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: 399882|回复: 253
go

Differential Requirements for Hematopoietic Commitment Between Human and Rhesus [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:58 |只看该作者 |倒序浏览 |打印
作者:Deepika Rajesha, Nachimuthu Chinnasamya,e, Shoukhrat M. Mitalipovf, Don P. Wolff,g,h, Igor Slukvinc,d, James A. Thomsonb,d, Aimen F. Shaabana,d 7 C+ d6 g, _% S! O3 \8 \
                  9 X- {& I: L% t
                  & w" d8 J6 a; c6 I$ _9 W
          & S' u! E) |% e
                         $ [  ]% q5 E8 P' T" C' W* h% R
            
) L/ [) Z! d# _, S            " j+ p! K8 n6 e* d" B) W1 }
            
/ q) N9 @! ~) _1 s& R            
, l) B# o; [, z8 K                      1 G5 V3 L' K! z, ]; Y$ t- u
        
% A6 E* e$ Q- I/ _; A& S; E        1 @+ t, @& O/ \" s- y2 [
        . K9 O4 j6 V+ n* x& K
          【摘要】) x6 ]! E8 n8 G3 i
      Progress toward clinical application of ESC-derived hematopoietic cellular transplantation will require rigorous evaluation in a large animal allogeneic model. However, in contrast to human ESCs (hESCs), efforts to induce conclusive hematopoietic differentiation from rhesus macaque ESCs (rESCs) have been unsuccessful. Characterizing these poorly understood functional differences will facilitate progress in this area and likely clarify the critical steps involved in the hematopoietic differentiation of ESCs. To accomplish this goal, we compared the hematopoietic differentiation of hESCs with that of rESCs in both EB culture and stroma coculture. Initially, undifferentiated rESCs and hESCs were adapted to growth on Matrigel without a change in their phenotype or karyotype. Subsequent differentiation of rESCs in OP9 stroma led to the development of CD34 CD45¨C cells that gave rise to endothelial cell networks in methylcellulose culture. In the same conditions, hESCs exhibited convincing hematopoietic differentiation. In cytokine-supplemented EB culture, rESCs demonstrated improved hematopoietic differentiation with higher levels of CD34  and detectable levels of CD45  cells. However, these levels remained dramatically lower than those for hESCs in identical culture conditions. Subsequent plating of cytokine-supplemented rhesus EBs in methylcellulose culture led to the formation of mixed colonies of erythroid, myeloid, and endothelial cells, confirming the existence of bipotential hematoendothelial progenitors in the cytokine-supplemented EB cultures. Evaluation of four different rESC lines confirmed the validity of these disparities. Although rESCs have the potential for hematopoietic differentiation, they exhibit a pause at the hemangioblast stage of hematopoietic development in culture conditions developed for hESCs. / G) g" O1 r* j# r  @) u5 H# \
          【关键词】 Embryonic stem cells Human Rhesus Hematopoiesis Hemangioblast Hematopoietic stem cell transplantation
  B9 H, G  O2 o                  INTRODUCTION. ^$ t' r+ _, ^% f
9 I9 p5 o9 b: h0 M( l2 @
Nonhuman primates, such as the rhesus macaque, have >90% DNA homology to humans and have long been used as models for studies on human behavior, reproductive biology, embryology, and various disease states . Given its similarities to humans, the rhesus macaque remains the primary in vivo model in which many clinically relevant questions regarding ESC-derived hematopoietic progenitor transplantation may be answered. Problems concerning microenvironmental induction/regulation of stem cell growth, specific allogeneic immune responses, and tumorigenesis cannot be satisfactorily addressed in xenogeneic or small animal allogeneic hosts. Furthermore, the homing and proliferation of human hematopoietic progenitors in the xenogeneic murine hematopoietic microenvironment may be greatly affected by disparate receptor/ligand and cytokine interactions. In addition, the limited proliferative demand placed on the transplanted cells as a result of the short life span of the mouse clouds the assessment of long-term hematopoietic stem cell (HSC) activity. Lastly, the absence of a reproducible allogeneic immune system prevents extrapolation of such xenogeneic transplantation studies to clinical applications. Hence, there is a crucial need to pursue study of ESC-derived hematopoietic progenitor transplantation in a well-characterized large animal allogeneic transplantation model that closely mimics the human hematopoietic system.7 {5 ?' @  c% R
& n3 S, c0 s3 Z3 I" O
However, despite success with human ESCs (hESCs), similar efforts to induce conclusive hematopoietic differentiation of rhesus ESCs (rESCs) using the same techniques have been unsuccessful. A few studies suggest limited hematopoietic development; however, the temporal emergence of CD45-positive cells and characteristic colony forming cells from differentiating rESCs has not been observed . Thus, the mechanisms underlying hematopoietic differentiation, expansion, and self-renewal are not well defined in monkey ESCs and, curiously, seem to differ from those of hESCs.1 j! @# F8 z, ]; o% Y# D
) l0 r, U7 l1 F. y
Given the importance of nonhuman primate ESCs, the current study pursues the differences in hematopoietic differentiation of rESCs compared with hESCs. Our results clearly demonstrate differential requirements for hematopoietic differentiation between these closely related species. Specifically, we show that rESCs differentiate to the bipotential stage of hematoendothelial development when using culture conditions developed for the hematopoietic differentiation of hESCs. Subsequent hematopoietic commitment is limited by an unclear mechanism. These findings emphasize the need to further study the differentiation of nonhuman primate ESCs as they relate to hESCs to gain essential information from this clinically relevant model." L+ N, A4 B# M; ?) U
* H) L( e) y2 S3 |! B1 C; p
MATERIALS AND METHODS& a1 @  b0 F: |2 e
) X2 d- }6 D% ?0 G6 ~
Embryonic Stem Cells( y1 S' ^0 J) ?. T) a1 {
; X3 e* s0 o( k$ S  D# K
The undifferentiated human embryonic stem cell line H9 (WiCell Research Institute, Madison, WI, http://www.wicell.org) was maintained by coculture with irradiated murine embryonic fibroblasts (MEFs) in Dulbecco's modified Eagle's medium (DMEM)/Ham's F-12 medium (F12) supplemented with 20% fetal bovine serum (FBS), 1% nonessential amino acids (NEAA), 1 mM L-glutamine (all from Invitrogen, Carlsbad, CA, http://www.invitrogen.com), 0.1 mM ß-mercaptoethanol (Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com), and 4 ng/ml human basic fibroblast growth factor (bFGF) (R&D Systems Inc., Minneapolis, http://www.rndsystems.com) .
" J( [2 E& O) f
6 j2 Q% u. F/ _" `; K! K7 bThe R366.4, R420, and R456 rhesus macaque ESCs derived from in vivo-flushed blastocysts  in DMEM/F12 (Invitrogen) supplemented with 20% FBS (HyClone, Logan, UT, http://www.hyclone.com), 1 mM glutamine, 0.1 mM ß-mercaptoethanol, and 1% NEAA (Invitrogen).! n0 j  o4 g+ G" |" e

4 s2 Y! [5 @7 `* [Undifferentiated CJ7 murine ESCs (kindly provided by Dr. Stuart Orkin, Boston, MA) were maintained by coculture with irradiated MEFs in gelatin-coated flasks in DMEM supplemented with 15% FBS (HyClone), 1 mM sodium pyruvate, 1% penicillin/streptomycin, 2 mM L-glutamine, 1% NEAA (all from Invitrogen), and 100uM 1-thioglycerol (Sigma-Aldrich).# r1 H% U, g" @7 e/ Q! h) I7 B

! p( \( K$ k  F2 ?, `' b/ X$ B. zMaintenance of Undifferentiated rESC and hESC Feeder-Free Cultures
6 ]3 k: b4 q. m
) @. k# [) y9 a3 Z4 `Initially, R366.4, R420, R456, ORMES-7, and H9 ESCs were maintained as undifferentiated cells by passage on irradiated MEFs (rESCMEF and hESCMEF). The cells were then adapted to feeder-free culture by allowing them to expand on Matrigel (BD Biosciences, San Diego, http://www.bdbiosciences.com)-coated plates as previously described by Xu et al. . rESC and hESC cultures growing on Matrigel (rESCMAT and hESCMAT) were maintained in MEF-conditioned media supplemented with bFGF at 4 ng/ml (R&D Systems).) w( R& x' R  b! X, Q) Z
$ G6 h% t4 A. G% }: y! i$ P3 M
Preparation of MEF-Conditioned Media2 s' h# C& S9 d& `2 Y0 ?4 s; `
0 t6 z- O; A) C5 k7 P9 L$ v
MEFs were harvested and irradiated at 40 Gy and seeded at 55,000 cells per cm2 in media containing 80% Knockout DMEM (KO-DMEM), 20% Knockout serum replacement, 1 mM L-glutamine, 0.1 mM ß-mercaptoethanol, and 1% NEAA (all from Invitrogen). MEF-conditioned medium (MEF-CM) was collected and supplemented with bFGF at 4 ng/ml. hESCMAT and rESCMAT cultures were fed with MEF-CM daily. Cultures were passaged before they became confluent by incubation in 200 units/ml collagenase IV for 5 minutes at 37¡ãC, dissociated, and then seeded back onto Matrigel-coated plates.
" ]3 x6 x+ K  S- A4 K
0 Q3 ~- K! D, @' ECoculture on OP9 Stromal Layer( _6 F1 X' S! @1 v+ y6 v) z

1 i2 e6 a! j9 z; _rESCMAT and hESCMAT cells were seeded on confluent OP9 layers as previously described . Briefly, OP9 cells were maintained in -minimal essential medium (MEM) containing 15% FBS (HyClone). The cells were allowed to reach confluence at least 3 days prior to the plating of rESCMAT and hESCMAT cells. On the day of plating, the medium was changed to MEM supplemented with 15% FBS (HyClone), 1 mM L-glutamine (Invitrogen), 50 µg/ml ascorbic acid (Sigma-Aldrich), 20% BIT9500 (Stem Cell Technologies, Vancouver, BC, Canada, http://www.stemcell.com), and 450 µM monothioglycerol. For the cytokine-supplemented OP9 cocultures, the following cytokines were added to the medium: 150 ng/ml stem cell factor (SCF), 150 ng/ml Flt-3 ligand (Flt-3L), 10 ng/ml interleukin (IL)-3, 10 ng/ml IL-6, 50 ng/ml granulocyte colony-stimulating factor (G-CSF), and 20 ng/ml bone morphogenetic factor (BMP-4) (all from R&D Systems). The cells were fed every fourth day and harvested on days 4¨C16 using collagenase IV.% E4 f1 @0 {9 _
7 D( B1 l8 K. J/ F# z  m
Embryoid Body Culture
7 A  J* N1 V3 {- [7 V) L
. [( s( z# R5 s2 ~9 t8 ?0 v* {6 [Undifferentiated hESCs and rESCs that were adapted to feeder-free growth on Matrigel-coated plates were harvested at confluence with collagenase IV. To promote EB formation, the cells were transferred to six-well low-attachment plates for an overnight incubation in KO-DMEM supplemented with 20% FBS (HyClone), 1% NEAA, 1 mM L-glutamine, and 0.1 mM mercaptoethanol (all from Invitrogen). The next day, cultures were fed fresh differentiation media alone (control) or were fed differentiation media supplemented with the following growth factors and cytokines: 150 ng/ml SCF, 150 ng/ml Flt-3L, 10 ng/ml IL-3, 10 ng/ml IL-6, 50 ng/ml G-CSF, and 20 ng/ml BMP-4 (all from R&D Systems). The media was changed every 4 days by transferring the EBs into a 15-ml tube and letting the aggregates settle for 5 minutes. The supernatant was aspirated and replaced with fresh differentiation media.
$ u3 j9 ]3 X+ t# B2 K
* _9 T/ O  n5 A+ H8 V( G9 e' }Undifferentiated murine CJ7 ESCs were maintained in feeder-free culture on gelatin-coated plates in the presence of LIF. For EB formation cells, were harvested using trypsin and were seeded in low-attachment plates in DMEM containing 15% FBS, (HyClone), 1 mM L-glutamine, 1% sodium pyruvate, 0.75% bovine serum albumin (fraction V), 450 µM monthioglycerol (all from Invitrogen), and 20% BIT9500 (Stem Cell Technologies). The next day, cultures were given fresh differentiation media alone (control) or differentiation media supplemented with 50 ng/ml SCF, 10 ng/ml IL-3, 10 ng/ml IL-6, 5 ng/ml G-CSF, 5 ng/ml granulocyte-macrophage colony-stimulating factor (GM-CSF), 10 ng/ml vascular endothelial growth factor (VEGF), 10 ng/ml thrombopoietin (TPO), and 10 ng/ml erythropoietin (EPO) (all from R&D Systems). The cells were fed every fourth day and harvested after 16 days of EB culture.
+ g$ Y8 E' N8 |6 g
5 a- e0 r8 u, y4 xFlow Cytometry
4 ]$ B: c3 y3 |; J2 Q/ @
* x6 D# i* G' \9 lCells were washed with media and treated with trypsin and collagenase IV (Invitrogen) for 20 minutes in a 37¡ãC incubator followed by washes with media and passage through a 70-µm cell strainer. The cells were resuspended at approximately 2 x 105 cells per milliliter and stained with the following fluorochrome-conjugated monoclonal antibodies: anti-human CD43 (L10), anti-human CD31 (WM-59), anti-human CD38 (HIT2), anti-nonhuman primate CD41 (HIP8), anti-human CD117 (YB5.B8) (all from eBiosciences, San Diego, CA, http://www.ebioscience.com); anti-human CD45 (D058¨C1283), anti-human (HI30), anti-human CD34 (563) (BD Biosciences); anti-human FLK-1 (89106), anti-human SSEA-4 antibody (MC-813-70), and anti-human Oct3/4 (240408) (all from R&D Systems). Human-rhesus cross-reactivity was confirmed using rhesus tissue samples. Nonviable cells were excluded with 7-aminoactinomycin D (BD Biosciences). Live cell analysis was performed on a FACSCalibur flow cytometer with Cell Quest software.  A) }0 u( `$ o) Q; e2 k

( h0 R9 V, T% ]For intracellular staining, cells were harvested using trypsin and collagenase IV, washed with phosphate-buffered saline (PBS), and fixed with 2% paraformaldehyde for 20 minutes on ice. The cells were washed with PBS and once with SAP buffer (PBS containing 2% fetal calf serum and 0.2% saponin). The cells were then stained with goat anti-Oct-3/4 (R&D Systems) in SAP buffer for 30 minutes at 4¡ãC and then washed twice with SAP buffer. The cells were then incubated with a fluorescein isothiocyanate (FITC)-conjugated, isotype-specific anti-rat IgG2b secondary antibody (BD Pharmingen, San Diego, http://www.bdbiosciences.com/pharmingen) for 30 minutes at 4¡ãC prior to washing and analysis on a FACSCalibur flow cytometer with Cell Quest software. For negative controls, the primary antibody was omitted.+ p+ s. D7 e* Z" N8 O8 U/ Q9 M+ }

) z0 ]  n( Y5 {  C* e1 IClonogenic Hematopoietic Progenitor Assay9 `' X& q8 H6 m4 F
0 K$ A% j5 B2 E% s0 l0 y$ m
Embryoid bodies were dispersed into single-cell suspensions using 1 mg/ml collagenase IV and 0.05% trypsin/EDTA. Viable cells were quantified, plated (3.0 x 105 cells per milliliter), and assayed in humidified chambers for hematopoietic CFCs using Human Methylcellulose Complete media (R&D Systems) containing 50 ng/ml SCF, 3 U/ml EPO, 10 ng/ml GM-CSF, and 10 ng/ml IL-3.& a. X* x, i" `% v& L* ~% }0 G* X
# ~2 N  V  }- K* R( m
Colony Histology
# x( J& h- M& @1 l# F6 h1 o! m
  o8 y$ \9 d) |) l4 S5 VIndividual colonies growing on methylcellulose were picked using a pulled-tip glass micropipette. The colony was placed in a 1.5-ml centrifuge tube with 1 ml of PBS. Cell clumps were dissociated by incubation with 0.05% trypsin for 5 minutes. The cells were then washed and resuspended in 300 µl of medium, mounted on Cytoclips (Thermo Electron Corporation, Waltham, MA, http://www.thermo.com), and centrifuged at 800 rpm for 5 minutes. Cells were fixed and stained with Wright-Giemsa reagents (Hema 3 stain; Fisher Scientific International, Hampton, NH, http://www.fisherscientific.com) according to the manufacturer's instructions.0 z7 Y' A% ?9 a9 @
; {+ q2 ~8 _. Q- j% S
For immunofluorescence staining, the cytospins were washed with twice with PBS and fixed for 10 minutes in PBS containing 2% paraformaldehyde. The fixed cells were washed with PBS and incubated initially with biotin-conjugated anti-VE-cadherin (16B1; eBioscience) and FITC-conjugated anti-CD45 (D058-1283; BD Biosciences) for 1 hour, washed five times, and incubated with streptavidin-Alexa Fluor 546 (Invitrogen) for another 45 minutes. Following the second staining step, the cells were washed and mounted using Antifade gold (Invitrogen). Images were acquired using an MRC 1024ES confocal microscope (Bio-Rad, Hercules, CA, http://www.bio-rad.com). Cells incubated with streptavidin-Alexa Fluor 546 alone and IgG1-FITC served as controls. For acetylated low-density lipoprotein staining, the colonies were plucked from methylcellulose and replated on a Matrigel-coated slide flask (Nalgene Nunc International, Rochester, NY) for 2 days. Aseptically, diI-acetylated low-density lipoprotein (Ac-LDL) (Biomedical Technologies, Stoughton, MA, http://www.btiinc.com) was diluted to 10 µg/ml in medium, added to the live cells, and allowed to incubate for 4 hours at 37¡ãC. The medium was removed, and the cells were washed several times with medium. The cells were mounted using mounted using Antifade gold (Invitrogen) and visualized using standard rhodamine excitation emission filters via confocal microscopy.  H, p' R9 k6 Q# c! x

* G2 X% j" T# ^5 B% V" j/ @& hQuantitative Real-Time Reverse Transcription-Polymerase Chain Reaction Analysis& y% j- b7 L0 S! r9 H

. v# `: l) Q: g* O, k, sTotal RNA was isolated from undifferentiated rESCs and 16-day rhesus EB cultures using the RNAqueous-4PCR Kit (Ambion, Austin, TX, http://www.ambion.com). RNA was treated with RNase-free DNase at the last step of the reaction. cDNA was synthesized using the Bio-Rad iScript cDNA synthesis kit. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed using iQ SYBR Green Supermix reagents and an iCycler thermal cycler and software (Bio-Rad). Rhesus-specific primers were as follows: 5'-GAAACCGCAAGGCATCTG-3' (forward) and 5'-CCCACAATTCCCGCTACC-3' (reverse) for GATA-1; 5'-CCACAGCCCTAGTATGAAAG-3' (forward) and 5'-TCACCGCATACAGAATCTAAG-3' (reverse) for GATA-2; 5'-AAAACAAAGGGCACAGCATC-3' (forward) and 5'-GAGACCAACGCAATTCATCA-3' (reverse) for SCL'; 5'-ATGCACGGCATCTGGGAATC-3' (forward) and 5'-GTCACTGTCCTGCAA-GTTGCTGTC-3' (reverse) for FLK-1; 5'-CCCTCTCCTGGGAGCATT-3' (forward) and 5'-AAAGAGAGGAAGGCTCTGGTG-3' (reverse) for RUNX-1; and 5'-ATCCCCCAATTCTCTGGAAC-3' (forward) and 5'-ATTGGGGAACTCCAGACACA-3' (reverse) for PU.1. All primers were tested and optimized for specificity with rhesus samples and nonreactivity with SYBR Green reagents using non-reverse-transcribed cDNA. Briefly, for each reaction, 12.5 µl of the SYBR Green PCR Master Mix (Bio-Rad) was mixed with 10 µM each primer (for each gene of interest or glyceraldehyde-3-phosphate dehydrogenase . The relative expression of each normalized target gene was compared with the GAPDH-normalized expression of the target gene. Fold change expression from undifferentiated rESCs was calculated as 2¨CCT, where CT = (CT of differentiated rESCs) ¨C CT (undifferentiated rESCs).
9 c4 z/ u$ y$ m7 K8 Q- H
. W& O9 I3 p" e/ [RESULTS
# j( N% y* H, l/ r) v# v' e: W9 Q; Q. j  ]) Q1 o
Undifferentiated rESCs Demonstrate a Phenotype Similar to That of hESCs When Expanded on MEF or Matrigel
7 s! ~/ j! [+ q* B
" u/ a) X$ \. q' H% LIn preliminary studies, we observed that the presence of even a small number of MEFs significantly inhibited the hematopoietic differentiation of rESCs. Therefore, undifferentiated rESCs (R366.4, R456, R420, and ORMES-7) and hESCs (H9) were expanded on irradiated MEFs (rESCMEF and hESCMEF) and adapted to feeder-free growth on Matrigel-coated plates (rESCMAT and hESCMAT). The rESCMAT maintained a normal karyotype after nearly 20 passages on Matrigel (Fig. 1A¨C1C). Each of the human and rhesus ESC lines has been shown to induce the development of teratomas following transplantation into immunodeficient mice . Figure 1D¨C1G provides sample histologic images of a single teratoma derived from the injection of undifferentiated ORMES-7 rESCs. Morphologic evidence is shown for cartilaginous (mesoderm), intestinal (endoderm), and neural (ectodermal) differentiation.( B+ N7 |/ U) r; C; p# `
+ z$ J7 f7 w8 d" r8 I5 `+ P
Figure 1. Feeder-free expansion of undifferentiated ESCs. Undifferentiated R366.4 rhesus ESCs (rESCs) were maintained on irradiated murine embryonic fibroblasts (A) or adapted to feeder-free conditions on Matrigel-coated plates (magnification, x25; scale bar = 50 µm) (B) without change in colony morphology. (C): Despite 20 passages on Matrigel, undifferentiated rESCs maintained a normal 42XY karyotype. (D¨CG): Histologic sections of teratomas formed by injection of undifferentiated ORMES-7 rESCs into SCID mice and examined at 15 weeks. (D): Low-power field demonstrating an overview of multiple cell types (magnification, x25). (E): Cartilage cells depicting mesodermal differentiation. (F): Gut epithelium depicting endodermal differentiation. (G): Neural tissue depicting ectodermal differentiation. Magnification (E¨CG), x100.
8 f: l0 Y: s0 j, `7 i" k9 q( w* |$ Q1 \9 Q4 U5 D" O& }8 r7 @
Following expansion on Matrigel, the undifferentiated ESCs were harvested and analyzed for the expression of antigens associated with pluripotency (c-Kit, SSEA-4, and Oct3/4) . The results showed that undifferentiated rESCs and hESCs were completely devoid of CD45 expression but expressed a low frequency of CD34  and FLK-1  cells, suggesting some heterogeneity among both rESCs and hESCs (Fig. 2A, 2B). Both human and rhesus ESCs maintained on Matrigel displayed a slightly higher frequency of the FLK-1  subset of cells in the culture but an otherwise similar differentiation profile compared with ESCs maintained on MEFs. As expected, the Oct-4, c-Kit, and SSEA-4 were highly expressed in the undifferentiated ESCs.
% w! Y  z/ C1 i& l
: `0 q- u1 x5 z" x) v. i2 GFigure 2. Analysis of undifferentiated rhesus ESCs (rESCs) and human ESCs (hESCs) for phenotypic markers associated with pluripotency. Undifferentiated H9 and R366.4 ESCs expanded on MEFs or on Matrigel were harvested and stained for antigens associated with pluripotency (CD117, SSEA-4, and Oct4) and those associated with hematoendothelial differentiation (FLK-1, CD34, CD45, CD41, CD38, and CD31). The frequency of each phenotype for either H9 (A) or R366.4 (B) undifferentiated ESCs was determined by flow cytometry. Each value represents the mean of three independent experiments ¡À SEM. Abbreviation: MEF, murine embryonic fibroblast." Z1 w* J8 B1 G( H7 D
* M9 \* t% g& i' Y) K3 h/ H( Z
rESCs Differentiating on OP9 Stroma Lack Phenotypic and Functional Hematopoietic Properties; U4 \/ K3 P# I4 _  \
) o# U/ h5 W3 i; }. B+ F4 W
Recently, Vodyanik et al.  demonstrated the generation of hematopoietic progenitor cells following coculture of hESCs with OP9 stroma. We compared the hematopoietic differentiation of rESCs with that of hESCs on OP9 stroma. As shown in Figure 3, rESCs demonstrated a limited capacity for hematopoietic differentiation compared with hESCs in OP9 stromal culture. Although CD34  cells could be detected at low levels, no CD45  cells were seen (
. D) W: ~9 j& v0 r
+ y+ |. W. w, {( X  [Figure 3. Hematopoietic differentiation of rESCs and hESCs in OP9 coculture. rESCmat (A, C) and hESCmat (B, D) cells were allowed to differentiate on confluent OP9 layers and harvested between day 4 and day 16. The cells were analyzed by flow cytometry for hematoendothelial differentiation (FLK-1, CD34, CD45, CD41, CD31, and CD38) and for the presence of undifferentiated ESCs (CD117). (A, C): Values represent the mean ¡À SEM of three individual experiments. (B, D): Values represent the mean of duplicate experiments. Abbreviations: hESC, human ESC; mat, Matrigel; rESC, rhesus ESC.
! o8 {; O* ]: B
% O. K+ _+ X* u% }5 A9 F7 B* ?0 ~Cytokine Supplementation of EB Cultures Leads to Improved Hematopoietic Differentiation of rESCs to a Lesser Degree Than in hESC Cultures. [# [" U! i5 Z+ E* i7 {: z+ F. Q
& I7 w, Z6 {# K4 Y
Chadwick et al.  demonstrated the hematopoietic differentiation of hESCs in cytokine-supplemented EB cultures. We compared the differentiation of rESCs and hESCs in EB culture with and without cytokine supplementation. Rhesus EB cultures without cytokine supplementation demonstrated low levels of CD34  (
& q3 d% s( v5 Z/ |1 E0 t$ R& |8 R, O: e  J! d
Figure 4. Hematopoietic differentiation of rESCs and hESCs as EB cultures in the presence or absence of cytokines. rESCmat and hESCmat cells were allowed to differentiate as EB cultures in the absence (A¨CD) and presence (E¨CH) of cytokines. The cells were harvested from day 4 to day 16 and analyzed by flow cytometry for hematoendothelial differentiation (Flk-1, CD34, CD45, CD41, CD31, and CD38) and for the presence of undifferentiated ESCs (SSEA-4 and CD117). (B, D): Values represent the mean of duplicate experiments. (A, C, E¨CH): Data shown represent the mean values ¡À SEM of three independent experiments. Abbreviations: hESC, human ESC; mat, Matrigel; rESC, rhesus ESC.0 f! B& `* O3 k% q
# A$ w6 ?9 x" o/ o- Z$ f
Rhesus EBs formed in the presence of cytokines (BMP-4, SCF, Flt-3 ligand, IL-3, IL-6, and GM-CSF) demonstrated a decrease in SSEA-4 frequency (60%¨C12%), with a concomitant rise in the levels of CD41  and CD34  cells. In addition, for the first time, detectable frequencies of CD45  cells were observed beginning at EB12 and continuing to EB22 (0.18%¨C0.98%) in the presence of cytokines (Fig. 4E, 4G). Human EBs revealed a robust hematopoietic differentiation with higher frequencies of CD34-positive (10.0 ¡À 1.7%), CD45-positive (20.0 ¡À 7.2%), and CD31-positive (18 ¡À 4.7%) cells in the presence of cytokines (Fig. 4F, 4H).8 Y2 J& ^0 V( Q* R9 R( `

2 G- m8 M/ A( g; g! j7 e  c1 }rESCs Demonstrate a Lower Capacity for Hematopoietic Differentiation in EB Culture Compared with hESCs or mESCs2 G. V1 u5 n$ F. W
* E; Q/ r/ l9 h
Using similar EB culture conditions, we compared the hematopoietic differentiation of rhesus EBs to both murine and human EBs. As shown in Figure 5A, both human and murine EBs demonstrated robust hematopoietic differentiation, as approximately one fourth of the cultured cells expressed CD45. Significantly lower levels of CD45 expression were observed in the differentiating rhesus EBs compared with the either the human or murine EBs. Surprisingly, the differentiation profile of H9 human EBs was closer to that of the CJ7 murine EBs than to those derived from the four different rESC lines tested (R366.4, R420, R456, and ORMES-7). To evaluate the possibility that the rESCs simply require more time in culture to demonstrate significant hematopoietic lineage commitment, rhesus EB cultures were maintained with cytokine supplementation for up to 7 weeks. As shown in Figure 5B, higher levels of CD34  cells were observed in all three lines of rESCs. However, these increases in CD34 expression were not associated with a hematopoietic lineage commitment, as the levels of CD45 decreased from 3.63% to 0.66% in R420 cells, whereas they remained relatively unchanged in the other two cell lines. In addition, there was a decrease in CD31 frequency in the R456 (from 4.4% to 0.7%) and R420 (from 3.48% to 0.32%) EBs. Thus, allowing the EBs to differentiate for longer periods of time enhanced CD34 expression but did not augment the levels of CD45  cells.3 n, y5 j2 ?  d8 [4 ?3 |

1 m" `: S: \4 I- w* TFigure 5. Comparison of hematopoietic differentiation in EB culture between human ESCs (hESCs), murine ESCs, and rhesus ESCs (rESCs). rESCs (R336.4, R420, R456, and ORMES-7), murine ESCs (CJ7), and hESCs (H9) were allowed to differentiate in cytokine-supplemented EB cultures. Human-specific cytokines were used for culturing human and rhesus EBs, whereas murine-specific cytokines were used for the CJ7 cells. (A): The EBs were harvested on day 16, and hematopoietic differentiation was assessed by flow cytometry. (B): Extended EB culture of rESCs (R336.4, R420, and R456) in the presence of cytokines for nearly 7 weeks, subsequently analyzed for hematopoietic differentiation by flow cytometry. (C): Coculture of R366.4 rESCs with OP9 stroma for 16 days in hematopoietic differentiation medium with or without cytokine supplementation. (D): Tracking of the frequencies of CD34  or CD45  cells during extended coculture of 366.4 rESCs in OP9 stroma. (A): Values represent the mean ¡À SEM of three independent experiments. (B¨CD): Values represent the mean of duplicate experiments.
8 v: t9 f3 u0 v
( Q) K  I6 m  f* Q/ q- k9 ^9 V7 E# A* |( d1 hNext, we examined the effect of cytokine supplementation on the differentiation of rESCs during OP9 coculture. As shown in Figure 5C, cytokine supplementation of OP9 stroma cocultures resulted in slightly lower frequencies of CD34  and CD41  cells and higher levels of FLK-1  cells. The increases in FLK-1 expression suggested enhanced development of hematopoietic mesoderm; however, CD45  cells remained at an undetectable level. Lastly, rESCs subjected to 4 weeks of OP9 coculture in the absence of cytokines demonstrated rapid expansion of CD34  cells (76% of mixed culture at 28 days), yet CD45 expression still remained undetectable (Fig. 5D).
8 y5 z1 J' U5 Z" ~* M' V) I, |  Z/ j1 `' R% [: Y4 p9 i$ ~
Rhesus EBs Express a Transcriptional Profile Consistent with Hematoendothelial Differentiation and Display Both Hematopoietic and Endothelial Differentiation in Semisolid Clonogenic Culture. m8 F. R* ~+ B0 O) q

, C9 M; A3 U& B) Z* ~- F1 oTo further characterize their hematopoietic development, rhesus EBs were studied for their expression of transcription factors critically associated with hematoendothelial development and subsequent lineage commitment (SCL/Tal-1, GATA-1, GATA-2, PU.1, and RUNX1). SCL/Tal-1 has been shown to play a fundamental role in the earliest stages of hematopoietic and endothelial development. In addition, blast colony-forming cells (BL-CFCs) (putative murine hemangioblasts) fail to form from SCL¨C/¨C ESCs .
+ O$ n2 e  B7 K& @; u" R9 J" D$ b: U6 w/ }5 Y* Q
The expression of these transcription factors was measured in three lines of undifferentiated rESCs and compared with day 16 rhesus EBs cultured in cytokine-enriched media. As shown in Figure 6A, all three lines of EBs demonstrated upregulation of factors associated with early hematoendothelial development, as evidenced by increases in GATA-1, GATA-2, SCL, and FLK-1 expression and a dramatic fall in Oct-3/4 expression. However, absence of hematopoietic lineage commitment was demonstrated by a lack of upregulation of either RUNX1 or PU.1 in two of the three lines studied. Interestingly, PU.1 appeared to be upregulated in the third group of rhesus EBs (R420), which also demonstrated the highest frequency of CD45  cells, as shown in Figure 5A.
. n( K+ i0 [: f  P/ e1 p6 ]0 A1 m8 S0 ~
Figure 6. Analysis of hematoendothelial colonies derived from rhesus EBs. Individual hematoendothelial colonies were picked from methylcellulose cultures of single-cell suspensions harvested from day 16 rhesus EBs cultured in cytokine-supplemented media. (A): Quantitative reverse transcription-polymerase chain reaction analysis of rhesus EBs for their expression of transcription factors critically associated with hematoendothelial development and subsequent lineage commitment (SCL/Tal-1, GATA-1, GATA-2, PU.1, and RUNX1). (B): Representative photomicrograph of hematoendothelial colony arising from methylcellulose culture of day 16 R420 EBs demonstrating a mixed colony of erythroid clusters, myeloid cells, and endothelial cells. (C, D): Wright stains (magnification, x100) of picked hematoendothelial colonies reveals morphology of macrophages, endothelial cells, and erythroid cells. (E, F): Double-stained microscopic images of cytospin preparation of individual hematoendothelial colonies. (G): Confocal fluorescence imaging (magnification, x25) reveals cells with either CD45 expression (FITC, green) or VE-cadherin expression (Alexa Fluor 546, red), confirming the existence of cells within the EB cultures capable of bipotential differentiation. (H¨CJ): Triple-stained confocal fluorescence images of individual hematoendothelial colonies (magnification, x25). (K): Merged image reveals distinct hematopoietic cells (CD45 FITC, green) in the background of endothelial cells (VE-cadherin PE-Cy5, purple; Ac-LDL, red). Abbreviations: Ac-LDL, acetylated low-density lipoprotein; FITC, fluorescein isothiocyanate; PE, phycoerythrin.8 p. J( K2 S+ Y) o4 r1 N

/ D7 I0 Z7 W  V$ A  X; QWhen plated in methylcellulose, robust hematopoietic colony formation was observed from both murine and human EBs (data not shown). However, as shown in Figure 6B, cells from rhesus EBs formed colonies of mixed erythroid, myeloid, and endothelial cell types, signaling the existence of bipotential hematoendothelial progenitors in the cultures reminiscent of BL-CFCs. The loosely adherent cells in the hematoendothelial colonies displayed erythroid or macrophage morphology on examination of Wright stains (Fig. 6C, 6D). To better characterize these mixed colonies, the cells were stained for antigens associated with endothelial (VE-cadherin and Ac-LDL) or hematopoietic (CD45) lineage commitment. As shown in Figure 6E¨C6G, small, rounded cells characteristic of hematopoietic morphology expressed only CD45 without VE-cadherin. Conversely, cells exhibiting endothelial morphology expressed VE-cadherin brightly without CD45 expression. Triple staining with CD45, VE-cadherin, and Ac-LDL is shown in Figure 6H¨C6K. Similar morphology was seen in cells with high uptake of Ac-LDL. Taken together, these findings indicate that exposure of the rhesus EBs to the current cytokine cocktail results in the abundant development of bipotential hematoendothelial progenitors with only limited commitment to definitive hematopoietic lineages. This "pause" in hematoendothelial differentiation illustrates a point of divergence from patterns of hematopoietic differentiation seen in murine and human EBs under similar conditions.
3 f! U2 C4 n$ ?' n
1 o+ L- L5 V0 h3 @" ~; f$ v, F( C7 R3 j, ZDISCUSSION
2 c3 }* `- u* h: s& j, ]8 z3 X
: ~4 [! D1 b& H) XProgress toward clinical application of ESC-derived hematopoietic progenitor cell transplantation requires rigorous evaluation in a clinically relevant animal model, such as monkeys. However, in contrast to hESCs, efforts to induce conclusive hematopoietic differentiation from rESCs have been unsuccessful . These peculiar differences in hematopoietic differentiation between human and nonhuman primate ESCs have not been consistently studied and are therefore poorly understood. Characterizing these functional differences will likely clarify the critical regulatory steps involved in the hematopoietic differentiation of ESCs. The need for a better understanding of rESC-derived hematopoiesis as it relates to human ESC biology provides the major impetus for this study.
( S1 \5 k7 B3 K; U' S
! _% ^1 S6 ^* o) pIn the present study, rESCs were adapted to feeder-free growth on Matrigel. The rESCMAT, like the hESCMAT , retained a normal karyotype despite nearly 20 passages of feeder-free growth. In OP9 stromal coculture, rESCs demonstrated no CD45 expression but developed CD34  cells, which gave rise to endothelial cell networks in subsequent methylcellulose culture. In the same conditions, hESCs exhibited convincing hematopoietic differentiation. In cytokine-supplemented EB culture, rESCs demonstrated improved hematopoietic differentiation, as evidenced by significant levels of CD34  and CD41  cells and detectable levels of CD45  cells. However, these levels remained dramatically lower than those for hESCs under identical culture conditions. Subsequent plating of cytokine-supplemented rhesus EBs in methylcellulose culture led to the formation of mixed colonies of erythroid, myeloid, and endothelial cells, confirming the existence of bipotential hematoendothelial progenitors in the cytokine-supplemented EB cultures. Evaluation of four independently isolated rESC lines confirms the validity of these disparities.; _) k% |1 n( p
! @1 {: I5 `* _! h$ o0 N6 R& d
Previous comparisons of murine and human hematopoietic ontogeny have revealed extensive homology between these otherwise disparate species. Surprisingly, ESCs derived from the more closely related rhesus macaque behave differently than their human counterparts in the same culture environment. These observations have been reported in previous studies of ESCs derived from the rhesus macaque and other nonhuman primates. Li et al. .
" L+ I! D' Q4 K. ^/ W( P) k. S9 j/ Q& H5 k0 S7 [$ ^
Recent reports have shown the generation of hematopoietic cells from cynomolgus ESCs (cyESCs), but the methodology used differs significantly from that used previously for hESCs. Hiroyama et al. . These findings imply the absence of an essential growth factor or the existence an inhibitor in OP9 coculture that limits hematopoietic commitment in cyESCs. This effect is not observed in OP9 coculture with hESCs as shown in Figure 3B.
9 D0 ~! W& S+ S- V$ G) i& R' h
. o  |/ j; K2 ?& @+ B3 `: [: ]In addition, a study of hematopoietic differentiation in common marmoset ESCs (cmESCs) . They concluded that cmESCs may possess a lower rate of intrinsic hematopoietic differentiation compared with hESCs. Interestingly, the authors found that when the cmESCs were transduced to overexpress Scl/Tal1 under the control of an EF1- promoter, a dramatic increase in the hematopoietic colony formation was observed. This was also associated with a significant rise in Gata-1 expression, possibly signaling a switch toward hematopoietic commitment., G, L: R' m& N" ~
' t' n  {- F  y- t, V% i
Similar conclusions were reached in the present study and are expanded to include multiple rhesus ESC lines in direct comparison with both human and murine ESCs. For the first time, the development of detectable quantities of CD45  cells confirms the existence of lineage-committed hematopoietic cells. However, their frequency is extremely low relative to both murine and human EB culture. The presence of FLK-1 and CD34 expression points to an arrest of hematopoietic commitment at the bipotential stage of hematoendothelial progenitor development, and subsequent differentiation reveals a bias toward endothelial differentiation. As shown in Figure 5B, allowing the rhesus EBs to differentiate for nearly 7 weeks did not change the above findings. Lastly, subculture of day 12 EB cultures by dissociation of the EBs and replating decreased the levels of CD45-positive cells and enhanced the endothelial differentiation (data not shown).
4 h6 w+ A" r& }  @) e! `/ N8 Q# ]- G) n' V
The observed differences between human and rhesus ESCs did not relate to the subtle differences in their initial procurement. Three of the rESC lines described in this study (R366.4, R420, and R456) were isolated from the inner cell mass of blastocysts produced in vivo. In contrast, one of the rhesus ESC lines (ORMES-7) and the human ESC line (H9) were derived from in vitro-derived rhesus or human blastocysts . The subsequent steps in the isolation and expansion of all ESCs derived from either species were identical. Since the results were consistent among all four lines of rESCs, we conclude that the disparities in hematopoietic differentiation between the rhesus and human ESC cultures were not simply due to the technique of blastocyst derivation.& Z8 F; K0 z' e/ T) K

! p, W  T! Z- w  O2 YPossible explanations for the observed arrest in the hematopoietic lineage commitment of differentiating rESCs include either the absence of an essential factor needed to augment hematopoietic differentiation or the existence of a specific inhibitory signal. In the case of OP9 stromal coculture, no trophic factors were added beyond those found in the fetal calf serum or provided by the stromal cells themselves. Although OP9 stroma is derived from the macrophage colony-stimulating factor-deficient osteopetrotic mouse and has been shown to secrete SCF and IL-7  and the endogenous levels of its receptor, CXCR4, might influence hematopoietic differentiation of rESCs on OP9 stroma and explain the differential stromal requirement between rESCs and hESCs. Future experiments designed to characterize the nature of the support provided by the OP9 stroma to human ESCs are currently under way and may provide clues regarding the key inductive or inhibitory factors influencing hematopoietic differentiation of rESCs.
1 F; r7 f* n' P
. w- Z& |( K* q4 I, A, [- `In the EB culture of rESCs, a supportive stromal layer is not used, thus eliminating any effects from direct contact or soluble factors arising from murine fibroblasts. However, an autologous stromal cell population also develops within the EBs that may impair hematopoietic differentiation . Future experiments designed to block TNF- and TGF-ß function are planned to assess the role of these factors in the hematopoietic differentiation of rhesus EBs.
( h' [. m* j5 C/ ]9 s% j! p
7 X' ]" {# |" q% CGrowing evidence shows that fibroblast growth factor (FGF) can positively regulate hematopoiesis by acting on various cellular targets, including stromal cells, early and committed hematopoietic progenitors, and possibly some mature blood cells . Thus, endogenous levels of FGFR within the rhesus EBs could specifically alter the differentiation of developing hemangioblasts. We are currently studying a number of candidate populations of hematoendothelial progenitors for their hematopoietic differentiation capacity following isolation and subculture.
3 F2 h! h* O( I! j& A. k& o+ O" Y% j9 B' B, J$ v
SUMMARY
+ z, O% ~+ R/ @' }8 z! g4 C# u5 |+ J
& w& O7 R) s9 z+ ?$ ^$ wIn summary, this direct comparison of hematopoietic differentiation between hESCs and rESCs illustrates critical differences in the biology of cultured ESCs between these closely related species. In light of the importance of a nonhuman primate model to clinical application of ESC-based therapy, these findings emphasize the need to develop reliable rESC-specific protocols for hematopoietic differentiation. An improved understanding of the factors regulating hematopoietic differentiation from rESCs will facilitate subsequent transplantation studies in allogeneic hosts.3 a# |( l+ [9 ^: C" a

9 z- f/ U" T! G1 IDISCLOSURES
; M  ~+ R, C$ ^0 O
1 {2 Y" |) r1 e+ ]The authors indicate no potential conflicts of interest.
% r8 P( {. w6 N5 a2 F. T: v' q8 c" _( ^% j# F6 F
ACKNOWLEDGMENTS+ R4 ?7 l8 y" N9 O! I* \
: {% q' R0 {% |# B5 r$ @
We thank Daniel Webster for assistance with the qRT-PCR assays, Rachel Lewis for technical assistance with the rESC cultures, Maxim Vodyanik for assistance with the hESC/OP9 coculture technique, and James Byrne for technical assistance with the in vivo teratoma formation. This work was supported in part by a grant to the University of Wisconsin Medical School under the Howard Hughes Medical Institute Research Resources Program for Medical Schools (A.F.S.) and by a Faculty Research Fellowship Award from the American College of Surgeons (A.F.S.).
1 ^. o* g0 h# }  k) j9 e          【参考文献】: G, T* b8 X. A6 I, {, [8 |

- a0 \, Z% x  C* S$ B) s  X
% B4 D1 c  I* ?4 xShi PA, Hematti P, von Kalle C et al. Genetic marking as an approach to studying in vivo hematopoiesis: Progress in the non-human primate model. Oncogene 2002;21:3274¨C3283.
2 f) ~3 O/ t. H1 Z8 M
+ O' W! \- M3 I+ C/ HHematti P, Obrtlikova P, Kaufman DS. Nonhuman primate embryonic stem cells as a preclinical model for hematopoietic and vascular repair. Exp Hematol 2005;33:980¨C986.
; v. v/ D3 o. u- F* s3 C: x7 n0 O& m3 @. j' @8 f0 W# f
Kiem HP, Sellers S, Thomasson B et al. Long-term clinical and molecular follow-up of large animals receiving retrovirally transduced stem and progenitor cells: No progression to clonal hematopoiesis or leukemia. Mol Ther 2004;9:389¨C395.8 p0 r+ ^3 j0 R9 \9 [3 F# b
4 k6 r# u/ T( F; ^; L9 X# L
Tarantal AF, Han VK, Cochrum KC et al. Fetal rhesus monkey model of obstructive renal dysplasia. Kidney Int 2001;59:446¨C456.: y3 r4 l  @( V3 u# t
$ L' K- Y  \# B( o
Roth GS, Mattison JA, Ottinger MA et al. Aging in rhesus monkeys: Relevance to human health interventions. Science 2004;305:1423¨C1426.
; L' H  X7 W  M# P1 u0 ]/ A+ T6 c+ V' y4 B) X
Hendrickx AG, Makori N, Peterson P. The nonhuman primate as a model of developmental immunotoxicity. Hum Exp Toxicol 2002;21:537¨C542.
! {* K! \2 ]" Y" i2 k0 U% K" l' [- E3 w* x/ S) U& ^
Gelowitz DL, Rakic P, Goldman-Rakic PS et al. Craniofacial dysmorphogenesis in fetally irradiated nonhuman primates: Implications for the neurodevelopmental hypothesis of schizophrenia. Biol Psychiatry 2002;52:716¨C720.
4 v( N7 W" D8 @7 Z( J5 ~' q4 U4 y
Cornblath DR, Dellon AL, MacKinnon SE. Spontaneous diabetes mellitus in a rhesus monkey: Neurophysiological studies. Muscle Nerve 1989;12:233¨C235." u0 \8 S4 C. u* Q* u' o" M
4 h* o4 U" B9 \) W+ l; M2 g( d
Barr CS, Schwandt ML, Newman TK et al. The use of adolescent nonhuman primates to model human alcohol intake: Neurobiological, genetic, and psychological variables. Ann N Y Acad Sci 2004;1021:221¨C233.
; l8 }5 d0 K4 }7 L: a( d  c! h: p% E9 B0 G# B
Abbott DH, Foong SC, Barnett DK et al. Nonhuman primates contribute unique understanding to anovulatory infertility in women. ILAR J 2004;45:116¨C131.
1 o* t+ o( S+ P$ @7 O# J5 g
: c5 j' x2 m* j" |Lu SJ, Quan C, Li F et al. Hematopoietic progenitor cells derived from embryonic stem cells: Analysis of gene expression. STEM CELLS 2002;20:428¨C437.
* G- \. p6 W8 o' O+ [' q& h' F" c/ v* m- E2 t9 b1 d
Lester LB, Kuo HC, Andrews L et al. Directed differentiation of rhesus monkey ES cells into pancreatic cell phenotypes. Reprod Biol Endocrinol 2004;2:42.
6 l! L+ d) g- r- b. r' B. j/ B- W& w' k, B4 R
Li F, Lu S, Vida L et al. Bone morphogenetic protein 4 induces efficient hematopoietic differentiation of rhesus monkey embryonic stem cells in vitro. Blood 2001;98:335¨C342.  x) _7 P; j. X% o9 F+ V. A
' J6 z& R9 {% Y2 m( t0 J! a
Umeda K, Heike T, Yoshimoto M et al. Development of primitive and definitive hematopoiesis from nonhuman primate embryonic stem cells in vitro. Development 2004;131:1869¨C1879.5 G6 p( q4 i) b* M/ X$ g$ ?
$ T# v4 P, n* Y& n/ a' H
Umeda K, Heike T, Yoshimoto M et al. Identification and characterization of hemoangiogenic progenitors during cynomolgus monkey embryonic stem cell differentiation. STEM CELLS 2006;24:1348¨C1358.
, Q: q, g! s# U8 P4 u5 v# v
; d9 [9 B& R! ?6 wHiroyama T, Miharada K, Aoki N et al. Long-lasting in vitro hematopoiesis derived from primate embryonic stem cells. Exp Hematol 2006;34:760¨C769.
' D9 f" v1 T/ E) @% P. h
, ~7 X$ W; ^8 p2 _  x2 ISasaki K, Nagao Y, Kitano Y et al. Hematopoietic microchimerism in sheep after in utero transplantation of cultured cynomolgus embryonic stem cells. Transplantation 2005;79:32¨C37.* H  _+ E8 F+ }* @
( ]! d9 a& W5 T
Asano T, Ageyama N, Takeuchi K et al. Engraftment and tumor formation after allogeneic in utero transplantation of primate embryonic stem cells. Transplantation 2003;76:1061¨C1067.7 C$ B) E$ N) D

( S1 O) i5 ]/ x% h; WThomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145¨C1147.
2 J8 |3 E/ q- i( i7 T$ z& D( H# Y$ a+ g5 W! r
Amit M, Carpenter MK, Inokuma MS et al. Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture. Dev Biol 2000;227:271¨C278., U. A* E) Q7 }, w* u

! N4 @; Z  \3 c) D6 x; P1 {; tThomson JA, Marshall VS. Primate embryonic stem cells., O$ H& A% G) y- T" ?

7 `& j' S2 V1 B3 \5 Q2 s; PMitalipov S, Kuo HC, Byrne J et al. Isolation and characterization of novel rhesus monkey embryonic stem cell lines. STEM CELLS 2006;24:2177¨C2186.
. `: i4 f- M/ A! p* U, B8 E( l6 w0 i! j2 A7 y; n/ P
Xu C, Inokuma MS, Denham J et al. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol 2001;19:971¨C974 comment 2002;20:119.
2 i# K& P6 y% _" G6 L% F& W% C' t0 S; I! E4 y+ T
Carpenter MK, Rosler E, Rao MS. Characterization and differentiation of human embryonic stem cells. Cloning Stem Cells 2003;5:79¨C88.
! e! N. k$ @, F7 ^5 g
9 X& u9 j$ V' C4 \$ \Vodyanik MA, Bork JA, Thomson JA et al. Human embryonic stem cell-derived CD34  cells: Efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential. Blood 2005;105:617¨C626.
5 t0 X5 z1 }5 ?2 N3 @/ s1 B) \& h( d! H
Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 2001;29:e45.
( C: g3 b: K! b" V& v* z3 t9 {$ Z9 Z- \* F7 O0 c9 J1 Q9 T+ W
Kuo HC, Pau KY, Yeoman RR et al. Differentiation of monkey embryonic stem cells into neural lineages. Biol Reprod 2003;68:1727¨C1735.7 I. O9 I+ b, h, L/ w9 {
0 G2 K' q3 u, O6 [3 b: e
Bavister BD, Wolf DP, Brenner CA. Challenges of primate embryonic stem cell research. Cloning Stem Cells 2005;7:82¨C94.
% B. r( u7 A; @# w! L  T0 \1 N. x; L( w5 ^
Mitalipov SM, Kuo HC, Hennebold JD et al. Oct-4 expression in pluripotent cells of the rhesus monkey. Biol Reprod 2003;69:1785¨C1792.1 {9 e6 j0 _/ @6 M0 r4 _# B
5 c# ~, p( E/ `! f# b
Draper JS, Pigott C, Thomson JA et al. Surface antigens of human embryonic stem cells: Changes upon differentiation in culture. J Anat 2002;200:249¨C258.9 O& h9 f. q; r% L8 `. x

0 X! g/ }+ @* \# k4 ZHenderson JK, Draper JS, Baillie HS et al. Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. STEM CELLS 2002;20:329¨C337.2 h( q: @: u; Z7 @- M
6 E* ~9 y' j# P3 c
Thomson JA, Kalishman J, Golos TG et al. Pluripotent cell lines derived from common marmoset (Callithrix jacchus) blastocysts. Biol Reprod 1996;55:254¨C259.1 t. N! X  E2 t# n8 T7 P
+ n% ~, p, y7 }  }5 B) b  b0 q
Nishikawa SI, Nishikawa S, Hirashima M et al. Progressive lineage analysis by cell sorting and culture identifies FLK1 VE-cadherin  cells at a diverging point of endothelial and hemopoietic lineages. Development 1998;125:1747¨C1757.6 F9 b: V; E9 f+ L, x

! m  G% k9 y- ?* }. P# NMikkola HK, Fujiwara Y, Schlaeger TM et al. Expression of CD41 marks the initiation of definitive hematopoiesis in the mouse embryo. Blood 2003;101:508¨C516.
- E, w4 B8 z* ^2 z" v
% {4 A6 N9 v! S2 Zde Pooter RF, Cho SK, Carlyle JR et al. In vitro generation of T lymphocytes from embryonic stem cell-derived prehematopoietic progenitors. Blood 2003;102:1649¨C1653., j; s4 W* s6 d- M" I  X4 y3 C
& o$ |- s- @1 X
Chadwick K, Wang L, Li L et al. Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood 2003;102:906¨C915.
; Y* |' W/ h. W1 V8 t
+ @2 w5 v+ C0 }: r  D1 MFaloon P, Arentson E, Kazarov A et al. Basic fibroblast growth factor positively regulates hematopoietic development. Development 2000;127:1931¨C1941.* o1 s+ V+ }' h% M* R
5 s5 j% [) ~# y& b0 e( a" F( o% G
Robertson SM, Kennedy M, Shannon JM et al. A transitional stage in the commitment of mesoderm to hematopoiesis requiring the transcription factor SCL/tal-1. Development 2000;127:2447¨C2459." q! F4 \6 \: ]- ^
, \, h6 D* b8 ?
Leonard MW, Lim KC, Engel JD. Expression of the chicken GATA factor family during early erythroid development and differentiation. Development 1993;119:519¨C531.8 S. j  X- z; a3 p4 V) |. t( r
* W" b) u  G" B: s0 ]( r" x3 X
Weiss MJ, Yu C, Orkin SH. Erythroid-cell-specific properties of transcription factor GATA-1 revealed by phenotypic rescue of a gene-targeted cell line. Mol Cell Biol 1997;17:1642¨C1651.
* q, B* Q6 m; O  K. L8 n" R% @
+ C; }8 O7 _1 g& M. jOrkin SH. Embryonic stem cells and transgenic mice in the study of hematopoiesis. Int J Dev Biol 1998;42:927¨C934.
9 V! C" A) y( \4 a* k' j
; M& T; Y$ Q% e3 y, YMinegishi N, Suzuki N, Yokomizo T et al. Expression and domain-specific function of GATA-2 during differentiation of the hematopoietic precursor cells in midgestation mouse embryos. Blood 2003;102:896¨C905.
4 w1 U! Q. \) m) {' @4 Y% N& `0 R1 F9 r* y# a8 y. {
Pal S, Nemeth MJ, Bodine D et al. Neurokinin-B transcription in erythroid cells: Direct activation by the hematopoietic transcription factor GATA-1. J Biol Chem 2004;279:31348¨C31356./ c# E# C6 {3 \$ u3 }
" {9 \: U1 d4 N. ~9 k. d
Cheng T, Shen H, Giokas D et al. Temporal mapping of gene expression levels during the differentiation of individual primary hematopoietic cells. Proc Natl Acad Sci U S A 1996;93:13158¨C13163.2 Q. L' E% t7 M/ m
( V# P7 I! B$ |0 o3 V& [+ c  v- ~3 N
Kitajima K, Tanaka M, Zheng J et al. Redirecting differentiation of hematopoietic progenitors by a transcription factor, GATA-2. Blood 2006;107:1857¨C1863.  e" D& y! V) O- `# q7 ~& q6 @
& q" F, `' t3 D) n: C: M8 S
Ling KW, Ottersbach K, van Hamburg JP et al. GATA-2 plays two functionally distinct roles during the ontogeny of hematopoietic stem cells. J Exp Med 2004;200:871¨C882.
, Z0 s( C6 Q( W7 c* A$ x: H
- |+ G7 X: V( w3 v5 f: |North TE, Stacy T, Matheny CJ et al. Runx1 is expressed in adult mouse hematopoietic stem cells and differentiating myeloid and lymphoid cells, but not in maturing erythroid cells. STEM CELLS 2004;22:158¨C168.# j9 p0 C1 M# L& I6 g& Z8 \
' ?9 U& J! I9 c( u" m  ^
Robin C, Ottersbach K, Durand C et al. An unexpected role for IL-3 in the embryonic development of hematopoietic stem cells. Dev Cell 2006;11:171¨C180.
8 ?& }; H) m, L# b9 h4 n* \4 [* s9 U* l% m' }% |2 R
Nakagawa M, Ichikawa M, Kumano K et al. AML1/Runx1 rescues Notch1-Null mutation-induced deficiency of para-aortic splanchnopleural hematopoiesis. Blood 2006;8:3329¨C3334.
2 _( o4 F: y6 b7 U7 g* Y$ x
" m: B9 ^' z- j: x; N! DLorsbach RB, Moore J, Ang SO et al. Role of RUNX1 in adult hematopoiesis: Analysis of RUNX1-IRES-GFP knock-in mice reveals differential lineage expression. Blood 2004;103:2522¨C2529.
- S1 y# ]6 i: f/ |5 j4 k& Z2 q
2 v7 h" r4 A4 K8 h! a1 Q8 VIchikawa M, Asai T, Saito T et al. AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis. Nat Med 2004;10:299¨C304.( g5 o" N2 w! a8 H# Y7 q

- d5 ^1 D2 `. I& |Cai Z, de Bruijn M, Ma X et al. Haploinsufficiency of AML1 affects the temporal and spatial generation of hematopoietic stem cells in the mouse embryo. Immunity 2000;13:423¨C431.4 z: m' }8 [6 `6 v0 P; ?! b
* P4 c5 \( H; Q8 h+ ?9 d
Wang Z, Skokowa J, Pramono A et al. Thrombopoietin regulates differentiation of rhesus monkey embryonic stem cells to hematopoietic cells. Ann N Y Acad Sci 2005;1044:29¨C40.9 {, `, Q6 _/ p8 P( L! V: Z+ v! S! n

: ?7 N" T8 d/ X8 ALu SJ, Li F, Vida L et al. Comparative gene expression in hematopoietic progenitor cells derived from embryonic stem cells. Exp Hematol 2002;30:58¨C66.
! ]- e0 Y3 V8 k! g+ z5 d. G, }  x$ N( G2 O; v
Kaufman DS, Lewis RL, Hanson ET et al. Functional endothelial cells derived from rhesus monkey embryonic stem cells. Blood 2004;103:1325¨C1332.
- ?5 N( M' X9 |$ f2 h
& F( K# ]5 @4 f- b, @7 o, S0 l3 AKurita R, Sasaki E, Yokoo T et al. Tal1/Scl gene transduction using a lentiviral vector stimulates highly efficient hematopoietic cell differentiation from common marmoset (Callithrix jacchus) embryonic stem cells. STEM CELLS 2006;24:2014¨C2022.: P- P" J% `' t" x
' T9 ^6 [4 @0 e/ H$ i
Thomson JA, Kalishman J, Golos TG et al. Isolation of a primate embryonic stem cell line. Proc Natl Acad Sci U S A 1995;92:7844¨C7848.
7 ~! R9 ]$ L* L8 S( N& r3 y% K1 w0 v6 \! Y/ e+ C
Pau KY, Wolf DP. Derivation and characterization of monkey embryonic stem cells. Reprod Biol Endocrinol 2004;2:41.* t# M# ^3 R1 X, |9 O

' D  L2 I7 c* H7 _! d1 _Wolf DP, Kuo HC, Pau KY et al. Progress with nonhuman primate embryonic stem cells. Biol Reprod 2004;71:1766¨C1771.
, g# t7 O0 d2 ^" \4 H* U% E, {1 q+ ~5 i4 R( I4 ~8 X
Cho SK, Webber TD, Carlyle JR et al. Functional characterization of B lymphocytes generated in vitro from embryonic stem cells. Proc Natl Acad Sci U S A 1999;96:9797¨C9802./ Q1 A0 q6 }" ?7 T, X$ a
8 z' x6 G1 K6 L; p; F& C8 e
Dennis JE, Charbord P. Origin and differentiation of human and murine stroma. STEM CELLS 2002;20:205¨C214.
% p1 _- v7 D' o/ e) t
6 v% i. t) Y% ]Nakano T, Kodama H, Honjo T. Generation of lymphohematopoietic cells from embryonic stem cells in culture. Science 1994;265:1098¨C1101.
, \2 D1 l3 L' u9 F4 t# c. V( c5 P# b
Feugier P, Li N, Jo DY et al. Osteopetrotic mouse stroma with thrombopoietin, c-kit ligand, and flk-2 ligand supports long-term mobilized CD34  hematopoiesis in vitro. Stem Cells Dev 2005;14:505¨C516.
- F% B% |, n& v- [2 b2 Z$ a. O, {
6 I' J1 _9 \: d1 y" `# L; I) JZhang H, Saeki K, Kimura A et al. Efficient and repetitive production of hematopoietic and endothelial cells from feeder-free monolayer culture system of primate embryonic stem cells. Biol Reprod 2006;74:295¨C306.3 }, B/ y) |) }* L2 A3 l
, o  O6 \8 d7 J6 u3 n
Eaves CJ, Cashman JD, Kay RJ et al. Mechanisms that regulate the cell cycle status of very primitive hematopoietic cells in long-term human marrow cultures. II. Analysis of positive and negative regulators produced by stromal cells within the adherent layer. Blood 1991;78:110¨C117.% @; |% K  @8 N: J$ h

. v' Z+ M( u+ i- r9 d. c3 e% e. R1 cCashman J, Clark-Lewis I, Eaves A et al. Stromal-derived factor 1 inhibits the cycling of very primitive human hematopoietic cells in vitro and in NOD/SCID mice. Blood 2002;99:792¨C799.
$ `: o& `9 G( S- F' i' o) E5 a2 o! u, |. U* U9 t0 Z
Van Ranst PC, Snoeck HW, Lardon F et al. TGF-beta and MIP-1 alpha exert their main inhibitory activity on very primitive CD34 2CD38- cells but show opposite effects on more mature CD34 CD38  human hematopoietic progenitors. Exp Hematol 1996;24:1509¨C1515.
3 t* C+ M9 g/ f" e
; F) d0 f- }1 Y, s8 Q, u& ~) LSnoeck HW, Weekx S, Moulijn A et al. Tumor necrosis factor alpha is a potent synergistic factor for the proliferation of primitive human hematopoietic progenitor cells and induces resistance to transforming growth factor beta but not to interferon gamma. J Exp Med 1996;183:705¨C710.
% `3 H  |& a# d% E
5 [/ W. t! c6 R+ r: \' fAllouche M. Basic fibroblast growth factor and hematopoiesis+ Y: V# _5 [( ]

! ]  R1 e+ Y- F$ e: r6 C; i" I  JBerthier R, Prandini MH, Schweitzer A et al. The MS-5 murine stromal cell line and hematopoietic growth factors synergize to support the megakaryocytic differentiation of embryonic stem cells. Exp Hematol 1997;25:481¨C490.
" _! C4 b+ F  B$ F0 P
4 ~7 M5 o& M, }, ^4 O' m* o$ A: MJohansson BM, Wiles MV. Evidence for involvement of activin A and bone morphogenetic protein 4 in mammalian mesoderm and hematopoietic development. Mol Cell Biol 1995;15:141¨C151.
( U5 m; [# n5 c7 s$ ^" e' |4 @) w9 R
Magnusson PU, Ronca R, Dell'Era P et al. Fibroblast growth factor receptor-1 expression is required for hematopoietic but not endothelial cell development. Arterioscler Thromb Vasc Biol 2005;25:944¨C949 comment 2005;25:883¨C886.
7 u0 A" g9 m& x+ ^
! l7 d- w2 [# K- R2 GAllouche M, Bayard F, Clamens S et al. Expression of basic fibroblast growth factor (bFGF) and FGF-receptors in human leukemic cells. Leukemia 1995;9:77¨C86.- m5 v7 T) H* E- K  H) S
5 e0 [% ^, L" @2 _. j
Moroni E, Dell'Era P, Rusnati M et al. Fibroblast growth factors and their receptors in hematopoiesis and hematological tumors. J Hematother Stem Cell Res 2002;11:19¨C32.
& }( y+ ]1 w- Z: y2 a
1 M$ m, Z6 f9 Q1 r4 uHuber TL, Zhou Y, Mead PE et al. Cooperative effects of growth factors involved in the induction of hematopoietic mesoderm. Blood 1998;92:4128¨C4137.

Rank: 1

积分
威望
0  
包包
8  
沙发
发表于 2009-3-19 09:48 |只看该作者
谢谢,资料很丰富

Rank: 2

积分
64 
威望
64  
包包
1734  
藤椅
发表于 2015-6-4 22:54 |只看该作者
我来看看!谢谢  

Rank: 2

积分
107 
威望
107  
包包
1889  
板凳
发表于 2015-6-28 18:39 |只看该作者
干细胞之家微信公众号
回帖是种美德.  

Rank: 2

积分
97 
威望
97  
包包
1738  
报纸
发表于 2015-7-3 19:18 |只看该作者
我想要`~  

Rank: 2

积分
70 
威望
70  
包包
1809  
地板
发表于 2015-7-25 09:10 |只看该作者
其实回帖算是一种没德德,所以我快成圣人了  

Rank: 2

积分
97 
威望
97  
包包
1738  
7
发表于 2015-8-9 22:36 |只看该作者
看贴回复是好习惯  

Rank: 2

积分
116 
威望
116  
包包
1832  
8
发表于 2015-8-10 09:53 |只看该作者
努力,努力,再努力!!!!!!!!!!!  

Rank: 2

积分
72 
威望
72  
包包
1942  
9
发表于 2015-8-11 16:35 |只看该作者
原来是这样  

Rank: 2

积分
56 
威望
56  
包包
1853  
10
发表于 2015-8-13 12:50 |只看该作者
表观遗传学
‹ 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, 2024-5-6 11:02

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.