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: 321750|回复: 262
go

Skeletal Myogenic Differentiation of Mesenchymal Stem Cells Isolated from Human [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:37 |只看该作者 |倒序浏览 |打印
Research Institute of Biotechnology, Histostem Co. Kangdong-gu, Seoul, Korea
6 V" f6 B: e6 x5 ~! {3 |
4 |4 j: u3 l0 A( Y* ?: zKey Words. Human umbilical cord blood ? Mesenchymal stem cells ? Immunophenotyping ? Myogenic differentiation* ~5 u! Z7 M7 V9 ]0 }! z; g& i
0 h+ S3 ?  e) W/ o. ^
Correspondence: Hoeon Kim, Ph.D., Research Institute of Biotechnology, Histostem Co. 518-4 Taijul Bldg, Doonchundong, Kangdong-gu, Seoul 134-060, Korea. Telephone: 82-2-470-9773; Fax: 82-2-470-6342; e-mail: hoeonkim@seoulcord.co.kr
# L+ l% ?4 \0 F
1 o% _7 r0 l- V5 m4 q. O( L7 sABSTRACT# Y: q# B" r* R  D2 z( v

& g  X: m& t  {& x9 UBone marrow (BM) has been regarded as a good source of both hematopoietic stem/progenitor cells and mesenchymal stem cells (MSCs) . These stem cells have the capacity for self-renewal and differentiating into cells of multiple lineages. MSCs derived from BM are capable of not only supporting hematopoiesis but also differentiating into mesodermal layer cells such as osteoblasts, chondrocytes, adipocytes, and myoblasts . However, the process to collect BM is invasive to donors and can cause complications such as infection, bleeding, and chronic pain, thereby limiting a wide application of BM-derived MSCs in tissue engineering and cell therapy.3 u6 N! T' U3 O
: Q3 v- i% s$ _3 R) S8 n- d
In recent decades, human umbilical cord blood (UCB) has been explored as an alternative source to BM for cell transplantation and cell therapy because of its hematopoietic and nonhematopoietic (mesenchymal) components. In contrast to BM aspiration, human UCB is obtained by a simple, safe, and painless procedure when the baby is delivered. Since the late 1980s, UCB has become an indispensable source of hematopoietic stem/progenitor cells for transplantation of hematopoietic stem cells to treat some hematological disorders . However, human UCB has been controversial for the presence of MSCs; some researchers successfully isolated MSCs from UCB, whereas others have not . Nevertheless, several groups reported that the UCB-derived MSCs could proliferate ex vivo and differentiate, at least, into osteoblasts and adipocytes . No evidence has shown yet that UCB-derived MSCs differentiate into skeletal myoblasts, but they are believed to have such a potential.
- Q; a; s& C. r$ @" O. j2 S- a: g. f8 {3 c8 ?1 Z
Myogenic differentiation is regulated by a family of myogenic regulatory factors (MRFs), including Myf5, MyoD, myogenin, and MRF4; MyoD and Myf5 are required for the determination of skeletal myogenic lineages, whereas myogenin and MRF4 are thought to regulate cell fusion and terminal differentiation . In postnatal life, the satellite cells located between muscle fiber sarcolemma and basal lamina are quiescent myoblasts, but they are fully determined to myogenic phenotype so that, once activated, they are capable of terminal differentiation . The quiescent satellite cells do not express transcription factors of a MRF family, whereas the activated ones exhibit a battery of molecular markers of Myf5, MyoD, and, to a lesser extent, myogenin. These satellite cells were once regarded as an ideal source for muscle regeneration and repair, but it turned out that they were few in injured muscle and that they were exhausted immediately during healing processes. A search for an alternative source with equivalent myogenic potential yielded MSCs not long ago when the BM-derived MSCs were shown to expand in vitro and differentiated successfully into myoblasts .
& w7 L! _4 L8 k5 ?# l7 q# U
" {) ?! Q% Z; F3 {/ UIn this paper, we report that fibroblast-like cells from human UCB, exhibiting mesenchymal phenotypes, are also able to differentiate into cells that express several skeletal muscle–specific genes. Our findings implicate that UCB is a potential source of MSCs for therapy of degenerative muscular diseases or muscle damage/loss from trauma.
, _4 U! V- k3 T$ }. E& M5 o9 H; Z/ U" M) W
MATERIALS AND METHODS. A$ r/ q8 r, z! l+ f9 E" H

8 M& d  W: g5 }+ DCharacteristics of UCB-Derived Adherent Cells/ {+ u; T# r  O4 L5 f
% Y- A* x$ d, z$ v: i  `
The mononuclear cells were obtained from UCB by Ficoll-Paque density gradient centrifugation and plated in the culture flasks. After 5 days of culture, nonadherent cells were removed by medium change. The adherent cells were small and rounded in shape. These cells grew larger and seemed to be comprised of heterogeneous cells, as judged by their appearance. The elongated cells began to appear among rounded cells between 8 and 15 days of culture, and they continued to grow to become fibroblast-like cells. By two or three passages of culture, the adherent cells became a population comprised mainly of bipolar fibroblast-like cells and could grow to confluency.( G! o7 O" i! ?6 g. o3 j9 d* Q* ^
, A1 u$ c4 ]- s* G
We examined the proliferation characteristics of the fibroblast-like cells at the fourth passage. The population-doubling time of cells is approximately 60 hours, as determined by viable counting. FACS analysis showed that 86% of cells were in the phase of G0/G1.
4 d" f3 q' @+ M  R0 E5 ?0 t" S1 O5 G0 M9 `, ~7 ~
Immunophenotyping of UCB-Derived Adherent Cells: Z% o" Q! V, f$ @$ G7 m

: d1 a( S' L, t9 M' `* }To characterize the adherent cell population derived from UCB, expression of a variety of CD markers and intracellular antigens like ASMA was examined by flow cytometry. Those adherent cells expressed CD13, CD29 (? 1 integrin), CD44, CD49e (5 integrin), CD54 (ICAM-1), CD90 (Thy-1), ASMA, CD105/SH2/endoglin, and CD73/SH3 (Fig. 1). Among these, SH2 and SH3 are well known as MSC-specific antigens. They expressed neither hematopoietic lineage markers such as CD34 nor monocyte-macrophage antigens such as CD14 and CD45 (Fig. 1). The lack of expression of CD14, CD34, and CD45 suggests that cell cultures were depleted of hematopoietic cells during subcultivation.
& {4 \! H( y' c7 _
7 P+ ?+ k% q; X- ?Figure 1. Immunophenotyping of umbilical cord blood–derived mesenchymal stem cells. Mesenchymal stem cells were detached, labeled with FITC- or phycoerythrin-conjugated monoclonal antibodies, and detected by flow cytometry. Relative number cells (counts) versus fluorescence intensity are presented. Abbreviations: ASMA, -smooth muscle actin; FITC, fluorescein isothiocyanate.
8 Y) V; S% M( P7 q
$ I; w8 M6 {% d  e  GThe adherent cells were also negative for expression of CD49d (4 integrin), CD106 (VCAM-1), and CD31 (an endothelial-related antigen) (Fig. 1). Similar to BM-derived MSCs, the cell population was positive for HLA class I but not for HLA DR (Fig. 1). All data above indicate that the adherent cells derived from UCB exhibit the phenotype of MSCs.
, r( t$ Y/ _+ {) U) x# H; _5 h7 L& |5 ?1 g$ I% m$ C7 Y  _% M
FACS and Reverse Transcription–PCR Analyses of Myogenic Differentiation. K8 ]3 o4 d$ |8 |7 h# q3 J2 S; T
! A- j0 C9 C  H+ g8 }$ o& G
A potential of UCB-derived MSCs differentiating into osteoblasts, chondrocytes, and adipocytes was demonstrated elsewhere . To investigate whether UCB-derived MSCs show a potential to differentiate into skeletal muscle cells, MSCs were cultured for up to 6 weeks in myogenic medium containing dexamethasone and hydrocortisone. At different time intervals, treated cells were observed by phase-contrast microscopy and then analyzed by flow cytometry with monoclonal antibodies against two muscle-specific transcription factors, MyoD and myogenin, as well as a skeletal protein, fast-twitch myosin. At week 1, MyoD and myogenin were expressed in approximately 8.7% and 90% of the treated cells, respectively, whereas non-treated cells remained unstained against anti-MyoD and anti-myogennin antibodies (Fig. 2A). However, the expression of MyoD and myogenin quickly vanished from week 2. This result is consistent with the fact that the two factors are involved in early myogenesis.
0 r' u8 o+ [! V; k+ g
( S, I1 B2 J5 vFigure 2. Myogenic differentiation of MSCs isolated from human umbilical cord blood. (A): After 1 and 2 weeks of induction in myogenic medium, cultures were analyzed by FACS for MyoD1 and myogenin. (B): Cells were incubated with myogenic medium for up to 6 weeks and were then analyzed by FACS for fast-twitch myosin. (C): MyoD1, myogenin, and myosin heavy chain mRNA levels were measured by reverse transcription–polymerase chain reaction during MSC differentiation in myogenic medium. For corresponding controls, cells were cultured in control medium (culture medium with no addition of dexamethasone and hydrocortisone). Abbreviations: FACS, fluorescence-activated cell sorting; MSC, mesenchymal stem cell./ z, M$ j# L+ R. x6 ~

: X3 f$ C& w. _6 Y, W3 K. J  WOn the other hand, fast-twitch myosin began to express only after 3 weeks of induction, and approximately 55.7% of treated MSCs at week 6 were visibly stained with monoclonal anti-skeletal myosin antibodies (Fig. 2B). This finding is also not surprising when considering that myosin is an element of skeletal muscle fibers that appears in late myogenesis.0 F& c2 l5 f5 J: q6 K, E

" ^6 n6 ~7 u- u, t9 j' n# J% kSkeletal myoblast differentiation of UCB-derived MSCs was also analyzed by semiquantitative reverse transcription (RT)-PCR of MyoD, myogenin, and MyHC. None of these factors were significantly expressed in the cells treated with nonmyogenic medium. In the case of the cells treated with myogenic medium, however, the mRNA levels of both MyoD and myogenin were significantly increased after 3 days (Fig. 2C). At week 1, the mRNA level of myogenin was highly increased to reach a presumed peak, whereas that of MyoD subsided quickly. The mRNA levels of both factors were almost abolished after 2 weeks (Fig. 2C). The mRNA of MyHC, on the other hand, appeared after 3 weeks of induction, and its expression steadily increased until the sixth week (Fig. 2C).
1 X* p) K, W2 i
! Q, s# _; G8 u, nImmunocytochemical Analysis of Myogenic Differentiation) [! G  g9 t$ j8 [& t) }5 o
) l4 L( F7 V7 v1 j- U0 X
To further confirm myogenic differentiation of UCB-derived MSCs, cells were examined immunocytochemically with monoclonal anti-MyoD, anti-myogenin, and anti-skeletal myosin antibodies. Figure 3A shows nuclear staining of MyoD and myogenin in treated cells. Consistent with our previous RT-PCR and FACS results, expression of myogenin was much higher than that of MyoD at 1 week of induction (Fig. 3A). Western blot analysis indicated that fast-twitch myosin, which appeared as a 200,000-dalton protein band, was highly expressed in the cells incubated for 6 weeks (Fig. 3B). Taken together with the data above, it is very likely that human UCB-derived MSCs are able to differentiate into skeletal myoblasts., `) W- i7 J8 W) P' i+ T

/ w& S0 O. A: X, e0 s! A2 }+ W" IFigure 3. Immunocytochemistry and Western blotting of umbilical cord blood–derived mesenchymal stem cells cultured in myogenic medium. (A): Immunocytochemical staining of the cells cultured in myogenic medium for 1 week. The cells were stained with MyoD or myogenin antibodies followed by a horseradish peroxidase (HRP)–conjugated secondary antibody and were colorized with the substrates. The control was stained only with the secondary antibody. (B): Western blot of the protein extract from the cells cultured in myogenic medium for 6 weeks. The blot was stained with a fast-twitch antibody followed by an HRP–conjugated secondary antibody. The level of -actin protein was used as a loading control.
7 F1 |! A% }+ J9 a! p" b
6 h3 k# m. p2 Z+ k. vDISCUSSION
7 _& ^! V5 @$ a, L7 E- m. M& b) _% M% W4 s* u: w( Y9 H
This research was supported in part by a grant (SC13032) from Stem Cell Research Center of the 21st Century Frontier Research Program funded by the Ministry of Science and Technology and a grant (01-PJ10-PG8-01EC01-0015) of Korea Health 21 R&D Project funded by the Ministry of Health and Welfare, Republic of Korea.$ y5 U7 `. q6 [' E! v: ?
/ y6 [1 |" x  g1 ~
REFERENCES
$ L- ?( Y3 o9 Z% w/ i- U7 D9 I
) L, Q. O' Z+ z, y& U$ aGolfier F, Barcena A, Harrison MR et al. Fetal bone marrow as a source of stem cells for in utero or postnatal transplantation. Br J Haematol 2000;109:173–181." ^% w4 J) f- `; L* M
1 e% v2 |& a6 Y2 l
Reyes M, Lund T, Lenvik T et al. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood 2001;98:2615–2625.( s, t- @4 v1 a7 C- ~

8 n- Y6 y3 L/ fAlbella B, Segovia JC, Guenechea G et al. Ex vivo expansion of hematopoietic stem cells. Methods Mol Biol 2003;215:363–373.
8 @( O/ l1 m- n: _8 p1 e) ~- V, Q! |' u
Jones EA, Kinsey SE, English A et al. Isolation and characterization of bone marrow multipotential mesenchymal progenitor cells. Arthritis Rheum 2002;46:3349–3360.
9 ]0 b: h0 L1 e- ?7 U3 _- C1 o  H
( ^% ~* x5 g8 X) i6 O. DSekiya I, Larson BL, Smith JR et al. Expansion of human adult stem cells from bone marrow stroma: conditions that maximize the yields of early progenitors and evaluate their quality. STEM CELLS 2002;20:530–541./ L+ f: v$ c2 c" d; u9 q

  {  u* U7 [% m' Y% w* z! T" `% vPittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–147./ @( B# R: |$ Q/ L1 v

. Z# K% ^$ w- X5 z" ^" Q2 vImabayashi H, Mori T, Gojo S et al. Redifferentiation of dedifferentiated chondrocytes and chondrogenesis of human bone marrow stromal cells via chondrosphere formation with expression profiling by large-scale cDNA analysis. Exp Cell Res 2003;288:35–50.( f7 u. S  z+ `7 D. |

( T; M# x, k  C7 A1 C" VWakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve 1995;18:1417–1426.
% o. K/ H7 D9 s+ o) j! B
9 z& {: u' v8 YCohena Y, Nagler A. Hematopoietic stem-cell transplantation using umbilical cord blood. Leuk Lymphoma 2003;44:1287–1299.
) m; S! C" ]8 N3 l1 T4 @0 U
8 h3 D9 j) D+ [0 ^# g6 ^, r2 a$ HLong GD, Laughlin M, Madan B et al. Unrelated umbilical cord blood transplantation in adult patients. Biol Blood Marrow Transplant 2003;9:772–780.$ B; U  b$ K6 C5 I# b

# Q& w! E6 Q3 u! yOoi J, Iseki T, Takahashi S et al. Unrelated cord blood transplantation for adult patients with de novo acute myeloid leukemia. Blood 2004;103:489–191.) }6 o3 [  [: U8 S
$ N8 f4 \8 O( Z; z; j0 Y9 w: v8 G
Frassoni F, Podesta M, Maccario R et al. Cord blood transplantation provides better reconstitution of hematopoietic reservoir compared with bone marrow transplantation. Blood 2003;102:1138–1141.
; q) d" T. P, V' e+ ?, R! g+ ]9 u# g# c% a, H
Bhattacharya A, Slatter M, Curtis A et al. Successful umbilical cord blood stem cell transplantation for chronic granulomatous disease. Bone Marrow Transplant 2003;31:403–405.
; l0 p8 |( t+ f# }5 Y" F) b0 D: ?# y& M" y& b
Laughlin MJ, Barker J, Bambach B et al. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors. N Engl J Med 2001;344:1815–1822.
- {. ?- r0 u5 T! `# }
& M/ N1 G3 m: }7 fWexler SA, Donaldson C, Denning-Kendall P et al. Adult bone marrow is a rich source of human mesenchymal "stem" cells but umbilical cord and mobilized adult blood are not. Br J Haematol 2003;121:368–374., G6 J/ m0 R  \+ p) O/ ~% u

2 i2 p( I. `6 Z; MMareschi K, Biasin E, Piacibello W et al. Isolation of human mesenchymal stem cells: bone marrow versus umbilical cord blood. Haematologica 2001;861099–1100./ M+ e5 g! n) y
6 \# ?% x7 A$ r5 ~9 G; d
Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 2000;109:235–242.
8 G7 Q3 B% x6 L9 |' a3 c  ]5 b6 H2 i; Y6 Z6 e
Goodwin HS, Bicknese AR, Chien SN et al. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow Transplant 2001;7:581–588.7 s7 n; a9 H) n
; w% j5 K. `. G" @7 \' ^
Romanov YA, Svintsitskaya VA, Smirnov VN. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC-like cells from umbilical cord. STEM CELLS 2003;21:105–110.
, y1 x  G7 B+ K" s, j
0 ~6 W9 z6 g4 oAurade F, Pinset C, Chafey P et al. Myf5, MyoD, myogenin and MRF4 myogenic derivatives of the embryonic mesenchymal cell line C3H10T1/2 exhibit the same adult muscle phenotype. Differentiation 1994;55:185–192.0 B3 v; O9 W8 F" ^) f

! Q2 z& ?5 w% PRohwedel J, Maltsev V, Bober E et al. Muscle cell differentiation of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents. Dev Biol 1994;164:87–101.
$ t) ]  p- I# {2 B; R) k
1 O4 B) \6 I! o4 J8 j" \Shimokawa T, Kato M, Ezaki O et al. Transcriptional regulation of muscle-specific genes during myoblast differentiation. Biochem Biophys Res Commun 1998;246:287–292.
) `  T+ H: P* S' t; P  _( v6 D7 a; E
Valdez MR, Richardson JA, Klein WH et al. Failure of Myf5 to support myogenic differentiation without myogenin, MyoD, and MRF4. Dev Biol 2000;219:287–298.
0 ?7 t' ]/ R; s2 t  y! {6 {2 n, G
# u# ^+ n2 F; z5 n# WRantanen J, Hurme T, Lukka R et al. Satellite cell proliferation and the expression of myogenin and desmin in regenerating skeletal muscle: evidence for two different populations of satellite cells. Lab Invest 1995;72:341–347.
  A, p% o" z$ `6 d+ W
$ E# B0 N& o% K9 x9 N3 W1 f# hSchultz E. Satellite cell proliferative compartments in growing skeletal muscles. Dev Biol 1996;175:84–94.
- `) d' j( C8 o0 d6 ^, m% I- |/ L* J& e6 R* Y
Zammit P, Beauchamp J. The skeletal muscle satellite cell: stem cell or son of stem cell? Differentiation 2001;68:193–204.
% |( c% B+ N5 X% S3 W: F2 D+ e3 }" }* D  L7 p& }" g" `8 d
De Angelis L, Berghella L, Coletta M et al. Skeletal myogenic progenitors originating from embryonic dorsal aorta coexpress endothelial and myogenic markers and contribute to postnatal muscle growth and regeneration. J Cell Biol 1999;147:869–878.9 M0 o  d; L3 ?2 D0 Z6 @
, a& F( ^5 p& f) \
Zuk PA, Zhu M, Mizuno H et al. Multilineage cells from human adipose tissue: implications for cell-based therdapies. Tissue Eng 2001;7:211–228.
5 z% S' w$ @/ r; s# _) Q7 t0 Y* y% q# ~) n- K8 C
Mizuno H, Zuk PA, Zhu M et al. Myogenic differentiation by human processed lipoaspirate cells. Plast Reconstr Surg 2002;109:199–209.
4 w/ t! j7 S2 C  [0 R' P+ h" N, m- Q7 l! f6 l' G
Zuk PA, Zhu M, Ashjian P et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 2002;13:4279–4295.
$ j5 ]& `, Z; T# a6 P( ~* x2 P0 X, y" G2 o* ?6 H  W% M
Michelagnoli MP, Burchill SA, Cullinane C et al. Myogenin: a more specific target for RT-PCR detection of rhabdomyosarcoma than MyoD1. Med Pediatr Oncol 2003;40:1–8.
7 }) V7 R8 I  ^9 f7 `6 B( D
& r/ ?: J5 h( R1 V) HUeda T, Araki N, Mano M et al. Frequent expression of smooth muscle markers in malignant fibrous histiocytoma of bone. J Clin Pathol 2002;55:853–858.
& a! v  U6 ^) d( ~, i
2 ]3 O* Z3 X  |( GEdmondson DG, Olson EN. Helix-loop-helix proteins as regulators of muscle-specific transcription. J Biol Chem 1993;268:755–758.(Eun Ji Gang, Ju Ah Jeong,)

Rank: 2

积分
136 
威望
136  
包包
1877  
沙发
发表于 2015-5-30 15:12 |只看该作者
一定要回贴,因为我是文明人哦  

Rank: 2

积分
61 
威望
61  
包包
1757  
藤椅
发表于 2015-7-22 00:00 |只看该作者
正好你开咯这样的帖  

Rank: 2

积分
97 
威望
97  
包包
1738  
板凳
发表于 2015-8-9 20:52 |只看该作者
干细胞之家微信公众号
端粒酶研究

Rank: 2

积分
75 
威望
75  
包包
2118  
报纸
发表于 2015-8-16 19:17 |只看该作者
小心大家盯上你哦  

Rank: 2

积分
116 
威望
116  
包包
1832  
地板
发表于 2015-9-11 22:36 |只看该作者
这贴子你会收藏吗  

Rank: 2

积分
69 
威望
69  
包包
1788  
7
发表于 2015-9-13 15:10 |只看该作者
我想要`~  

Rank: 2

积分
161 
威望
161  
包包
1862  
8
发表于 2015-9-18 09:18 |只看该作者
谢谢哦  

Rank: 2

积分
68 
威望
68  
包包
1752  
9
发表于 2015-10-16 11:27 |只看该作者
…没我说话的余地…飘走  

Rank: 2

积分
129 
威望
129  
包包
1788  
10
发表于 2015-10-29 18:08 |只看该作者
说嘛1~~~想说什么就说什么嘛~~  
‹ 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, 2024-5-1 07:40

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.