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: 262232|回复: 249
go

Tumor Necrosis Factor Promotes Human T-Cell Development in Nonobese Diabetic/Sev [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:36 |只看该作者 |倒序浏览 |打印
a Immunology Department, Weizmann Institute of Science, Rehovot, Israel;
: C, H9 L7 N$ |' }& D) ~9 M- ?
4 P! X  d; z' B) t/ \# \- cb Etablissement Francais du Sang Bourgogne/Franche-Comte, Besancon, France;# L! Z0 V9 Y# \; O9 |

; K* V' K2 u; wc Gene Therapy Institute, Hadassah University Hospital, Jerusalem, Israel;% j# c' u3 Z/ n; N7 ~% [

- y9 ~# F! q" C- ud Hematology and Bone Marrow Transplantation Department, Chaim Sheba Medical Center, Tel-Hashomer, Israel;
& _& h  Z: A: E, ^7 F
; K, J, g: |3 ?# }5 |8 Ae Department of Obstetrics and Gynecology, Assaf-Harofeh Medical Center, Zerifin, Israel;# K& U' `7 v; B" T! k$ {

6 ^5 `# k, ?, Z: B+ Q9 Jf Institut de Genetique Moleculaire de Montpellier, Montpellier, France- ^" c  e3 l* X& A5 r9 {, G4 P7 T

6 Q; A( W# @( K) W' e& D  i; ~Key Words. TNF ? Transplantation ? T lymphopoiesis ? Cord blood ? Mobilized peripheral blood cells ? Hematopoietic stem cells! h( _9 j: c  p& w- @

# ?& Q$ k' H0 y0 J* ZCorrespondence: Tsvee Lapidot, Ph.D.,Weizmann Institute of Science, Department of Immunology, PO Box 26, Rehovot, 76100, Israel. Telephone: 972-8-9342481; Fax: 972-8-9344141; e-mail; Tsvee.Lapidot@weizmann.ac.il
/ w& v' B, P& S3 C, Z0 x* b6 T) q( @5 X; A: k/ t
ABSTRACT
- M4 S  x9 t3 G  q
/ {* \+ L- A- F$ h/ _1 L$ pT-cell progenitors develop from hematopoietic stem cells (HSCs) that migrate from the bone marrow (BM) to the thymus, where they undergo a sequential program of proliferation, differentiation, and selection to generate mature functional T-cells for export to the peripheral blood and tissues . This process continues throughout adult life, although at a reduced rate . Limitation of this process is a major cause for early immune dysfunction in adult patients undergoing HSC transplantation. Depressed T-cell development in these patients has been related mainly to low levels of thymic function in adults . Understanding the mechanisms of human T-cell development and function is thus important to enhance immune reconstitution after clinical HSC transplantation.
6 R; a8 x5 Z6 n4 g& P1 j% u
7 Z2 b, u! s; g1 O7 L1 uTo better elucidate these processes, a variety of experimental systems for studying human T lymphopoiesis have been used . One of the in vitro strategies is the fetal thymus organ culture (FTOC), in which primitive human progenitors are seeded onto lymphoid-depleted murine fetal thymic lobes . This approach enables studies to be performed within the thymic tissue, but development of mature CD8  thymocytes is rare , and differentiation is uncoupled from the in vivo–occurring migration and homing of cells. Severe combined immunodeficient (SCID) and nonobese diabetic (NOD)/SCID mice have been used as a functional preclinical model for in vivo engraftment of human hematopoietic progenitor cells. This system identified a small population of primitive hematopoietic cells capable of repopulating transplanted recipients, defined as SCID-repopulating cells (SRCs) . However, using these models, studies of T-cell reconstitution after HSC transplantation are limited because of the low and slow development of human T cells . A defect in migration of human T-cell progenitors to the mouse thymus was suggested by van der Loo et al. , who succeeded in inducing homing of human cells to the thymus of NOD/SCID mice previously transplanted with enriched human CD34  cells by G-CSF and stem cell factor (SCF)–induced mobilization.
% x' N+ P4 K) O& T. k
) j/ {+ k0 I$ X) N+ VOther studies using in vivo models include the SCID-hu model, in which human CD34  cells are coimplanted with T cell–depleted human fetal thymus . Hence, both FTOC and SCID-hu models are based on the availability of a fetal thymic tissue. In another model, extrathymic T-cell development can be achieved in the athymic immunodeficient bnx mouse by cotransplantation of human BM CD34  stem and progenitor cells together with human BM stromal cells engineered to produce human interleukin (IL)-3 and IL-7 . However, significant numbers of human T cells can be recovered from the BM of these mice only 4–6 months after transplantation, and there is no human B-cell development in these mice.0 x$ Y) K% {8 w; G# E, s
4 J  \% M8 L3 F" m! Z
Models that were recently developed are based on the inhibition of natural killer (NK) cell activity in NOD/SCID and Rag2 knockout mice either by neutralizing antibodies against the murine IL-2R  or by knockout of the IL-2Rg gene . In these models, human T cells develop in the murine thymus, and mature peripheral T cells begin to emerge 8–12 weeks after transplantation of enriched human cord blood (CB) CD34  cells.6 l  ]" E: @( E5 w- z9 I8 i) X4 y7 B

0 J8 K: Q1 Y6 V) K# CExtrathymic differentiation of T cells plays an important role in immune reconstitution in the absence of efficient thymic function. Strober and colleagues  have reported the presence of early murine T-cell progenitors in the adult mouse BM that generate CD4  and CD8  T-cell receptor (TCR)-?  T cells in an extrathymic pathway, supported by the marrow microenvironment. Similarly, extrathymic differentiation of human T cells in mouse BM was documented both in the bnx/hu model  and after transplantation of human CB CD34  progenitor cells transduced with an active form of Notch into NOD/SCID recipient mice .9 E' v# _0 i! f* d  O) [

8 j" g+ d5 ?" O8 M) {The cytokine tumor necrosis factor  (TNF), produced by a variety of cell types, including thymic stromal cells, was found to be associated with critical events leading to T-lineage commitment and differentiation. In mice, TNF induces CD25 (IL-2R) expression on early immature thymocytes and is required for additional thymocyte maturation to single-positive (SP) CD4 and CD8 cells . Weekx et al.  showed that TNF also promotes differentiation of human BM CD34 -enriched cell populations into T cells in the FTOC system. However, T-cell differentiation and selection, as well as the thymus architecture, are normal in TNF knockout mice. Interestingly, there is a threefold increase in B-cell numbers in the thymus of these mice .
, }5 E3 M* d. x
) w: _: X4 H: V6 sThe present study was designed to determine whether TNF can augment human T-cell development in vivo. We established a system that rapidly generated human T cells in vivo, based on TNF pretreatment to irradiated NOD/SCID mice before transplantation of either human CB mononuclear cells (MNCs) or, even more clinically relevant, adult mobilized peripheral blood (MPBL).6 N- W4 P1 w* N
. y: D+ |. a4 m; k% Q- i
MATERIALS AND METHODS
; C3 z5 C. j  s" m; L2 Y
6 V, k+ n+ \& d, |- s0 a9 ~. Y" s' t; TTNF Promotes Reconstitution of Human T Lymphocytes from CB SRC with a Concomitant Reduction in Human Immature B Lymphocytes in the BM of NOD/SCID Recipient Mice* r. w# c- O7 K: @, @$ Z
! _' l3 X, a4 V! N5 U7 D1 v* H& v
The potential involvement of TNF in human T-cell development was studied by injection of TNF into sublethally irradiated NOD/SCID mice and transplantation of human CB MNCs 4 hours later. The activity of human TNF on the mice was confirmed by examination of adhesion molecule expression in the murine BM. We found that human TNF upregulated inter cellular adhesion molecule (ICAM)-1 expression (twofold increase) on murine BM cells in vivo within 8 hours (data not shown). One month after transplantation, BM cells of recipient mice were analyzed for expression of the human CD45 marker to evaluate total human cell engraftment in TNF-treated mice. Levels of total human cell engraftment in TNF-treated mice were similar to those detected in untreated mice (50 ± 9.7% and 47.4 ± 7.9%, respectively; n = 8). However, flow cytometric analyses showed the emergence of a CD45high cell population in the BM of TNF-treated mice, which was not apparent in the BM of untreated mice, parallel to a reduction in immature CD19  B cells (Fig. 1A). We additionally analyzed the CD45high cells by staining with CD4, CD8, and CD3 markers of human T cells and found that these cells were T lymphocytes (Fig. 1B; CD3 staining is not shown). Some of these cells were CD4 CD8  double-positive (DP), phenotype characteristic to immature T cells, strongly suggesting that TNF treatment induced human T-cell development. The frequency of human T-cell engraftment, determined by the presence of more than 0.5% DP cells in the murine BM, was 46% (27 of 59) in TNF-pre-treated recipient mice versus 25% (11 of 43) in control recipient mice.  y4 B, U+ `2 Q

& a1 \8 U& q$ e0 h; r7 @Figure 1. Human cell engraftment in the BM of CB MNC-transplanted NOD/SCID. NOD/SCID mice were sublethally irradiated (375 cGy) and were either pretreated or not (control) with 0.5 μg TNFper mouse injected i.p. Four hours later, mice were transplanted with 20 x 106 human CB MNC per mouse (i.v.). BM cells were collected from individual recipient mice 1 month after transplantation, stained with human-specific monoclonal antibodies, and analyzed by flow cytometry for the presence of human cells. Gates were set on side-scatter (SSC) versus forward light scatter (FSC) and on viable cells. (A): Representative analysis of human cells in an individual recipient mouse as assessed by the percentages of CD45  human hematopoietic cells and CD19 CD45  B cells. (B): Presence of human T cells in representative BM sample as assessed by monitoring CD4 and CD8 expression. (C): Percentage of human lymphocytes engrafted in murine BM. Each dot represents one mouse, and bars indicate the mean value. (D): Cells recovered from the murine BM were differentiated in vitro with human stem cell factor plus interleukin-15 (100 ng/ml each) for 10 days and then stained for the expression of the CD56 natural killer cell marker. Percentages of human cells are indicated. *Significant difference in T- and B-cell engraftment between TNF-pretreated mice and untreated mice (p = .01). Abbreviations: BM, bone marrow; CB MNC, cord blood mononuclear cell; NOD/SCID, nonobese diabetic/severe combined immunodeficient; TNF, tumor necrosis factor.
+ L5 E( N' I) ?: j
; K6 N8 X, j# D/ _" a, E" _  TIn preliminary experiments performed to determine the optimal protocol for promoting T-cell repopulation, we injected TNF at different time points before, during, and after transplantation of total MNCs, CD3 -depleted MNCs, or CD34 -enriched cells (data not shown). Analyses of the different protocols examined demonstrated that human T-cell engraftment was obtained only when total MNCs were transplanted together with TNF administration 4 hours before transplantation. Therefore, we established this procedure as the standard protocol for our subsequent transplantation experiments. TNF-treated recipient mice rarely showed any signs of graft-versus-host disease (GVHD) within 2 months after transplantation.2 R" y6 @# A* k7 }: Z  a" q8 E2 t
0 _$ Y; Z& q# Q; p
T-cell engraftment after TNF treatment led to a concomitant reduction in the levels of immature human B cells (Fig. 1A). As demonstrated in Figure 1C, the percentages of developing human B cells in the BM were 31 ± 5.5% (n = 7) in control mice compared with 12.6 ± 3.9 (n = 7) in TNF-treated mice (p = .01). The ability of human lymphoid progenitors from the BM of TNF-treated mice to differentiate ex vivo into CD45 CD56  NK cells in response to IL-15 and SCF was also lower than that of progenitors from control untreated mice (Fig. 1D).% _' k% H1 P  _
1 m4 D- y. G. W' z. w
TNF Treatment In Vivo Leads to the Emergence of DP Pre-T Cells 2–3 Weeks after Transplantation
3 y9 P4 a$ v: x+ c' Z4 V" m) v8 J# ]; v; \
To determine whether the human T cells detected in mouse BM 1 month after transplantation were derived from the expansion of transplanted mature T cells that homed to the BM or, alternatively, whether they developed in the mouse, we followed their kinetics in the BM of the recipient mice. A low percentage of human T cells that homed to the BM was detected 24 hours after transplantation (
4 X- ]& i8 ^( H  ?( j4 u0 T5 M" A9 L( D8 L
Figure 2. Kinetics of human T-cell development in the BM of transplanted mice. Irradiated nonobese diabetic/severe combined immunodeficient mice were either pretreated or not (control) with TNF before transplantation of 20 x 106 cord blood mononuclear cells. BM cells were collected from individual recipient mice at various time points, and the presence of human T cells was monitored by staining with human-specific CD4/CD8 surface-marker monoclonal antibody. Representative flow cytometric analyses are shown, and the percentages of single-positive and double-positive T cells are indicated. Each plot represents different mouse. Abbreviations: BM, bone marrow; TNF, tumor necrosis factor; w, week.1 m# t' A- `7 H5 d) \: C- h7 E, P* h

3 j6 J% |- ]1 }9 w. j, h7 W9 o0 \0 XEngrafted Human T Cells Are Derived from Immature CD34 -Enriched Cells
( |/ Q' B" Z. r' K/ C% c/ m, k9 m- S! D0 v* ^: C5 f
To additionally substantiate that the reconstituted T cells originated from immature CD34  progenitors, we performed sex-mismatch studies using Y chromosome as a marker. Enriched CB CD34  cells from male donors were separated by two columns and determined to be devoid of mature CD3  T cells (Fig. 3A). These cells were then cotransplanted into NOD/SCID mice together with CB CD34– MNCs from a female donor. One month after transplantation, the CD4/8 DP and SP subsets recovered from the BM of the recipients were separated by antibody-magnetic bead column and used for PCR amplification of Y-chromosome DNA-specific sequences. Figure 3B shows that Y-chromosome sequences are present in the separated human T cells, as well as in the human T-negative separated cells. Such approach shows that the engrafted human T cells were mostly of male origin, indicating that they derived from the immature CD34  progenitor cells. Of note, transplantation of 20 million CD34– MNCs (which include approximately 4 million mature T cells) or transplantation of purified, 4 million mature T cells did not engraft or, in rare cases, led to only negligible levels of human cell engraftment only by MNC, most probably because of low CD34  cell contamination. In addition, enriched CD34  cells transplanted alone did not give rise to T cells within 1 month after transplantation (data not shown).
# Y8 }5 d  G- f# z7 \
) K. _3 A4 D7 z; qFigure 3. Detection of CD34 -derived T cells in the BM of NOD/SCID recipient mice. Irradiated NOD/SCID mice pre-treated with TNF were transplanted with 2 x 105 human cord blood male CD34 -enriched cells together with 20 x 106 female CD34– MNCs. (A): Representative flow cytometric analysis of the purity of male CD34 -separated cells. The purified cells are devoid of CD3  mature T cells. (B): One month after transplantation, the human CD4/CD8 double- and single-positive BM subsets were separated. DNA was extracted and assessed for the presence of Y chromosome–specific sequences by polymerase chain reaction. The resulting products resolved on a 1.6% agarose gel are shown. A band with the predicted size of 150 bp is detected in the positively selected BM T cells as well as in the remaining T-negative cells, indicating the colonization of the BM by progenitors of male origin. As a positive control, DNA from CD34  cells of male donors is shown, and as a negative control, CD34– female MNC DNA is shown. The H2O lane shows a control using Y chromosome–specific primers and no template DNA. Abbreviations: BM, bone marrow; MNC, mononuclear cell; NOD/SCID, nonobese diabetic/severe combined immunodeficient; TNF, tumor necrosis factor.
3 e' h7 G6 N/ \$ T3 d7 n1 N  y  [  O. R# j& }  ~# ]
TNF-Pretreatment In Vivo Enhances Human T-Cell Reconstitution from G-CSF–Induced MPBL Transplanted Cells5 t% Z% H8 D+ D( v

) `1 M4 j1 E" y3 w0 ?" f3 _1 U0 BWe next examined whether TNF also enhances T-cell engraftment in hematopoietic repopulating cells derived from sources other than CB. We thus chose to study adult G-CSF–induced MPBLs, because they are the major source of HSCs for clinical transplantations. The experimental protocol of TNF pretreatment was identical to that used for CB cells. The results show that TNF enhanced both total and T-lineage engraftment from human MPBL (total engraftment, 4.4 ± 1.6% in TNF-treated mice versus 1.7 ± 1% in mice without TNF treatment, Fig. 4A), but the levels of engraftment were in general lower than those detected in CB MNC-transplanted mice. The high variability in levels of engraftment was probably attributable to variability in SRC frequency and expression of CXCR4, a receptor critically involved in homing and engraftment, in different adult donors in general . However, the trend was similar in all experiments. As shown in Figure 4B, human MPBL transplanted into TNF-treated mice gave rise to human T cell engraftment, whereas myeloid and B cells were rarely found. The frequency of human T-cell engraftment, determined by the presence of greater than 0.5% DP cells in the murine BM, was 43% (6 of 14) in TNF-pretreated mice versus 21% (3 of 14) in control mice. It thus appears that TNF-treatment has a beneficial effect on human T-cell reconstitution after MPBL as well as CB transplantation in NOD/SCID mice." e& F4 ]  r3 J' B5 r0 z8 z
. R' w: L+ p( M, V6 e  @
Figure 4. Human cell engraftment in the BM of MPBL-transplanted NOD/SCID mice. NOD/SCID mice were sublethally irradiated (375 cGy) and were either pretreated or not (control) with 0.5 μg TNF per mouse injected i.p. Four hours later, mice were transplanted with 20 x 106 human adult MPBL per mouse i.v., and BM cells were collected from individual recipient mice 1 month after transplantation. (A): Total human cell engraftment within BM of recipient mice determined by CD45 staining. Each dot represents one mouse, and bars indicate the mean value of 11 mice from three independent experiments. (B): Representative flow cytometric analysis of individual recipient mouse (percentages are indicated). Abbreviations: BM, bone marrow; MPBL, mobilized peripheral blood; NOD/SCID, nonobese diabetic/severe combined immunodeficient; TNF, tumor necrosis factor., v0 H7 B( i& F% T

- j7 w. P0 G% J* t) x& S. G  }Human T Lymphocytes Reconstitute the Hematopoietic Organs and the Peripheral Circulation of TNF-Treated NOD/SCID Recipients/ R# {1 k, E$ a# j1 a6 a* x1 S

/ ]* j# V) F" x& K8 H$ fTo assess whether reconstitution of human T cells in NOD/ SCID mice after TNF treatment leads to T-cell repopulation in lymphoid and hematopoietic tissues other than BM, we studied the engraftment of human T lymphocytes in the spleen, thymus, and peripheral blood (PB) of recipient mice 1 month after transplantation of either CB MNC or MPBL (Fig. 5). As shown in Figure 5A, human T- cell repopulation was not restricted to the BM; human T cells were also detected in spleen and PB of TNF-treated recipient mice. Most important, we detected immature human T cells also in the thymus of these mice, the major organ for T-cell development. Levels of T-cell engraftment were generally similar in all of the organs examined (Fig. 5B), although engraftment in the murine thymus appeared at lower levels. Human cell engraftment levels were higher in mice transplanted with CB MNCs compared with those transplanted with MPBL in all organs examined. In mice that were not treated with TNF and did not show human T cells in their BM, human T cells could not be detected in their spleen, thymus, or PB (data not shown).
0 F+ L* V8 f2 q' J0 N! C# ^& A' P
Figure 5. Human cell engraftment in murine organs of TNF-pretreated NOD/SCID mice. Cells were collected from individual recipient mice 1 month after transplantation of human CB MNC or MPBL, stained with human-specific mAbs, and analyzed for the presence of human cells. (A): Representative flow cytometric analysis of human cells in the BM, spleen, thymus, and PB of NOD/SCID mouse transplanted with CB MNC (percentages are indicated). (B): Percentage of human CD4  and CD8  single- and double-positive cells in the BM, spleen, and thymus of recipient mice transplanted with either CB MNC or MPBL. Each bar represents the mean ± standard error of the mean of at least seven mice transplanted with CB MNC and three mice transplanted with MPBL. *Significant difference in T-cell engraftment between CB and MPBL for BM (p = .05). Abbreviations: BM, bone marrow; CB MNC, cord blood mononuclear cell; MPBL, mobilized peripheral blood; NOD/SCID, nonobese diabetic/severe combined immunodeficient; PB, peripheral blood; TNF, tumor necrosis factor.& l  v% {9 J* f) G& E; H4 o
/ k3 j; _( d% a! c1 G1 A' `
Phenotypic Characterization of the Engrafted Human T Lymphocytes in TNF-Treated NOD/SCID Mice- f* f+ n: i0 s( c

9 [' Q0 A7 p- B& ZTo further characterize the nature of the T cells engrafted in TNF-pretreated recipient mice, T lymphocytes from the BM and thymus were analyzed for additional markers of T cells. The phenotype of human T lymphocytes from a representative experiment is shown in Figure 6. Most of the T cells expressed CD3 on their surface. Mature SP CD4 CD3  and CD8 CD3  T cells were rapidly detectable within 3 weeks after transplantation. Further phenotypic analysis showed that the human CD3  cells expressed TCR?, and no substantial levels of TCRcells were detected. Importantly, expression of CD1a, a marker of immature thymocytes that disappears on terminal differentiation, was detected only in the thymus at low levels (Fig. 6, thymus). Phenotypic analyses for naive (CD45RA ) and memory (CD45RO ) cells in the BM demonstrated an abundance of CD45RO  memory cells when CB MNCs were transplanted. In the case of MPBL transplantation, all of the engrafted human T cells were CD45RO  (data not shown). Table 1 summarizes the results of the relative percentages of SP and DP T-cell populations, calculated in relation to the total T-cell engraftment levels. When CB MNCs were transplanted, the levels of CD8 SP and CD4 SP T cells in the murine BM were mostly similar, although there was variability from mouse to mouse. When MPBLs were transplanted, the levels of CD8 SP cells were higher than CD4 SP cell levels (2:1 ratio).
3 l( p' o" C, p9 Z3 A4 Y5 Y9 Q
& B5 I- y: d0 `' _2 V" ?Figure 6. Phenotypical analysis of human T cells engrafted in CB nonobese diabetic/severe combined immunodeficient recipient mice. BM and thymus cells from tumor necrosis factor–pretreated mice transplanted with CB mononuclear cells were collected 1 month after transplantation, stained with human-specific monoclonal antibody, and analyzed by flow cytometry for different markers of human T cells. Representative results are presented (percentages are indicated). Abbreviations: BM, bone marrow; CB, cord blood; TCR, T-cell receptor.
' M, u. n% f$ O! q- n% t5 A# H5 D2 E& a6 T) r$ h0 z" }1 c5 Y
Table 1. Proportion of human CD4 and CD8 single- and double-positive T cells in tumor necrosis factor–treated nonobese diabetic/severe combined immunodeficiency recipient mice
& c+ i5 {. ~8 L7 t: T3 s) c# H  }6 `& V5 Y/ c
To examine the clonality of human T cells engrafted in TNF-pretreated NOD/SCID mice, hypervariable complementarity determining region 3 size distribution of 24 variable regions of ? chain (V?) of the TCR subsets was analyzed by the immunoscope method. Analysis of TCR-V? expression exhibited a wide range of TCR-V? usage in the BM and spleen of some TNF-pretreated recipient mice. Results of T cells from the BM of one recipient mouse are shown in Figure 7. The results were variable with greater and reduced diversity in individual mice. Nevertheless, the data demonstrate the reconstitution of human T cells expressing multiple TCR-V? chains.0 p! t* |# \2 k. g! E! Z
- U! u7 U% d! y
Figure 7. CDR3 size distribution of human TCR-V? subsets of T cells developed in TNF-treated mice. The TCR-V? repertoire was assessed by analysis of TCR CDR3 size distribution (Immunoscope profiles) of BM and spleen cells obtained from TNF-pretreated mice 4 weeks after transplantation of human mononuclear cells. Twenty-four PCR products were generated by RT-PCR with 24 different TCR-V? subfamily-specific primers and one constant consensus primer (C?), followed by a run-off reaction with a fluorescent C? primer. The graphs represent fluorescence intensity in arbitrary units plotted against the amino acid size of CDR3. The size distributions within nine TCR-V? families from BM of one recipient mouse are shown. A polyclonal profile is observed for TCR-V? families 3, 7, 8, 9, 13a, 14, and 18, and a skewed profile is observed for TCR-6b and 11. Abbreviations: BM, bone marrow; CDR3, complementarity determining region 3; RT-PCR, reverse transcription–polymerase chain reaction; TCR, T-cell receptor; TNF, tumor necrosis factor.
) D' z( m, X5 X) M; [: l! w
; T3 l/ v1 w5 M1 ^" J; \( |5 i5 v) [We sought to evaluate development of T cells by analysis of TRECs, which are formed during the TCR rearrangement process that T cells undergo during maturation in the thymus, in additional experiments with seven human donors. Because recently differentiated T cells contain a certain TREC content, quantification of this TREC content is considered a measure of T lymphopoiesis. DNA was isolated from human T cells separated from the BM and spleen of recipient mice 1 month after transplant and analyzed for TREC content by quantitative PCR (numbers of human T cells detected in the murine thymus in this stage were not sufficient to perform TREC analysis). TRECs were detected in T cells isolated from most samples, including spleen as well as BM (Table 2). Nevertheless, levels of TRECs were approximately 5 to 60 per million cells, which is relatively very low, suggesting that these cells have undergone multiple rounds of division after differentiation, as previously observed in both murine and human transplantation studies in which T cells differentiated from HSCs . Alternatively, we cannot rule out that these low levels are also attributable to proliferation of the transplanted mature T cells.+ Z5 G! F; p0 O4 O" J3 [

: Z/ n% q$ l5 z9 Y; z+ Q+ {8 iTable 2. TREC content of human CD4 and CD8 cells recovered from tumor necrosis factor–treated recipient mice1 D8 _1 \4 |6 t) W  I
, m; k9 ^7 u& C" z9 E7 `! Y# P
DISCUSSION
8 ~1 y. ^6 [% k% V1 z' w' ?9 W" E6 x* o. {0 ^7 I: T
This work was supported in part by a grant from AFIRST France-Israel grant agency. We would like to thank Professor Dov Zipori for fruitful discussions and to Loya Abel for helpful technical assistance.& Y" c/ {* H1 v7 W$ @
3 Z1 B7 R1 h) C
REFERENCES! J( {& ?9 l1 Q, Z0 n- }. f
* y; _& r# h, n% M5 U2 ?
Spits H. Development of alphabeta T cells in the human thymus. Nat Rev Immunol 2002;2:760–772.
2 D9 g/ A6 g7 l9 C! l- c/ `, ^9 X( S
Jamieson BD, Douek DC, Killian S et al. Generation of functional thymocytes in the human adult. Immunity 1999;10:569–575." ~! O  V& m. B; o+ J
5 o/ Q3 i6 ?2 d+ H- A5 o
Kalwak K, Gorczynska E, Toporski J et al. Immune reconstitution after haematopoietic cell transplantation in children: immunophenotype analysis with regard to factors affecting the speed of recovery. Br J Haematol 2002;118:74–89.$ Y+ `" V% _7 D2 t; ]0 H# Z

1 c3 o7 i2 q* r( }Roux E, Dumont-Girard F, Starobinski M et al. Recovery of immune reactivity after T-cell–depleted bone marrow transplantation depends on thymic activity. Blood 2000;96:2299–2303.6 ^9 i: s2 R' L" S. b

, ~" }5 m" Z" k  B1 V' ^Plum J, De Smedt M, Verhasselt B et al. Human T lymphopoiesis: in vitro and in vivo study models. Ann N Y Acad Sci 2000;917:724–731.0 ]( g  c& V& e6 n3 D, [* u
! x/ K; ]' w( _; m: i! c+ ]
Globerson A, Kollet O, Abel L et al. Differential effects of CD4  and CD8  cells on lymphocyte development from human cord blood cells in murine fetal thymus explants. Exp Hematol 1999;27:282–292.
: |2 d" v; O2 f/ K' ?8 b) u; K" S  L+ e+ u8 e
Hare KJ, Jenkinson EJ, Anderson G. In vitro models of T cell development. Semin Immunol 1999;11:3–12.
! y" r3 `- o5 H" G9 Z8 ~- }) x% j' s8 y
Blom B, Res P, Noteboom E et al. Prethymic CD34  progenitors capable of developing into T cells are not committed to the T cell lineage. J Immunol 1997;158:3571–3577.
* L1 o9 t$ i9 O& X8 m
/ z- L8 F, n' x, pLapidot T, Pflumio F, Doedens M et al. Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice. Science 1992;255:1137–1141.
1 Q; S5 U. s  D3 O; x$ u
' p5 {. I% E1 u! N/ UVormoor J, Lapidot T, Pflumio F et al. Immature human cord blood progenitors engraft and proliferate to high levels in severe combined immunodeficient mice. Blood 1994;83:2489–2497.
/ {8 ?& ]5 [4 K" x1 m- r
0 z# y/ L. u) l* ^! Y/ N% o; CPflumio F, Izac B, Katz A et al. Phenotype and function of human hematopoietic cells engrafting immunedeficient CB17-severe combined immunodeficiency mice and nonobese diabetic-severe combined immunodeficiency mice after transplantation of human cord blood mononuclear cells. Blood 1996;88:3731–3740.+ e/ u' B( o+ ?* z) A' B6 d7 D
6 P( I0 ?& M1 w
Larochelle A, Vormoor J, Hanenberg H et al. Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: implications for gene therapy. Nat Med 1996;2:1329–1337.7 s0 S+ ]- F$ R7 W- u- x7 g

* h* ^! G2 M! a6 y6 MCashman JD, Lapidot T, Wang JC et al. Kinetic evidence of the regeneration of multilineage hematopoiesis from primitive cells in normal human bone marrow transplanted into immunodeficient mice. Blood 1997;89:4307–4316.9 B% J8 T' r: Q) t' X
6 ~5 a/ |# E/ i& b2 u
Hogan CJ, Shpall EJ, McNulty O et al. Engraftment and development of human CD34 -enriched cells from umbilical cord blood in NOD/LtSz-scid/scid mice. Blood 1997;90:85–96." ~6 ]. Y5 O) d' p% k
7 i' B: \& c/ Q7 ?8 {" P
van der Loo JC, Hanenberg H, Cooper RJ et al. Nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mouse as a model system to study the engraftment and mobilization of human peripheral blood stem cells. Blood 1998;92:2556–2570.
2 [) G4 O" R7 G, a; M  D, f; M
9 s. W5 U3 S" f1 g4 N. s: m1 kYusarov S, Kollmann TR, Kim A et al. Severe combined immunodeficiency mice engrafted with human T cells, B cells, and myeloid cells after transplantation with human fetal bone marrow or liver cells and implanted with human fetal thymus: a model for studying human gene therapy. Blodd 1997;89:1800–1810.
1 O1 K: }, z9 L- Q. H& A# c: I
, S& c, o# ^6 ?9 ?% W( C2 t6 jTsark EC, Dao MA, Wang X et al. IL-7 enhances the responsiveness of human T cells that develop in the bone marrow of athymic mice. J Immunol 2001;166:170–181.6 d6 a9 ~( N6 A- r9 Y0 f7 H
( x# Z0 Y3 X! z1 A8 g
Kerre TC, De Smet G, De Smedt M et al. Adapted NOD/SCID model supports development of phenotypically and functionally mature T cells from human umbilical cord blood CD34  cells. Blood 2002;99:1620–1626.
/ l  n! @3 C* V- J" i
% Y5 W" P+ k+ D) c+ SYahata T, Ando K, Nakamura Y et al. Functional human T lymphocyte development from cord blood CD34  cells in nonobese diabetic/Shi-scid, IL-2 receptor gamma null mice. J Immunol 2002;169:204–209.
" P3 s7 D* |6 l+ Q8 {) `6 s9 k1 y. t4 @: O# H6 |8 y" O
Traggiai E, Chicha L, Mazzucchelli L et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science 2004;304:104–107.
* s( f) ]& F/ ~4 [5 e8 e3 e! j+ y. E4 B( M+ c" b+ Z. K
Garcia-Ojeda ME, Dejbakhsh-Jones S, Weissman IL et al. An alternate pathway for T cell development supported by the bone marrow microenvironment: recapitulation of thymic maturation. J Exp Med 1998;187:1813–1823.1 ]" E; ^/ Y/ B
: K# `+ s+ ~$ Z, f# R3 |7 ^: X
Dejbakhsh-Jones S, Garcia-Ojeda ME, Chatterjea-Matthes D et al. Clonable progenitors committed to the T lymphocyte lineage in the mouse bone marrow: use of an extrathymic pathway. Proc Natl Acad Sci U S A 2001;98:7455–7460.
+ @- A1 `& G  h: X8 e3 i. y9 R( A; i# |0 v5 r/ v% Z
De Smedt M, Reynvoet K, Kerre T et al. Active form of Notch imposes T cell fate in human progenitor cells. J Immunol 2002;169:3021–3029.
" }) Y. N& ~% ^9 V+ ]
% |' }( V" r6 I$ _1 D& f% U; k+ DZuniga-Pflucker JC, Di J, Lenardo MJ. Requirement for TNF-alpha and IL-1 alpha in fetal thymocyte commitment and differentiation. Science 1995;268:1906–1909.
/ F9 m" _$ q( `) R9 k3 O. j! n# l3 p) E5 J3 o9 a' Y6 ]5 ^" r' f
Weekx SF, Snoeck HW, Offner F et al. Generation of T cells from adult human hematopoietic stem cells and progenitors in a fetal thymic organ culture system: stimulation by tumor necrosis factor-alpha. Blood 2000;95:2806–2812.
7 {( U# s6 a$ l( U# a; y6 {# B) Y9 r8 \* Z( [
Grech AP, Riminton DS, Gabor MJ et al. Increased thymic B cells but maintenance of thymic structure, T cell differentiation and negative selection in lymphotoxin-alpha and TNF gene-targeted mice. Dev Immunol 2000;8:61–74.: I5 t4 d' y# _2 A$ U0 X# T

, K- h: u: p! N0 K% ]8 G" \Crisa L, Cirulli V, Smith KA et al. Human cord blood progenitors sustain thymic T-cell development and a novel form of angiogenesis. Blood 1999;94:3928–3940.
! b+ z! q5 f8 J1 U) Z
* \4 E9 y$ D& Q! X7 C! b) FPannetier C, Even J, Kourilsky P. T-cell repertoire diversity and clonal expansions in normal and clinical samples. Immunol Today 1995;16:176–181.
5 D4 h4 {; {6 F6 H4 T
5 `! w  \7 U% y( M: E. Y/ kArden B, Clark SP, Kabelitz D et al. Human T-cell receptor variable gene segment families. Immunogenetics 1995;42:455–500.
* L1 s. p& k% F; j9 V# [% h! N
' l' B6 n4 R  k  k( \Peled A, Petit I, Kollet O et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science 1999;283:845–848.
: [# J7 a5 [5 X* u/ u2 b! X
- I+ z1 o& F0 qVoermans C, Kooi ML, Rodenhuis S et al. In vitro migratory capacity of CD34  cells is related to hematopoietic recovery after autologous stem cell transplantation. Blood 2001;97:799–804.. x2 q5 x9 ~( M. l5 a

. n9 _$ p8 T" I. h! JBroers AE, Posthumus-van Sluijs SJ, Spits H et al. Interleukin-7 improves T-cell recovery after experimental T-cell-depleted bone marrow transplantation in T-cell-deficient mice by strong expansion of recent thymic emigrants. Blood 2003;102:1534–1540.. l6 b' p' L7 B3 }  D$ s
8 ^4 c1 p* V5 ?# v
Hazenberg MD, Otto SA, de Pauw ES et al. T-cell receptor excision circle and T-cell dynamics after allogeneic stem cell transplantation are related to clinical events. Blood 2002;99:3449–3453.' _! Q3 J. M3 S* a* E3 d+ C

, W5 W& O) f4 K8 l9 M% m9 C/ t9 hTsukasaki K, Miller CW, Kubota T et al. Tumor necrosis factor alpha polymorphism associated with increased susceptibility to development of adult T-cell leukemia/lymphoma in human T-lymphotropic virus type 1 carriers. Cancer Res 2001;61:3770–3774.# q- Y% n+ Z, R8 ]: b

6 N! s  \. k. [* N6 G6 s6 g' qMarino MW, Dunn A, Grail D et al. Characterization of tumor necrosis factor-deficient mice. Proc Natl Acad Sci U S A 1997;94:8093–8098.: W7 R, P$ [/ P* V6 {
; y, t3 B5 P+ Y5 k5 F8 b/ d( h
Ameloot P, Takahashi N, Everaerdt B et al. Bioavailability of recombinant tumor necrosis factor determines its lethality in mice. Eur J Immunol 2002;32:2759–2765.
7 d) W2 @* Z% `3 ^( X6 p. Y! U! M2 X' u+ ]7 A$ _& N
Lewis M, Tartaglia LA, Lee A et al. Cloning and expression of cDNAs for two distinct murine tumor necrosis factor receptors demonstrate one receptor is species specific. Proc Natl Acad Sci U S A 1991;88:2830–2834.  S$ s' ?/ i# E
; ^3 O( h/ r  O( \
Tartaglia LA, Rothe M, Hu YF et al. Tumor necrosis factor’s cytotoxic activity is signaled by the p55 TNF receptor. Cell 1993;73:213–216.+ S4 q. l4 R" k5 e- Z6 \$ `& F9 F

. m. D( O! q  |. \Tartaglia LA, Weber RF, Figari IS et al. The two different receptors for tumor necrosis factor mediate distinct cellular responses. Proc Natl Acad Sci U S A 1991;88:9292–9296.& ^1 w, Y8 G# i+ R7 P7 H9 h

7 O% Z# r& q. R6 P+ BPui JC, Allman D, Xu L et al. Notch1 expression in early lymphopoiesis influences B versus T lineage determination. Immunity 1999;11:299–308./ V* k7 a( V! X( J3 J) Q
1 o5 }) T  @& _* H
Hogan CJ, Shpall EJ, McNiece I et al. Multilineage engraftment in NOD/LtSz-scid/scid mice from mobilized human CD34  peripheral blood progenitor cells. Biol Blood Marrow Transplant 1997;3:236–246." J. N) h5 i& ^3 }
7 P( \2 H4 u& g. g* M7 f+ c
Hess DA, Levac KD, Karanu FN et al. Functional analysis of human hematopoietic repopulating cells mobilized with granulocyte colony-stimulating factor alone versus granulocyte colony-stimulating factor in combination with stem cell factor. Blood 2002;100:869–878.6 J/ t" `) q& w$ X) o

' e4 _. C8 w" C* D) uWang JC, Doedens M, Dick JE. Primitive human hematopoietic cells are enriched in cord blood compared with adult bone marrow or mobilized peripheral blood as measured by the quantitative in vivo SCID-repopulating cell assay. Blood 1997;89:3919–3924.# G% g' d# l( x, p' \5 o9 h* F7 t
; M8 b$ A. V6 K4 l' L5 z7 [6 i4 A2 ?
Garcia S, Dadaglio G, Gougeon ML. Limits of the human-PBL-SCID mice model: severe restriction of the V beta T-cell repertoire of engrafted human T cells. Blood 1997;89:329–336.: Y- P2 G' ^3 @
7 q' }7 m+ G+ s' O1 f) M* m+ w' ?
Tary-Lehmann M, Saxon A, Lehmann PV. The human immune system in hu-PBL-SCID mice. Immunol Today 1995;16:529–533.
! w* m# R+ B7 N: z) `7 P, ^- [
Suzuki K, Oida T, Hamada H et al. Gut cryptopatches: direct evidence of extrathymic anatomical sites for intestinal T lymphopoiesis. Immunity 2000;13:691–702.
) r9 N( l0 z$ O# y' Z3 f6 A) M- e8 w$ p- ^6 m/ p9 b
Rocha B, Vassalli P, Guy-Grand D. Thymic and extrathymic origins of gut intraepithelial lymphocyte populations in mice. J Exp Med 1994;180:681–686.
' T2 @# @1 g! J/ W5 Z. u3 f7 c  J; f$ d9 B1 C3 |" n' n# @/ f
Sato K, Ohtsuka K, Hasegawa K et al. Evidence for extrathymic generation of intermediate T cell receptor cells in the liver revealed in thymectomized, irradiated mice subjected to bone marrow transplantation. J Exp Med 1995;182:759–767.
* J; P1 L. p8 H: o9 ?4 M3 _# r  i* h6 ^# w3 m( n3 n' D
Antica M, Scollay R. Development of T lymphocytes at extrathymic sites. J Immunol 1999;163:206–211.# ?2 o- p) i2 y2 o/ A# h$ R

* L  X7 |0 p  {' s8 |9 H8 V3 dBlais ME, Louis I, Corneau S et al. Extrathymic T-lymphocyte development. Exp Hematol 2003;31:349–354.
# B- A0 `. o" u; t) B) f
' q6 \1 D" z9 b: ]  N+ Q+ UKitchen SG, Korin YD, Roth MD et al. Costimulation of naive CD8  lymphocytes induces CD4 expression and allows human immunodeficiency virus type 1 infection. J Virol 1998;72:9054–9060.7 o+ z) P5 a% J0 p6 e0 U$ @4 V: e
3 O5 ~! Z+ Y. _9 q
Hazenberg MD, Borghans JA, de Boer RJ et al. Thymic output: a bad TREC record. Nat Immunol 2003;4:97–99.(Sarit Samiraa, Christophe)

Rank: 2

积分
79 
威望
79  
包包
1769  
沙发
发表于 2015-5-26 15:10 |只看该作者
胚胎干细胞

Rank: 2

积分
161 
威望
161  
包包
1862  
藤椅
发表于 2015-6-2 10:01 |只看该作者
加油啊!!!!顶哦!!!!!  

Rank: 2

积分
132 
威望
132  
包包
1727  
板凳
发表于 2015-7-10 13:42 |只看该作者
干细胞之家微信公众号
这年头,分不好赚啊  

Rank: 2

积分
116 
威望
116  
包包
1832  
报纸
发表于 2015-9-7 18:49 |只看该作者
哈哈,顶你了哦.  

Rank: 2

积分
77 
威望
77  
包包
1730  
地板
发表于 2015-9-12 03:46 |只看该作者
不错,支持下  

Rank: 2

积分
77 
威望
77  
包包
1730  
7
发表于 2015-9-19 21:47 |只看该作者
正好你开咯这样的帖  

Rank: 2

积分
116 
威望
116  
包包
1832  
8
发表于 2015-10-14 21:27 |只看该作者
我的啦嘿嘿  

Rank: 2

积分
80 
威望
80  
包包
1719  
9
发表于 2015-10-29 08:27 |只看该作者
楼上的话等于没说~~~  

Rank: 2

积分
97 
威望
97  
包包
1738  
10
发表于 2015-11-9 11:01 |只看该作者
神经干细胞
‹ 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, 2024-4-24 23:09

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.