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: 273602|回复: 234
go

Neural Cell Adhesion Molecule Contributes to Hemopoiesis-Supporting Capacity of [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:48 |只看该作者 |倒序浏览 |打印
a 1st Department of Pathology,: X# C* D& w4 |+ C; I% S3 ]

" M9 m1 o9 d: c% E9 a# m$ _& J4 t* Yb Regeneration Research Center for Intractable Diseases,( o1 R+ n8 H. `: D$ i% r' o
' v% Z0 R# o2 q& i# V5 f
c Department of Transplantation for Regeneration Therapy (Sponsored by Otsuka Pharmaceutical Co., Ltd.), Kansai Medical University, Moriguchi, Osaka, Japan;
. u# z" D' E! ~# `
( C6 A  A, W  F& |1 H/ Y0 Id Department of Biotechnology, Kyoto Institute of Technology, Kyoto, Japan;/ k! @' z" Q! x$ b) V9 V+ Z

7 s  T2 H( @* N# m1 v0 ]e Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin, China;
- u" a) ~% ]$ z
8 \& K% h4 A/ h+ [  u. e4 ~' tf Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, California, USA
6 J; |7 s2 s3 G# ?! o& t. u: |9 z- y4 o/ ~: y
Key Words. Neural cell adhesion molecule ? Pluripotent hemopoietic stem cells ? Bone marrow stromal cells ? PA6 ? Mouse( o/ F7 F0 u1 G7 P1 ~' `$ U/ m
6 P+ d9 `+ T( }4 |" Y5 y  l& ?( o- t, I
Correspondence: Susumu Ikehara, M.D., Ph.D., First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City, Osaka 570-8506, Japan. Telephone: 81-6-6993-9429; Fax: 81-6-6994-8283; e-mail: ikehara@takii.kmu.ac.jp
% h4 p# u3 Z. G3 t3 H/ P
( G6 s  K6 O# w" C8 x6 q) K+ zABSTRACT3 p( b7 w1 o3 p' g* H) X4 C
' }/ H: n& ~6 y. b( q- |( u" x
In adult mammals, hemopoiesis is restricted to the extravascular compartment of the bone marrow (BM), where pluripotent hemopoietic stem cells (P-HSCs) and their clonogenic progeny associate intimately with distinctive stromal cell elements comprising the hemopoietic microenvironment or stem cell niches . Stromal cells produce various growth factors, cell adhesion molecules, and matrix proteins that contribute to the formation of stem cell niches, which govern the homing, growth, survival, and differentiation of HSCs . Growth factors produced by stromal cells include cytokines (e.g., monocyte colony-stimulating factor , interleukin-6, leukemia inhibiting factor , thrombopoietin, hepatocyte growth factor, and transforming growth factor ? ), ligands of receptor tyrosine kinase (e.g., stem cell factor  and Flt-3 ligand ), bone morphogeneic protein 4 , and sonic hedgehog . However, in vitro, it is difficult to enhance the self-renewal or expansion of P-HSCs and immature progenitor cells without stromal cells, even if all known exogenous growth factors and other materials are added to the culture . In contrast, long-term hemopoiesis can be maintained by only coculturing HSCs with stromal cells . Our recent reports have also demonstrated that successful BM transplantation depends on the cotransplantation of donor stromal cells ; stromal cells migrate into the recipient BM and spleen, where they support hemopoiesis. These findings have shaped the view that stromal cell–hemopoietic cell interactions in the marrow microenvironment are crucial for physiological hemopoiesis.
3 k4 B1 [6 q- q1 S- D: a
: B; b1 O, g( o! F1 e' J7 V  @Current data suggest that two classes of molecules produced by stromal cells contribute to stromal cell–hemopoietic cell interactions and regulate hemopoiesis: hemopoietic growth factors and CAMs . However, stem cell niches and the mechanisms involved in controlling hemopoiesis remain largely unknown, although Zhang et al.  and Calvi et al.  have now confirmed that osteoblasts have a function in stromal cell–hemopoietic cell regulation in animals. To clarify the mechanisms underlying cell-to-cell interactions between HSCs and stromal cells, we took advantage of our previously established monoclonal antibodies (mAbs) against BM stromal cells . These mAbs were produced by inducing neonatal tolerance in rats using mouse BM stromal cell lines MC3T3-G2/PA6 (PA6) and PA6-mutant (PA6-M); the former has the capacity to support HSCs, whereas the latter has no such capacity. An anti-PA6 mAb inhibits pseudoemperipolesis and suppresses the proliferation of HSCs, suggesting that the anti-PA6 mAb reacts with molecules responsible for the interaction between HSCs and stromal cells.
( J+ P! R7 C" B& u. j  q8 ^: H! ~, u4 n4 [/ e; W
In the present study, we established a fetal BM-derived stromal cell line (FMS/PA6-P) expressing a high level of PA6 molecules, which has high hemopoiesis-supporting capacity. We report herein that neural cell adherion molecule (NCAM) is the molecule that reacts with anti-PA6 mAb and is related to the hemopoiesis-supporting capacity of stromal cells.0 k; g7 H; h: Y, w3 E5 m7 e3 `

' `: n. s" ~6 e( QMATERIALS AND METHODS
( k2 W: `5 P9 D- E6 ]! j4 I; e& [+ W" Z, d4 c8 P- f  h0 l1 l
Establishment and Characterization of the FMS/PA6-P Cell Line7 \/ D. U3 X$ w- f$ y

+ n% g+ P- w" C% o0 I# {- MPA6-positive adherent cells sorted from the third passage of the fetal BM adherent cells were cultured in a flask containing DMEM supplemented with 10% FBS and LIF (10 ng/ml). Interestingly, although the cells were dormant for about 2 months, several cells showing the same morphology began to proliferate thereafter and formed several large colonies 2 weeks later. They were subcultured, and a cell line (FMS/PA6-P) was thus established. The FMS/PA6-P cell line showed fibroblastic morphology, although BM adherent cells showed heterogeneous morphology. The FMS/PA6-P cell line proliferated rapidly with a doubling time of 1.5 days and stopped dividing at confluency. The FMS/PA6-P cell line exhibited continuous growth and retained a stable morphology for more than 25 passages. The FMS/PA6-P cell line was negative for hematolymphoid and endothelial lineage markers (Fig. 1). However, the FMS/PA6-P cell line reacted with anti-CD44, Sca-1, vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and very late antigen 4 (VLA-4) mAbs (Fig. 1), which is characteristic of hemopoiesis-supporting stromal cells . Most interestingly, a high expression of molecules recognized by the anti-PA6 mAb was observed in FMS/PA6-P cells until approximately the 20th passage.
. }) p' y( W9 x. t7 ^4 W- Y5 p) @, h4 |0 x7 [! S0 ]
Figure 1. Flow cytometric analysis of FMS/PA6-P cell line. The FMS/PA6-P cells of different passages were collected by trypsin-EDTA treatment and stained with fluorescent isothiocyanate anti–ICAM-1, VCAM-1, CD11c, Mac-1, Sca-1, CD45, CD3, CD4, CD8, and PE anti-CD11a, CD31, VEGFR-2 (BD Biosciences Pharmingen). They were also stained with anti-PA6, CD44, VLA-4 mAbs, followed by PE anti-rat immunoglobulin G. The stained cells were analyzed using a FACScan. The FMS/PA6-P cell line reacts with anti-PA6, CD44, Sca-1, VCAM-1, ICAM-1, and VLA-4 mAbs but not with mAbs against surface markers of hematolymphoid cells and endothelial cells. The closed profile indicates the cells stained with isotype-matched control mAbs. Abbreviations: ICAM-1, intercellular adhesion molecule 1; mAB, monoclonal antibody; PE, phycoerythrin; Sca-1, stem cell antigen-1; VCAM-1, vascular cell adhesion molecule 1; VEGFR-2, vascular endothelial growth factor receptor 2; VLA-4, very late antigen 4.# u) \  }( D" B5 ?
  y# ]; W/ N- ?1 ?# x
Hemopoiesis-Supporting Capacity of the FMS/PA6-P and Other Stromal Cell Lines In Vitro
% ?0 x4 N) \; o2 M8 X3 x! B, k  U0 M8 q2 S
When HSCs of B6 mice were cocultured with the FMS/PA6-P cell layers without cytokines, they crawled under the stromal layer and proliferated. The proliferating cells demonstrated a cobblestone-like appearance and were referred to as CAFCs. To compare the capacity of FMS/PA6-P (eighth or 22nd), MS-5, and PA6 to sustain long-term hemopoiesis, HSCs were cultured on a monolayer of these cell lines without adding exogenous cytokines. After 7-day culture, not only were the counts of cobblestone colonies significantly higher, but the size was also larger in cultures of HSCs on FMS/PA6-P cells (eighth) than other stromal cell lines (data not shown). Hemopoiesis can be maintained for only 8 weeks on the FMS/PA6-P (22nd), MS-5, and PA6, whereas a high number of cobblestone colonies were still observed in the culture on the FMS/PA6-P (eighth) cells thereafter, and hemopoiesis could be maintained on the FMS/PA6-P (eighth) up to 26 weeks. Furthermore, as shown in Table 1, HSCs cocultured with FMS/PA6-P (eighth) showed an approximately 34.7-fold increase in total cells and 8.2-fold increase in Sca-1  CD45  cells after 4 weeks. The numbers of total cells and Sca-1  CD45  cells were significantly higher in cultures with FMS/PA6-P (eighth) than those with FMS/PA6-P (22nd), MS-5, or PA6.% v* V( a( y) I. X

0 g0 C7 u4 {3 C2 XTable 1. Different hematopoiesis-supporting capacities of stromal cell lines
( _' Y0 [2 {0 F$ m! D8 }1 v
9 n. q# a8 |% W, N3 M; sRequirement for Cell-to-Cell Contact Between HSCs and Stromal Cells
' x$ I! a5 \, L' m6 J- e
# j7 y5 T, ^) C8 eWe examined the effect of interruption of cell-to-cell contact between HSCs and stromal cells on the expansion of HSCs. As shown in Figure 2, the numbers of total hemopoietic cells, Sca-1 CD45  cells, and total CFU-C were significantly lower in non-contact cultures than those in contact cultures; HSCs cocultured with the FMS/PA6-P cell line showed approximately 2.3- and 5.3-fold increases in total hemopoietic cells and Sca-1 CD45  cells, respectively. These results demonstrated that the direct cell-to-cell contact between HSCs and FMS/PA6-P cells is essential for the maximum expansion of HSCs and progenitor cells.
- X& H+ p1 Q& W7 s; o. ]2 z' M& G# L6 v' Q
Figure 2. Effect of interruption of stromal cell contact on ex vivo expansion of HSCs. HSCs (3 x 104) were cultured on a monolayer of FMS/PA6-P (contact) or on a transwell insert placed above the stromal layer (noncontact) for 10 days and then analyzed. Data are expressed as the mean ± SD of five wells. *p & W  q+ }% \4 H" h

4 T0 }! n6 w; O& uExpression of Cytokines and PA6 Molecule in Various Stromal Cell Lines7 r& |6 M+ r1 Q0 M4 A; v( w% ^

; a( }% z: n* ]9 R& G# ~9 ^To further analyze the mechanism regulating the hemopoiesis-supporting ability, we first examined the cytokine expressions in various cell lines. As shown in Figure 3A, not only Flt3-L  and basic fibroblast growth factor  (which promote cell growth) but also LIF  and TGF-?1  (which negatively regulate hemopoiesis) are found in all of the stromal cell lines. In contrast, the expression level of SCF, macrophage inflammatory protein 1, insulin-like growth factor-type 1, and granulocyte-CSF (G-CSF) differed greatly among these cell lines. No difference was observed between the eighth and 22nd passage of FMS/PA6-P, although the eighth passage showed better supporting ability than the 22nd passage. The cell line FMS/NS showed no expression of G-CSF and SCF. Accordingly, it seems that both G-CSF and SCF are responsible for the hemopoiesis-supporting ability. However, G-CSF is not produced from MS-5 and PA6, indicating that G-CSF is not an essential cytokine for supporting capacity. Therefore, the differences in cytokine expression are not enough to explain the better hemopoiesis-supporting capacity in FMS/PA6-P (eighth).
' J& {7 K2 Z4 ]9 K: l" L+ N4 o; U9 d- W0 D, C- ~& O
Figure 3. Expressions of PA6 molecule and cytokines in various stromal cell lines or BM adherent cells. (A): mRNA expression of cytokines in various stromal cell lines. The FMS/PA6-P (eighth and 22nd), MS-5, PA6, FMS/NS cells cultured in 25-cm2 flasks were irradiated under the same conditions as we had investigated the in vitro hemopoiesis-supporting effect of these stromal cell lines. Expression of the indicated cytokines in various stromal cell lines was analyzed using reverse transcription–polymerase chain reaction. (B): Flow cytometric analysis of PA6 expression in various cell lines and BM adherent cells. The closed profile indicates the cells stained with isotype-matched control antibodies. (C): Western blotting analysis of PA6 expression in various cell lines. PA6 molecules were expressed in a higher amount in the cell lines and BM adherent cells having the ability to support hemopoiesis than in those without such ability, and the most supportive cell line (FMS/PA6-P ) expressed the highest level of PA6 molecule. Abbreviations: bFGF, basic fibroblast growth factor; BM, bone marrow; Flt3-L, Flt-3 ligand; G-CSF, granulocyte-colony stimulating factor; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; IGF-1, insulin-like growth factor type 1; LIF, leukemia inhibiting factor; MIP-1, macrophage inflammatory protein1; SCF, stem cell factor; TGF-?1, transforming growth factor ?1.
' w0 X/ O1 X' l/ `+ f( Y
  b2 V" q0 |2 ~" gWe next examined the expression of PA6 molecule in the FMS/PA6-P of different passages and other stromal cell lines by flow cytometry. As shown in Figure 3B, a high level of PA6 expression was observed in FMS/PA6-P until approximately the 20th passage, whereas a decrease in this level was observed thereafter. In contrast, no significant change was detected in the expression level of CD44, Sca-1, VCAM-1, ICAM-1, and VLA-4 among the FMS/PA6-P cells cultured for different periods (data not shown). Furthermore, although PA6 expression was observed in MS-5 and PA6 stromal cell lines, the expression level in MS-5 was lower than that in FMS/PA6-P cells (eighth) and the expression level in PA6 was even lower and was comparable with that in FMS/PA6-P cells (22nd). In the stromal cell lines showing no hemopoiesis-supporting capacity (FMS/NS), the expression level of PA6 was very low.) }( z: {4 V( ^

, L( p8 _% S8 g# B% O8 yThe PA6 molecule in various stromal cell lines was further detected by Western blotting. A molecule of approximately 140 kDa reacted with anti-PA6 mAb, and the most supportive cell line, FMS/PA6-P (eighth), expressed the highest level of PA6 (Fig. 3C), suggesting that the level of PA6 expression is positively correlated to the hemopoiesis-supporting capacity of the stromal cells.
) N0 X: o: {* H* z/ M" I, k
* n& o3 x: w; x1 g5 N2 \4 A9 `Inhibitory Effect of Anti-PA6 Antibody for HSC Proliferation on FMS/PA6-P Cell Line
3 Z" r  A3 `+ S  n) K& o* O; t. o3 \& ]& t; M/ C
When 1 and 0.2 μg/ml anti-PA6 mAb were added to the culture medium and 3 x 103 HSCs was inoculated on the FMS/PA6-P monolayer, the 3H-TdR uptake was only 23% and 21% of that of the isotype control (Fig. 4), respectively, reflecting that the proliferation of HSCs was suppressed to a great extent by the anti-PA6 mAb, although not completely.$ [4 G! s8 {3 H, Z4 a9 h! d
+ H; k' }7 u% }( l5 Y0 `* D% E7 V
Figure 4. Inhibitory effect of anti-PA6 mAb or anti-NCAM mAb on the proliferation of HSCs. Anti-PA6 mAb or anti-NCAM mAb at 0.2 and 1 μg/ml can significantly inhibit the proliferation of HSCs. *p
: A) @( r' F3 `% y' D1 ^$ t5 |
( G) l5 n- c4 [6 W& {) UNCAM is the Molecule That Reacts with Anti-PA6 Antibody8 y+ z# d2 i2 @
# n& N8 P8 d5 K, n! _) C7 Y; v
Since the PA6 gene has a deduced size of more than 3 kb, it is difficult to clone. We therefore attempted to obtain some peptide information on the PA6 protein using MALDI-TOF peptide mass fingerprinting. The molecules that react with anti-PA6 mAbs were identified by affinity chromatography on immobilized Sepharose columns coupled with anti-PA6 mAbs. The eluted materials were separated by SDS-PAGE and visualized by silver staining. A dominant protein band with an apparent molecular weight of 140 kDa was confirmed by Western blotting to be the molecules that react with anti-PA6 mAbs (Figs. 5A, 5B). Four of the 140-kDa protein bands were excised and digested with trypsin. The resulting peptides were analyzed by MALDI-TOF peptide mass fingerprinting. Sequences of the tryptic peptides exhibited 100% homology with NCAM (Table 2). Furthermore, anti-PA6 mAb does not react with the PA6 molecule after pretreatment with anti-NCAM mAb (Fig. 5C), indicating that the PA6 molecule is NCAM. Indeed, as shown in Figure 6, NCAM expression patterns of the FMS/PA6-P (eighth and 22nd), MS-5, PA6, and FMS/NS cell lines were similar to PA6 expression patterns of these cell lines in Figure 3B, confirming that the expression level of NCAM is positively correlated to the hemopoiesis-supporting capacity of stromal cell lines. This finding was further confirmed by an inhibitory effect of anti-NCAM mAb on the proliferation of HSCs cocultured with FMS/PA6-P cells, which is compatible with the inhibitory effect of anti-PA6 mAbs on the proliferation of HSCs (Fig. 4). In addition, although in very low amount, 180-kDa and 120-kDa protein bands were also detected among the proteins identified by affinity chromatography using anti-PA6 mAbs when the film was exposed for longer periods (data not shown).- i# S0 v+ U7 Y! u8 @, N
  l& q  Z9 ?8 Q, ]
Figure 5. Identification of the molecule that reacts with anti-PA6 mAb. (A): The FMS/PA6-P cell lysates were first precleared by immobilized bovine serum albumin columns and then loaded onto immobilized anti-PA6 mAb columns. Bound proteins were eluted with 0.5 M NH4OH, lyophilized, and subjected to SDS-PAGE using a 5%–20% polyacrylamide gel. After silver staining, a dominant 140-kDa protein band was noted to react with anti-PA6 mAb. (B): One percent of eluted proteins was subjected to SDS-PAGE at the same time, transferred onto polyvinylidene difluoride membrane, and analyzed by Western blotting. A 140-kDa protein band was revealed to react with anti-PA6 mAb with the highest intensity. (C): Inhibitory effect of anti-NCAM mAb against binding of anti-PA6 mAbs to PA6 molecule. Anti-PA6 mAb does not react with PA6 molecule on FMS/PA6-P cell line after pretreatment with anti-NCAM mAb. (1): Cells were only stained with anti-PA6 mAbs followed by PE anti-rat IgG. (2, 3): Cells were first incubated with (2) anti-NCAM mAb or (3) normal mouse IgG and then stained with anti-PA6 mAbs followed by PE anti-rat IgG. The stained cells were analyzed using a FACScan. Broken lines indicate the cells stained with isotype-matched control mAbs. Abbreviations: IgG, immunoglobulin G; mAb, monoclonal antibody; NCAM, neural cell adhesion molecule; PE, phycoerythrin; SDS-PAGE, sodium dodecyl sulfate–polyacrylamide gel.6 v3 Z! q2 z, Q* x

4 N! k6 @; h- k3 _Table 2. Identification of mouse NCAM protein by MALDI-TOF analysis9 G& h( ~! N" J6 N" M& d( j( d- c
+ r. z$ y' s$ X2 P) b5 P
Figure 6. NCAM expression in various stromal cell lines. The FMS/PA6-P cells (eighth and 22nd passages), MS-5, PA6, and FMS/NS were stained with anti-NCAM monoclonal antibody, followed by phycoerythrin anti-mouse immunoglobulin G. The expression patterns of NCAM in various stromal cell lines were similar to those of PA6 molecule. The closed profile indicates the cells stained with iso-type-matched control antibodies. Abbreviation: NCAM, neural cell adhesion molecule., B5 Q) X1 ]2 u7 R# i
9 Q/ [/ B% o- }
NCAM Contributes to Hemopoiesis-Supporting Capacity of Stromal Cells
9 m' ?) ~! U0 o1 w' r8 b
, B) F9 {) w" b3 l: iTo investigate the role of NCAM in the hemopoiesis-supporting capacity of stromal cells, we repressed the expression of NCAM by an approach based on siRNA . Transfected NCAM siRNA specifically reduced NCAM expression in FMS/PA6-P cells expressing a high level of NCAM by 60% at the protein level 48 hours after infection, as measured by flow cytometry (Fig. 7A) and by nearly 80% at the mRNA level 24 hours after infection, as measured by RT-PCR (Fig. 7B). The maximum silencing was observed at 96 hours at the protein level (71%) (Fig. 7A) and 72 hours at the mRNA level (90%) (Fig. 7B). This finding confirmed that NCAM silencing occurred by reducing mRNA stability. Four days after transfection, NCAM expression in the NCAM siRNA-transfected FMS/PA6-P cells began to increase slightly. However, these cells maintained a low level of expression at least 13 days after transfection (data not shown).
( {9 i# A2 N+ X" C3 ?
+ e- a4 M6 C8 _Figure 7. Hemopoiesis-supporting activity of NCAM. (A, B): Suppression of NCAM expression by siRNA. (A): The FMS/PA6-P cells (10th passage), transfected with NCAM or control siRNA at the times indicated, were trypsinized and stained with anti-NCAM monoclonal antibody, followed by phycoerythrin anti-mouse immunoglobulin G. The tained cells were then analyzed using a FACScan. The broken lines indicate the cells stained with isotype-matched control antibodies. (B): Total RNA was extracted from the above cells at the times indicated and subjected to reverse transcription–polymerase chain reaction with NCAM and GAPDH primers. (1): FMS/PA6-P cells transfected with control siRNA; (2, 3, 4): FMS/PA6-P cells transfected with NCAM siRNA 24, 48, and 72 hours later. (C): Repression of NCAM in FMS/PA6-P cells suppresses LTC-IC frequency. The LTC-IC frequency is 1 out of 198 ± 21 HSCs in control siRNA-transfected FMS/PA6-P cells and 1 out of 614 ± 45 HSCs in NCAM siRNA-transfected FMS/PA6-P cells. (1): FMS/PA6-P cells transfected with control siRNA; (2): FMS/PA6-P cells transfected with NCAM siRNA. Error bars indicate the SD. (D): Repression of NCAM in FMS/PA6-P cells suppresses the formation of long-term CAFCs. NCAM siRNA-transfected FMS/PA6-P cells were irradiated, cultured with 1 x 103 Lin–Sca-1  HSCs, and retransfected weekly with siRNA. CAFCs were counted after 5 weeks. (1): FMS/PA6-P cells transfected with control siRNA; (2): FMS/PA6-P cells transfected with NCAM siRNA. Results are reported as a ratio of the value from control siRNA-transfected cells and are the mean ± SD from 12 wells. *p
' M( V% B* Q0 Z, ?. `2 W9 T% F( H: C
To assess the effect of NCAM silencing on the hemopoiesis-supporting capacity of stromal cells, the cells were irradiated 96 hours after transfection (the time of maximum silencing), and HSCs of serial cell dilutions (1 x 102 to 3 x 104 per well) were then inoculated. After 5 weeks of culture, the LTC-ICs were quantified by in vitro colony assay of all the cells collected from each well. The LTC-IC frequency was 1 of 198 ± 21 HSCs in the control siRNA-transfected FMS/PA6-P cells and 1 of 614 ± 45 HSCs in the NCAM siRNA-transfected FMS/PA6-P cells (Fig. 7C). The frequency of long-term CAFCs (Fig. 7D) was also suppressed by repression of NCAM in the FMS/PA6-P cells. Taken together, these results indicate that NCAM functions on the maintenance of HSCs.( [' U$ q1 j+ q2 B' S  A2 b5 ^
$ [( W$ W& j6 T9 l5 Q
DISCUSSION
+ f; c7 S: G# T% W6 t, a9 Z  t1 O- }' a8 l
This work was supported by a grant from Haiteku Research Center of the Ministry of Education, a grant from the Millennium program of the Ministry of Education, Culture, Sports, Science and Technology, a grant from the Science Frontier program of the Ministry of Education, Culture, Sports, Science and Technology, a grant-in-aid for scientific research (B) 11470062, grants-in-aid for scientific research on priority areas (A)10181225 and (A)11162221, and Health and Labor Sciences research grants (Research on Human Genome, Tissue Engineering Food Biotechnology), the 21st Century Center of Excellence Program (Project Leader), the Ministry of Education, Culture, Sports, Science and Technology, a grant from the Department of Transplantation for Regeneration Therapy (Sponsored by Otsuka Pharmaceutical Company, Ltd.), a grant from Molecular Medical Science Institute, Otsuka Pharmaceutical Co., Ltd., and a grant from Japan Immunoresearch Laboratories Co., Ltd. (JIMRO). We thank S. Miura for conducting FACS sorting and K. Yasaka for helping to analyze peptide mass. We also thank Hilary Eastwick-Field and K. Ando for manuscript preparation.! {! N7 ?; e4 o+ z

% F4 k+ {" s9 a5 Q6 B, [8 w7 mDISCLOSURES
: @( f' u% `8 \; t3 S/ ^; q: `3 }" m9 `* y7 P4 l
The authors indicate no potential conflicts of interest.* e0 Q+ ~- R" J( ?2 R. R2 M

; j# R. R( s5 l) T! VREFERENCES3 G0 B7 e$ @2 j2 J

. k4 S# ^$ a; B7 r  PDorshkind K. Regulation of hemopoiesis by bone marrow stromal cells and their products. Annu Rev Immunol 1990;8:111–114.6 G9 u$ K( n- A! v& ?* q* G- u
8 B) B1 N2 G% B: Q- K5 m, {* O
Trentin JJ. Influence of hematopoietic organ stroma (hematopoietic inductive microenvironments) on stem cell differentiation. In: Gordon AS, ed. Regulation of Hematopoiesis. New York: Appleton-Century Crofts, 1970:161–170.
# s8 u0 p; W; S5 G' ]8 E1 p( s& U' Q; S. p8 ]7 L
Weissman I. Developmental switches in the immune system. Cell 1994;76:207–210.0 n* V# ]  k4 J+ b1 @# U

; r" G4 Z, e( G& {; C! U4 XChabannon C, Torok-Storb B. Stem cell-stromal cell interactions. Curr Top Microbiol Immunol 1992;77:123–136.+ s1 }' ^  S9 O0 j6 q* `( K

% J; e# B) a, ?! \+ U, m8 VTavassoli M, Minguell JJ. Homing of hemopoietic progenitor cells to the marrow. Proc Soc Exp Biol Med 1991;196:367–373.! C' d  B4 R6 o% i8 ^, H0 f( \" y

. o. Q( l. L* p; L" gVarnum-Finney B, Purton LE, Yu M et al. The Notch ligand, Jagged-1, influences the development of primitive hematopoietic precursor cells. Blood 1998;91:4084–4091.
8 C, ~5 Z1 D9 Z6 r' O' c; ]% D" U
Bhatia M, Bonnet D, Wu D et al. Bone morphogenetic proteins regulate the developmental program of human hematopoietic stem cell. J Exp Med 1999;189:1139–1148.
. p5 ~, a, ~6 q+ ]: h% Z! x9 S- }
Bhardwaj G, Murdoch B, Wu D et al. Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nat Immunol 2001;2:172–180.& s' ]8 \; A# D+ J; |0 K8 l

$ i% C8 L( e: `" nBernstein ID, Andrews RG, Zsebo KM. Recombinant human stem cell factor enhances the formation of colonies by CD34  and CD34  lin– cells, and the generation of colony-formation cell progeny from CD34  lin– cells cultured with interleukin-3, granulocyte colony-stimulating factor, or granulocyte-macrophagecolony-stimulating factor. Blood 1991;77:2316–2321.* f% Y% s! o/ u: M# g# B# U+ L
; w$ I+ v% j5 X7 j7 T4 x. l* P( M
Lowry PA, Zsebo KM, Deacon DH et al. Effects of rhSCF on multiple cytokine-responsive HPP-CFC generated from Sca-1  Lin– murine hematopoietic progenitors. Exp Hematol 1991;19:994–996.7 c* n  U+ }9 v) v, J* _# V
! G- N# [5 V- S) |+ i' {
Li CL, Johnson GR. Stem cell factor enhances the survival but not the self-renewal of murine hematopoietic long-term repopulating cells. Blood 1994;84:408–414.
+ m& u! L  ?# b/ D
" w# u) H- q, _4 ^Ogawa M, Nishikawa S, Ikuta K et al. B cell ontogeny in murine embryo studied by a culture system with the monolayer of a stromal cell clone, ST2: B cell progenitor develops first in the embryonal body rather than in the yolk sac. EMBO J 1988;7:1337–1343.) \! R0 Q: Q  k7 B& c6 t( o
' r& h3 h& X" D" ]1 t
Kodama HA, Amagai Y, Koyama H et al. In vitro hemopoiesis within a microenvironment created by MC 3T3-G2/PA6 preadipocytes. J Cell Physiol 1984;118:233–240., a+ _3 e. ~3 k# O9 O

0 E; o$ G6 |- f5 P. h# SDexter TM, Allen TD, Lajtha LG. Conditions controlling the proliferation of hematopoietic stem cells in vitro. J Cell Physiol 1977;91:335–344.. ^5 o* H/ X% N

# B+ t# [" }$ [Itoh K, Tzuka H, Sakoda H et al. Reproducible establishment of hemopoietic supportive stromal cell lines from murine bone marrow. Exp Hematol 1989;17:145–153.
  i0 v* V4 S7 |9 D2 }, A7 w* @* M1 W* A* X
YeZQ, Burkholder JK , Qiu P etal. Establishment of an adherent cell feeder layer from human umbilical cord blood for support of long-term hematopoietic progenitor cell growth. Proc Natl Acad Sci U S A 1994;91:12140–12144.- E5 @& S) e" F% A- N8 M3 c
4 K; L# k0 I; `$ r  O2 a
Nishikawa M, Ozawa K, Tojo A et al. Changes in hematopoiesis-supporting ability of C3H10T1/2 mouse embryo fibroblasts during differentiation. Blood 1993;81:1184–1192.  R# F  W* p  k' _/ l2 s  E" U

8 x5 T+ z* ^% S/ ~0 Y; UIssaad C, Croisille L, Katz A et al. A murine stromal cell line allows the proliferation of very primitive human CD34  /CD38– progenitor cells in long-term cultures and semisolid assays. Blood 1993;81:2916–2924.
* t9 {% o) s8 u+ Z* z- L4 U5 X: P  l& \5 u+ _8 j" M6 ^
Hisha H, Nishino T, Kawamura M et al. Successful bone marrow transplantation by bone grafts in chimeric-resistant combination. Exp Hematol 1995;23:347–352.
" |& b4 Y: @# {. ~  x9 E# f9 |& j& T+ P+ ?2 a, g
Ishida T, Inaba M, Hisha H et al. Requirement of donor-derived stromal cells in the bone marrow for successful allogeneic bone marrow transplantation. J Immunol 1994;152:3119–3127.2 u6 B/ e" r* d* y$ h
9 O( Z  l1 O3 p# z1 U/ O: a
Hashimoto F, Sugiura K, Inoue K et al. Major histocompatibility complex restriction between hematopoietic stem cells and stromal cells in vivo. Blood 1997;89:49–54.
- n! R, ^: b6 [) P: d  ~. }* Z6 H
Sugiura K, Inaba M, Hisha H et al. Requirement of major histocompatibility complex-compatible microenvironment for spleen colony formation (CFU-S on day 12 but not on day 8). STEM CELLS 1997;15:461–468.
3 o( K+ ^+ ~# U9 r, y  e  x; H7 E$ c. K. h0 M4 A$ P! O+ a
Toksoz D, Zsebo KM, Smith KA et al. Support of human hematopoiesis in long-term bone marrow cultures by murine stromal cells selectively expressing the membrane-bound and secreted forms of the human homologue of the steel gene product, stem cell factor. Proc Natl Acad Sci U S A 1992;89:7350–7354.
3 m# h# J' a6 m; D4 _. p5 z/ Y% p+ Q( x) t' V3 }( T
Hannum C, Culpepper J, Campbell D et al. Ligand for FLT3/FLK2 receptor tyrosine kinase regulates growth of hematopoietic stem cells and is encoded by variant RNAs. Nature 1994;368:643–648.
( R* Q! L3 h# @
) a* C9 ~9 {5 ?0 QSatoh M, Mioh H, Shiotsu Y et al. Mouse bone marrow stromal cell line MC3T3-G2/PA6 with hematopoietic-supporting activity expresses high levels of stem cell antigen Sca-1. Exp Hematol 1997;25:972–979.% i! z7 i% d& _- B, E& R- R
& d* c, @' p- ~: S
Ueno H, Sahita-Ishikawa M, Morikawa Y et al. A stromal cell-derived membrane protein that supports hematopoietic stem cells. Nat Immunol 2003;4:457–463.4 V" P" n- i0 C6 }8 f: L) a
2 \8 @: ^2 \5 N7 O# {( n5 Q  p
Zhang J, Niu C, Ye L et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 2003;425:836–841.3 C' ]* U! L; ]% R' ~

, u: n: R5 Q, m$ YCalvi LM, Adams GB, Weibrecht KW et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 2003;425:841–846.
# {( M2 s: R& s+ K
  E4 s" e4 n+ U% ~6 f2 P6 EIzumi-Hisha H, Than S, Ogata H et al. Monoclonal antibodies against a preadipose cell line (MC3T3-G2/PA6) which can support hemopoiesis. Hybridoma 1991;10:103–112.) s# ?6 i8 i% b& Z/ T1 {# v

; `4 z4 W% s  L6 J# h! o7 W+ N7 i! QJiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41–49.
+ I. o& o, a, l! S3 @
. `6 u* |0 y" W8 }; cFan TX, Hisha H, Jin TN et al. Successful allogeneic bone marrow transplantation (BMT) by injection of bone marrow cells via portal vein: stromal cells as BMT-facilitating cells. STEM CELLS 2001;19:144–150.
" F* U2 a( x8 ?3 A( r; Q- Y( u: K( `( S: \; e8 a, g
Itoh K, Tezuka H, Sakoda H et al. Reproducible establishment of hemopoietic supportive stromal cell lines from murine bone marrow. Exp Hematol 1989;17:145–153.4 {' }% }9 a7 R9 m2 o8 _1 p
5 o1 p, R% L; l% T; q* ^
Kodama H, Amagai Y, Koyama H et al. A new preadipose cell line derived from newborn mouse calvaria can promote the proliferation of pluripotent hemopoietic stem cells in vitro. J Cell Physiol 1982;112:89–95.& [* H, Q, J) v  k) N
9 x# @4 u. r; c% Z) `
Tse KF, Marrow JK, Hughes NK et al. Stromal cell lines derived from LP-BN5 murine leukemia virus-infected long-term bone marrow cultures impair hematopoiesis in vitro. Blood 1994;84:1508–1518.! C& B1 @/ h  G3 H! W& u
8 T6 D$ v+ O0 N/ q' I! ^
Pessina A, Neri MG, Mineo E et al. Expression of B cell markers on SR-4987 cells derived from murine bone marrow stroma. Exp Hematol 1997;25:536–541.2 R' B6 J* S8 m2 ?+ i2 M

5 L2 B9 L) F2 u! qTanaka T, Akira S, Yoshida K et al. Targeted disruption of the NF-IL-6 discloses its essential role in bacteria killing and tumor cytotoxicity by macrophages. Cell 1995;80:353–361.1 f. C% O. Y2 Y. T; z& k4 b
& {! g+ I6 T1 [  c6 d
Hana V, Murphy LJ. Interdependence of epidermal growth factor and insulin-like expression in the mouse uterus. Endocrinology 1994;135:107–112.
5 h) }# ~7 w5 r  D, X5 S; |. W- `1 j7 V
Wang Z, Ren S, Melmed S. Hypothalamic and pituitary leukemia inhibitory factor gene expression in vitro: a novel endotoxin-inducible neuro-endocrine interface. Endocrinology 1996;137:2947–2953.1 d+ V$ {2 D' U4 Z8 `' j

9 R) q# u: `8 L8 z) O: |) Z1 y; ySimmons PJ, Masinovsky B, Longenecker BM et al. Vascular cell adhesion molecule-1 expressed by bone marrow stromal cells mediates the binding of hematopoietic progenitor cells. Blood 1992;80:388–395.
" s, ]. b* t( \8 V+ n
, S: G  a  _; C! |) Y" HTeixido J, Hemler ME, Greenberger JS et al. Role of beta 1 and beta 2 inte-grins in the adhesion of human CD34hi stem cells to bone marrow stroma. J Clin Invest 1992;90:358–367., g9 l% s: Q0 k. M0 O  \2 c
4 b6 a8 p% l9 ]$ m6 V  o3 ^# s3 V$ t
Oostendorp RA, Spitzer E, Brandl M et al. Evidence for differences in the mechanisms by which antibodies against CD44 promote adhesion of erythroid and granulopoietic progenitors to marrow stromal cells. Br J Haematol 1998;101:436–445.4 Q" h% N/ S& Q

0 C, J1 m4 l. E0 e" HHidalgo A, Robledo MM, Teixido J. CD44-mediated hematopoietic progenitor cell adhesion and its complex role in myelopoiesis. J Hematother Stem Cell Res 2002;11:539–547.2 Y" R# H% J' K1 _5 N9 a9 Z
7 g- H6 ~" V+ w$ }( v/ s1 ~
Haylock DN, Horsfall MJ, Dowse TL et al. Increased recruitment of hematopoietic progenitor cells underlies the ex vivo expansion potential of FLT3 ligand. Blood 1997;90:2260–2272.0 l6 b+ W: e  I1 }/ i

: c6 F, o7 A$ s$ Z( L$ K* w* ~) d6 wHudak S, Hunte B, Culpepper J et al. FLT3/FLK ligand promotes the growth of murine stem cells and the expansion of colony-forming cells and spleen colony-forming units. Blood 1995;85:2747–2755.
$ Z; J9 m- P" L: F6 x4 C2 R+ U) l" n' ?; ~2 t, {2 a% T# Q2 x, E
Huang S, Terstappen LW. Lymphoid and myeloid differentiation of single human CD34 , HLA-DR , CD38– hematopoietic stem cells. Blood 1994;83:1515–1526.
4 u; ]& `/ @) B" D4 V8 m2 f$ V4 I# X& q: f) t* e1 l
Gabbianelli M, Sargiacomo M, Pelosi E et al. "Pure" human hematopoietic progenitors: permissive action of basic fibroblast growth factor. Science 1990;249:1561–1564.
, @9 e) a7 r  `% c: h+ S2 o3 f, q. R9 ^$ b" P
Liebermann DA, Hoffman B. Differentiation primary response genes and proto-oncogenes as positive and negative regulators of terminal hematopoietic cell differentiation. STEM CELLS 1994;12:352–369.* G" A/ I9 ^5 \9 m* m
& y. v# [! |/ B4 ~( J
Cashman JD, Clark-Lewis I, Eaves AC et al. Differentiation stage-specific regulation of primitive human hematopoietic cycling by exogenous and endogenous inhibitors in an in vivo model. Blood 1999;94:3722–3729.% `' Y0 P9 Y1 n& t2 }9 d
9 N! F% ]! K6 O+ N) j% p
Sitnicka E, Ruscetti FW, Priestley GV et al. Transforming growth factor beta1 directly and reversibly inhibits the initial cell division on long-term repopulating hematopoietic stem cells. Blood 1996;88:82–88.2 V3 Q! v& d$ l' ~$ m1 `
1 t0 x  I. k4 `7 {9 z
Elbashir SM, Harborth J, Lendeckel W et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature 2001;411:494–498.7 b# W* E: T# l* n! t7 V: K: q

  T! I5 B0 \# K7 q7 [* HCunningham BA, Hemperly JJ, Murray BA et al. Neural cell adhesion molecule structure, immunoglobulin-like domains, cell surface modulation, and alternative RNA splicing. Science 1987;236:455–459.$ |1 u! M2 \% S+ u; ]

) d6 y! k0 N2 F' Y6 ~- B! RNiethammer P, Delling M, Sytnyk V et al. Cosignaling of NCAM via lipid rafts and the FGF receptor is required for neuritogenesis. J Cell Biol 2002;157:521–532.
1 \8 u0 s9 y- }4 ^2 o" n
1 y: q0 `0 F% ~2 ?. q, \7 U: uCrossin KL, Krushel LA. Cellular signaling by neural cell adhesion molecules of the immunoglobulin superfamily. Dev Dyn 2000;218:260–279.
' q6 N# `- l* `5 R3 g: `* E# J! w+ q; i+ ^
Schachner M. Neural recognition molecules and synaptic plasticity. Curr Opin Cell Biol 1997;9:627–634.1 k* P+ R& j% K7 s9 b

+ s$ y4 _% P3 E6 [Ronn LC, Hartz BP, Bock E. The neural cell adhesion molecule (NCAM) in development and plasticity of the nervous system. Exp Gerontol 1998;33:853–864.' F& Y8 [. s1 h3 S9 F, y4 b  W2 i

  N8 k" j, O; lBixby JL, Pratt RS, Lilien J et al. Neurite outgrowth on muscle cell surface involves extracellular matrix receptors as well as Ca2 -dependent and -independent cell adhesion molecules. Proc Natl Acad Sci U S A 1987;84:2555–2559.
1 c* E/ q2 R8 j, d: V( G" N, B' e: X, T, y$ m- O" z
Doherty P, Cohen J, Walsh FS. Neurite outgrowth in response to transfected N-CAM changes during development and is modulated by polysialic acid. Neuron 1990;5:209–219.4 V* C# d) @0 V7 @6 O! f8 S' E7 c& \1 H- I

, u& n; u% C6 x- T2 S8 c, FToba Y, Horie M, Sango K et al. Expression and immunohistochemical localization of heparan sulphate proteoglycan N-syndecan in the migratory path way from the rat olfactory placode. Eur J Neurosci 2002; 15:1461–1473.% n, C, n6 V, b3 L) y
% Z: u  `. n0 v) S9 @% @
Gupta P, Oegema TR Jr., Brazil JJ et al. Structurally specific heparan sulfates support primitive human hematopoiesis by formation of a multimolecular stem cell niche. Blood 1998;92:4641–4651.! j! M1 T% d# ~4 ~7 E4 M+ E
. \+ c. h5 C5 b# r7 E0 ^+ z+ w' j
Warrens AN, Simon AR, Theodore PR et al. Cross-species compatibility of intercellular adhesion molecule-1 (CD54) with its ligands. Transplantation 2000;69:394–399.(Xiaoli Wanga,e, Hiroko Hi)

Rank: 2

积分
129 
威望
129  
包包
1788  
沙发
发表于 2015-5-27 10:01 |只看该作者
不知道说些什么  

Rank: 2

积分
161 
威望
161  
包包
1862  
藤椅
发表于 2015-6-25 07:51 |只看该作者
回复一下  

Rank: 2

积分
72 
威望
72  
包包
1942  
板凳
发表于 2015-6-25 13:28 |只看该作者
干细胞之家微信公众号
支持一下  

Rank: 2

积分
69 
威望
69  
包包
1788  
报纸
发表于 2015-7-11 12:59 |只看该作者
端粒酶研究

Rank: 2

积分
69 
威望
69  
包包
1788  
地板
发表于 2015-7-29 09:36 |只看该作者
…没我说话的余地…飘走  

Rank: 2

积分
98 
威望
98  
包包
1756  
7
发表于 2015-8-3 12:08 |只看该作者
免疫细胞治疗  

Rank: 2

积分
88 
威望
88  
包包
1897  
8
发表于 2015-8-6 09:43 |只看该作者
先顶后看  

Rank: 2

积分
69 
威望
69  
包包
1788  
9
发表于 2015-10-2 12:41 |只看该作者
严重支持!

Rank: 2

积分
104 
威望
104  
包包
1772  
10
发表于 2015-10-6 10:01 |只看该作者
干细胞之家是国内最好的干细胞网站了
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-5-4 07:36

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.