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: 262721|回复: 237
go

Functional Expression of HGF and HGF Receptor/c-met in Adult Human Mesenchymal S [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:38 |只看该作者 |倒序浏览 |打印
a Interdisciplinary Center for Clinical Research on Biomaterials, IZKF BIOMAT, Aachen, Germany;
1 j" C- m, G' s; m+ W4 b" j: ?2 T7 n% B
b Institute of Pathology, University Hospital, Aachen, Germany
4 h' D+ |+ P+ X1 _8 b7 |( Y+ \$ {) }4 {) Q* F
Key Words. HGF ? Mesenchymal stem cells ? Cell migration ? Mobilization
) h& j5 A+ I* d2 K2 {6 l- M! H4 U) i% l- o5 F' C: e  f9 L
Willi Jahnen-Dechent, Ph.D., IZKF BIOMAT, University Hospital, Pauwelsstrasse 30, D-52074 Aachen, Germany. Telephone: 49-241-80-80163; Fax: 49-241-80-82573; e-mail: willi.jahnen@rwth-aachen.de
0 e! R# P, M9 c
$ \: P9 p; B' D, ]1 z. k  G* FABSTRACT6 v7 i5 Z- |' S) w5 I. z
9 S" j) k( H! z9 j5 @
A classic concept of tissue repair holds that inflammatory cells enter the damaged tissue and signal resident tissue-specific progenitor cells (e.g., parenchymal cells, fibroblasts) for mitosis. Several studies suggest that multipotent (bone marrow) mesenchymal stem cells can also contribute to tissue repair after mobilization, migration, and engraftment of the damaged tissue . In addition, circulating immature cells seem to participate in regeneration of many different tissues . The role of mesenchymal stem cells in tissue remodeling was shown in different in vivo models〞for example, for hepatic regeneration , muscle regeneration , and infarcted myocardium . Not surprisingly mesenchymal and circulating stem cells have attracted increasing attention because they hold great therapeutic potential for endogenous tissue repair and tissue engineering.' N' e* X7 O! |# _1 w$ T; }# d- n
- R& v: \' h: n3 X$ ?) L
Since the original report from Friedenstein et al. , a number of different protocols have been defined to isolate multipotent adult mesenchymal stem cells from bone marrow specimen . Unlike hematopoietic stem cells, human mesenchymal stem cells (hMSC) adhere to cell culture plastic, which is exploited for their isolation . HMSC express CD105 and CD73 but not the lineage-specific surface antigens CD14, CD34, and CD45 . Markers specific for hMSC are not known. Therefore, putative hMSC isolates have to be verified by their capacity to differentiate at least into adipocytes, chondrocytes, and osteoblasts. In addition, bone marrow-derived mesenchymal stem cells can be differentiated in vitro into bone marrow stromal cells and into endothelial, myogenic, hepatic, and neurogenic cells. Cell transplantation studies in human patients and in animals have demonstrated that bone marrow-derived cells can colonize most organs. The colonization was much enhanced by inflammation accompanying, for example, graft rejection or infarction. Hence, successful engraftment of several organs was enhanced by irradiation , chemical injury, and genetic diseases  or following infarction . Despite major advances in MSC biology, our knowledge of the signals required for MSC mobilization and migration to the injured tissue site lags behind the extensive experience with hematopoetic stem cells, which are in routine clinical use. Therefore we sought to determine which factors may be responsible for mobilization of hMSC. Hepatocyte growth factor/scatter factor (HGF/SF) is a multipotent growth factor that exerts a mitogenic, motogenic, and morphogenic response on cells expressing c-met, the cellular HGF receptor. HGF/SF is essential in paracrine signaling of mesenchymal and epithelial cells, particularly during embryogenesis, repair, and carcinogenesis . In malignant and transfected cells autocrine stimulation has been described . In pathology, HGF/SF has been shown to induce tumor cell invasion in tissues .4 |, C7 v6 y$ L% K
9 B7 `/ n4 @8 h( j" H
Studies on HGF and c-met expression in bone marrow cells were reported by Takai et al. . These authors demonstrated that bone marrow stromal cells constitutively express HGF and promote hematopoiesis. In addition, expression of c-met by stromal cells suggested an autocrine stimulation of stromal cells by HGF. However, it was not determined if HGF and c-met were also expressed by hMSC. This study was undertaken to address this question, including functional aspects of HGF and c-met-like cell migration and proliferation. We demonstrate that HGF and c-met are constitutively expressed by hMSC and that the expression of HGF is downregulated by transforming growth factor-? (TGF-?). Furthermore, HGF exerted a strong chemotactic stimulus on hMSC, which may be further enhanced by autocrine signaling through the HGF c-met pathway.2 w! U! n. Z  Q% ~, T% v1 d+ G

; Q) E. i  x9 X8 BMATERIALS AND METHODS# d2 M4 ?2 l6 j. T! C1 H5 L& D5 \

! a# i! k+ @$ S& n4 T8 |* z: c% iTotal RNA was extracted using guanidinium thiocyanate (RNeasy Kit; Qiagen; Hilden, Germany; http://www.qiagen.com). Reverse transcription was accomplished with 5 μg of total RNA using the "first-strand cDNA synthesis kit" (Amersham Pharmacia Biotech; Buckinghamshire, UK; http://www.amershambiosciences.com) with FPLCpureTM murine reverse transcriptase. PCR was carried out under the following conditions: denaturation at 95~C for 1 min, annealing at 54~C (HGF) or 60~C (c-met) for 1 min, extension at 72~C for 1 min (30 cycles), and a final extension at 72~C for 10 min. PCR amplicon size (266 bp for HGF and 440 bp for c-met) was analyzed by electrophoresis on a 2% agarose gel and visualized with ethidium bromide. The oligonucletoide primer and the GenBank/EMBL identifiers of template sequences were as follows:" E; d4 S5 P+ F  ?
' Q# m+ Q7 }5 Y) K9 ?# h
c-met (NM000245)
2 _0 k: Z) h/ \, N0 X
; p) a! Z) k' s: r1 M1 H# @forward (nt 1398–1423 ): 5'-AGAAATTCATCA GGCTGTGAAGCGCG-3'$ b; L- H  f. }5 q, P
! m( W  i% [) Y1 W
reverse (nt 1814–1838 ): 5'-TTCCTCCGATCG CACACATTTGTCG-3'
" Y0 x. j5 B/ G! M7 J  v: o. i: o# w# d8 \% F
HGF (XM168542)6 n2 P' G* v" F

! a  i. d# M! f6 ^, [2 Sforward (nt 548–568): 5'-GGTAAAGGACGCAGC TACAAG-3'# G2 a$ b6 e# o( r

+ s* B5 M1 k4 ^) r5 Q" nreverse (nt 794–814): 5'-ATAACTCTCCCCATTGC AGGT-3'* d$ A; g, d( j2 D5 K' ~8 y* e

" A# }9 w  q1 T2 A; [Immunohistochemistry& ^8 k- t$ f( I" Q( p. E! T9 \
! m/ Z& i# _  f7 R6 j/ A
Subconfluent hMSCs were grown in chamber slides and fixed for 30 min using 0.5% paraformaldehyde (in PBS). After washing with wash buffer (Dulbecco’s phosphate-buffered saline solution A , 0.5% BSA in PBS), unspecific protein-binding capacity was blocked for 15 min using blocking buffer (PBS with 5% BSA). Cells were incubated for 45 min at RT with the first antibody, diluted 1:50 in PBSSA-NP-40 (1:200 dilution of nonidet P-40  in PBSSA), washed three times for 10 min in PBSSA, followed by an incubation with the secondary antibody (1:250 in PBSSA) for 30 min at RT and again three washing steps. FITC-conjugated streptavidin was added (1:250 in PBSSA) for 30 min, protected from light. Finally, cells were washed three times with PBSSA for 10 min and mounted in 4',6'-diamidino-2-phenylindole hydrochloride (DAPI)-containing mounting medium.4 Q8 m: R, L  S3 R" [1 H! e

: J- R: {4 ~6 a$ z- ^/ YPolyclonal rabbit c-met antibody (primary antibody) was purchased from Santa Cruz Biotechnology (Santa Cruz, CA; http://www.scbt.com), and secondary antibody (biotin-conjugated goat anti-rabbit) and FITC-conjugated streptavidin from DAKO. As a negative control, cells were incubated with the first antibody, which had been preincubated overnight at 4~C with a specific blocking peptide (Santa Cruz Biotechnology).# Z1 F6 {9 e, F) T' g8 g+ E8 J
6 i, u  V2 O7 ^' I# {9 H
Western Blotting& S: F+ i6 @4 I/ a) i
$ P6 ?. O8 S% ]" Y% L( L/ x- m
Mesenchymal stem cells were lysed with insect cell lysis buffer (PharMingen International; Hamburg, Germany; http://www.pharmingen.com), and protein concentration was determined using the BCA-Kit (Pierce Biotechnology; Rockford, IL; http://www.piercenet.com). Ten μg of protein lysate and protein marker (Perfect Protein AP WB Marker; Novagen; Darmstadt, Germany; http://www.emdbiosciences.com) were separated on 4%-12% SDS gels (NuPAGE; Karlsruhe, Germany) at 70 V for 2.5 hours. Membranes were fixed with methanol, and after electroblotting (60 min, 150 mA) on polyvinylidene fluoride membrane (Bio-Rad; Munich, Germany; http://www.bio-rad.com), the membranes were blocked overnight with low-fat milk at 4~C. Immunostaining was accomplished by incubation with rabbit polyclonal antibody against c-met (1:200, Santa Cruz Biotechnology) for 90 min at room temperature. After 1 hour of incubation with alkaline phosphatase (AP)-conjugated secondary antibody (1:5,000; Roche; Mannheim, Germany; http://www.roche.com) at room temperature, staining was developed using Sigma Fast 5-bromo,4-chloro,3-indoyl phosphate/nitroblue tetrazolium (BCIP/NBT) tablets (Sigma). As a negative control, the first antibody was preincubated with a fivefold excess of blocking peptide in a small volume of PBS at 4~C overnight./ m$ p5 r  r/ M2 r; }8 Z' h

9 B8 \* V  Q: B" V; kHGF-ELISA( @* O  i0 b* N3 q* y

# a/ N6 Q; Z5 o. P8 Y  ^. rHGF concentration in hMSC-conditioned media was measured by Quantikine human HGF-ELISA (R&D Systems; Minneapolis, MN; http://www.rndsystems.com), which is based on a sandwich enzyme immunoassay technique with a precoated HGF-specific antibody. To this end, cells were seeded in 96-well plates (20,000 cells/well) and stimulated for 24 hours with TGF-?3 (10, 1, and 0.1 ng/ml), interleukin-1? (IL-1?) (10, 1, and 0.1 ng/ml), and bFGF (10, 1, and 0.1 ng/ml). Unstimulated cells served as a control. Supernatants were harvested and stored at -70~C until they were analyzed.: c: ?- N& Y- \/ ^* V2 {
0 k) @1 K5 ]4 m# i" i( D
Scratch Assay ? hMSC were grown to confluency in a six-well plate (Becton Dickinson). A scratch in the cell layer was made with a pipette tip over the total diameter of 34.5 mm. HGF was added at 0, 25, 50, and 75 ng HGF/ml medium. Closure of this "wound" was documented photographically (Axiovert 25; Zeiss; Cologne, Germany; http://www.zeiss.com) after 24 hours, and cells in four segments of the scratched area, each of 320 μm x 320 μm, were counted.# `3 a, R& S- C$ z

  f1 x8 T5 A$ G2 X; i9 Z: a- kBoyden-Chamber Assay ? For analysis of cell motility, 1 x 105 hMSCs/ml were seeded in the top compartment of a Boyden chamber (NeuroProbe; Gaithersburg, UK; http://www.neuroprobe.com). The bottom compartment contained different HGF concentrations and was separated from the top compartment by a polycarbonate membrane with 8 μm pores (Corning; Düsseldorf, Germany; http://www.corning.com). Cells were allowed to migrate for 16 hours at 37~C in a humidified atmosphere. After removing cells from the upper side of the membrane with cotton swabs, membranes were fixed, stained with hematoxylin (Merck; Darmstadt, Germany; http://www.merck.com), and transferred onto glass slides. Cells on the bottom side of the membrane were counted in five different highpower fields. Each analysis was performed in triplicate.
" v  h: x  u$ k
( s( p) a! X* w9 T6 k0 e: |' ^Cell Proliferation Assay ? Cells were seeded in 24-well plates (3,000 cells per well) and stimulated for 24 hours with HGF (0, 25, and 50 ng HGF/ml medium) in low serum (2% FCS). The medium containing 20% FCS was used as positive control. Proliferation was measured by detection of ATP content of the cells with a luciferase detection system (ViaLight HS; BioWhittaker; Verviers, Belgium; http://www.biowhittaker.be). This bioluminescent method uses luciferase, which catalyzes the formation of light from ATP and luciferin. The emitted light intensity is linearly related to the ATP concentration . ATP content was measured 1, 2, 3, 4, and 7 days after stimulation. The medium with HGF was renewed after day 4.; e, _, h) Q' \3 ]

: `  n; O5 d* bRESULTS& _$ X. E9 b, F( K' i, u

6 N/ v% p1 |5 }2 X2 ZAdult pluripotent stem cells are thought to reside in many tissues. They can be isolated from skin  and adipose tissue , and a baboon study has shown that infused stem cells can colonize a wide range of tissues〞for example, gastrointestinal, kidney, lung, liver, thymus, and skin . However, the amount of stem cells in most tissues is exceedingly low, which leads to the hypothesis that injured tissues produce appropriate cues for engraftment and, furthermore, that the local microenvironment would stimulate the differentiation of engrafted (stem) cells into functional, specialized cells . This begs the question of which signals are necessary and sufficient for stem cell mobilization and recruitment. HGF is well known to augment cell migration, scattering (i.e., "scatter factor"), and proliferation of many different cell types, predominantly of epithelial cells. Furthermore, HGF is considered to be a humoral mediator of organogenesis and morphogenesis of various tissues and organs, as well as regeneration of organs, tumor invasion, and metastasis .( N# q+ ]) n* `* U

2 @2 v( ^5 M+ cIn the bone marrow, HGF is known as an important hematopoietic regulator . Synthesis and release of HGF precursors could be part of an autocrine mechanism. Pre-HGF has to be activated by proteolytic cleavage〞for example, through serine proteases like uPA and tPA, which are also released by hMSC (Neuss et al., unpublished observations). Tissue repair and wound healing are regulated by soluble mediators provided by inflammatory cells. Therefore, we investigated the effect of bFGF, IL-1, and TGF-? on the production of HGF. TGF-?, which is known to be growth inhibitory for epithelial cells, downregulates the production of HGF by hMSC. This effect is not attributed to cell death after TGF-? stimulation (data not shown). In contrast, basic fibroblast growth factor, which stimulates blood vessel formation and angiogenesis , and the proinflammatory cytokine IL-1? have no significant influence on the expression of HGF (determined by ELISA). Nevertheless, bFGF upregulates the production of the pre-HGF-activating serine protease tPA in hMSC (data not shown). Thus, production of mature HGF is regulated by the microenvironment, depending on processes like tissue damage or inflammation. Activated monocytes or macrophages, which are replete at damaged tissue sites, are known to produce HGF/c-met . On a more general note, HGF concentration is increased at sites of tissue damage . After partial hepatectomy in the rat, HGF levels are even measurably elevated in the blood . Taken together, these increases in local and systemic HGF could provide a key chemotactic signal to mobilize and attract hMSC for tissue repair.* e$ c! L, f) T6 ~* e: g

3 N/ n4 y& Y- l( k. j. |Here we identified HGF/SF as a potent regulator of hMSC function, regulating migration and proliferation in vitro. Our study demonstrates the production of HGF and the expression of the HGF receptor c-met by a defined stromal cell population, human mesenchymal stem cells. These cells were shown by FACS analysis to lack any lineage-specific surface marker expression, including CD4, CD14, CD34, and CD117. In addition, the cells were capable of differentiation into adipocytes, chondrocytes, and osteoblasts. Accordingly, these cells represent the bone marrow stromal cell population described by the groups of Pittenger  and Haynesworth  as MSC. Migration of hMSC was increased by HGF in Boyden chamber assays (chemotactic effect), whereas proliferation of hMSC was negatively influenced. Furthermore, HGF promoted the repopulation of a cell-free "wound" in a cell monolayer wounding model. We attribute this repopulation mainly to the exogenously added HGF, since hMSC did not upregulate HGF production after the cell monolayer was wounded (data not shown).
+ X& Z; o, x3 B3 z) n2 l5 m' t  C
Expression of HGF and c-met was also reported by Takai  in a bone marrow stromal cell population, but this population was not further characterized. Bone marrow stromal cells are widely used as feeder layers for long-term cultures of hematopoietic stem cells. Due to different isolation and culture procedures, these cells are less homogenous than the MSC used here. It is possible, however, that hMSCs are also contained in bone marrow stromal cells and that, in fact, hMSCs are the source of HGF required for hematopoiesis in the coculture system. This conclusion is supported by the fact that our hMSC produced similar amounts of HGF as did the feeder cells described by Takai et al.  and, furthermore, that neither the bone marrow stromal cells nor our hMSC showed increased proliferation in response to HGF. The amount of HGF expressed by hMSC (0.7–2 ng/ml) is higher than the normal HGF serum concentration (0.24–0.33 ng/ml ). As mentioned, HGF concentration can be elevated in the blood after wounding occurs . We suggest that increased systemic HGF may mobilize bone marrow or tissue resident hMSC to colonize damaged target organs. According to our in vitro results, the elevated HGF should also inhibit hMSC proliferation. We speculate that the local environment will ultimately determine hMSC differentiation into mature tissue cells, as is the case in embryonic development.% _( Q3 @2 ]/ j& M' I& B$ P- q

+ Q- n6 S9 h8 vHGF could possibly help to mobilize autologous hMSC and to direct possible allogeneic hMSC in future stem cell therapy. The role of HGF and its possible role in dysregulation of wound healing (like that described for TGF-? and other growth factors in diabetic mice, for instance)  have to be further clarified. The observation that ischemic or traumatic rat brain extracts induced production of HGF in hMSC  also supports the idea of HGF as an important factor for tissue repair by (transplanted or autologous) hMSC. The notion that HGF/c-met signaling may be involved in hMSC mobilization and recruitment to damaged tissues is entirely compatible with previous reports that this important regulator of cell motion and differentiation is critically involved in normal development of epithelial tissues , as well as the metastatic spread of tumor cells .
4 X1 C$ ]& B1 V/ r; s0 L5 R/ ^
$ G. R( N4 `! c0 E+ G; K7 FACKNOWLEDGMENT
2 }/ w, T8 z* g  d; ]7 h
- `" M4 J; T+ v; C: F1 N# k* These authors contributed equally.
, K2 j5 `, {( q: X+ x& H
! i: h- X$ }! N. `0 W9 {REFERENCES
: x  R- b# m3 P3 r2 k8 H" I+ n+ h& a! i2 O, |2 G8 b' ^) A0 z
Springer ML, Brazelton TR, Blau H et al. Not the usual suspects: the unexpected sources of tissue regeneration. J Clin Invest 2001;107:1355–1356.! K( v& `" M" [* A: z

  o! Y0 }3 L& {: ~Jackson KA, Majka SM, Wang H et al. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest 2001;107:1395–1402.% p0 X% @! \% |( ^

" J/ U* A( m! MHillebrands JL, Klatter FA, van den Hurk BM et al. Origin of neointimal endothelium and ?-actin-positive smooth muscle cells in transplant arteriosclerosis. J Clin Invest 2001;107:1411–1422.9 J+ }# y. V" P  W, q7 ^8 d5 K& X
+ ], c! b8 X' \
Petersen BE, Bowen WC, Patrene KD et al. Bone marrow as a potential source of hepatic oval cells. Science 1999;284:1168–1170.2 O2 `+ s* L. c- @8 u
- J5 `. ]9 s& \! _6 K
Jiang Y, Jahagirdar BN, Reinhardt RL et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;18:41–49./ h# g0 \3 N' n$ {& l
& M2 X, w, ^5 b3 V
Ferrari G, Cusella-De Angelis G, Coletta M et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science 1998;279:1528–1530.( t6 J; v2 W: [( i& n3 \" H( P5 L

! f: ]' _& ?+ r, j3 G2 t7 z( \De Bari C, Dell’Accio F, Vandenabeele F et al. Skeletal muscle repair by adult human mesenchymal stem cells from synovial membrane. J Cell Biol 2003;160:807–809.
, \8 j, g4 M4 f; w2 o. [! c
0 |* ^* n0 y6 m" AToma C, Pittenger MF, Cahill KS et al. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2002;105:93–98.
, K4 X  x7 T* a+ y' i8 o1 X2 q$ j2 G; a
Shake JG, Gruber PJ, Baumgartner WA et al. Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg 2002;73:1919–1925.
7 h( ^, a. V9 m: J: [% a) G9 w% j8 X% z
Friedenstein AJ, Petrakova KV, Kurolesova AI et al. Heterotropic transplants of bone marrow: analysis of precursor cells for osteogenic and heterotropic tissues. Transplantation 1968;6:230–247.& |& p! s* R: ]- j! u! u/ V: H
& j: v$ S, |0 X, _9 P  b
Quirici N, Soligo D, Bossolasco P et al. Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol 2002;30:783–791.; e# X$ i% {: J6 ?4 p3 ?

) _; }# ~" G& {1 DReyes M, Lund T, Lenvik T et al. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood 2001;98:2615–2625.1 O6 h& r" I: Y' x

& p$ Q1 K# }$ o1 ~, E; y" X6 z3 m- RHaynesworth SE, Goshima J, Goldberg VM et al. Characterization of cells with osteogenic potential from human marrow. Bone 1992;13:81–88.
& ]/ \' c5 Z2 r+ w
4 A) t5 S% D4 F8 D) q0 P4 }Pittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–146.
+ m+ W2 f3 L" [/ q4 b) c
: k( h' c$ H% I/ g2 b/ _9 ?6 hKrause DS, Theise ND, Collector MI et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 2001;105:369–377." j/ _0 i3 K. x0 q% X

' a9 b. q+ I( V/ mStephens P, Hiscox S, Cook H et al. Phenotypic variation in the production of bioactive hepatocyte growth factor/scatter factor by oral mucosal and skin fibroblasts. Wound Rep Reg 2001;9:34–43.
8 |) j0 t* P! e" m' {0 y  o* z1 Y7 i
+ m2 u$ k$ O0 W& Y+ D' `Stuart KA, Riordan SM, Lidder S et al. Hepatocyte growth factor/scatter factor-induced intracellular signalling. Int J Exp Path 2000;81:17–30.
  F- M* l9 ?; s
  A0 o) a! J0 {Qian LW, Mizumoto K, Maehara N et al. Co-cultivation of pancreatic cancer cells with orthotropic tumor-derived fibroblasts: fibroblasts stimulate tumor cell invasion via HGF secretion whereas cancer cells exert a minor regulative effect on fibroblasts HGF production. Cancer Lett 2003;190:105–112.
, A2 M' M2 B- ?( A
- A) d; S" F( }$ n# V; BTakai K, Hara J, Matsumoto K et al. Hepatocyte growth factor is constitutively produced by human bone marrow stromal cells and indirectly promotes hematopoiesis. Blood 1997;89:1560–1565.
) X  {) @3 q( }& W) z
1 I3 D( p  ~( e9 kSchuldiner M, Yanuka O, Itskovitz-Eldor J et al. Effects of eight growth factors on the differentiation of cells derived from human embryonic stem cells. PNAS 2000;97:11307–11312.
+ \2 i& c- y/ B/ M, M% V
7 N8 ?2 s1 X7 [9 WCrouch S, Kozlowski R, Slater K et al. The use of ATP bioluminescence as a measure of cell proliferation and cytotoxicity. J Immunol Methods 1993;160:81–88.1 b- L9 _' F9 ]: e8 @
+ E6 U( D# o1 T- I
Prockop DJ, Sekiya I, Colter DC. Isolation and characterization of rapidly self-renewing stem cells from cultures of human marrow stromal cells. Cytotherapy 2001;3:393–396.3 ?. b. b' ?3 e6 C

: S% Z" v: K, yToma JG, Akhavan M, Fernandes KJ et al. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol 2001;3:778–784.
7 C' s  e- o8 o9 X9 [) a8 X, ?/ u/ }! _  _* [0 b1 e
Zuk PA, Zhu M, Mizuno H et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001;7:211–228.
1 p* ^( f* Z9 F+ C" R, Z$ r4 X& K
4 c% P" t2 I- I9 J$ u1 XDevine SM, Cobbs C, Jennings M et al. Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood 2003;101:2999–3001.
) {5 R5 g. H1 t- R  A+ x0 s+ T+ G: d: y( ]1 @1 K& i3 ~2 A' V* H2 F
Matsumoto K, Date K, Ohmichi H et al. Hepatocyte growth factor in lung morphogenesis and tumor invasion: role as a mediator in epithelium-mesenchyme and tumor-stroma interactions. Cancer Chemother Pharmacol 1996;38:42–47." x" Q5 G! }$ B

: L# k5 i& t7 k8 w6 RKmiecik TE, Keller JR, Rosen E et al. Hepatocyte growth factor is a synergistic factor for the growth of hematopoietic progenitor cells. Blood 1992;80:2454–2457.3 ]- A/ E8 H& Z' {# s

7 f# F, ?+ p: B# sNishino T, Hisha H, Nishino N et al. Hepatocyte growth factor as a hematopoietic regulator. Blood 1995;85:3093–3100.& t& V% c0 @9 d
- U4 b( g. L3 Y: J" g) P
Chesnoy S, Lee PY, Huang L. Intradermal injection of transforming growth factor ? gene enhances wound healing in genetically diabetic mice. Pharm Res 2003;20:345–350.. t0 A/ B5 g7 H9 x
0 u, F5 {4 y% a' k! m; v: A
Beilmann M, Odenthal M, Jung W et al. Neoexpression of the cmet/hepatocyte growth factor-scatter factor receptor gene in activated monocytes. Blood 1997;90:4450–4458.
# `' |& y2 n/ m* k
. V% n9 W: i8 h% I9 k6 G1 ]Cowin AJ, Kallincos N, Hatzirodos N et al. Hepatocyte growth factor and macrophage-stimulating protein are upregulated during excisional wound repair in rats. Cell Tissue Res 2001;306:239–250.
1 S) |5 X% ^/ k. Y
$ I! `' D2 |4 d/ V4 w( NWilson SE, Chen L, Mohan RR et al. Expression of HGF, KGF, EGF and receptor messenger RNAs following corneal epithelial wounding. Exp Eye Res 1999;68:377–397.1 [5 R7 n1 o. H
, n0 G4 E8 t8 I$ S& u( w# a* [
Yoshida S, Yamaguchi Y, Itami S et al. Neutralization of hepatocyte growth factor leads to retarded cutaneous wound healing associated with decreased neovascularization and granulation tissue formation. J Invest Dermatol 2003;120:335–343.; ^* a3 ^/ H+ W! Y1 U( A0 g; t
! T! [1 ]7 C) v, h  T
Pediaditakis P, Lopez-Talavera JC, Petersen B et al. The processing and utilization of hepatocyte growth factor/scatter factor following partial hepatectomy in the rat. Hepatology 2001;34:688–693.
3 s9 o9 m9 H7 k$ L0 c, X1 k/ d/ D5 ^, u6 R- V7 d
Soeki T, Tamura Y, Shinohara H et al. Serum hepatocyte growth factor predicts ventricular remodeling following myocardial infarction. Circ J 2002;66:1003–1007.
% O1 Z+ f* g/ ?- j8 n
/ ?4 d. x* A9 U' Q+ ?Claxton MJ, Armstrong DG, Boulton AJ. Healing the diabetic wound and keeping it healed: modalities for the early 21st century. Curr Diab Rep 2002;2:510–518.
1 N5 m) B' B) V3 @. Y$ W2 D+ s( {) f% p3 P
Chen X, Li Y, Wang L et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology 2002;22:275–279.9 O1 I  a" u/ R6 ]( l( d+ n1 x
6 e' ?% \+ e/ J# ~
Chen X, Katakowski M, Li Y et al. Human bone marrow stromal cell cultures conditioned by traumatic brain tissue extracts: growth factor production. J Neurosci Res 2002;69:687–691.; P& ]$ U( ]+ k6 \* n& G

2 \8 I. B7 \, s9 I  H) H0 {) YBirchmeier C, Gherardi E. Developmental roles of HGF/SF and its receptor, the c-met tyrosine kinase. Trends Cell Biol 1998;8:404–410.3 W3 I2 i. ]8 S0 h' b1 p9 V9 k0 A
9 W; d7 l7 J5 y  c3 O* Z! h
Jeffers M, Rong S, Vande Woude GF. Hepatocyte growth factor/scatter factor-met signaling in tumorigenicity and invasion/metastasis. J Mol Med 1996;74:505–513.(Sabine Neussa,b,*, Eva Be)

Rank: 2

积分
107 
威望
107  
包包
1889  
沙发
发表于 2015-6-15 08:35 |只看该作者
写得好啊  

Rank: 2

积分
101 
威望
101  
包包
1951  
藤椅
发表于 2015-6-15 13:18 |只看该作者
干细胞治疗  

Rank: 2

积分
84 
威望
84  
包包
1877  
板凳
发表于 2015-7-31 19:18 |只看该作者
干细胞之家微信公众号
支持~~  

Rank: 2

积分
129 
威望
129  
包包
1788  
报纸
发表于 2015-8-17 07:02 |只看该作者
长时间没来看了 ~~  

Rank: 2

积分
98 
威望
98  
包包
2211  
地板
发表于 2015-9-10 15:54 |只看该作者
抢座位来了  

Rank: 2

积分
68 
威望
68  
包包
1752  
7
发表于 2015-9-12 02:01 |只看该作者
风物长宜放眼量  

Rank: 2

积分
79 
威望
79  
包包
1769  
8
发表于 2015-9-12 20:39 |只看该作者
支持你一下下。。  

Rank: 2

积分
136 
威望
136  
包包
1877  
9
发表于 2015-9-14 07:43 |只看该作者
回个帖子支持一下!

Rank: 2

积分
77 
威望
77  
包包
1730  
10
发表于 2015-9-29 11:54 |只看该作者
内皮祖细胞
‹ 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 07:59

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.