whether the expression of FasL by the donor cells was of physiological significance, lin¨C BMC were transplanted into wild-type and Fas-defective (lpr) allogeneic hosts (Fig. 1B). The deficient engraftment in lpr recipients (p
. P2 Z. @9 T- k, R7 ?( F( U5 e5 H0 v( J, s  d" i
Figure 1. FasL plays a role in allogeneic cell engraftment. (A): Mice conditioned with 850 rad total body irradiation (TBI) were injected with 107 allogeneic lin¨C bone marrow cells (BMC) (H2KdH2Kb). After 48 hours, the bone marrow of the recipient mice was harvested, and the expression of Fas and FasL were determined in reference to the donor-host origin and lineage marker expression (n = 8). (B): Wild-type and lpr mice conditioned with 850 rad TBI were transplanted with 2 x 106 lin¨C BMC from BALB/c donors. The levels of donor chimerism were measured in the peripheral blood at 3 weeks post-transplantation (n = 6). (C): Radiation-conditioned recipients (850 rad) were injected with 106 naïve lin¨C BMC in conjunction with 106 lin¨C BMC or splenocytes coated with FasL protein and a control group transplanted with 2 x 106 naïve lin¨C BMC (n = 5). The levels of donor chimerism determined in the peripheral blood at 3 weeks post transplantation. Abbreviation: FasL, Fas ligand; spl, splenocytes; X, 106 naïve lin¨C bone marrow cells.
* r( K, o# k8 j/ [6 j
. l$ X& k( y3 aEctopic Expression of FasL Enhances the Engraftment of Allogeneic Hematopoietic Cells
# P' `1 H& a9 O9 q9 R( ^" w# f* |, W4 W4 t) S
The enforced expression of FasL in bone marrow and dendritic cells improves the efficiency of engraftment . We expressed the FasL chimeric protein on the surface of splenocytes and lin¨C BMC via biotinylation and transplanted these cells along with 106 naïve lin¨C BMC into irradiated allogeneic hosts (H2KdH2Kb). The levels of engraftment were significantly improved by expression of the FasL protein on the lin¨C BMC (p
( l# k2 Y1 ~  Y2 B: Z# J1 n+ g. J9 Q2 S+ `) ^
To determine whether expression of FasL on all the grafted lin¨C BMC cells would impact engraftment, the protein was adsorbed on the surface of donor cells with almost absolute efficiency (Fig. 2A). Transplantation of 1.5 x 106 FasL-decorated allogeneic lin¨C BMC resulted in superior levels (p
$ o" H8 P! K& e3 z+ F- n/ |
6 r7 C& s5 B( ?& wFigure 2. Expression of FasL on all donor cells improves allogeneic cell engraftment. (A): Streptavidin-FasL chimeric protein was efficiently adsorbed on the surface of bone marrow cells (BMC) via biotinylation. The protein was detected with an anti-FasL monoclonal antibody in flow cytometry. (B): B6 recipients conditioned with 850 rad total body irradiation were injected with 106 naïve (n = 7) and FasL-decorated (n = 10) lin¨C BMC from BALB/c donors. Chimerism was determined in the peripheral blood by flow cytometry at 3 weeks post-transplantation. (C): Mice transplanted with unmanipulated and FasL-coated lin¨C BMC proceeded to develop full chimerism at 16 weeks post-transplantation (n = 10). (D): At 14 weeks after primary transplantation, the chimeric mice served as donors of whole BMC to secondary irradiated hosts (n = 5). Abbreviations: FasL, Fas ligand; PE, phycoerythrin.
) d6 @) M" \- z1 z
4 D! g" ]% o; P" P- C! h& bExpression of FasL Protein Blocks Alloreactivity in a Fas-Dependent Manner
  X7 T4 I, a; P& |8 {: V; U9 C
! I$ g6 S: a; N' N& GThe likely mechanism in support of allogeneic cell engraftment is AICD-mediated inhibition of the alloresponses by the overexpressed FasL protein. To provide direct evidence for the immunoinhibitory role of FasL, cells coated with the protein were injected intraperitoneally into allogeneic hosts. The responses of recipient splenocytes were assayed after 7 days in a mixed lymphocyte reaction. FasL-decorated lin¨C BMC and splenocytes specifically blocked alloreactive responses, and the responses to third party antigens remained intact (Fig. 3A, 3B). FasL-decorated splenocytes were more effective than lin¨C BMC in their inhibition of alloreactive T-cell responses. This may be due to the prevalence of professional antigen-presenting cells in the spleen, capable of effectively activating alloreactive T cells that become sensitive to FasL-mediated killing through AICD .1 R# a, ~$ @- u0 c0 p  b

! [' b/ Y: \8 D. n, b3 Q. [: aFigure 3. FasL chimeric protein abrogates alloreactivity early after transplantation. (A): B6 mice (H2Kb) injected with 8 x 106 naïve or FasL-coated allogeneic lin¨C bone marrow cells (BMC) (H2Kd) were evaluated in a 5-day mixed lymphocyte reaction (MLR) assay at 7 days post-transplantation (n = 6). Splenocytes of B10.BR mice (H2Kk) served as third-party antigens. (B): The spleens of mice injected with 8 x 106 naïve or FasL-coated allogeneic splenocytes (H2KdH2Kb) were evaluated in MLR (n = 5). (C): Irradiated (2,000 rad) lin¨C BMC from green fluorescent protein mice coated with FasL chimeric protein were injected into allogeneic BALB/c hosts irradiated with 650 rad. Five days later, the organs were harvested, and the presence of FasL on the donor cells was assayed by flow cytometry (n = 5). Abbreviations: Allo, allogenic; FasL, Fas ligand; Spleno, splenocytes; Syn, syngeneic.# ?) p( ]9 @/ ^8 c0 I% C

7 W0 d; @. l2 `We determined the time frame of FasL protein activity on the surface of lin¨C BMC in view of the short survival of allogeneic splenocytes in vivo . To avoid dilution of the protein by cell division, the donor lin¨C GFP  BMC were decorated with FasL, irradiated, and then transplanted into sublethally irradiated allogeneic hosts (B6.GFPBALB/c). Approximately 15% of the FasL-coated cells were recovered from the bone marrow, spleen, liver, and lung at 5 days after injection, and 30%¨C50% of these cells retained the FasL protein on their surface, as determined with an anti-streptavidin antibody (Fig. 3C). Loss of the protein from the surface of donor cells recovered from these organs was consistent with an apparent half-life time of approximately 4 days, suggesting that FasL is particularly important in the very early stages of hematopoietic cell engraftment.% [4 R+ w3 C9 c: ?! M5 R
* ^* t3 s4 N9 k- O. B1 u
Transient Display of Ectopic FasL Protein Improves Syngeneic Cell Engraftment
1 {( _% @1 x/ W  l- T5 U8 m. ~. }; _. ~0 X! S# p
The veto activity of FasL-decorated lin¨C BMC  may operate at the systemic level, where it prevents destruction of prehomed BMC by the host immune system, or in the bone marrow, where it may remove the residual host cells that survived irradiation. The residual host cells include immunocytes, HSPC, and bone marrow stroma. To determine whether the engraftment-facilitating advantage of cells expressing FasL in allogeneic hosts operated only through the abrogation of alloreactive responses (systemic and intra-bone marrow), we performed syngeneic transplants, where immune modulation would not be expected to be a relevant variable in the engraftment process. Surprisingly, decoration of BMC with FasL improved (p
# K" w1 A& F, v/ J# h# K3 _7 ?4 N" K2 e( {  f1 z" y& n8 t$ _
Figure 4. FasL chimeric protein enhances syngeneic cell engraftment. (A): Mice conditioned with 850 rad total body irradiation were transplanted with 5 x 105 naïve and FasL-coated lin¨C BMC from syngeneic donors (CD45.1CD45.2). Donor chimerism was measured in the peripheral blood at 3 weeks post-transplantation. (B): After syngeneic transplants, the short-term engraftment was superior in recipients of FasL-coated cells, and all the mice proceeded to develop full donor chimerism at 16 weeks post-transplantation (n = 11). (C): Wild-type recipients were transplanted with 5 x 105 syngeneic (CD45.2CD45.1) lin¨C BMC from FasL-defective (gld) donors and from wild-type donors with and without FasL protein expression (n = 6). Abbreviations: BMC, bone marrow cells; FasL, Fas ligand.
& ]; {+ x) }& V. ~, Y4 G( T! t/ w7 a+ T
, T5 ^! V2 y0 t' V2 ?The Primary Target of the Donor Cell FasL Is the Bone Marrow Stroma3 z) q  f) {5 z4 v3 ^, C
4 {$ s# r9 ]& c& @; p$ ^: x6 `
To ascertain that the engraftment-facilitating effect of FasL operated through Fas signaling, we performed transplants of cells from wild-type mice into Fas-defective lpr recipients (n = 8). The engraftment advantage achieved by expression of the FasL protein on the surface of the cellular allografts was lost, indicating that the mechanism involved a competent Fas signaling pathway in the host (Fig. 5A). These results were further proof that the enhanced engraftment of FasL decorated cells was a specific effect of FasL and not an artifact of ex vivo cell manipulation. As expected, syngeneic transplants of unmanipulated and FasL-coated cells into lpr mice led to similar levels of engraftment (Fig. 5B).
/ H7 K3 B1 M/ V$ @
3 D5 z8 P% K- G9 _Figure 5. The primary target of donor cell FasL is the host marrow stroma. (A): B6/lpr recipients conditioned with 850 rad were injected with 1.5 x 106 naïve and FasL-coated lin¨C BMC from allogeneic donors (H2KdH2Kb). Chimerism was determined in the peripheral blood by flow cytometry at 3 weeks post-transplantation (n = 6). (B): Expression of FasL in 5 x 105 lin¨C BMC transplanted into syngeneic lpr recipients (CD45.1CD45.2) had no significant effect on engraftment at 3 weeks (n = 6). (C): Transplantation of 107 whole BMC from GFP donors into myeloablated lpr recipients resulted in full donor (GFP ) chimerism in bone marrow at 6 weeks post-transplantation (n = 8). (D): Stromal cultures of the GFP/lpr chimeras were predominantly of the lpr (GFP¨C) host phenotype after gating out CD45  and CD11c  cells. The data are representative of cultures from five transplanted mice. (E): Full GFP/lpr chimeras (Fas GFP  BMC and Fas¨CGFP¨C stroma) served as recipients of naïve and FasL-coated lin¨C BMC from CD45.1 mice (n = 6). Chimerism was determined in the peripheral blood at 3 weeks post-transplantation. Abbreviations: BMC, bone marrow cells; FasL, Fas ligand; GFP, green fluorescent protein.
: ?/ @0 ~8 Z+ b4 L& U" ~5 Y; U
8 t9 B# d! f5 R3 YThe evidence of a nonimmunogenic mechanism involved in the engraftment-supporting effect of FasL led us to seek the specific cellular targets of FasL-decorated cells in the host (stroma or residual host HSPC and immunocytes that survived irradiation). We generated a chimeric mouse with Fas¨C stroma and Fas  hematopoietic cells. This mouse was created by transplantation of 107 whole BMC from GFP donors into myeloablated (950 rad TBI) lpr recipients. At 6 weeks post-transplantation, these mice displayed full donor chimerism in the peripheral blood and the bone marrow (Fig. 5C). To ascertain that the marrow stroma was of the lpr host phenotype, the marrow aspirate was plated in long-term cultures. The predominant phenotype of the cells that grew in culture was of the lpr host (GFP¨C) origin, after gating out the CD11c  and CD45  cells (Fig. 5D). Sublethally-irradiated (850 rad) chimeras served as recipients of a second transplant of syngeneic CD45.1 cells. The levels of hematopoietic chimerism were similar after transplantation of naïve and FasL-coated lin¨C BMC (Fig. 5E), similar to the loss of FasL-mediated engraftment advantage in lpr recipients (Fig. 5B). By elimination, these data indicated that the primary target of donor cell FasL was the mouse stroma, rather than veto activity on residual hematopoietic cells in the host bone marrow.
8 W- u7 j$ q; [, w+ u6 w% y4 N& a2 @! D+ X7 H
DISCUSSION+ W: y0 M' \0 u. E1 n. i$ _4 y: o* L
- L- w4 e; c: y7 }+ {
The results of this study unravel the complex involvement of the Fas/FasL interaction in the early stages of hematopoietic stem and progenitor cell engraftment. The robust constitutive expression of the receptor and its ligand in donor cells suggested that this interaction is an important event in early engraftment. We recently demonstrated that the bone marrow-homed donor cells that upregulated Fas expression were insensitive to apoptosis mediated by this receptor (submitted manuscript). Thus, Fas was not involved in suppression of donor cell activity and viability, as had been previously reported . The insensitivity of lineage-negative donor cells to Fas-mediated apoptosis allowed them to counterattack the host alloreactive immune cells using constitutively expressed FasL. This activity was markedly enhanced by enforced expression of the noncleavable FasL protein on the surface of cells. This engraftment advantage is mediated, at least in part, by FasL-mediated interaction between the donor cells and the host marrow stroma through nonimmunogenic mechanisms. Thus, FasL augmented hematopoietic cell engraftment by at least two distinct mechanisms.
: P/ g% x3 \, Y; r# \8 E
( ?7 B* h4 m5 z1 o1 X% v( VThe immunogenic mechanism of FasL-mediated effects involved counterattack of the host alloreactive responses by presentation of the donor antigens in conjunction with FasL as an apoptotic trigger  and makes this approach attractive for clinical applications.# H7 L' n0 J0 b" S# ~
: e2 L  x# o9 `7 M% L- H
The early immunomodulation achieved by expression of FasL protein on the surface of HSPC in concert with the insensitivity of these cells to Fas-mediated apoptosis endows the hematopoietic progenitors themselves with a potent regulatory activity against the host alloreactive responses, as previously suggested .
3 j" A( h. P# A% s/ D  @
8 G( f+ s" ?9 K: W8 [In addition to potent immune regulatory activity, the donor cell FasL was also involved in nonimmunogenic interactions in the host bone marrow. We have no evidence to support the possibility that the donor cells removed residual host bone marrow cells to free niches for engraftment . Reconstitution of lpr mice with wild-type bone marrow cells created a chimera with Fas¨C stroma and Fas  hematopoietic cells. In these chimeras, the engraftment deficit observed in the lpr mice was not restored. Thus, FasL apparently supported engraftment in the syngeneic setting by interaction with the marrow stroma. This interaction involves distinct mechanisms from the suppression of graft rejection in the allogeneic transplants.
: r, u  a8 t6 O
. {2 t2 d3 {3 |4 kFas signaling appears to be involved in several functions of the marrow stroma that host engraftment and likely converges with additional cytokine-mediated signals in directing and regulating the activity of the grafted cells ." C: K8 ^$ z2 L( A3 q

% P; r" F; @% C3 g$ b! w4 \# VIn summary, the proposed regulatory mechanisms mediated by FasL include abrogation of allorejection, interaction with the bone marrow stroma, apoptosis, and stimulatory signals in hematopoietic cells. The insensitivity of the donor cells and bone marrow cellular components of the host to Fas-mediated apoptosis make FasL an attractive approach to augment the success of stem cell transplants. The use of a protein with a limited life span in vivo markedly enhanced engraftment and alleviated the need of large numbers of donor cells to achieve efficient immunomodulation and hematopoietic reconstitution.% g4 E7 s2 f4 k; V( j' y* y

, Q1 N& K7 n$ d0 W2 ?9 y# F. {DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST
' m/ b9 U6 }+ j4 F, V% N% |
* T+ X, f: K6 c8 MH.S. and E.S.Y. own stock in, have acted as consultants to, have performed contract work for, served as officers or members of the Board for, and have financial interests in ApoImmune Inc. (Louisville, KY).& |$ H! j! w) t1 f/ L8 `

3 g% c) w8 p9 B' eACKNOWLEDGMENTS! O& l6 m$ g# F0 ?" }7 I2 H
0 N' P8 c9 o0 I/ d' ]) \; z
This work was funded by Grants from the United States-Israel Binational Science Foundation (2003276 to N.A., I.Y., H.S., E.S.Y.), the Frankel Trust for Experimental Bone Marrow Transplantation (I.Y., J.S., N.A.), JDRF Innovative Grant 5-2005-1102 (to N.A.), ADA 1-05-JF-56, NIH R21: HL080108-01 (to E.S.Y.), and NIH R01:AI47864, NIH R21:AI057903 (to H.S.). The chimeric FasL protein technology used in this manuscript is licensed from the University of Louisville by ApoImmune Inc., Louisville, KY, for which H.S. serves as CSO and H.S. and E.S.Y. have significant equity in the company.
! F: F2 I1 t6 |* R- ^          【参考文献】* }5 N2 {3 n- A4 R- p% a+ i6 i6 F
7 s: A: v% A5 K& J( {* W' ]( O
5 P/ t* T/ b$ s. s# Z: T8 z, X# w3 W
Rathmell JC, Cooke MP, Ho WY et al. CD95 (Fas)-dependent elimination of self-reactive B cells upon interaction with CD4  T cells. Nature 1995;376:181¨C184.7 d8 {" ]& @% M; ]6 M" {! m

! Y1 s/ u% w5 y. Y0 cKurts C, Heath WR, Kosaka H et al. The peripheral deletion of autoreactive CD8  T cells induced by cross-presentation of self-antigens involves signaling through CD95 (Fas, Apo-1). J Exp Med 1998;188:415.
7 a1 u5 }8 j8 X# q" x: g+ n* ?4 j! c; `0 J( @& W
Hanabuchi S, Koyanagi M, Kawasaki A et al. Fas and its ligand in a general mechanism of T-cell-mediated cytotoxicity. Proc Natl Acad Sci U S A 1994;91:4930¨C4934.
% F! y1 \: b/ J& [% M( x7 r) m0 p0 |  V& g) {' n$ O" M5 m
Ju ST, Cui H, Panka DJ et al. Participation of target Fas protein in apoptosis pathway induced by CD4  Th1 and CD8  cytotoxic T cells. Proc Natl Acad Sci U S A 1994;91:4185¨C4189.
$ A  p1 q$ a6 S: Q
( D& v  A! i6 K2 i4 g6 xLu L, Thomson AW. Manipulation of dendritic cells for tolerance induction in transplantation and autoimmune disease. Transplantation 2002;73 (suppl 1):S19¨CS22.+ \8 H- s! G1 T' e% l
$ o$ A/ [, @+ M1 O! [4 J
Bohana-Kashtan O, Civin CI. Fas ligand as a tool for immunosuppression and generation of immune tolerance. STEM CELLS 2004;22:908¨C924.
3 S- o1 F/ v1 R; F
7 Q+ q! w" T4 W# S8 l) @Askenasy N, Yolcu ES, Yaniv I et al. Fas-ligand as a double-edged immunomodulator to induce transplantation tolerance. Blood 2005;105:1396¨C1404.
/ {4 ~" W: E3 X% `: [4 s( U' Y
8 k! R2 b) \8 A) YZhang HG, Liu D, Heike Y et al. Induction of specific T-cell tolerance by adenovirus-transfected, Fas ligand-producing antigen presenting cells. Nat Biotechnol 1998;16:1045¨C1049.; [5 `- {2 E3 G" W

8 O, A& W* E* i( u  mMatsue H, Matsue K, Walters M et al. Induction of antigen-specific immunosuppression by CD95L cDNA-transfected ¡®killer' dendritic cells. Nat Med 1999;5:930¨C937./ b$ y5 {+ g/ C

8 \. _: |' H" }5 \# N; ], p: @) G  WZhang HG, Su X, Liu D et al. Induction of specific T cell tolerance by Fas ligand-expressing antigen-presenting cells. J Immunol 1999;162:1423¨C1430.0 @! A, y( L: u! w# z
) f! F7 e( w) p2 a" b& x
Georgantas RW 3rd, Leong KW, August JT. Antigen-specific induction of peripheral T cell tolerance in vivo by codelivery of DNA vectors encoding antigen and Fas ligand. Hum Gene Ther 2000;11:851¨C858.6 N9 E1 ]: @4 Y* d( M' S# T- u
7 `% _8 J1 b8 v3 n/ I
Min WP, Gorczynski R, Huang XY et al. Dendritic cells genetically engineered to express Fas ligand induce donor-specific hyporesponsiveness and prolong allograft survival. J Immunol 2000;164:161¨C167.
- U4 s* y5 k8 S" w* p2 Q. O+ v; T" M7 P# s
Yolcu ES, Askenasy N, Singh N et al. Cell membrane modification for rapid display of proteins as a novel means of immunomodulation: FasL-decorated cells prevent islet graft rejection. Immunity 2002;17:795¨C808.
! `4 H) C- H' ~0 f  J% l' i& |1 g$ y- U$ ^, y
Askenasy N, Yolcu E, Wang Z et al. Display of Fas-ligand protein on cardiac vasculature as a novel means of regulating allograft rejection. Circulation 2003;107:1525¨C1531.
2 }% W, ~: z$ o7 m1 i
  b2 e: r& Z; q: L0 sGeorgantas RW 3rd, Bohana-Kashtan O, Civin CI. Ex vivo soluble fas ligand treatment of donor cells to selectively reduce murine acute graft versus host disease. Transplantation 2006;82:471¨C478.
( d5 H1 X' ]  o: Z! @) s3 ?& X; K9 S# ?5 l, X, U( X
George JF, Sweeney SD, Kirklin JK et al. An essential role for Fas ligand in transplantation tolerance induced by donor bone marrow. Nat Med 1998;4:333¨C335.
5 E( e* `/ B5 r7 i
1 B/ q4 f" a" y1 z5 I; n% JReich-Zeliger S, Zhao Y, Krauthgamer R et al. Anti-third party CD8 CTLs as potent veto cells: Coexpression of CD8 and FasL is a prerequisite. Immunity 2000;13:507¨C515.1 ]& b/ I2 u* I: H0 `: j* n
- h; z" ~2 `7 M8 x0 @' t$ j% `
Goldstein DR, Thomas JM, Kirklin JK et al. Indefinite allograft survival mediated by donor bone marrow is dependent on the presence of a functional CD95 (Fas) gene in recipients. J Heart Lung Transplant 2001;20:1132¨C1135.
. j) w0 |; t4 v! C4 u) K' I5 {: l& G. _8 p! ~/ ^, l- K
Rich RF, Green WR. Characterization of the Fas ligand/Fas-dependent apoptosis of antiretroviral, class I MHC tetramer-defined, CD8  CTL by in vivo retrovirus-infected cells. J Immunol 2002;168:2751¨C2758.( E& M% d* Y; }+ y/ e0 {5 b/ _+ }
5 E5 X# h7 Z. X9 b# Q8 w
Baker MB, Riley RL, Podack ER et al. Graft-versus-host-disease-associated lymphoid hypoplasia and B cell dysfunction is dependent upon donor T cell-mediated Fas-ligand function, but not perforin function. Proc Natl Acad Sci U S A 1997;94:1366¨C1371.4 o! \8 H- f' F; x1 I
$ J( K/ i8 F$ e6 k
Jiang Z, Podack E, Levy RB. Major histocompatibility complex-mismatched allogeneic bone marrow transplantation using perforin and/or Fas ligand double-defective CD4( ) donor T cells: Involvement of cytotoxic function by donor lymphocytes prior to graft-versus-host disease pathogenesis. Blood 2001;98:390¨C397.; P4 \: ^% v. K

0 M* W- h- t9 T% r; V1 W: ^9 PIwasaki T, Hamano T, Saheki K et al. Effect of graft-versus-host disease (GVHD) on host hematopoietic progenitor cells is mediated by Fas-Fas ligand interactions but this does not explain the effect of GVHD on donor cells. Cell Immunol 1999;197:30¨C38.
8 m$ C2 d$ V3 ?0 `8 r- b% x* y
3 ^: H9 D8 L0 |; a. ivan den Brink MR, Moore E, Horndasch KJ et al. Fas ligand-deficient gld mice are more susceptible to graft-versus-host-disease. Transplantation 2000;70:184¨C191.: n) {. ~# z9 o2 k3 a3 X. z

3 u3 M* D) s% C- i- n6 I* U( k7 fLepri E, Delfino DV, Migliorati G et al. Functional expression of Fas on mouse bone marrow stromal cells: Upregulation by tumor necrosis factor  and interferon . Exp Hematol 1998;26:1202¨C1208.1 Z, \# [- }4 O( p8 X4 y
0 {" _4 |+ i! t0 B  m
Stahnke K, Hecker S, Kohne E et al. CD95 (APO-1/FAS)-mediated apoptosis in cytokine-activated hematopoietic cells. Exp Hematol 1998;26:844¨C850.+ W  b+ b0 n2 b1 Y2 d7 z: I

8 i8 ^+ W: Z3 r# K- `. MBarcena A, Muench M, Song KS et al. Role of CD95/Fas and its ligand in the regulation of the growth of human CD34   CD38¨C fetal liver cells. Exp Hematol 1999;27:1428¨C1439.
8 y( L* I& l1 t
, r6 w* j( B& `1 Z% T& g7 l5 AJosefsen D, Myklebust JH, Lynch DH et al. Fas ligand promotes cell survival of immature human bone marrow CD34 CD38- hematopoietic progenitor cells by suppressing apoptosis. Exp Hematol 1999;27:1451¨C1459.
, b" m. G1 r( F7 L" W+ l5 ]0 U! o/ u4 D# i, V1 S2 j  U% ~2 Y
Saheki K, Fujimori Y, Takemoto Y et al. Increased expression of Fas (APO-1, CD95) on CD34  haematopoietic progenitor cells after allogeneic bone marrow transplantation. Br J Haematol 2000;109:447¨C452.' K# D5 Q0 \6 k0 R# G- D9 ]

$ ]- B5 m: |; }8 R/ Q) ~Bryder D, Ramsfjell V, Dybedal I et al. Self-renewal of multipotent long-term repopulating hematopoietic stem cells is negatively regulated by Fas and tumor necrosis factor receptor activation. J Exp Med 2001;194:941¨C952.
3 c% l( o0 G: v- s' A% N; }. f- k( |% q+ X( D; U
Dybedal I, Yang L, Bryder D et al. Human reconstituting hematopoietic stem cells up-regulate Fas expression upon active cell cycling but remain resistant to Fas-induced suppression. Blood 2003;102:118¨C126.
; w( t* q( u: ]2 O9 U0 r9 N/ u1 V, J) D
Bachar-Lustig E, Li HW, Gur H et al. Induction of donor-type chimerism and transplantation tolerance across major histocompatibility barriers in sublethally irradiated mice by Sca-1( )Lin(-) bone marrow progenitor cells: Synergism with non-alloreactive (host x donor)F(1) T cells. Blood 1999;94:3212¨C3221.
& d* u1 X6 d* ^* `$ a. v& s2 d6 T3 y3 p+ y6 G5 `8 M- {
Rachamim N, Gan J, Segall H et al. Tolerance induction by "megadose" hematopoietic transplants: donor-type human CD34 stem cells induce potent specific reduction of host anti-donor cytotoxic T lymphocyte precursors in mixed lymphocyte culture. Transplantation 1998;65:1386¨C1393.& G2 C% ]. ?5 P- K( R6 h

# c( s, \) Y/ m: GGur H, Krauthgamer R, Berrebi A et al. Tolerance induction by megadose hematopoietic progenitor cells: Expansion of veto cells by short-term culture of purified human CD34( ) cells. Blood 2002;99:4174¨C4181.
- d  G4 T, _, J
  D% p4 Y: Z1 Z& [Gur H, Krauthgamer R, Bachar-Lustig E et al. Immune regulatory activity of CD34  progenitor cells: Evidence for a deletion-based mechanism mediated by TNF-alpha. Blood 2005;105:2585¨C2593.
- l+ J/ f. C4 ?* H) u' c+ }' l4 D. O7 a* j
Whartenby KA, Straley EE, Kim H et al. Transduction of donor hematopoietic stem-progenitor cells with Fas ligand enhanced short-term engraftment in a murine model of allogeneic bone marrow transplantation. Blood 2002;100:3147¨C3154.2 ?/ t  ?* U6 C9 F& w
. k5 N5 K. [, ~! F  D5 j. X9 i
Stein J, Yaniv I, Askenasy N. Critical early events in hematopoietic cell seeding and engraftment. Folia Histochem Cytobiol 2005;43:191¨C195.2 J# V& y8 k6 G3 [' W  i
( d* O3 l" T4 [
Singh NP, Yolcu ES, Askenasy N et al. ProtExTM: A novel technology to display exogenous proteins on the cell surface for immunomodulation. Ann N Y Acad Sci 2005;1056:344¨C358.
1 r% Y2 |* _" O; b. T2 a: _, q% a7 q' ^0 x( _2 K# R6 X0 s
Raddatz G, Deiwick A, Sato T et al. Inhibition of cytotoxic alloreactivity by human allogeneic mononuclear cells: Evidence for veto function of CD2 cells. Immunology 1998;94:101¨C108.
9 V5 i- _7 D1 A5 Q7 ^) y
  F3 w- u0 G: @8 wReich-Zeliger S, Gan J, Bachar-Lustig E et al. Tolerance induction by veto CTLs in the TCR transgenic 2C mouse model. II. Deletion of effector cells by Fas-Fas ligand apoptosis. J Immunol 2004;173:6660¨C6666.
/ u0 U/ F5 f* v2 N1 O& e6 L  l3 c5 x9 ~+ v5 K
Askenasy N, Yolcu ES, Shirwan H et al. Characterization of adhesion and viability of early seeding hematopoietic cells in the host bone marrow in vivo and in situ. Exp Hematol 2003;31:1292¨C1300.
( y& s2 U7 A. A% R2 E
$ ~, |8 t2 r8 W1 f# P9 s8 PAskenasy N, Stein J, Yaniv I et al. The topologic and chronologic patterns of hematopoietic cell seeding in host femoral bone marrow after transplantation. Biol Blood Marrow Transplant 2003;9:496¨C504.1 m/ P& u  q+ J' _5 I% T1 P9 e
! \& W4 R4 V  n* W1 {- F- B9 l
Askenasy N, Farkas DL. In vivo imaging studies of the effect of recipient conditioning, donor cell phenotype and antigen disparity on homing of haematopoietic cells to the bone marrow. Br J Haematol 2003;120:505¨C515.
9 ]- u: b- J9 E0 m9 n) Z4 m5 j8 L  Y9 D) n2 J% L
Jones RJ, Celano P, Sharkis SJ et al. Two phases of engraftment established by serial bone marrow transplantation in mice. Blood 1989;73:397¨C401.. b6 X# |4 m/ v6 U, T8 I
9 R$ R4 @: B  G
Harrison DE, Stone M, Astle CM. Effects of transplantation on the primitive immunohematopoietic stem cell. J Exp Med 1990;172:431¨C437.
, w. [% v/ u9 }- V! G! d
% d: f# L) Y- ]2 e& i3 wHoney K, Bemelman F, Cobbold SP et al. High dose bone marrow transplantation induces deletion of antigen-specific T cells in a Fas-independent manner. Transplantation 2000;69:1676¨C1682.
/ b$ }3 Z' o% f* g0 o
: F4 {" d9 S. }/ N/ RChrobak P, Gress RE. Veto activity of activated bone marrow does not require perforin and Fas ligand. Cell Immunol 2001;208:80¨C87.$ Y. Z1 T4 A2 _2 a. d" t* h; C
  y6 ]' s- Z2 S" K. A- a3 S# S
Reisner Y, Reich-Zeliger S, Bachar-Lustig E. The role of veto cells in bone marrow transplantation. Current Opinion in Organ Transplantation 2006;11:366¨C372.6 d2 ?7 l! k3 t$ q1 \! f

; Y5 Z9 I6 T4 y" w" Y! }Quesenberry PJ, Colvin GA, Abedi M. Perspective: Fundamental and clinical concepts on stem cell homing and engraftment: A journey to niches and beyond. Exp Hematol 2005;33:9¨C19.' x5 A# m. F# K2 R# _! |0 J# F
" f/ J- I) ^0 c' J3 ~$ D
Yaniv I, Stein J, Farkas DL et al. The tale of early hematopoietic cell seeding in the bone marrow niche. Stem Cells Dev 2006;15:4¨C16." r! Z1 K4 Y3 y2 _/ C! w* u5 R

: w1 `  u4 W/ oGupta P, Blazar BR, Gupta K et al. Human CD34( ) bone marrow cells regulate stromal production of interleukin-6 and granulocyte colony-stimulating factor and in crease the colony-stimulating activity of stroma. Blood 1998;91:3724¨C3733.
+ R  _# z3 t1 `, T8 i& o& @. ]8 L+ _3 v
Moore MA. Cytokine and chemokine networks influencing stem cell proliferation, differentiation and marrow homing. J Cell Biochem Suppl 2002;38:29¨C38.
( ?: Q5 H3 F, b& b
! r0 B) p) Q7 ~) z* p! wSato T, Selleri C, Anderson S et al. Expression and modulation of cellular receptors for interferon-gamma, tumour necrosis factor, and Fas on human bone marrow CD34  cells. Br J Haematol 1997;97:356¨C365.9 _( L9 N+ {" O. M; q" A2 r
. e- S/ S' A, ^2 S* f+ b( l* _
Wajant H, Pfizenmaier K, Scheurich P. Non-apoptotic Fas signaling. Cytokine Growth Factor Rev 2003;14:53¨C66.
0 }: d# o3 F4 Y0 R+ q
" e4 f" m, _5 z7 X2 }* g" WAggarwal BB, Singh S, LaPushin R et al. Fas antigen signals proliferation of normal human diploid fibroblast and its mechanism is different from tumor necrosis factor receptor. FEBS Lett 1995;364:5¨C8.
5 S2 z+ [! X& B' f( m5 p. G/ w* ~/ W3 ?6 x, O! m+ m
Biancone L, Martino AD, Orlandi V et al. Development of inflammatory angiogenesis by local stimulation of Fas in vivo. J Exp Med 1997;186:147¨C152.
2 C, i/ K+ m' ?( ^" e! N
: N9 _5 C7 R7 g% lAhn JH, Park SM, Cho HS et al. Non-apoptotic signaling pathways activated by soluble Fas ligand in serum-starved human fibroblasts: Mitogen-activated protein kinases and NF-kappa B-dependent gene expression. J Biol Chem 2001;276:47100¨C47106.1 f# Y+ I4 Y8 O, E8 b2 ?+ `+ e! A

) j0 W. A) `& b) V9 i. YKitaura H, Nagata N, Fujimura Y et al. Effect of IL-12 on TNF-alpha-mediated osteoclast formation in bone marrow cells: Apoptosis mediated by Fas/Fas ligand interaction. J Immunol 2002;169:4732¨C4738.: U0 J4 e! F0 t) J; D

7 |9 E: t- _9 I( ~1 Q3 ^Park H, Jung YK, Park OJ et al. Interaction of Fas ligand and Fas expressed on osteoclast precursors increases osteoclastogenesis. J Immunol 2005;175:7193¨C7201.$ S9 R4 l7 V9 D3 g
1 v7 w* h8 M' L( F' T9 R
Desbarats J, Newell MK. Fas engagement accelerates liver regeneration after partial hepatectomy. Nat Med 2000;6:920¨C923.0 O3 V- ~" h" y- o: J# ]8 z

  w( Z' c+ y" r& `5 TFabricius D, Bonde S, Zavazava N. Induction of stable mixed chimerism by embryonic stem cells requires functional Fas/FasL engagement. Transplantation 2005;79:1040¨C1044.

Rank: 2

积分
75 
威望
75  
包包
2118  
沙发
发表于 2015-5-27 10:18 |只看该作者
先顶后看  

Rank: 2

积分
116 
威望
116  
包包
1832  
藤椅
发表于 2015-5-31 11:38 |只看该作者
彪悍的人生不需要解释。  

Rank: 2

积分
69 
威望
69  
包包
1788  
板凳
发表于 2015-6-1 10:01 |只看该作者
干细胞之家微信公众号
佩服佩服啊.  

Rank: 2

积分
162 
威望
162  
包包
1746  
报纸
发表于 2015-7-26 10:18 |只看该作者
顶的就是你  

Rank: 2

积分
104 
威望
104  
包包
1772  
地板
发表于 2015-7-28 18:05 |只看该作者
要不我崇拜你?行吗?  

Rank: 2

积分
136 
威望
136  
包包
1877  
7
发表于 2015-9-9 09:35 |只看该作者
我是来收集资料滴...  

Rank: 2

积分
80 
威望
80  
包包
1719  
8
发表于 2015-9-11 06:23 |只看该作者
神经干细胞

Rank: 2

积分
97 
威望
97  
包包
1738  
9
发表于 2015-10-5 15:58 |只看该作者
你加油吧  

Rank: 2

积分
70 
威望
70  
包包
1809  
10
发表于 2015-10-12 17:43 |只看该作者
昨天没来看了 ~~  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2025-5-23 07:01

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.