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: 369012|回复: 224
go

Isolation and Characterization of Novel Rhesus Monkey Embryonic Stem Cell Lines [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:01 |只看该作者 |倒序浏览 |打印
作者:Shoukhrat Mitalipova, Hung-Chih Kuob, James Byrnea, Lisa Cleppera, Lorraine Meisnerc,d, Julie Johnsonc,d, Renee Zeierc, Don Wolfa,e,f
. n9 j3 z9 w$ r- S                  4 }: O& S+ Z+ ~$ m" u3 k. Y4 |) j' i
                  
/ v0 g9 M# Q8 U8 x         
/ P$ B! I' m" A* S8 E4 _* z                         2 X) I& p$ o2 U2 g3 |  d
            9 M% t4 H& h! x/ X1 C- S0 r
            
. @3 G( P" [' i6 N6 B# R) x. d            : y) k; t/ m/ w5 }* H
            ; N1 q6 A6 C8 o. W5 ^8 K
                     
  r& r5 C# D4 y, g        + w+ h3 P# Q5 I& ?- _/ N
        
. `* O5 \. _* P  t        
: i: [  W; Y+ }          【摘要】* h& Y6 Q1 H: E  ]# ~
      ESCs are important as research subjects since the mechanisms underlying cellular differentiation, expansion, and self-renewal can be studied along with differentiated tissue development and regeneration in vitro. Furthermore, human ESCs hold promise for cell and tissue replacement approaches to treating human diseases. The rhesus monkey is a clinically relevant primate model that will likely be required to bring these clinical applications to fruition. Monkey ESCs share a number of properties with human ESCs, and their derivation and use are not affected by bioethical concerns. Here, we summarize our experience in the establishment of 18 ESC lines from rhesus monkey preimplantation embryos generated by the application of the assisted reproductive technologies. The newly derived monkey ESC lines were maintained in vitro without losing their chromosomal integrity, and they expressed markers previously reported present in human and monkey ESCs. We also describe initial efforts to compare the pluripotency of ESC lines by expression profiling, chimeric embryo formation, and in vitro-directed differentiation into endodermal, mesodermal, and ectodermal lineages.
1 ~6 s  z5 i6 `% g7 t2 O2 {4 n. w4 l          【关键词】 Embryonic stem cells Chimera Karyotype Rhesus monkey Transcriptome
9 S$ B  X  T' Q4 F1 \0 ^$ K                  INTRODUCTION
) q. Q. m5 e% E! W3 @6 ?3 h; C6 h% z
ESCs are important as research subjects since the mechanisms underlying cellular differentiation, expansion, and self-renewal can be studied along with differentiated tissue development and regeneration in vitro. Stem cells also provide the underpinning for the field of regenerative medicine and offer hope for the treatment of a wide range of human conditions that can be attributed to the loss or malfunction of specific cell types. Finally it is becoming increasingly apparent that stem cells could ultimately serve as a source of viable gametes . Nonhuman primates (NHPs), especially old world macaques, are valuable animal models because of their extensive use in biomedical research, their close phylogenetic relationship to humans, and, hence, their clinical relevance. Although the development of cell-based therapies can and does benefit from experimentation in rodent models, potential applications of human ESCs in regenerative medicine must be pioneered in NHP species prior to or in parallel with human research for scientific and ethical reasons.! P: y. T" q3 ~9 F! F
: |! c, s# \1 O7 C! }& N
Although more than 100 human ESC lines have been isolated and are either registered by NIH or in the scientific literature . The ESCs derived from humans and NHPs share similar growth, morphologic characteristics, and marker expression compared with each other and marked differences compared with the mouse. Furthermore, although interline differences are beginning to emerge in primate ESC lines, specific properties have not yet been adequately assessed in any species.) t: g- E3 V& V1 |; }/ L% s: @
( P8 h/ T$ g! Z. }
We have been interested in rhesus monkey ESC lines, initially with the intent of comparing lines established from in vitro derived versus in vivo derived blastocysts, as the latter represents a gold standard unavailable in the human . Here, we summarize our experience in the establishment of 18 ESC lines from rhesus monkey preimplantation embryos generated by the application of the assisted reproductive technologies at the Oregon National Primate Research Center. The newly derived monkey ESC lines were maintained in vitro without losing their chromosomal integrity, and they expressed markers previously reported present in human and monkey ESCs. We also describe initial efforts to compare the pluripotency of ESC lines by expression profiling, chimeric embryo formation, and in vitro directed differentiation into endodermal, mesodermal, and ectodermal lineages.
% V# l+ w- A2 U( ~# X, i5 Y0 @- S2 H7 J7 ~. N2 L/ G* w
MATERIALS AND METHODS8 h% y3 J+ q& r

* m5 T: b# N5 y$ d5 X: `# kOvarian Stimulation, Recovery of Rhesus Macaque Oocytes, Fertilization by Intracytoplasmic Sperm Injection, and Embryo Culture
" e$ h) S% y. |  U' b" z1 K* B( ~/ z: n) x! E
Controlled ovarian stimulation and oocyte recovery has been described previously . Briefly, sperm were diluted with 10% polyvinylpyrrolidone (1:4; Irvine Scientific, Santa Ana, CA, http://www.irvinesci.com), and a 5-µl drop was placed in a micromanipulation chamber. A 30-µl drop of TH3 was placed in the same micromanipulation chamber next to the sperm droplet, and both were covered with paraffin oil. The micromanipulation chamber was mounted on an inverted microscope equipped with Hoffman optics and micromanipulators. An individual sperm was immobilized, aspirated into an ICSI pipette (Humagen, Charlottesville, VA, http://www.humagenivf.com), and injected into the cytoplasm of a metaphase II-arrested oocyte, away from the polar body. After ICSI, injected oocytes were placed in 4-well dishes (Nalge Nunc International Co., Naperville, IL, http://www.nalgenunc.com) containing protein-free HECM-9 medium covered with paraffin oil and cultured at 37¡ãC in 6% CO2, 5% O2, and 89% N2. Embryos at the eight-cell stage were transferred to fresh plates of HECM-9 medium supplemented with 5% fetal bovine serum (FBS) (HyClone, Logan, UT, http://www.hyclone.com) and cultured for a maximum of 9 days, with medium change every other day.+ H/ z& l, c9 u& T2 v( B# |3 e+ r
  K5 t7 ~7 e0 w
ESC Derivation and Culture4 }6 S' x. \0 ?+ c) f0 s9 j
$ L. B# u4 \, x$ e4 {% }
Zonae pellucidae of selected expanded blastocysts were removed by brief exposure (45¨C60 seconds) to 0.5% pronase in TH3 medium. For immunosurgical isolation of inner cell masses (ICMs) , zona-free blastocysts were exposed to rabbit anti-rhesus spleen serum (a gift from Dr. J.A. Thomson) for 30 minutes at 37¡ãC. After extensive washing in TH3, embryos were incubated in guinea pig complement reconstituted with HECM-9 (1:2, vol/vol) for an additional 30 minutes at 37¡ãC. Partially lysed trophectodermal cells were mechanically dispersed by gentle pipetting with a small-bore pipette (125 µm in inner diameter; Stripper pipette; Midatlantic Diagnostics Inc., Marlton, NJ, http://www.midatlanticdiagnostics.com) followed by the rinsing of ICMs three times with TH3 medium. Isolated ICMs were plated onto Nunc four-well dishes containing mitotically inactivated feeder layers consisting of mouse embryonic fibroblasts (mEFs) and cultured in either Dulbecco's modified Eagle's medium (DMEM) with glucose and without sodium pyruvate (Invitrogen, Carlsbad, CA, http://www.invitrogen.com) supplemented with 1% nonessential amino acids (Invitrogen), 2 mM L-glutamine (Invitrogen), 0.1 mM ß-mercaptoethanol, and 20% FBS or DMEM/Ham's F-12 medium (DMEM/F12) (Invitrogen) with the same supplements but 15% FBS. ICMs that attached to the feeder layer and initiated outgrowth were manually dissociated into small-cell clumps with a microscalpel and replated onto new mEFs. After the first passage, colonies with ESC-like morphology were selected for further propagation, characterization, and low temperature storage. The medium was changed daily, and ESC colonies were split every 5¨C7 days manually or by disaggregation in collagenase IV (1 mg/ml, at 37¡ãC for 2¨C3 minutes; Invitrogen) and replating collected cells onto dishes with fresh feeder layers. Cultures were maintained at 37¡ãC, 3% CO2, and the balance air or 3% CO2, 5% O2, and 92% N2.; z) F1 O" E& h5 k, {

  T- e# @3 B( ~9 n: GEmbryoid Body Formation and In Vitro Differentiation of ESCs6 Z! X* M  V; e% q6 s) J

  D6 M( B4 {/ P: _8 y1 u% c5 h, O( fFor embryoid body (EB) formation, entire ESC colonies were loosely detached from feeder cells and transferred into feeder-free, 6-well, Ultra Low adhesion plates (Corning Costar, Acton, MA, http://www.corning.com/lifesciences) and cultured in suspension in ESC medium for 5¨C7 days. To induce further differentiation, EBs were transferred into collagen-coated, 6-well culture dishes (Becton Dickinson and Company) to allow attachment. To induce neuronal differentiation, medium was replaced with serum-free DMEM/F12 containing ITS supplement (insulin, transferrin, and sodium selenite; Invitrogen) and fibronectin (5 µg/ml; Invitrogen) . Differentiation into cardiac cells or retinal pigment epithelium was initiated by EB formation in suspension as described above. EBs were then plated into collagen-coated dishes, and cultures were maintained in ESC medium for 2¨C4 weeks.6 O/ f4 ]9 ?0 z, n; Y9 P! o
8 r$ f: g- d( x& a6 z3 o5 I( t
Immunocytochemical Procedures
, a' t- u8 t! u* k  L
, ^+ E$ M* q7 D3 e' P! F" n5 ?Undifferentiated and differentiated ESCs were fixed in 4% paraformaldehyde for 20 minutes. After permeabilization with 0.2% Triton X-100 and 0.1% Tween-20, nonspecific reactions were blocked with 10% normal goat serum (Jackson Immunoresearch Laboratories, West Grove, PA, http://www.jacksonimmuno.com). Cells were then incubated for 40 minutes in primary antibodies, washed three times, and exposed to secondary antibodies conjugated with fluorochromes (Jackson Immunoresearch Laboratories) before costaining with 2 µg/ml 4',6-diamidino-2-phenylindole for 10 minutes, whole-mounting onto slides, and examination under epifluorescence microscopy.
6 l' g! p; {7 L0 J3 u6 P  D6 e7 ]. O/ k. Y$ q6 \8 T
Primary antibodies were from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, http://www.scbt.com) (OCT-4, stage-specific embryonic antigen , and cardiac transcription factors GATA-4 and myocyte enhancer factor 2), ImmunoStar, Inc. (Hudson, WI, http://www.immunostar.com; serotonin), and R&D Systems Inc. (nestin). Musashi1 was kindly provided by Dr. H. Okano (Keio University, Tokyo, Japan).9 p! v% m) m2 }3 l# x
# F1 H7 F1 g0 Y# N
Reverse Transcription-Polymerase Chain Reaction
5 p4 m" x/ y& q& H. p4 I* o
: E, b3 {* s0 l+ t/ F) q) ^Total RNA was extracted from ESCs and ESC-derived differentiated phenotypes using RNA purification kit (Invitrogen) according to the manufacturer's instructions. Total RNA was treated with DNase I before cDNA preparation using SuperScript III First-Strand Synthesis System for reverse transcription-polymerase chain reaction (RT-PCR) (Invitrogen) according to the manufacturer's instructions. The first strand cDNA was further amplified by PCR using individual primer pairs for specific genes. The sequence, annealing temperature, and cycle number of each pair of primers are listed in supplemental online Table 1. All PCR samples were analyzed by electrophoresis on 2% agarose gel containing 0.5 µg/ml ethidium bromide.: @: b& j& ~+ i) A0 F0 m

9 o, I1 k- H6 ]1 ~3 P# E+ N/ m5 aTable 1. Transcriptome analysis of rhesus monkey ESC lines
$ Y& Y0 F) g( `% r. B1 ]8 W7 o3 j: Z1 Z- D+ @) K3 W" p; d3 u
Microarray Analysis
8 E5 U, x" k3 r( p
5 X/ b! x2 j- M# WTotal RNA was isolated as indicated above. Labeling, hybridization, and scanning was performed according to standard Affymetrix protocols (more details are given in Affymetrix GeneChip Expression Analysis Technical Manual, http://www.affymetrix.com). A microarray analysis was performed on a rhesus macaque Affymetrix GeneChip with 52,865 probe sets representing over 20,000 genes (protocols and Minimum Information About Microarray Experiment information are given in supplemental online data). The normalized microarray data were further analyzed using GeneChip Operating System (GCOS) 1.2. MAS-5 statistical analysis was performed to calculate the signal log ratio (SLR) for each probe set to determine the percentage of the transcriptome that significantly varied (p
* I0 T4 p: k5 g# P5 |. t0 _  u; j2 c; a; l5 ~" D  e
Cytogenetic Analysis( q8 S$ V  S7 a
8 v% d4 g) t; X: M5 o: P
Mitotically active ESCs in log phase were incubated with 120 ng/ml ethidium bromide for 40 minutes at 37¡ãC, 5% CO2, followed by 120 ng/ml colcemid (Invitrogen) treatment for 20¨C40 minutes. Cells were then dislodged with 0.25% trypsin and centrifuged at 200g for 8 minutes. The cell pellet was gently resuspended in 0.075 M KCl solution and incubated for 20 minutes at 37¡ãC, followed by fixation with methanol:glacial acetic acid (3:1) solution. Fixed cells were dropped on wet slides, air-dried, and baked at 90¡ãC for 1 hour. G banding was performed using trypsin-EDTA and Leishman stain (GTL) by immersing slides in 1x trypsin-EDTA with two drops of 0.4 M Na2HPO4 for 20¨C30 seconds. Slides were then rinsed in distilled water and stained with Leishman stain for 1.5 minutes, rinsed in distilled water again, and air-dried. For GTL-banding analysis, 20 metaphases were fully karyotyped under an Olympus BX40 microscope equipped with x10 and x100 plan-apo objectives. Images were then captured, and cells were karyotyped using a CytoVysion digital imaging system (Applied Imaging Corporation, San Jose, CA, http://www.aicorp.com).! X' r4 V% A8 j
/ p9 f# U: q' n- F
Chimeric Embryo Production. A- Y8 l  M) |' h
# _/ C: U/ S3 V: |/ Z# u, w+ b% U, e
Disaggregated ESC clumps (2¨C3 cells per clump) were labeled with the membrane-specific red fluorescent dye PKH-26, according to the manufacturer's directions, to identify and track ESCs within chimeric embryos. Cleavage stage (4¨C8-cell) rhesus monkey embryos were produced by ICSI as described above. A total of 15¨C25 labeled cells was injected into the perivitelline space of recipient embryos to ensure close contact with blastomeres. Injected embryos were cultured to the blastocyst stage, and the incorporation of cells into the chimeric embryo was monitored by epifluorescence microscopy every other day throughout preimplantation development. Cell numbers in blastocysts were quantitated by confocal microscopy.
; j- Z* H) S4 g" z! W3 x3 {; `6 L$ G/ w( z% o
RESULTS! R" J7 D: T' V5 ]
- U/ R* h) P# i5 \! j  y0 }/ m
Derivation and Propagation of Novel Rhesus Monkey ESC Lines
, c" `1 D5 H+ ?! D* E% a0 `0 e3 D: g; f  Z6 k
Expanded blastocysts with good morphology containing distinct, prominent ICMs were selected for ESC line derivation experiments. The majority (approximately 70%) of individually plated ICMs usually attached to the feeder layer within 2¨C3 days and initiated three-dimensional outgrowths consisting of tightly packed cells. After 5¨C7 days of culture, outgrowths were manually excised from the plates, dissociated into 5¨C10 smaller clumps with a microscalpel, and replated onto fresh mEFs. Approximately 50% of the replated ICMs generated one or two ESC-like colonies within 3¨C7 days, consisting of cells with a high nuclear to cytoplasmic ratio and prominent nucleoli (Fig. 1A). These colonies were isolated and passaged onto fresh feeders manually or with collagenase treatment for an additional 5¨C10 passages to generate sufficient cell numbers for cryopreservation, characterization, karyotyping, and in vitro differentiation studies. Eighteen ESC lines (designated Oregon rhesus monkey embryonic stem 1¨C18 . DMEM/F12 supported faster growth rates (passage interval, 5¨C7 days) than DMEM (8¨C10 days), even though the FBS supplementation in DMEM/F12 was lower than that of DMEM. The efficiency of ESC line isolation was also significantly higher (p
( y& [. A+ W1 _* {8 g: |; f  s3 V) ]6 e9 C' O* Q/ p
Figure 1. Rhesus monkey ESC morphology, showing undifferentiated and differentiated markers detected by immunocytochemistry. (A): Phase-contrast micrograph of an ESC colony growing on mouse embryonic fibroblasts (mEFs). (B): ESCs grown on mEFs were fixed with paraformaldehyde and labeled with primary antibodies against specific markers and secondary antibodies conjugated with Cy3 fluorochrome (red, right panel) and costained with DAPI (blue, left panel). Note that only colonies of ESCs are positive for tested markers of stem cells, whereas feeder cells were negative. (C): Differentiation of monkey ESCs into ectodermal lineages. a, Nestin expression in progenitor cell populations. b, Phase-contrast micrograph of monkey ESC-derived neuronal phenotypes. c, Expression of neuronal marker TujIII. d, Phase-contrast micrograph of pigmented clusters of retinal pigment epithelial cells. e, Polygonally shaped morphology of pigmented and surrounding non-pigmented epithelial cells. f, Pigmented and nonpigmented epithelial cells coexisted with other unidentified nonepithelial cells. g, Multilayered cultures with pigmented or nonpigmented epithelial cells growing on the top layers and nonepithelial, highly organized cells growing under epithelial layers. h, Both pigmented and nonpigmented monkey ESC-derived epithelial cells strongly reacted with a monoclonal antibody to CRALBP. Scale bars = 100 µm (a, b, c, f, g, and h), 50 µm (e), 200 µm (d). Abbreviations: CRALBP, cellular retinaldehyde binding protein; DAPI, 4',6-diamidino-2-phenylindole; SSEA, stage-specific embryonic antigen; TRA, tumor-related antigen; TujIII, ß-III-tubulin.
; J# I. C8 ~3 }
! y" t& w0 @- y& d. g% |Table 2. Genes with the greatest average fold change in monkey ESCs compared with the somatic cell control
3 K0 H1 G8 o0 ]. ~! J0 h2 N8 P) J; L& ~! X# Q& e, y
Although DMEM/F12 was associated with higher derivation efficiency and improved ESC growth properties, pH stability was problematic under 5% CO2 conditions. Maintenance of medium pH in the optimal 7.2¨C7.4 range was achieved, however, when the CO2 level was reduced to 3%. Recognizing that optimal growth and development of preimplantation stage monkey embryos from which ESCs are derived is achieved under reduced oxygen levels , culture in 5% O2 was evaluated. Cells grew at a rate similar to controls; however, a lower incidence of spontaneous differentiation was noted (results not shown). Notable differences in growth rates were observed between ORMES cell lines with passage intervals ranging between 5 and 7 days for fast-growing (ORMES-6, -7, and -13) and between 14 and 21 days for slow-growing lines (ORMES-4 and -12) under comparable conditions. The latter were also characterized by poor survival after conventional freezing. The slow growth rate of ORMES-4 was evident during initial ESC line establishment steps and was maintained at later passages. In contrast, the initial slow growth rate of ORMES-12 was overcome during extensive propagation.
9 y% u$ V! H. a
0 x  {7 f5 I0 i# C7 ~7 c  `! vMonkey ESCs are extremely sensitive biological materials, and reproducible propagation of the undifferentiated phenotype is associated with rigorous optimization of culture conditions and protocols. Quality control of media and media components, plasticware, and equipment generally involved with ESC culture was critically important in trouble-free maintenance of ESCs. For instance, toxic effects of several brands or lots of culture dishes were observed uniquely with monkey ESCs (results not shown), as other cell types such as fibroblasts did not display negative effects. ORMES cell lines were also sensitive to mEF quality. Feeder plates prepared from early passage mEFs (passages 2¨C3) supported better undifferentiated cell growth than those prepared from later passages (passage 5¨C10). We also found that monkey ESCs were sensitive to FBS preparations, necessitating that each new lot be tested for the ability to support ESC growth before purchase. In contrast to human ESCs , we found no noticeable effect of bFGF on rhesus monkey ESC growth. When growth medium was supplemented with 4 ng/ml bFGF, undifferentiated growth on mEFs and passage intervals of several tested ORMES cell lines were similar to controls. Several ORMES cell lines were adapted to grow in medium supplemented with 20% knockout serum replacement instead of FBS, resulting in slightly slower growth rates and minor changes in morphology.
2 C- \3 `" T' q0 P' j! d3 e$ h6 O8 e0 `5 w* z! a" {
In Vitro Characterization of Monkey ESC Lines) N+ y9 B' g2 n4 O! O6 c

3 k6 z; S* p- d. {& T3 ?All established ESC lines expressed key pluripotent markers, including OCT-4, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, and alkaline phosphatase as detected by immunocytochemistry (ICC) or immunohistochemistry (Fig. 1B). In addition, monkey ESCs also reacted to antibodies against other pluripotency-related products, including NANOG, the forkhead transcriptional regulator FOXD3, and phosphatidyl-anchored cell surface glycoprotein THY-1. SSEA-1 was not expressed in these lines (results not shown). RT-PCR analysis was used to further assess expression of genes characteristic of pluripotent cells, including POU5F1 (OCT4), NANOG, FOXD3, DPPA5, CRIPTO, acidic zinc finger gene REX-1, transcription factor SOX-2, growth factors FGFR-4 and LEFTYA, telomerase-related TERF1, TERF2, and TERT, ABC transporter BCRP-1/ABCG-2, and the gap junctional components CONNEXINS 43 and 45. All ORMES cell lines expressed these genes, with the exception of the REX-1 gene (supplemental online Fig. 1). RT-PCR with four previously published primers  and one set of primers based on the human REX-1 gene sequence failed to amplify any product in monkey ESCs. Overall, our results demonstrate that the ORMES cell lines express markers previously reported present in mouse (except SSEA-1), human, and monkey ESCs.
  k, f+ I; v- u- O7 j* n; x
$ R+ j) D2 M( f: o# {Microarray Analysis
9 _# _7 e$ S% [# P5 H1 }* ]' s
6 G/ E, @5 D. v3 `% g% m, jTo further investigate the transcriptional activity of monkey ESCs and begin defining "stemness" genes, we conducted genome-wide expression profiling of several ORMES cell lines. Initially, two samples from a single ESC line (ORMES-6) at passage 41 were collected from the same culture dish (ORMES-6A and ORMES-6B) and compared with each other and with a culture of adult rhesus monkey skin fibroblasts. Only minimal variation (1.2%) was observed between the two ORMES-6 samples. However, significant variation was observed when each of these samples was compared with the somatic cell control (25.4% and 26.1%, respectively).
% p# k: f. @# X+ R# O* V! I& s2 D: O6 l- {
Next, the transcriptomes of five ORMES cell lines were compared with each other using GCOS and to adult rhesus monkey skin fibroblasts (Table 1). The transcriptome of each ORMES cell line was consistently different from the calculated average by approximately 11%, a value that was remarkably consistent. The transcriptome of each ORMES cell line was different from the control somatic cell line by 25%¨C29%. The percentage of the transcriptome with a change of more than twofold was only 4% for each ORMES cell line compared with the ESC average, but it was still over 17%¨C18% compared with the somatic control. This filtered result suggests that the majority of the differences observed between ORMES cell lines were relatively small. Despite differences, all analyzed ORMES cell lines consistently and strongly expressed a subset of stem cell marker genes. We calculated the average fold change for each probe set and then selected 25 ontologically identifiable genes with the greatest average fold change compared with the somatic cell control (Table 2).( K  f9 `4 {" x, |
- s, b& K' F8 x9 o/ ^. k( ^
Several stem cell markers were highly expressed in all five ORMES cell lines, as expected, including NANOG, LIN-28, PODXL, POU5F1, and GDF-3 (Table 2 in bold). Subsequent semiquantitative RT-PCR analysis was used to validate expression of NANOG and POU5F1 in ORMES cell lines (results not shown).
3 q  c5 R" ?, }6 W, L
0 ]) i+ m" b3 ~" ]& jCytogenetic Analysis+ G; u3 ?  a* P% x9 k/ T

& ?' E! D, n. p4 p( SDetailed G-banding analysis of ORMES cell lines revealed that 15 were karyotypically normal, with a diploid set of 42 chromosomes. The anticipated even distribution of male (n = 9) and female (n = 9) lines was observed (Table 3). ORMES-1 carried a stable, balanced 11;16 translocation, and ORMES-2 had balanced 5;19 and 1;18 translocations based on the presence of these abnormalities at early and subsequent passage numbers. ORMES-5 demonstrated a pericentric inversion involving chromosome 1.
+ T0 u1 Q0 B  o! b+ K; Z* j  H2 r7 v. j
Table 3. Derivation conditions and karyotype of rhesus monkey ESC lines 1¨C18; all lines were cultured on mEFs; o8 v! P9 m8 z* A( ^
7 [0 p4 }9 v; G4 A: y0 g" o- q0 `
ESC Line Pluripotency
$ ]8 R0 ?7 v" L9 m6 v4 n( q9 r+ a1 ~, L7 N
To determine whether newly established ESC lines can give rise to cell lineages representative of all three embryonic germ layers, we applied in vitro approaches designed to induce differentiation into neural, retinal (ectodermal), cardiac (mesodermal), and pancreatic (endodermal) lineages via EB formation.$ U3 _. O7 u$ m
: I) i* j) w5 R* P3 e4 @
Cardiac Differentiation6 n9 o- c: _$ o( V7 ]
9 `; ?: `: H  U/ w
Mesodermal differentiation of several ORMES cell lines involved EB production in suspension culture for 5¨C7 days followed by plating them onto collagen-coated dishes for adherent culture. Approximately 7¨C14 days after plating, attachment, and further spontaneous differentiation, contracting cell aggregates were observed in all tested ORMES cell lines (Table 4). Analysis of these aggregates by ICC revealed expression of cardiac-specific markers, including cTnI and cTnT, -MHC, sMHC, SERCA2, ANP, tropomyosin, -actinin, MLC-2V, MLC-2A, and cardiac transcription factors GATA-4 and myocyte enhancer factor 2 (supplemental online Fig. 2a¨C2d) .8 }! c2 i1 `7 |. ?) T8 b

* A8 R: M- \% @1 Z  {9 e; F$ Y2 YTable 4. In vitro differentiation of ORMES cell lines: _8 a( `% i! ]3 b

" i2 _; N& z" i/ t6 W. _Figure 2. Contribution of Oregon rhesus monkey embryonic stem 1 cells in the preimplantation development of chimeric monkey embryos. (A): Approximately 10¨C15 disaggregated ESCs were injected into monkey embryos at the 4¨C8-cell stage. (B): Epifluorescence microscopy of injected ESCs labeled with the membrane-specific fluorescent dye PKH-26. (C): Hatching blastocyst developed following injection of ESCs. (D): Epifluorescence microscopy of the same blastocyst showing incorporation of fluorescent cells into both the trophectoderm and inner cell mass.  H6 P+ e0 `0 U( U8 G

( t4 I& C+ ^7 K. ^+ p) bNeural Differentiation
! \$ ^( [% U: h, f) B8 J4 Y
9 @* f. h1 h+ h, JDifferentiation into neuronal phenotypes was induced using a stepwise approach originally developed for mouse ESCs and adapted by us for primate cells . Between 10% and 30% of cells in the differentiated population expressed the glial cell marker GFAP.
% @, M. U  r/ l& j8 b+ p- C. u1 w
, q4 Z4 ]% w* z# G) qRetinal Differentiation8 m  o9 J+ Q: [
& [# d& m6 F% j
Differentiation into dark pigmented epithelial cells resembling retinal pigment epithelial (RPE) was observed during EB formation in suspension culture for 5¨C7 days followed by plating EBs onto collagen-coated dishes to allow attachment and adhesion as described above for cardiac differentiation. After approximately 2¨C3 weeks of adherent culture, clusters of darkly pigmented cells were observed (Fig. 1C, d) that grew in size and eventually become visible to the naked eye at a density of 2¨C3 clusters per 35-mm plate. Pigmented epithelial cells appeared polygonally shaped, similar to those observed in cultured retinal cells derived from human ESCs (Fig. 1C, e) . Both pigmented and nonpigmented epithelial cells in these cultures expressed CRALBP (Fig. 1C, h), whereas bestrophin expression was correlated with the level of pigmentation.
) K, p' m  p/ E! l6 t/ p. c2 k. U% N; G! C  A- F0 v
Pancreatic ß-Cell Differentiation7 S* w( T: t0 }, ^% s
, y' ?7 t6 a" F; W' H# s6 j, G
Populations of progenitor cells expressing nestin were subjected to protocols using exendin-4 and nicotinamide to redirect the differentiation into endocrine phenotypes .
# [7 Z9 t5 g' n. |  `; L. R5 @2 a' ]  D, b8 }
Chimera Production
% F) y! ^: k' a9 t3 k  p
2 ]: G5 p* P3 d' bIn an effort to further define the potency of monkey ESCs, we documented the participation of ORMES-1 cells in the preimplantation development of chimeric monkey embryos. Approximately 10¨C15 ESCs labeled with PKH-26 were injected into monkey embryos at the 4¨C8-cell stage produced by ICSI, and the resulting chimeric embryos were cultured in vitro to the blastocyst stage. In five replicates involving 29 embryos, development progressed to the compact morula (19 of 29; 65%) and blastocyst (12 of 29; 41%) stages at rates comparable to nonmanipulated control counterparts. Incorporation of fluorescent cells was observed in all chimeric embryos throughout the preimplantation development period up to the day 8 expanded blastocyst stage with evidence of integration into both the trophectoderm and ICM (Fig. 2). As determined by confocal microscopy, total cells numbers in chimeric (n = 8) and noninjected control (n = 6) blastocysts were not significantly different (527 ¡À 45 , respectively; p > .07; Mann-Whitney test). An ESC contribution to chimeric embryos was evidenced by the presence of 49 ¡À 7 (SEM) fluorescent cells. One clinical pregnancy was established following the transfer of 15 chimeric embryos at various preimplantation stages into five recipients that aborted at 5 weeks.
  r9 x5 v  W4 v9 w4 Y8 k
9 l5 s6 R2 h+ d- ]" FDISCUSSION
, X; y: q; ]% A1 y0 m- n) L9 f: x; Y1 Z& g) U. ]9 {7 c' E" }" V0 G
ESC line banking efforts provide several insights into factors affecting the isolation and propagation of primate ESC lines. Although the average efficiency of derivation (27 ¡À 6) was comparable to the reported experience with human and nonhuman primate ESCs . Unquestionably, embryo quantity is another factor in the equation for success.
4 N6 D  L& `8 `6 d/ H4 l. `4 ]$ |
- @% X. Y( b: r+ N: _ICC analysis confirmed expression of well-defined pluripotent markers in these novel monkey ESC lines, corroborated by PCR analysis. These genes include POU5F1, NANOG, TERT, FOXD3, SOX2, TERF1, TERF2, and LEFTYA; however, in contrast to mouse ESCs, REX-1 gene expression was not detected in undifferentiated ORMES cell lines using several primer sets designed for the human sequence. Microarray expression data confirmed the absence of REX-1 transcripts in ORMES cell lines, and it is worth noting the inconsistency in REX-1 expression among several human ESC lines, suggesting that expression of this gene is not essential for self-renewal of primate ESCs .
+ D$ O3 i6 l# @, e# G9 s
6 s4 h4 v. X$ ?3 p3 \6 T& hAnother characteristic of ESCs is their ability to maintain normal karyotype during prolonged culture in vitro. Cytogenetic analysis of ORMES cell lines revealed that the majority of lines maintained a normal karyotype with a diploid set of 42 chromosomes. Both male (n = 9) and female (n = 9) cell lines were equally represented, in agreement with the conclusion that primate pluripotent cell lines from both sexes can be efficiently derived and maintained in vitro . Notably, these changes were associated with exposure to an extended feeder-free culture system on Matrigel or to enzymatic passaging using collagenase, trypsin, or cell-dissociation buffer. Thus, it is possible that the collagenase-based splitting technique used for ORMES-1 to -6 may have contributed to the karyotypic abnormalities in ORMES-1, -2, and -5.3 N% ?* }! C- u& s, ~- J7 X5 J6 T

2 ^6 x9 [( ~9 M; ^Despite close similarities in the morphology, marker expression properties, and karyotypes of ORMES cell lines, important differences are also apparent. Because of the implications of this observation to the long-term objective of cell-based therapeutic applications, we have begun a comparison of monkey ESC lines. Differences in growth rates and survival following cryopreservation were noted during the establishment of some ORMES cell lines that persisted during more extensive propagation. In contrast, growth rates of other cell lines changed over time at later passages, perhaps reflecting adaptation to culture conditions, inadvertent selection of a different phenotype, or even genetic and/or epigenetic changes.
4 H4 L% j3 ^3 [4 z9 {# N( T+ }, o% p6 I
With regard to the gene expression profile, ORMES cell lines exhibited similar but not identical transcriptomes. Expression profiling by rhesus monkey Affymetrix oligonucleotide array revealed that 11%¨C12% of the transcriptomes from the five different ORMES cell lines demonstrated significant changes in gene expression compared with the pooled ESC average (a shared significant homology in gene expression of 88%¨C89%). Approximately 4% of the compared transcriptomes demonstrating a fold change greater than two (Table 1). The majority of this transcriptional variation must be biologically significant, as we observed only 1.2% transcriptional variation between biological replicates that was likely introduced during the RNA purification and microarray analysis process (RNA amplification, cDNA processing, washing, scanning, etc.). The shared significant homology in gene expression between the five different rhesus ESC lines examined at 88%¨C89% is comparable with microarray comparisons between human ESC lines, where the shared significant homology in gene expression is 85% . Despite the observed transcriptome differences between ESC lines, all of the ORMES cell lines examined strongly expressed key markers necessary to maintain a stem cell state, including POU5F1, NANOG, LIN-28, PODXL, and GDF-3 (Table 2). RT-PCR analysis confirms that NANOG and POU5F1 are present only in undifferentiated and not in differentiated ORMES cells. This suggests that it may not be the entire transcriptome that dictates the embryonic stem cell state, but only a subset of key stemness genes. Perhaps as long as key stemness genes, such as NANOG, LIN-28, PODXL, POU5F1, and GDF-3, are strongly expressed, the ESC transcriptomes possess a degree of biological plasticity that can accommodate a limited degree of transcriptional variation while still maintaining an undifferentiated embryonic stem cell state.
( S5 ?! v$ W  j* C9 z0 P/ s1 q- \0 n$ q$ M
The ultimate measure of pluripotency is demonstrating ESC participation in chimeric animals, a well-defined and frequently exercised option in mice . Our results demonstrated that chimeric blastocysts could be produced readily in the rhesus monkey by ESC injection, setting the stage for future potency determination in chimeric fetuses or offspring. Although the fluorescent signal from PKH-26 made it impossible to trace ESC fate in chimeric fetuses or offspring during long-term studies, creating genetically modified ESC lines carrying reporter genes, such as eGFP, would appear feasible given adequate resources., G) j$ s/ t8 Z" q4 n3 W+ J) q
4 A. s5 w7 u3 c* L% O
In conclusion, we have summarized our experience in generating 18 new ESC lines from rhesus monkey blastocysts generated in vitro. This bank of ESC lines, augmented by in vivo-derived lines  that can serve as a gold standard, represents a valuable resource. Future plans include continued interline comparative studies and creating additional MHC-typed and xeno-free monkey ESC lines in anticipation of transplantation trials.4 L* t/ S0 t$ ?3 ?# R: C
. `" s. s$ r0 w, P
DISCLOSURES6 t) m8 ^0 l; C' C" w$ T, J' \
% Z  a2 G! G9 H) T) J6 l
D.W. has acted as a consultant for the NIH within the last 2 years.
$ X7 M& K+ d) l) o/ R1 c- A  n: n' A
ACKNOWLEDGMENTS
# p4 g3 Q4 V- D( `4 V8 e- s& {9 }; t0 o* R
We acknowledge the Division of Animal Resources, the Endocrine Services Core, and the Molecular Biology Core at the Oregon National Primate Research Center and Affymetrix Microarray Core at the Oregon Health and Science University for their assistance and technical services. We are grateful to Cathy Ramsey, Christine Gagliardi, Carrie Greenberg, Michelle Sparman, and Ching-Yu Chuang for technical assistance; Dr. John Fanton and Darla Jacobs for laparoscopic oocyte retrieval; Julianne White for administrative support; and Joel Ito for help with illustration materials. The Assisted Reproductive Technologies/ESC core facility assisted by providing semen samples, media, and mEF feeder layers for ESCs. Rabbit anti-rhesus spleen whole serum was developed by and obtained from Dr. J.A. Thomson, Wisconsin National Primate Research Center, University of Wisconsin-Madison. This study was supported by NIH Grants RR00163 (Dr. D. Dorsa, Oregon Health & Science University), HD18185 (Dr. R.L. Stouffer, Oregon Health & Science University), and RR15199 (D.W.) and by an Academia Sinica of Taiwan grant (H.-C.K.).+ D0 P2 u5 T5 H
          【参考文献】% L/ E0 z( Z6 d* Z

1 u1 R- W) c: N( ~( E" u& ~5 H( n' R
Hubner K, Fuhrmann G, Christenson LK et al. Derivation of oocytes from mouse embryonic stem cells. Science 2003;300:1251¨C1256.5 }9 N  F9 ?3 L, b/ H( a! f

; S4 C! w+ b  G, uToyooka Y, Tsunekawa N, Akasu R et al. Embryonic stem cells can form germ cells in vitro. Proc Natl Acad Sci U S A 2003;100:11457¨C11462.
( t6 t- J2 o! D; n
) L3 o! t/ z1 ]Geijsen N, Horoschak M, Kim K et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 2004;427:148¨C154.+ @$ ?/ V  X' F. N
4 x2 m3 _+ |3 X5 x5 |
Thomson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145¨C1147.6 `/ [2 W5 ^4 \6 H& X# [

* \6 y) X# T3 n7 p# S/ nPickering SJ, Braude PR, Patel M et al. Preimplantation genetic diagnosis as a novel source of embryos for stem cell research. Reprod Biomed Online 2003;7:353¨C364.0 P# X$ J/ i$ o

) j' E5 d1 m2 c( n, qMitalipova M, Calhoun J, Shin S et al. Human embryonic stem cell lines derived from discarded embryos. STEM CELLS 2003;21:521¨C526.
4 m. M# ^% K4 D) u) d0 K, b- Y  K5 C
Cowan CA, Klimanskaya I, McMahon J et al. Derivation of embryonic stem-cell lines from human blastocysts. N Engl J Med 2004;350:1353¨C1356.
3 v" u" S3 Y+ @# g- R0 E( l/ _
0 f+ i3 q+ |+ c/ D, l+ QHeins N, Englund MC, Sjoblom C et al. Derivation, characterization, and differentiation of human embryonic stem cells. STEM CELLS 2004;22:367¨C376.
( o8 d* W* E( Y0 U7 Q5 _  i$ u' R4 b
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.
8 G8 b, \& Y  x/ U+ w* X8 u: N. v% t: r4 `
Marshall VS, Waknitz MA, Thomson JA. Isolation and maintenance of primate embryonic stem cells. Methods Mol Biol 2001;158:11¨C18.; Y" s: ]5 H$ p" F
( J* w" Z/ B' v& a
Thomson JA, Kalishman J, Golos TG et al. Pluripotent cell lines derived from common marmoset (Callithrix jacchus) blastocysts. Biol Reprod 1996;55:254¨C259.
% v) X+ C! i! b, T* |" g& L' y- @7 L8 t1 ]0 m$ v+ J! X
Sasaki E, Hanazawa K, Kurita R et al. Establishment of Novel Embryonic Stem Cell Lines Derived from the Common Marmoset (Callithrix jacchus). STEM CELLS 2005;23:1304¨C1313.- W5 I3 B( P; ]7 @
* a# Y8 o( E6 S) z3 @
Suemori H, Tada T, Torii R et al. Establishment of embryonic stem cell lines from cynomolgus monkey blastocysts produced by IVF or ICSI. Dev Dyn 2001;222:273¨C279.
8 L3 h5 i& f: M+ N) S1 k6 [
" ]; [6 H7 |! e! l0 }7 Z! tCibelli JB, Grant KA, Chapman KB et al. Parthenogenetic stem cells in nonhuman primates. Science 2002;295:819.
* N$ x' t2 T% o- x- K
3 L! F4 }, r& l& pBavister BD, Wolf DP, Brenner CA. Challenges of primate embryonic stem cell research. Cloning Stem Cells 2005;7:1¨C13.7 }) L" b1 }( K9 B

% B1 F/ B& K3 \3 C( h1 SFujimoto A, Mitalipov SM, Kuo HC et al. Aberrant genomic imprinting in rhesus monkey ESCs. STEM CELLS 2006;24:595¨C603.
2 k! ?( }$ x8 m3 \1 W6 }/ t( D+ O
Fujimoto A, Mitalipov SM, Clepper LL et al. Development of a monkey model for the study of primate genomic imprinting. Mol Hum Reprod 2005;11:413¨C422.
2 K& N( _$ W) d! \
: G" }, F$ T" d- Q5 f& qZelinski-Wooten MB, Hutchison JS, Hess DL et al. Follicle stimulating hormone alone supports follicle growth and oocyte development in gonadotrophin-releasing hormone antagonist-treated monkeys. Hum Reprod 1995;10:1658¨C1666.) ?0 i  j! Z8 ]9 j
! \6 t* n3 U% f! ~( ]6 j9 {
Bavister BD, Yanagimachi BD. The effects of sperm extracts and energy sources on the motility and acrosome reaction of hamster spermatozoa in vitro. Biol Reprod 1977;16:228¨C237.
- \! k' }  \: ?1 b5 F; q- C0 R1 K1 v; |8 G4 H/ \& \5 h$ u
McKiernan SH, Bavister BD. Culture of one-cell hamster embryos with water soluble vitamins: Pantothenate stimulates blastocyst production. Hum Reprod 2000;15:157¨C164.
' Z' O, d8 q: {* }& |0 ^8 J0 z5 [& V# I2 m" x( n" Y
Wolf DP, Thormahlen S, Ramsey C et al. Use of assisted reproductive technologies in the propagation of rhesus macaque offspring. Biol Reprod 2004;71:486¨C493.
6 a, N: }8 s1 \6 {+ R8 {1 s* s5 }# ~3 U1 @% G! v( q
Solter D, Knowles BB. Immunosurgery of mouse blastocyst. Proc Natl Acad Sci U S A 1975;72:5099¨C5102.
( _0 h" _+ h" C- X
, i2 o# R- i: O5 _3 P. z1 bKuo HC, Pau KY, Yeoman RR et al. Differentiation of monkey embryonic stem cells into neural lineages. Biol Reprod 2003;68:1727¨C1735.* ~6 w. P% V3 z' W( a( G
6 B4 r5 N; L* ^) D
Lester L, Kuo H-C, Andrews L et al. Directed differentiation of rhesus monkey ESCs into pancreatic cell phenotypes. Reprod Biol Endocr Online 2004;2:42.: e. j4 ?) Y; j
1 h! l8 D6 x3 b3 j' y
Ginis I, Luo Y, Miura T et al. Differences between human and mouse embryonic stem cells. Dev Biol 2004;269:360¨C380.
- f5 v/ ?& E  O! l) {: u+ X& {& r  I4 n7 N. i) e5 x7 d1 p1 B
Kuo H-C, Chuang C-U, Lin H et al. In vitro characterization and transplantation of non-human primate embryonic stem (ES) cell-derived cardiomyocytes (CM). Paper presented at: 3rd Annual Meeting of the International Society for Stem Cell ResearchJune 23¨C25, 2005San Francisco, CA.
9 ^8 v* A! Q, ?1 U$ o" M& e5 |* ]4 n
% v+ d  E5 ]$ P5 E4 u+ o* SKuo H-C, Wolf D. Differentiation of rhesus monkey embryonic stem cells into cardiomyocyte phenotypes. Paper presented at: ARTs in Action in Non-human Primates. Post-conference Symposium Associated with the 2004 IETS Annual MeetingJanuary 10¨C13, 2004Portland, OR.
+ Z  P4 s# }& C* c$ k) j% a9 W5 l* Z( f; V; G
Salli U, Reddy AP, Salli N et al. Serotonin neurons derived from rhesus monkey embryonic stem cells: Similarities to CNS serotonin neurons. Exp Neurol 2004;188:351¨C364.
# d2 e2 G; q" e% j
4 p3 @& f3 ~" Z# ZKlimanskaya I, Hipp J, Rezai KA et al. Derivation and comparative assessment of retinal pigment epithelium from human embryonic stem cells using transcriptomics. Cloning Stem Cells 2004;6:217¨C245.! U3 P% x9 M6 X3 g) x2 d$ ?

8 Q" k/ a9 w. b# N! e2 t" D. pBakall B, Marmorstein LY, Hoppe G et al. Expression and localization of bestrophin during normal mouse development. Invest Ophthalmol Vis Sci 2003;44:3622¨C3628.3 n7 s. ?5 u: N& m+ ~7 _; M

% g; d9 s/ E$ Y$ F. w% S3 QRichards M, Tan SP, Tan JH et al. The transcriptome profile of human embryonic stem cells as defined by SAGE. STEM CELLS 2004;22:51¨C64.! K4 W! c% ^' W
0 l* |) E7 N" t: d0 L$ R) k
Robertson EJ, Evans MJ, Kaufman MH. X-chromosome instability in pluripotential stem cell lines derived from parthenogenetic embryos. J Embryol Exp Morphol 1983;74:297¨C309.. Q( |/ K4 e. q  f9 W

) T! Q7 r4 ?+ n  OZvetkova I, Apedaile A, Ramsahoye B et al. Global hypomethylation of the genome in XX embryonic stem cells. Nat Genet 2005;37:1274¨C1279.
* F% m9 Y+ U% l# J% ]+ w# o
7 L& @! }  i- B4 z7 IVrana KE, Hipp JD, Goss AM et al. Nonhuman primate parthenogenetic stem cells. Proc Natl Acad Sci U S A 2003;100 (suppl 1):11911¨C11916.
1 X0 X( z; [) b7 u3 p8 B
  ^% D$ I) X, O  n+ W* ]+ VDraper JS, Smith K, Gokhale P et al. Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nat Biotechnol 2004;22:53¨C54.3 q. e( d! h1 M: ~3 g+ c5 Q8 t
* s0 ~4 v) L& F) L. c) e
Mitalipova MM, Rao RR, Hoyer DM et al. Preserving the genetic integrity of human embryonic stem cells. Nat Biotechnol 2005;23:19¨C20.( w" B  m2 o3 Z2 M9 F+ c

$ G, i/ v  X( q0 K% WBrimble SN, Zeng X, Weiler DA et al. Karyotypic stability, genotyping, differentiation, feeder-free maintenance, and gene expression sampling in three human embryonic stem cell lines derived prior to August 9, 2001. Stem Cells Dev 2004;13:585¨C597.# x+ V/ g2 R) ~1 s' h

  _# i; f$ k$ d! eBhattacharya B, Miura T, Brandenberger R et al. Gene expression in human embryonic stem cell lines: Unique molecular signature. Blood 2004;103:2956¨C2964.- V% b6 b/ T8 c% f! k/ K  m4 @

0 y& e) g. p1 D; F7 v3 aBradley A, Evans M, Kaufman MH et al. Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines. Nature 1984;309:255¨C256.2 `( ]3 y! |6 ^) K% v& }( ]4 w, `5 ~

$ L3 u& i0 Z  @3 c: R: XRho GJ, Kang TY, Kochhar HP et al. Effect of blastomere sex and fluorescent labelling on the development of bovine chimeric embryos reconstituted at the four-cell stage. Mol Reprod Dev 2001;60:202¨C207.

Rank: 2

积分
129 
威望
129  
包包
1788  
沙发
发表于 2015-5-30 10:09 |只看该作者
在线等在线等  

Rank: 2

积分
61 
威望
61  
包包
1757  
藤椅
发表于 2015-6-16 16:10 |只看该作者
呵呵,等着就等着....  

Rank: 2

积分
107 
威望
107  
包包
1889  
板凳
发表于 2015-7-1 20:43 |只看该作者
干细胞之家微信公众号
脂肪干细胞

Rank: 2

积分
69 
威望
69  
包包
1788  
报纸
发表于 2015-8-12 20:42 |只看该作者
肿瘤干细胞

Rank: 2

积分
75 
威望
75  
包包
2193  
地板
发表于 2015-8-15 17:17 |只看该作者
初来乍到,请多多关照。。。  

Rank: 2

积分
129 
威望
129  
包包
1788  
7
发表于 2015-9-13 01:10 |只看该作者
牛牛牛牛  

Rank: 2

积分
118 
威望
118  
包包
1769  
8
发表于 2015-9-20 17:10 |只看该作者
世界上那些最容易的事情中,拖延时间最不费力。  

Rank: 2

积分
84 
威望
84  
包包
1877  
9
发表于 2015-10-2 17:29 |只看该作者
天啊. 很好的资源

Rank: 2

积分
64 
威望
64  
包包
1769  
10
发表于 2015-10-29 13:10 |只看该作者
干细胞抗衰老  
‹ 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, 2024-6-9 04:00

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.