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: 345919|回复: 220
go

Anatomical Compartments Modify the Response of Human Hematopoietic Cells to a Mi [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:08 |只看该作者 |倒序浏览 |打印
作者:Yasuo Nagasawaa, Brent L. Woodb, Linlin Wanga, Ingrid Lintmaera, Wenjin Guoa, Thalia Papayannopouloua, Michael A. Harkeyc, Cynthia Nourigatc, C. Anthony Blaua 3 y; Y) C$ G9 j7 p4 q
                  
) t% w. t5 Q9 M* ]* O, o                  1 F  w+ A7 [5 I4 [- ^3 t
         
1 L# e+ ?' F; |. X% i% V) I! O                         & w3 n2 R+ r/ x( V
            & f4 b" f/ E3 c- W. W: x
            & G8 {* Y$ ]. m$ p' l# m
            
! _6 \) s, [* ^! O7 a- ~8 e6 h+ ?            
; ]- A! U" H; `! E( D                     
: @9 O- q6 R# ~5 u        
; m$ f; p1 Z* y# L' |, @1 n9 p* B5 X        
+ q6 l+ S$ c$ i7 s        / r/ Q) L- }6 S# ~7 g% Y" \8 l# c% R6 \
          【摘要】3 a1 m3 b" `0 B9 Z! l3 I' w8 D7 {
      Methods for specifically regulating transplanted cells have many applications in gene and cell therapy. We examined the response of human cord blood CD34  cells to a specific mitotic signal in vivo. Using a conditional signaling molecule (F36VMpl) that is specifically activated by an artificial ligand called a chemical inducer of dimerization (CID), human hematopoietic cells transplanted into immune deficient mice were induced to proliferate. Only differentiating erythroid precursors and multipotential and erythroid progenitors (colony-forming unit ) responded; however, the nature of the response differed markedly between bone marrow and spleen. In the marrow, F36VMpl induced a 12- to 17-fold expansion of differentiated erythroid precursors and a loss of CFU-mix and BFUe. In the spleen, F36VMpl induced a marked rise in BFUe and CFU-mix and, relative to marrow, a much less prominent rise in more mature red cells. Clonal analysis was most consistent with the interpretation that the spleen and bone marrow differentially regulate the response of human progenitors to a mitotic signal, possibly influencing progenitor expansion versus differentiation. These findings establish CIDs as in vivo growth factors for human hematopoietic cells. " a& b$ R) @: z' d+ a2 W
          【关键词】 Gene therapy In vivo selection Regulation Hematopoiesis
# d5 G! z+ ?  a                  INTRODUCTION8 T* k' F, D. i

2 F5 c9 ?# _" Q8 jA challenge for gene and cell therapy is to develop safe and effective methods for controlling transplanted cells, allowing their number or function to be titrated to a desired therapeutic endpoint. One method for controlling cells uses conditional signaling molecules that can, upon activation, elicit death, proliferation, differentiation, or other desired functions .
0 a% ~' z5 d0 v4 y8 ~. T2 P$ m4 a5 N" J" J
Methods for regulating cell proliferation most commonly rely on growth factor receptors, modified to substitute their binding site for endogenous ligand with the binding site for a CID. Insensate to endogenous ligand, these modified receptors are specifically activated by the CID, which acts as a growth factor for genetically modified cells. Studies of CID-regulated in vivo selection have identified the signaling domain of the thrombopoietin receptor, mpl, as a potent inducer of CID-dependent hematopoietic cell growth. Results from three different animal species (mice, dogs, and baboons) have been reported  (C.A. Blau, unpublished results). These findings indicate that environmental signals that are absent in cell culture can significantly modulate the response to F36VMpl signaling in vivo. Therefore, prior to a clinical trial, it would be useful to evaluate the response of human hematopoietic cells to F36VMpl signaling in vivo.( p$ p2 m* Y  q1 K( h" b  ]
7 B: ?  S6 W  D0 m$ \+ ~
MATERIALS AND METHODS
7 |/ J% E) r8 V6 j" H  Z
- @' r& [5 u8 I) NConstruction of Lentiviral Transfer Vectors and Preparation of Viral Vector Stocks8 I- ~0 n  j- s- l, ^0 w5 g

- a0 q5 Z$ I$ C! |6 @- v( eThe lentivirus vector encoding F36VMpl was assembled using a lentivirus construct pLHGAFMIG (W. Guo and C.A. Blau, unpublished results; details available upon request), and a construct containing hPGK-GFP (kindly provided by L. Naldini, Universit¨¤ Vita-Salute School of Medicine, Milan, Italy) . Titer, determined in HT1080 cells, was 4¨C9 x 108 IU/ml.) d0 z8 x$ C2 Q$ U) y9 i
) l2 g4 U" Z# D
Cells* o1 M/ z5 E$ @: T2 t' [
: F7 R  B4 F$ G4 Q; @
Umbilical cord blood was collected under an Institutional Review Board-approved protocol from the obstetrical unit of the University of Washington Medical Center. CD34  cell selection and cryopreservation were performed as described previously.
: i; v, l5 h5 z- C" z0 ?# B
# P( g" d1 Y: [  [* n2 B$ c: XMice
5 A) s- l; d3 b' J  w$ }& y' U1 u% J0 {! y
NOD/SCID/ß2Mnull mice , provided by the Core Center of Excellence in Molecular Hematology at the Fred Hutchinson Cancer Research Center, were bred in a specific pathogen-free facility. All procedures were performed according to protocols approved by the Animal Care and Use Committees of the University of Washington and the Fred Hutchinson Cancer Research Center.
1 C, l  K4 ]1 H* k+ i" P' T1 F6 j9 N" D2 [% |( H
Gene Transfer and Transplantation3 ?2 q9 J/ [* D3 Q
2 p: U7 V. d% q  y/ i
Transductions were performed essentially as described  interleukin 6, 100 ng/ml rh stem cell factor, 100 ng/ml rh FLT-3 ligand, and 20 ng/ml rh thrombopoietin; Peprotech, Rocky Hill, NJ, http://www.peprotech.com) and vector at a concentration of 5 x 105 cells/ml (multiplicity of infection: 10) for 20 hours in a 37¡ãC, 5% CO2 incubator. Following transduction, cells were injected via the tail vein into sublethally irradiated (3.5 Gy) NOD/SCIDß2Mnull mice at a cell dose of 2 x 105 cells per mouse. To determine the efficiency of gene transfer, transduced cells were washed with IMDM and plated in IMDM with 10% fetal bovine serum (FBS) with 20 ng/ml rhIL3, 20 ng/ml rhIL6, and 100 ng/ml recombinant human stem cell factor (rhSCF). After 5 days, the percentage of GFP  cells was assessed by flow cytometry.
7 q9 }& r7 f6 ^/ b
- r& c1 j6 _" d( jEx Vivo Expansion4 V/ Q4 M9 ^2 G! p9 |# T- E

9 ~; o) C6 F$ k7 ZFollowing lentiviral transduction, CD34  cord blood cells were cultured in IMDM containing 10% FBS, 100 U/ml penicillin, and 100 µg/ml streptomycin either in the presence or in the absence of AP20187 (100 nM; ARIAD Pharmaceuticals, Inc., Cambridge, MA, www.ariad.com/regulationkits) in a 37¡ãC, 5% CO2 incubator.
; j. ^+ [/ J& ?1 ~; r- d4 m/ @8 Q3 N
CID Administration
4 c/ j, I# i* S" W; M3 i: R; L; r4 O" r+ Z2 e" b- @5 P' b: n+ \  z
Lyophilized AP20187 was solubilized in 100% ethanol to produce a 5 mg/ml stock that was stored at ¨C20¡ãC. AP20187 was diluted fresh on the day of injection in sterile water containing 10% PEG 400 (Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com) and 1.44% Tween 80 (Sigma-Aldrich). AP20187 was administered via daily intraperitoneal injection. (0.2 mg/mouse, approximately 10 mg/kg).
0 ]8 J) D2 h- @' T9 X3 B
% b3 J0 U- B. W0 U1 e2 _Flow Cytometry6 |% @$ c  @) ~/ T- b! n

- D" [6 T7 r& {4 M  xTransduced cells were washed and resuspended in phosphate-buffered saline containing 1% FBS. The presence of human cells in mouse tissues was determined using flow cytometric analysis of cells harvested from the bone marrow, spleen, and blood after labeling with monoclonal antibodies against human CD45, CD71, CD33, CD19, CD34, and CD41a (Becton, Dickinson and Company, Franklin Lakes, NJ, http://www.bd.com) and analyzed on a Becton Dickinson LSRII. Syto41 (Molecular Probes Inc., Eugene, OR, http://probes.invitrogen.com) was used to identify nucleated cells. Nucleated erythroid cells were defined as positive for syto41 and bright CD71 without CD45. Myelomonocytic cells were defined as positive for CD45 and CD33, and B cells were positive for CD45 and CD19 without CD33.
* m$ z( }' {! \, N1 F
/ {' D- s& H4 s6 L; b; x+ }. RColony Assays' J% U- ?& q% l0 p) o' j
& H% }/ t4 k. K8 E! c2 f" H
Bone marrow and spleen cells were plated in methylcellulose medium (Stem Cell Technologies, Vancouver, BC, Canada, http://www.stemcell.com) containing 0.92% methylcellulose in IMDM with 30% FBS, 2 mM L-glutamine, 10¨C4 M 2-mercaptoethanol, and 1% bovine serum albumin with 3 U/ml rh erythropoietin (Amgen, Thousand Oaks, CA, http://www.amgen.com), 10 ng/ml rh-granulocyte macrophage colony-stimulating factor, 10 ng/ml rhIL3, and 50 ng/ml rhSCF (Peprotech), at concentrations of either 5 x 104 marrow cells or 1 x 106 spleen cells per ml. Cultures were plated in triplicate and incubated in a highly humidified 37¡ãC, 5% CO2 incubator. Colonies were scored by light and fluorescence microscopy 14 days later (MZ16FA; Leica, Heerbrugg, Switzerland, http://www.leica.com). The GFP plus filter set contained a 480/40-nm exciter filter and a 510 LP barrier filter.
' l" G3 v8 E! W! g, U0 Z  Z' h
2 y) u6 J; [$ H  cSingle Colony Linear Amplification-Mediated Polymerase Chain Reaction6 A; X. ]! B3 C5 \6 _

7 b3 [7 I3 ?( G3 ]' xColony DNA was prepared by transferring a colony, in minimal volume, from the methylcellulose matrix to 100 µl of lysis buffer containing 10 mM Tris, pH 7.5, 0.1% Triton X-100, and 500 µg/ml Proteinase K. The lysate was digested at 55¡ãC for 2 hours and heat-denatured at 99¡ãC for 10 minutes. Genomic DNA was purified from sorted and unsorted bone marrow and spleen cells using a QiaAmp DNA Blood Mini Kit (Qiagen, Valencia, CA, http://www1.qiagen.com).
4 L  h% ?0 h! N8 }
; y. {$ m) c0 k% D- i9 x7 g* eInsertion site analysis was performed using a three-arm modification of linear amplification-mediated polymerase chain reaction (LAM-PCR) (M.A. Harkey and C.A. Blau, manuscript submitted). Reactions were conducted with either 10 µl of colony DNA (100¨C1,000 cells), or 50¨C100 ng of column-purified DNA. All amplifications were done with Advantage II Polymerase Mix and buffer system (BD Biosciences, San Diego, http://www.bdbiosciences.com) using the following cycling conditions: melt at 95¡ãC for 20 s, anneal at 60¡ãC for 45 s, elongate at 68¡ãC for 90 s. First, single-stranded copies of the viral 3'LTR-host junction region were made by 100 cycles of linear amplification, using a biotinylated LTR primer (biotin-5'AAGCCTCAATAAAGCTTGCC). The product was bound to streptavidin-coated magnetic M280 Dynabeads (Dynal Biotech, Oslo, Norway, http://www.invitrogen.com/dynal). Subsequent reactions and washes (3 x 200 µl after every reaction) took place on this matrix. Matrix-bound DNA was resuspended in 20 µl of random-priming reaction containing 500 nM dNTPs, 100 ng/µl random hexamers, 2¨C5 units Klenow enzyme (New England Biolabs, Beverly, MA, http://www.neb.com), and manufacturer-supplied buffer at 1x and incubated at 37¡ãC for 60 minutes. Products were divided into three equal arms, and digested with Tsp509I, HaeIII, or RsaI (New England Biolabs). DNA that remained bound to the matrix was then ligated to a double-stranded anchor primer using Fast Link Ligase (Epicenter, Biotechnologies, Madison, WI, http://www.epibio.com). For the Tsp509I arm, component primers GACCCGGGAGATCTGAATTCAGTGGCACAGCAGTTAGG and AATTCCTAACTGCTGTGCCACTGAATTCAGATC were used, yielding a Tsp509I-compatible end. For the HaeIII and RsaI arms, a blunt-ended anchor primer was used, with CCTAACTGCTGTGCCACTGAATTCAGATC as the second component. The double-stranded junction fragments, containing host DNA flanked by LTR and anchor-primer (AP) sequences, were eluted from the matrix in 5 µl of 0.1 N NaOH and amplified by two rounds of nested PCR. Two microliters of the eluate was amplified in a 50-µl PCR with LTR primer 1 (TGAGTGCTTCAAGTAGTGTGTGC) and AP 1 (GACCCGGGAGATCTGAATTC). This reaction was diluted 100-fold in water, and 2 µl of template was amplified by 30 cycles of nested PCR, using LTR primer 2 (ACTCTGGTAACTAGAGATCC) and AP 2 (GATCTGAATTCAGTGGCACAG). The LAM-PCR products were separated by electrophoresis on Novex 4¨C20% polyacrylamide TBEgels (Invitrogen, Carlsbad, CA, http://www.invitrogen.com), stained with ethidium bromide, and visualized by ultraviolet light fluorescence.
9 X* Z! A  q* B  R' O  N3 _$ `
6 f# S, y2 o) H9 G- jClonal Tracking by Clone-Specific PCR/ z& y, t1 X$ {' v# H6 t

! q, F+ e7 h, P! rIn order to track specific lentivirus-containing clones, PCR primers were designed to amplify the viral host junction of the inserted provirus. LAM-PCR products from spleen colonies were separated on 3% agarose gels, and individual bands excised and cloned into the TOPO-TA vector, pCR4 (Invitrogen). The cloned inserts were sequenced from flanking M13-F and M13-R primer sites in the vector using Big Dye fluorescent dideoxynucleotide chemistry (Amersham Biosciences, Piscataway, NJ, http://www.amersham.com) and an ABI Prism 310 Genetic Analyzer (Applied BioSystems, Foster City, CA, http://www.appliedbiosystems.com). Sequences were screened for the presence of LTR and anchor-primer motifs, and the intervening sequence was compared to the human genome using the University of California-Santa Cruz BLAT service (http://genome.ucsc.edu/cgi-bin/hgBlat). Primers were designed from the host sequence to amplify the host-LTR junction in combination with our LTR primers. The insertion site for clone 7 is human chromosome 2: 68,683,670. The insertion-site¨Cspecific tracking primers are ATGTTATTATGATAATATGTAAAGGG and TATAATTTCTAATAGGGTAAATATCC, corresponding to chromosome 2: 68,683,306 ¨C 68,683,329, and 68,683,381¨C68,683,406, respectively. The insertion site for clone 9 is human chromosome 17: 31,324,273. The insertion site-specific tracking primers are CCTGAGTTACTGGGACTATAGG and AAGCACATGTATGCTCTCATGGG, corresponding to chromosome 17: 31,323,801¨C31,323,822 and 31,323,936¨C31,323,958, respectively. The presence of clones 7 and 9 in colony or bulk DNAs was determined by nested PCR (with the cycling conditions described above) using LTR primers 1 and 2 and the appropriate clone-specific primers. The nested priming approach was necessary to eliminate nonspecific amplification observed with single primer sets. Round 1 amplification was performed at 15 cycles for the clone 9 insertion and 30 cycles for clone 7. Round 2 amplification was performed at 25, 30, 35, and 40 cycles for both clones. A parallel PCR for an intron of the single-copy mouse GAB2 gene was used to confirm the presence of host DNA. The GAB2 primers were GAGACAGTGAGGAGAACTATGTCC and CACACTTGGTAATGCACAGCTTCC. Cycling conditions for GAB2 were as follows: melt at 94¡ãC for 10 seconds, anneal/elongate at 68¡ãC for 1 minute.
: F) H- c# N& x3 ]* @  N5 t
- q! y  h0 E% gSplenectomies
. k$ t/ [0 E; V& l2 x0 `8 {( E2 `5 _- {, Y
NOD/SCID mice were anesthetized with a 500-µl intraperitoneal injection of Avertin made by dissolving 2.5 g of 2,2,2-tribromoethanol in 5 ml of 2-methyl-2-butanol (both Sigma-Aldrich), then adding distilled water to a final volume of 200 ml, and then sterile filtering. The mouse was placed in a supine position, the abdomen was shaved and disinfected, and a 2-cm incision was made just below the left costal margin to expose the spleen. The spleen was exteriorized and excised with blunt forceps and scissors. The wound was then closed with Super Glue.% w( S/ ^0 d5 C7 S+ D8 V- X3 E
9 U9 K9 \6 _0 v! C
Statistical Analyses# l1 Y  N& i8 F8 C' n$ a1 U" o

* b3 G4 m9 v8 [3 X  ]1 h# zAll comparisons were made using Student¡¯s t test.7 G$ [5 @* c/ C

# W! v1 L$ w: c; G6 c9 k  KGlobin Immunofluoresence
1 ~# `5 L+ b" P; k. B
3 B/ t( }0 d* i1 H0 @2 @0 ^Sorted CD71  cells from mice transplanted with F36VMpl-transduced cord blood CD34  cells and treated with a 2-week course of CID were resuspended in fetal calf serum and used to produce cytocentrifuge smears. Air-dried slides were methanol-fixed, washed, and incubated with an anti- monoclonal antibody  in a humidified chamber at 37¡ãC for 1 h. Slides were washed and developed by incubating with a fluorescein isothiocyanate-conjugated goat anti-mouse secondary antibody. The slides were subsequently examined by fluorescence microscopy.9 m1 ?' {1 L) G( e/ B. [' b

1 O: c  }, J. |; Z5 E$ z/ FRESULTS
7 v; v- y! m* X+ V1 ?$ D& }
' G5 A4 _! \3 M3 o5 h& yLentivirus-Mediated Gene Transfer Confers CID Responsiveness In Vitro+ ?) m+ `/ h8 g2 c2 J2 q5 s

( `4 q- L( w! h- VTo improve gene transfer into cells capable of repopulating immune-deficient mice .
& @2 x5 T. V; g" Z& y+ e0 f0 H! N& c, B% |  k6 X) ]) T/ V- v* R
Figure 1. F36VMpl-mediated selection of transduced human erythroid cells. (A): Cord blood CD34  cells were transduced with the F36VMpl lentivirus vector and cultured for 14 days in the presence or absence of AP20187 (100 nM). Numbers of total cells (closed circles, x10¨C4), GFP  burst forming units-erythroid (triangles, x 10¨C1), and GFP  CFU-mix (diamonds, x10¨C) in the presence of AP20187 and total cells in the absence of AP20187 (open circles) are depicted over time. (B¨CI): representative experiment showing that the F36VMpl response is restricted to human erythroid cells. (B¨CF): The percentages presented are relative to all nucleated cells in the femur. (B): Mean percentages of human non-erythroid (CD45 ) and erythroid (CD71 ) cells relative to total nucleated cells in the femurs of chemical inducer of dimerization (CID)-treated ( ) and non-CID-treated (¨C) mice either 2 weeks or 3 weeks post-transplant. Mice evaluated at week 2 post-transplant had received CID for 7 days, whereas mice evaluated at week 3 post-transplant had received CID for 14 days. The femurs of CID-treated mice ( ) contained a significantly higher percentage of human hemopoietic cells at week 3 compared to non-CID-treated controls (¨C) due to a preferential expansion of CD71  erythroid cells (p = 0.04). This expansion in CD71  cells was entirely attributable to an increase in erythroid cells expressing GFP (C) (p =.04). Responses were not observed in myeloid (CD33 ) (D) or B lymphoid (CD19 ) lineages (E) or among CD34  cells (F). Week 2 results reflect mean values from four CID-treated ( ) and four non-CID-treated (¨C) mice; week 3 results indicate mean values from four CID-treated ( ) and five non-CID-treated (¨C) mice. Error bars (B¨CF) denote standard errors of mean. p values were obtained using a t test. Results at week 3 were confirmed in three independent experiments. (G): Wright Giemsa stain of GFP  CD71  cells sorted from the marrow of a CID-treated mouse at week 3 (1,000x). (H): Fluorescent histograms of human erythroid cells from four CID-treated NOD-SCIDß2mnull mice (blue lines, right) and three untreated controls (red lines, left). The mean fluorescent intensity of the GFP  population is more than two decades higher in CID-treated mice compared to controls. (I): Staining of a marrow section (top, x100 objective; bottom, x400) from a CID-treated mouse evaluated at week 3 shows localization of GFP expression in an island of normoblasts (i) and absence of GFP expression in a cluster of myeloid cells (ii). Staining was performed using a rabbit polyclonal antibody directed against GFP and a secondary antibody conjugated to horseradish peroxidase. Brownish stain indicates GFP positivity. (J): Immunostaining with an anti- globin antibody demonstrates that erythroid cells responding to CID express fetal hemoglobin. Green fluorescence denotes the presence of  globin. Abbreviation: GFP, green fluorescent protein.  a7 k! A4 w" V7 \% C) \
8 J1 g8 i  V6 I
In Vivo Selection of Transduced Human Erythroid Cells
& l: W. y' r! j6 P& c6 m; r' `# d1 A8 z& W" e+ f1 p
NOD/SCID/ß2 microglobulin-null (NOD/SCID/ß2Mnull) mice have been used to identify short-term reconstituting SCID-repopulating cells (SRCs), which contribute to blood cell production within the first 3 months post-transplantation, and long-term¨Creconstituting SRCs, which engraft beyond 6¨C8 weeks post-transplantation . Short-term reconstituting SRCs predominantly contribute to erythroid cell production at week 3 post-transplantation, whereas by week 6, human red cell production virtually ceases, replaced by a predominance of human-origin B cells.5 v) K8 U# D. Y6 B+ z

1 I6 ^. H+ H& ~# l6 _An initial study using transduced CD34  cells confirmed previous reports showing that human erythroid engraftment declines precipitously beyond week 3 post-transplantation (data not shown) , making it necessary to evaluate the effects of CID treatment within this time frame. Mice transplanted with transduced CD34  cells were treated, beginning 1 week later, with daily injections of AP20187. CID-treated and control vehicle-treated mice were evaluated after 7 or 14 days of AP20187 (or control vehicle), 2¨C3 weeks post-transplant, respectively. Although no effect of CID exposure was evident after 7 days (week 2), the 14-day course of AP20187 (week 3) produced a significant response. The fraction of human erythroid (CD71 ) cells relative to all nucleated cells in the femurs of CID-treated mice was 24.0%, compared to 6.6% in non-CID-treated controls, a 3.6-fold increase (Fig. 1B) (p ; m3 U6 J1 b4 T3 @5 }/ x# ?% J1 i

9 F% x: X, w# i) ICID Treatment Induces the Emergence of BFUe and CFU-Mix in the Spleen# d: ]2 }) i7 i1 U: S# i$ v

4 M0 O5 S+ e% M5 XContrasting the expansion of morphologically identifiable erythroid cells in the marrow at week 3, marrow progenitors fell (Fig. 2A; experiments not shown), mirroring the cell culture results (Fig. 1A). In contrast, GFP  BFUe and CFU-mix were consistently higher in the spleens of CID-treated mice compared with those of controls (Fig. 2A). The effect of CID treatment was especially dramatic among BFUe and CFU-mix capable of colony formation in the presence of erythropoietin (epo) alone (Fig. 2A). Epo-dependent BFUe or CFU-mix was never observed in the spleens of non-CID-treated mice. The vast majority of epo-dependent BFUe and CFU-mix were intensely GFP positive (96% in each of two experiments). The strict correlation between CID exposure and the appearance of epo-responsive BFUe and CFU-mix in the spleen defines these colony-formers as specific indicators of the CID response.* D: k6 l( L' |! k1 B" D

3 x. L2 t/ }8 E) R9 _; N" oFigure 2. Differential effects of F36VMpl signaling on progenitors in the femur and spleen. Unfractionated mononuclear cells from the femurs and spleens of CID-treated ( ) and non-CID-treated (¨C) mice were plated, and GFP-positive colonies were scored. Mice were analyzed 3 weeks post-transplant, following a 14-day course of AP20187 (or vehicle control). Photomicrographs show representative morphologies of GFP  BFUe (top) and CFU-mix (bottom). Note spherical morphology of BFUe. (A): Total numbers of GFP  BFUe and CFU-mix per femur in four CID-treated ( ) and four non-CID-treated (¨C) mice. Results reflect mean values. Error bars depict standard errors of mean. Colony assays were performed in IL-3, SCF, GM-CSF, and erythropoietin (Epo) or Epo alone. Results using Epo, GM, IL-3, and SCF were confirmed in three independent experiments. Results using Epo alone were confirmed in two independent experiments. Results from only a single representative experiment are shown. (B): Linear amplification-mediated polymerase chain reaction (LAM-PCR) profiles of three epo-responsive spleen colonies (colonies 1¨C3) in each of four CID-treated mice. LAM-PCR was performed using three different restriction enzymes, Tsp509I (top panel), HaeIII (middle panel), and RsaI (bottom panel) as described in Materials and Methods. Arrows designate internal control bands (see Materials and Methods). Colonies 2 and 3 from mouse 10 show an identical pattern with all three enzymes, indicating common ancestry. Numbers in each lane show the estimate number of transgene insertions based on results from all three enzymes. Abbreviations: BM, unfractionated bone marrow cells from the same mice; Epo, erythropoietin; GM-CSF, granulocyte macrophage colony-stimulating factor; IL-3, interleukin 3; MW, molecular weight; SCF, stem cell factor.' v* b( g6 t, u; p
4 W- u2 D% H: ~2 l+ g
Multiple Clones Contribute to the CID Response
  h* P$ u: D& g! x) B2 v" V( t' S6 U" N4 X, u: L, M! Q
The causal link between CID treatment and the appearance of epo-responsive BFUe and CFU-mix in the spleen provided an opportunity to examine the clonality of the progenitor response. Provirus insertion site analysis of individual epo-responsive BFUe or CFU-mix colonies was performed using LAM-PCR with the restriction enzyme Tsp509I. Twelve epo-responsive spleen colonies (BFUe or CFU-mix) from four CID-treated mice suggested 10 distinct provirus insertion patterns (Fig. 2B). To confirm these findings, independent LAM-PCRs were performed using two other restriction enzymes (HaeIII and RsaI), which revealed that one pair of colonies sharing a similar LAM-PCR pattern with Tsp509I digestion were in fact distinct clones. LAM-PCR patterns suggested that colonies emerging in response to CID contained 1¨C4 provirus copies.) u8 q' ~2 T0 s( G9 ^! u6 B+ e3 r
2 }' E' l/ v  i
More extensive clonal tracking was performed in a single CID-treated mouse (Fig. 3). A total of 55 spleen-derived colonies were examined, 24 from cultures containing IL-3, SCF, GM-CSF, and epo and 31 from cultures containing epo alone. Of 55 total colonies, 36 yielded evaluable LAM-PCR patterns.
% o4 c9 B: `, y3 J2 c6 e; h: Z5 C) b
' R3 z6 k/ ~* v$ n8 u* FFigure 3. Clonal expansion of progenitors in the spleen and apparent lack of relationship to the marrow red cell response. (A): Schematic depiction of clonal relationships among CID responsive progenitors in the spleen. Linker amplification-mediated-polymerase chain reaction (LAM-PCR) using Tsp509I was performed on 55 colonies from the spleen of one CID-treated mouse (Fig. 1, week 3) that was evaluated 21 days post-transplantation, immediately after completing a 14-day course of CID. Results show evaluable insertion patterns shared by two or more colonies. Red circles depict burst-forming units-erythroid (BFUe). Purple circles depict CFU-mix. Circles enclosed with solid lines portray progenitors identified in this experiment. Circles enclosed with dashed lines depict parental colony forming cells whose existence was not demonstrated, but is inferred. Top, LAM-PCR patterns shared by either three (clone 12) or two (clones 5 and 7) CFU-mix. Middle, LAM-PCR patterns shared by BFUe and CFU-mix. Three colonies derived from clone 9 were identified in cultures containing erythropoietin (epo) only (solid downward arrows), and two colonies were identified from cultures containing IL-3, GM-CSF, SCF, and epo (dashed arrows pointing to right). Bottom: LAM-PCR pattern shared by 2 BFUe. The inferred colony could be either a CFU-mix or a BFUe. (B): Tracking of insertions 9 (top panel) and 7 (middle panel) in selected colonies from the same mouse depicted in A and in sorted GFP  erythroid (CD71 ) and non-erythroid (CD71¨C) cells in the marrow. Numbers indicate the assigned LAM-PCR pattern from A and data not shown. X, colony that yielded a nonevaluable pattern by LAM-PCR. Bottom panel: GAB2 control primers show abundant DNA in wells from sorted marrow cells. (C): The spleen is not required for the CID-induced expansion of transduced human erythroid cells. Left panel: F36VMpl-transduced human CD34  cells were transplanted into splenectomized or control NOD-SCID mice, followed 1 week later by a 5-day course of either CID ( ) or vehicle only (¨C). The duration of CID treatment in this experiment was curtailed due to excessive deaths in both CID and vehicle-treated mice in the splenectomized group. After 9 days (3 weeks post-transplant), mice were sacrificed, and the frequency of GFP-positive cells among the CD71  population was compared between the two groups. Responses obtained following the 5-day course of CID were similar to those of nonsplenectomized NOD-SCID mice treated with a 13-day course of CID (right panel). Each column represents mean values from three mice. Error bars depict standard errors of mean. Abbreviation: GFP, green fluorescent protein.3 j1 c6 C7 P, g' V, \4 |9 r
' }7 Y8 S# A" u# T$ V( B  _- T
Since the epo-dependent colonies were only seen in the spleens of CID-treated mice (Fig. 2A), they are a direct consequence of the CID response. Of the 31 colonies from epo-only cultures, 25 yielded evaluable LAM-PCR patterns. Six were CFU-GMs, and none of these shared LAM-PCR patterns with BFUe. The remaining 19 BFUe and CFU-mix contained 11 different insertion patterns. Five colonies (four BFUe and one CFU-mix) contained unique patterns. Of the remaining 14 colonies (Fig. 3A), four pairs contained common insertions (one BFUe/BFUe, two CFU-mix/CFU-mix, and one BFUe/CFU-mix), and two triplets contained shared insertions (one BFUe/BFUe/CFU-mix and one CFU-mix/CFU-mix/CFU-mix). Additional progeny from clone 9 were identified in a BFUe and a CFU-mix colony obtained from cultures containing multiple growth factors. These findings demonstrate that F36VMpl activation promotes a clonal in vivo expansion of BFUe and CFU-mix in the spleen." X# ~. d4 X5 Z

) c! c$ [- W5 L5 X+ X4 R  D  |5 PDistinct Ancestry of CID-Responsive Erythroid Cells in the Marrow and CID-Responsive BFUe and CFU-Mix in the Spleen
8 E7 x- y& D- \% a* x7 e8 }
* l* K# p) a& T- p2 ATo gain further insight into a possible ancestral relationship between CFU-mix and BFUe generated in the spleens of CID-treated mice and differentiating erythroid cells in their marrows, we compared LAM-PCR banding patterns. A shared LAM-PCR pattern between spleen progenitors and differentiating marrow erythroid cells would have two potential explanations. First, BFUe and CFU-mix in the spleen might give rise to differentiated red cell progeny that migrate to the marrow. Second, if BFUe and CFU-mix appearing in the spleens of CID-treated mice had originated in the marrow, then traces of their ancestors or progeny may have been left behind.
5 p3 {% ^' @% c# `% _$ L- k6 Q9 T
& c6 d  m. a. S6 IWe first looked in the bone marrows of the four mice depicted in Figure 2B. Although no shared LAM-PCR patterns were observed, marrow mononuclear cells (Fig. 2B, lanes labeled BM) gave rise to much less distinct patterns compared with the single colonies, consistent with the inability of LAM-PCR to accurately reflect the contributions of individual clones within a complex mixture (M.A. Harkey and C.A. Blau, manuscript submitted). To more accurately establish whether a progenitor/progeny relationship existed between CID-responsive progenitors in the spleen and erythroblasts in the marrow, we sequenced insertions from two clones (clones 7 and 9) from the mouse examined in Figure 3A. Specific primers to detect these insertions were used both to verify the accuracy of the LAM-PCR-designated clonal assignments and to determine whether clones 7 and 9 contributed to red cell progeny in the marrow of the same mouse. PCR products of the predicted size were detected in all colonies previously assigned to clones 7 and 9 (Fig. 3B). Clone 9 primers also detected minor bands of appropriate size in colonies previously assigned to clones 5 and 11; however, these low-intensity bands were likely the result of cross contamination during the colony picking procedure. Neither clone 7 nor clone 9 insertions were detectable in sorted GFP  CD71  cells from the marrow of the same mouse. These findings strongly suggest the absence of a clonal relationship between erythroid and mixed progenitors in the spleens and erythroblasts in the marrows of CID-treated mice. Unexpectedly, a faint band of the appropriate size was obtained from sorted nonerythroid (GFP  CD71¨C) cells using the clone 7-specific primers (Fig. 3B). Since both of the clone 7 progenitors identified in the spleen were CFU-mix (Fig. 3A), it is possible that nonerythroid progeny of these progenitors were able to escape the spleen and migrate to the marrow, whereas the erythroid progeny appeared not to traffic from the spleen to the marrow.
5 n% n, z  t. x# N0 d- _2 U! R2 D. L* b. D/ N
The Spleen Is Dispensable for the CID Response3 z$ }6 Y$ {' z3 T! T

% p( r3 G7 r5 tTo confirm that the CID-induced appearance of BFUe and CFU-mix in the spleen is not required for the emergence of differentiated erythroid cells in the marrow, we evaluated splenectomized mice. Due to the frailty of NOD-SCIDß2null mice, we tested the effect of splenectomy on the F36VMpl response in NOD-SCID mice. Splenectomized mice tolerated the transplantation procedure much less well than their nonsplenectomized counterparts. Among 20 splenectomized mice, 10 mice (5 in each group) died between 9 and 16 days post-transplant. By comparison, 18 of 20 nonsplenectomized mice survived, with both deaths occurring among control-injected animals. To minimize further loss of mice, injections were curtailed to 5 days. Both splenectomized and nonsplenectomized mice were analyzed 3 weeks post-transplant, 9 days after completing CID or control injections. To test whether the shorted duration of CID treatment might reduce the magnitude of the response, a cohort of mice was treated with a 13-day course of CID. The percentage of CD71  cells that expressed GFP trended higher in the marrows of CID-treated mice, and was not diminished by splenectomy (Fig. 3C). Among nonsplenectomized animals, responses to the 5- and 13-day courses of CID treatment were similar (Fig. 3C, right).3 n$ J. b- c4 A: `/ |

4 U; k* m2 S/ n. w0 S8 }3 VEvaluating the Persistence of the Response to F36VMpl Signaling
1 P/ a9 A" H7 b2 A3 Z9 t/ z0 n) P2 d- C  ?; ?4 U4 h, _
Mice treated with CID between weeks 1 and 3 showed a dramatic decline in GFP  erythroid cells by week 8, falling from up to 50% of all nucleated cells in the femur at week 3 to 5 H4 V( a' M9 I0 s
' E4 F$ I' \3 [0 |' K
Figure 4. Persistence of the F36VMpl response. (A): GFP 71  cells as a percentage of total mononuclear cells in the femurs of CID-treated ( ) and non-CID-treated (¨C) mice at week 3 and in CID-treated mice at week 8. Despite a marked expansion of GFP  erythroid cells in CID-treated mice at week 3, these cells are no longer evident in the marrow by week 8. (B¨CD): Persistence of CID effects at week 8. A 14-day course of CID ( ) or vehicle (¨C) had been given between weeks 1 and 3. (B): The percentage of CD71  cells expressing GFP at week 8 post-transplantation was higher in the femurs (white bars) but not in the spleens (gray bars) of mice that had received CID. (C): Erythroid and mixed progenitors capable of forming colonies in the presence of epo alone remained higher in the spleens of mice that had received CID. Hatched boxes indicate GFP  colonies. (D): The percentage of GFP  BFUe was higher in the spleens of mice that received a 14-day course of CID between weeks 1 and 3 post-transplant (¡À) or a 3-day course of CID given at week 7 (¨C/ ) compared to controls (¨C/¨C). Differences in the percentages of GFP-positive BFUe in the spleens of CID-treated mice in either the  /¨C or the ¨C/  groups are significantly different from the ¨C/¨C group. (E): From the same experiment depicted D, CID treatment either between weeks 1 and 3 or at week 7 had no effect on the percentage of GFP  CD19  B cells at week 8. (D, E): Error bars denote standard deviations for  /¨C group and show individual values for ¨C/¨C and ¨C/  groups. Numbers indicate mice per group. Abbreviations: Epo, erythropoietin; GFP, green fluorescent protein.
" }0 a0 |! O7 N1 ]& ?/ Q: e
8 X# @4 C8 ?' f; m* _6 `, f9 {* z" vErythroid Specificity of the F36VMpl Response% }6 |. k7 G$ d

2 ^* D0 Q  O, J1 B4 E% VIn view of our previous finding that B cells respond prominently to F36VMpl signaling in a dog model , we re-examined human B cells at 8 weeks post-transplant. Three mice that had received a two-week course of CID between weeks 1 and 3 were compared to two mice that received a single 3-day course of CID on week 7 and to two mice that had not received CID. Both groups of CID-treated mice exhibited a significantly higher fraction of GFP  BFUe in their spleens at week 8 compared to controls (Fig. 4D), whereas the fraction of GFP  B cells was indistinguishable between the CID-treated mice and controls (Fig. 4E). The absence of an effect of CID treatment between weeks 1 and 3 on transduced B cells at week 8 was confirmed in a second independent experiment (not shown).6 s2 m% l* k( Y6 d

& @1 O: I. Y1 n9 |7 ?" I9 w0 jDISCUSSION
* V8 x% ?) R: Z+ y3 g1 f. P" T; b7 S8 W6 K: T0 w* F
Our results demonstrate that human hematopoiesis can be regulated in vivo using a conditionally active growth factor receptor. Similar approaches may be useful in any situation where it is desirable to retain control over transplanted cells . Despite its panhematopoietic expression pattern, F36VMpl signaling in human hematopoietic cells induces proliferation that appears restricted to transduced erythroblasts and their progenitors. Flow histograms document that CID treatment induces the preferential expansion of cells expressing high levels of the linked GFP marker, a potentially advantageous feature in the context of a linked therapeutic gene.
% ]; e% p, x9 [" `- u" K: Y- V2 k/ q$ m+ I& M" L# Z
In a previous study, we found that baboons exhibited a very modest hematopoietic response to F36VMpl signaling both in vitro and in vivo . Although it remains possible that human hematopoietic cells may behave differently than described here in humans, this can only be definitively ascertained in a clinical trial.
3 y* x3 Z/ ~! C
1 @9 i+ G7 Z# o3 FThe responses of hematopoietic stem cells (HSCs) and progenitors to mitotic signaling can be significantly modulated by environmental factors. A case in point is chronic myelogenous leukemia (CML), where bcr-abl-expressing HSCs dominate hematopoiesis in vivo but are rapidly out-competed by normal progenitors in suspension culture .* X" i$ J6 r! B  X

* q- W5 o9 q; ~Controversy exists as to whether the anatomical compartment in which a HSC or progenitor resides can influence its response to a mitogenic stimulus. It is not known whether adult human peripheral blood HSCs and progenitors inherently differ from bone marrow HSCs in their capacity for engraftment and repopulation. This question has been especially difficult to address in humans due to the confounding influence of growth factors, which are uniformly administered prior to collection of peripheral blood HSCs and are rarely used prior to marrow HSC collection , suggesting an inherent biological difference between progenitors in these compartments.
: }/ Y4 B2 U" c
; O1 z! l- a' Z) R$ w0 tResults presented here suggest that the compartment in which a human hematopoietic progenitor resides can influence its response to a mitotic stimulus. F36VMpl signaling induced a consistent decline in marrow BFUe and CFU-mix and a consistent rise in spleen BFUe and CFU-mix. Although we cannot exclude the possibility that BFUe and CFU-mix trafficked from the marrow to the spleen in response to F36VMpl signaling, this appears highly unlikely based on 1) our consistent inability to detect circulating progenitors in the blood in response to CID treatment (data not shown); 2) tracking studies that demonstrate clonal relationships among CFU-mix, BFUe, and between CFU-mix and BFUe in the spleen (Fig. 3A); 3) the inability of tracking studies to find any clonal relationships between spleen CFU-mix/BFUe and marrow erythroid cells. Had a progenitor been induced to travel from the marrow to the spleen in response to CID treatment, it seems reasonable to expect some trace of its former presence in the marrow.
: }  h" I. a6 V1 X: A/ i1 p/ ]
% ~" ?1 u4 D9 z+ V2 i6 o2 a: `3 QTaken together, our findings are most consistent with the interpretation that the response of human hematopoietic progenitors to a mitotic signal can be influenced by the anatomical compartment in which they reside. Although we have not excluded the possibility of differences in AP20187 uptake, retention, or clearance between marrow and spleen, an environmental influence over the F36VMpl response is in keeping with our previous findings that growth factors can modulate the F36VMpl response in cultured human cord blood CD34  cells .# {5 _" {4 g2 g. g

' D' l( O; U" {7 N1 s  Z, wAlthough the apparent epo-dependence of the CID-responsive spleen progenitors provided a means to examine the clonality of the CID response, this growth pattern is not characteristic of BFUe or CFU-mix. We have previously shown that F36VMpl-transduced human CD34  cells can form small BFUe in the presence of CID alone and that CID-dependent colony formation is augmented, both in colony size and colony number, by the addition of growth factors, including epo . We therefore conclude that this unusual pattern of colony growth likely reflects the combined effect of CID that is retained within the colony-forming cell at the time of plating and epo added to the culture medium.
/ G' s$ \/ M2 E/ U0 v3 I+ w
+ K5 y" F. f7 h) }+ `% zOur results show that, analogous to the effect of epo on red blood cells, CIDs can act as growth factors for F36VMpl-transduced red blood cells. Although the need for repeated CID treatments poses an encumbrance and entails an additional risk of toxicity due to the need for chronic intermittent drug exposure, reversibility also has considerable conceptual appeal in view of the risk of leukemia associated with gene insertion .5 Q& P! J0 E2 T3 f, o) L

9 K' x- u& E3 j% ^ACKNOWLEDGMENTS  h1 V1 o7 E& p
* J4 z" P3 _9 b. S1 u: K
We acknowledge the assistance of Sue Chacona with cord blood collections, and expert technical assistance of Amanda Melbye, Betty Mastropaolo, Marilyn Skelly, Allen Gown, and Greg Levin. This work was supported by NIH grants DK52997, DK61844, and HL53750.
5 |' v1 W* Y& {6 m9 J1 {: t' N6 _1 h7 ?5 p4 a. o3 G/ q
DISCLOSURES# J. g. w% p/ g3 L) ~5 Q

" m( a! }" @3 T5 w) VC.A.B. is named as an inventor on a patent application related to the work that is described in the present study that is owned by the University of Washington and is licensed to ARIAD Pharmaceuticals.' \( j. B; Z- D& O
          【参考文献】
3 z2 J) E& |, T  B& m+ ` 5 C2 X5 C1 |# O5 K# I% B0 }' U" b
4 [% f2 _2 L4 M% Y- I
Neff T, Blau CA. Pharmacologically regulated cell therapy. Blood 2001; 97:2535¨C2540.
" H' i" a$ W5 R  z% D6 `2 L- b: t& v2 o# D( r
Spencer DM, Wandless TJ, Schreiber SL et al. Controlling signal transduction with synthetic ligands. Science 1993;262:1019¨C1024.& V  ], C+ }4 l2 A% s; Z

/ O, j+ H0 x4 z. mJin L, Zeng H, Chien S et al. In vivo selection using a cell growth switch. Nat Genet 2000;26:64¨C66./ y/ E% J: }3 G$ n. Z- }
, F/ w, e' }9 I* _' P+ ?8 v( P
Neff T, Horn PA, Valli VE et al. Pharmacologically regulated in vivo selection in a large animal. Blood 2002;100:2026¨C2031.
, ~) V% t! `4 E# m# d0 r5 n
8 U+ ~- u7 q8 ?+ B+ ]Hacein-Bey-Abina S, Le Deist F, Carlier F et al. Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy. N Engl J Med 2002;346:1185¨C1193.
# O6 }; G. d& }, j3 m8 a5 {4 t( Q$ g  e0 |' c( b- w0 c
Aiuti A, Slavin S, Aker M et al. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science 2002;296:2410¨C2413.& }5 K( c6 V7 T% L9 _$ w
+ w7 p0 F/ {3 d# `; W6 e; w3 F; c
Richard RE, De Claro RA, Yan J et al. Differences in F36VMpl-based in vivo selection among large animal models. Mol Ther 2004;10:730¨C740.& I2 c! Y* t6 I$ z

; y1 o3 `. L' K9 y2 R4 \. OGao Z, Golob J, Tanavde VM et al. High levels of transgene expression following transduction of long-term NOD/SCID-repopulating human cells with a modified lentiviral vector. STEM CELLS 2001; 19:247¨C259.
' d/ z6 Z* _4 H) o$ s3 z4 g6 }  d0 o$ I5 |
Kaneko S, Nagasawa T, Nakauchi H et al. An in vivo assay for retrovirally transduced human peripheral T lymphocytes using nonobese diabetic/severe combined immunodeficiency mice. Exp Hematol 2005;33: 35¨C41.
0 o1 B( X0 F! X1 Q3 ^- P# q* z8 e3 ], l' {3 P
Richard RE, Wood B, Zeng H et al. Expansion of genetically modified primary human hemopoietic cells using chemical inducers of dimerization. Blood 2000;95:430¨C436.
! `) ~" _$ j* I! g: `9 E& P( L; u( k7 k; |( C5 [! {
Jin L, Siritanaratkul N, Emery DW et al. Targeted expansion of genetically modified bone marrow cells. Proc Natl Acad Sci U S A 1998;95: 8093¨C8097., o( \$ K) C- O, p+ M2 w; _9 J" {

7 T. `# H* Q5 ]+ Q1 R- ]* b+ sRichard RE, Blau CA. Small-molecule-directed mpl signaling can complement growth factors to selectively expand genetically modified cord blood cells. STEM CELLS 2003;21:71¨C78.2 p' W+ g- r  Y) _8 Q8 t

9 @! z2 d. `, X- Z& MFollenzi A, Ailles LE, Bakovic S et al. Gene transfer by lentiviral vectors is limited by nuclear translocation and rescued by HIV-1 pol sequences. Nat Genet 2000;25:217¨C222.
" H3 S5 P1 {2 g  |9 t
, b) y6 a$ [) n: BAilles L, Schmidt M, Santoni de Sio FR et al. Molecular evidence of lentiviral vector-mediated gene transfer into human self-renewing, multi-potent, long-term NOD/SCID repopulating hematopoietic cells. Mol Ther 2002;6:615¨C626.
* j* ^( `6 R& F3 y# g9 Q( w9 N* @
4 w; f4 }( n9 {7 i0 WDull T, Zufferey R, Kelly M et al. A third-generation lentivirus vector with a conditional packaging system. J Virol 1998;72:8463¨C8471.& [7 D! p- W2 n

+ i9 |5 v4 ]" ]1 M) s0 AGlimm H, Eisterer W, Lee K et al. Previously undetected human hematopoietic cell populations with short-term repopulating activity selectively engraft NOD/SCID-beta2 microglobulin-null mice. J Clin Invest 2001;107:199¨C206.5 R; C% ~# y3 W, l9 R0 b

- Q4 F3 U, e: l% ~Stamatoyannopoulos G, Farquhar M, Lindsley D et al. Monoclonal antibodies specific for globin chains. Blood 1983;61(3):530¨C539.
! U4 o3 @% `# `9 p9 P* ?3 G1 j, |
Mazurier F, Gan OI, McKenzie JL et al. Lentivector-mediated clonal tracking reveals intrinsic heterogeneity in the human hematopoietic stem cell compartment and culture-induced stem cell impairment. Blood 2004; 103:545¨C552.; `' r8 B, ~! t
" X7 M+ X: t8 T1 W5 I
Imren S, Fabry ME, Westerman KA et al. High-level beta-globin expression and preferred intragenic integration after lentiviral transduction of human cord blood stem cells. J Clin Invest 2004;114:953¨C962.+ W  D( u4 e, T
, I/ F$ f  T& t, F
Neildez-Nguyen TM, Wajcman H, Marden MC et al. Human erythroid cells produced ex vivo at large scale differentiate into red blood cells in vivo. Nat Biotechnol 2002;20:467¨C472.5 V( i# b( w" J" h+ f

# n  ^: D3 z0 F4 h) |Richard RE, Weinreich M, Chang KH et al. Modulating erythrocyte chimerism in a mouse model of pyruvate kinase deficiency. Blood 2004;103:4432¨C4439.2 Q* h, i* Z- G, u

, I# E5 |. M/ a3 n) E3 xEmery DW, Tubb J, Nishino Y et al. Selection with a regulated cell growth switch increases the likelihood of expression for a linked gamma-globin gene. Blood Cells Mol Dis 2005;34:235¨C247.5 K9 O. x7 T3 S- u  G: K& j! R# x
+ h& e% _6 |4 i+ j; ], ^
Petzer AL, Eaves CJ, Barnett MJ et al. Selective expansion of primitive normal hematopoietic cells in cytokine-supplemented cultures of purified cells from patients with chronic myeloid leukemia. Blood 1997;90:64¨C69.
& S$ v; E7 G0 p$ j/ A8 A7 e' P7 N
, F- `$ N3 x( qBroudy VC. Stem cell factor and hematopoiesis. Blood 1997;90:1345¨C1364.5 l1 e% u8 g& T
5 z6 e# S: e9 g) a/ q) g* M
Bodine DM, Seidel NE, Zsebo KM et al. In vivo administration of stem cell factor to mice increases the absolute number of pluripotent hematopoietic stem cells. Blood 1993;82:445¨C455., n' [: `/ z4 ?+ \  e% t

1 l: M7 U5 J! |: ]) G: BElfenbein GJ, Sackstein R. Primed marrow for autologous and allogeneic transplantation: A review comparing primed marrow to mobilized blood and steady-state marrow. Exp Hematol 2004;32:327¨C339.- c, U1 ]. q8 c1 A6 [2 g& j) \
# T* L, M+ K0 N7 |5 l; p
Verfaillie CM, Bhatia R, Steinbuch M et al. Comparative analysis of autografting in chronic myelogenous leukemia: Effects of priming regimen and marrow or blood origin of stem cells. Blood 1998;92:1820¨C1831.
* b! m! F- V1 `( }- w; F( t, j
% t; y) E0 j1 O- o! f1 w4 g. m* i+ xBlau CA, Peterson KR, Drachman JG et al. A proliferation switch for genetically modified cells. Proc Natl Acad Sci U S A 1997;94:3076¨C3081.
* e! m4 i; ?2 G5 r& r  a; S6 H0 i4 U, n4 d1 r
Hacein-Bey-Abina S, von Kalle C, Schmidt M et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003;302:415¨C419.

Rank: 2

积分
64 
威望
64  
包包
1782  
沙发
发表于 2015-6-2 17:39 |只看该作者
是楼主原创吗  

Rank: 2

积分
97 
威望
97  
包包
1738  
藤椅
发表于 2015-6-18 12:17 |只看该作者
真好。。。。。。。。。  

Rank: 2

积分
64 
威望
64  
包包
1734  
板凳
发表于 2015-7-27 17:54 |只看该作者
干细胞之家微信公众号
初来乍到,请多多关照。。。  

Rank: 2

积分
89 
威望
89  
包包
1794  
报纸
发表于 2015-8-4 17:10 |只看该作者
今天临床的资料更新很多呀

Rank: 2

积分
64 
威望
64  
包包
1734  
地板
发表于 2015-9-29 17:50 |只看该作者
干细胞从业人员  

Rank: 2

积分
75 
威望
75  
包包
2193  
7
发表于 2015-10-11 14:42 |只看该作者
青春就像卫生纸。看着挺多的,用着用着就不够了。  

Rank: 2

积分
104 
威望
104  
包包
1772  
8
发表于 2015-10-12 09:35 |只看该作者
声明一下:本人看贴和回贴的规则,好贴必看,精华贴必回。  

Rank: 2

积分
77 
威望
77  
包包
1964  
9
发表于 2015-10-13 16:18 |只看该作者
不管你信不信,反正我信  

Rank: 2

积分
162 
威望
162  
包包
1746  
10
发表于 2015-10-21 12:10 |只看该作者
谢谢分享  
‹ 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, 2024-4-20 14:16

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.