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: 454238|回复: 233
go

Murine but Not Human Mesenchymal Stem Cells Generate Osteosarcoma-Like Lesions i [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:54 |只看该作者 |倒序浏览 |打印
作者:Susana Aguilara,b, Emma Nyec,d, Jerry Chane, Michael Loebingera, Bradley Spencer-Denec,d, Nick Fiske, Gordon Stampc,d, Dominique Bonnetb, Sam M. Janesa作者单位:aCentre for Respiratory Research, Rayne Institute, University College London, London, United Kingdom; * a" n; d; r5 a% J1 i4 T" p0 K
                  
; J) \. @/ v3 O2 w' h' z! k' X                  
/ i; k  h0 L7 B/ ^% N' F; d          2 ~$ x3 o  X/ D* x$ c4 }- j2 ~
                        
( T, ~$ R0 ?8 B! @% ]& P4 o6 ]            
* t7 W1 p. j" ]* j            
* }- M- W5 k, ^              ~! V- Y6 \" X; X% [* n# u
            
# L! V- c% u. _! K4 L! `; Q                     
* W  H: }+ i, w) W2 N% S% @        ! I9 A* u1 ]7 U/ J# x% W& l9 f  C
        
- Q9 r/ H' B- t8 z6 {        
: V7 M6 W) y7 |% i          【摘要】; p, @% Z( u: M$ }
      Murine mesenchymal stem cells are capable of differentiation into multiple cell types both in vitro and in vivo and may be good candidates to use as cell therapy for diseased or damaged organs. We have previously reported a method of enriching a population of murine MSCs that demonstrated a diverse differentiation potential both in vitro and in vivo. In this study, we show that this enriched population of murine mesenchymal stem cells embolize within lung capillaries following systemic injection and then rapidly expand within, and invade into, the lung parenchyma, forming tumor nodules. These lesions rarely contain cells bearing the immunohistochemical characteristics of lung epithelium, but they do show the characteristics of immature bone and cartilage that resembles exuberant fracture callus or well-differentiated osteosarcoma. Our findings indicate that murine mesenchymal stem cells can behave in a manner similar to tumor cells, with dysregulated growth and aberrant differentiation within the alveolar microenvironment after four passages. We demonstrate that unlike human MSCs, MSCs from different mouse strains can acquire chromosomal abnormalities after only a few in vitro passages. Moreover, other parameters, such as mouse strain used, might also play a role in the induction of these tumors. These findings might be clinically relevant for future stem cell therapy studies., p3 A9 Y' ?& n6 A) B0 k* J

$ y7 O3 w( X1 k/ r) T: @* ?: B1 m5 E0 gDisclosure of potential conflicts of interest is found at the end of this article. 5 }$ U* D9 r/ K' r1 `
          【关键词】 Tumor Mesenchymal stem cell Lung Cell therapy Osteosarcoma
9 `' ?# s: ]1 S) g8 u2 Y+ @1 ]; i                  INTRODUCTION9 N+ u0 B, c( n: k; Z' u

9 V: [" {) H7 G* k% d, s3 D, ?3 `! GMSCs (also known as marrow stromal cells) are bone marrow-derived stem cells with the potential to differentiate into bone, cartilage, and fat .
0 V9 F% U; v2 n+ a0 _; c6 I$ c5 n
In vivo experiments have confirmed expectations of traditional MSC differentiation by showing repair of bone and cartilage after local injection using injury models .
. n4 x2 x: U* y: w+ j/ l2 Z: [  `6 E" A" d2 p8 m/ h  }" k2 \
After venous injection, bone marrow-derived cells (BMDC) must pass through the lungs. Crucial to the regeneration of lung epithelium or replacement of genes in genetic disorders, BMDC must engraft and differentiate into an epithelial phenotype. Previous studies suggest higher engraftment of BMDC into the lungs compared with other organs  was seen only after a pulmonary insult but not in undamaged controls.
4 ^6 n/ ^8 R: l, y, C+ s5 V
4 o% O# C2 R$ Z) h- f) PMurine MSC transplantation has led to both lung engraftment as type 2 pneumocytes and attenuation of lung damage . These cells were transduced with a lentiviral construct carrying eGFP, allowing the tracking of engrafted MSC progeny. Although we saw some evidence for differentiation of these cells into multiple cell types, including airway epithelial cells, the mice became short of breath and had to be sacrificed after 28 days. This study examines in detail the kinetics of MSC engraftment in the lung parenchyma and their subsequent differentiation potential./ T9 e8 n' Q- @0 m& o& B! [/ h$ ?0 b
) f& ?) f6 o% K
We found that after systemic delivery of MSCs, cells embolized in lung parenchyma capillaries. These cells both transmigrated and divided over the next 24 hours. However, far from showing the expected local differentiation pattern, the majority of cells expanded to form tumors of immature disorganized bone resembling well-differentiated osteosarcoma or exaggerated callus. However, there were no definitive features of neoplastic transformation, such as atypical nuclei, tumor necrosis, or areas of undifferentiated sarcoma. The lesions expanded rapidly, destroying the lung parenchyma, and in several cases led to recipient death by 28 days. Importantly, we found that the formation of these tumors appears specific to murine MSCs. The same type of experiments using MSCs derived from human fetal blood underwent similar initial engraftment but subsequent clearance from the lungs. The precise mechanism of the tumor development is not clear; however, we demonstrated that murine MSCs acquired significant chromosomal abnormalities after four in vitro passages, whereas we did not detect any abnormalities in human MSCs after six or more passages in vitro. Our results show that an enriched population of murine MSCs can be expanded in vitro for cell therapy studies, but the cells are chromosomally unstable, and this may contribute, in part, to tumor formation after systemic injection.
, o1 w0 h0 y0 {& f* r  w/ A% `6 m
MATERIALS AND METHODS2 M4 x1 O4 z$ x3 M. Z

& s5 K4 E1 \4 @; T& jMurine MSC Isolation, Purification, and Expansion) ^9 H, }) ?1 E$ _

/ D3 }4 z$ e; c/ p+ vMSCs from NOD/SCID bone marrow were used for initial experiments because of the ease of culturing MSCs without the presence of contaminating hematopoietic cells in this mouse strain  F1) mice. Bone marrow cells were collected by flushing the femurs, tibias, and iliac crests from 8¨C12-week-old mice with phosphate-buffered saline (PBS) supplemented with 2% fetal bovine serum (FBS) (Gibco, Paisley, U.K., http://www.invitrogen.com). Red blood cell-depleted bone marrow mononuclear cells were plated at a density of 106 cells per cm2 in murine mesenchymal medium with murine mesenchymal supplements (Stem Cell Technologies, Vancouver, BC, Canada, http://www.stemcell.com), supplemented with 100 IU/ml penicillin and 100 µg/ml streptomycin (Gibco). Half the culture medium was changed at day 3 to remove some nonadherent cells. Whole medium was subsequently replaced weekly. The cells were grown for 2¨C3 weeks until almost confluent. Adherent cells were then detached by 0.25% trypsin-EDTA and replated using a 1:3 dilution factor. Subsequent passaging and seeding of the cells was performed at a density of 5,000 cells per cm2. MSCs were enriched at passages 2 and 3 by elimination of cells stained with rat anti-mouse CD45-CyChrome and CD11b-PE (BD Biosciences, Oxford, U.K., http://www.bdbiosciences.com). The negative fraction (35%¨C40% of adherent cells) was sorted using the FACSVantage (Becton, Dickinson and Company, Oxford, U.K., http://www.bd.com) and expanded before transduction and injection as described below. At injection, MSCs were resuspended in PBS, 2% fetal calf serum with 1 mM EDTA and filtered to avoid cell aggregates./ x+ Q& C3 e( o( h2 ]) `

3 `9 L( Y' E! X) w- ~0 OHuman Adult and Fetal MSC Sample Collection1 w8 m" i' y2 u
: Q( K' |9 Z: d1 J! n9 @
Human adult bone marrow cells were purchased from Stem Cell Technologies. Fetal blood collection was approved by the Research Ethics Committee (Hammersmith and Queen Charlotte's Hospitals) in compliance with national guidelines regarding the use of fetal tissue for research purposes (Polkinghorne Commission Recommendations). All women gave written informed consent for collection and use of human tissues. Murine experiments were carried out under full ethical approval from the host institutions and the appropriate Home Office license.
& y  {3 H: E* }
" f1 X' o& {- W  G4 [: VFetal blood (100 µl) was obtained by ultrasound-guided cardiac aspiration (10 weeks of gestation) before a clinically indicated termination of pregnancy. Fetal gestational age was determined by crown-rump length measurement on ultrasound.
: N  k' s0 Z$ `7 J9 h; I' e' {) ^) o4 }2 T
Culture of Adult and Fetal Blood MSCs
# M+ A- Y7 y8 g9 f. w, l! }$ L1 X; c5 S3 n
Fetal blood was plated in 100-mm dishes at 105 nucleated cells per milliliter, and bone marrow mononuclear cells were plated at a density of 106 cells per cm2. Both were cultured in MSC growth medium consisting of 10% FBS (Stem Cell Technologies) in Dulbecco's modified Eagle's medium (Sigma-Aldrich, Poole, Dorset, U.K., http://www.sigmaaldrich.com) supplemented with 2 mM L-glutamine, 50 IU/ml penicillin, 50 mg/ml streptomycin (Gibco-BRL, Gaithersburg, MD, http://www.gibcobrl.com) at 37¡ãC in 5% CO2. After 3 days, nonadherent cells were removed, and the medium was replaced. After 10 days, adherent cells were then detached by 0.25% trypsin-EDTA (Stem Cell Technologies) and replated at a density of 5,000 cells per cm2, expanded, and cultured to confluence in 75-cm2 flasks. Adult BM-derived MSCs and fetal blood-derived MSCs underwent 4 and 5 passages, respectively, before being transduced.
+ \) V  g- ?- t% t$ @6 G
6 w/ r# N& t+ p" }" HIn Vitro Lentivirus-Mediated Gene (eGFP) Transfer into Murine and Human MSCs' R4 c4 H1 x' X- ~6 o. u' E* O" W

* l6 L" o& M( J9 W. D6 s6 [% @1 lThe HIV-1-based self-inactivating lentiviral vector (pHRSINcPPT-SEW), carrying the eGFP reporter gene under the control of the spleen focus-forming virus long terminal repeat, was used to transduce murine and human adult MSCs. For transduction, 1 x 104 purified MSCs from passage 4 were seeded into individual wells of a 12-well plate. The following day, virus particles were added at a multiplicity of infection (MOI) of 50 to murine MSCs and at an MOI of 10 to human adult MSCs, and transductions were performed for 20 hours./ e2 z8 K5 w" H# o+ L" J
4 a8 D8 @! J' }9 I4 z. l% A
Fetal MSCs were transduced with a lentivirus encoding the eGFP reporter gene driven by hPGK at an MOI of 12.5 as previously described .0 K! Y) h# S: M2 ?' v

- i6 |' L- D& |3 }' l- OTransduced MSCs were washed several times after virus removal with culture medium to avoid viral contamination before infusion into mice. Flow cytometry assessment of the transduced cells at day 5 showed expression of eGFP expression >95% for murine MSCs .) S5 V3 r# o; X4 e
/ C. o+ f7 |; r+ r. H5 ^1 x+ Y1 s
Adoptive Transfers' B6 X' R* z  M; V
' X8 m, L& G1 d: H+ d$ E0 j$ s; C
A total of 39 NOD/SCID mice ages 8¨C12 weeks old received donor NOD/SCID MSCs in four independent in vivo studies. eGFP-MSCs (2 x 106), obtained 4 days post-virus removal to minimize further expansion, were delivered intravenously by tail vein injection into each sublethally irradiated mouse (375 cGy using a 137Cs source). Pairs of mice were then sacrificed on days 1, 2, 7, 14, and 28 postinfusion, and lungs were collected in three experiments. In the fourth experiment, nine NOD/SCID mice were used; five of them were injected with eGFP-MSCs (2 x 106), and four were injected with nontransduced MSCs (2 x 106). The fourth experiment also included nine NOD/SCID mice that were injected with MSCs from Rosa26-LacZ mice (0.35 x 106 and 2 x 106 cells) after sublethal irradiation (375 cGy). MSCs from Rosa26-LacZ mice were also injected (0.5 x 106 and 2 x 106 cells) into syngenic mice after sublethal irradiation of 500 cGy or no irradiation. For the human fetal MSCs, a total of 16 NOD/SCID mice were injected with 2 x 106 cells after sublethal irradiation (375 cGy). Human adult MSCs were injected (1 x 106 and 2 x 106 cells) into 12 NOD/SCID mice after sublethal irradiation of 375 cGy and into 5 NOD/SCID mice with no irradiation.
& S3 ]% i$ h. ~9 Q$ {' k8 u# y" S( ?
Tissue Processing and Immunohistochemistry
! \  ~' _% {, |  ]" H# Z& I% S) D/ ?8 M7 f, X3 g- n
Tissues were fixed in 10% neutral buffered formalin (NBF) and embedded in paraffin. Sections (4 µm thick) were stained with H&E or Alizarin Red or immunostained using the avidin-biotin-peroxidase or alkaline phosphatase technique.' e4 C5 s7 E: r2 N' ]! Y

) D& i: p0 ]7 D5 g5 T6 j3 {For all the mice used in the fourth experiment, lungs were inflated with 1% paraformaldehyde, fixed in NBF, and embedded in paraffin. For human cells and the MSCs from Rosa-LacZ mice, the left main bronchus was ligated after lung inflation, and the left lung was excised and embedded in optimal tissue compound for frozen sectioning.
8 K! o: G9 C; ~6 U* x; e" t7 `4 X4 G# n. c" y% ~
The sections were immunostained with anti-eGFP antibody (rabbit polyclonal; 1:500; Molecular Probes Inc., Leiden, The Netherlands, http://probes.invitrogen.com), anti-Ki67 (rat monoclonal; 1:25; DAKO, Cambridgeshire, U.K., http://www.dako.com), anti-AE1/AE3 (mouse monoclonal; 1:100; DAKO), anti-endomucin (V.7C7 rat IgG2a polyclonal; 1:500; generously provided by Dr. Dieter Vestweber, Institute of Cell Biology, University of Munster, Munster, Germany), anti-TTF-1 (mouse monoclonal; 1:50; Novocastra Ltd., Newcastle upon Tyne, U.K., http://www.novocastra.co.uk), anti-osteocalcin (goat polyclonal; 1:100; Santa Cruz Biotechnology Inc., Heidelberg, Germany, http://www.scbt.com), and anti-collagen type II (mouse monoclonal; 1:25; Biocarta, San Diego, http://www.biocarta.com)." W4 c# y8 D5 u
- x# q" Y% V  y, z. b8 F( b
The endomucin antibody required no antigen retrieval. For Ki67, eGFP, TTF-1, and osteocalcin, sections required microwave antigen retrieval (sodium citrate, pH 6, for 10 minutes), and sections were quenched for endogenous peroxidase (with 1.6% H2O2) and when necessary for endogenous alkaline phosphatase (2 mM levamisole). For AE1/AE3, sections were treated with protease (Streptomyces griseus) antigen retrieval (0.04% in PBS at 37¡ãC for 10 minutes; Sigma-Aldrich), and for osteocalcin, sections were treated with pepsin antigen retrieval (1 mg/ml in 50 mM Tris-HCl, pH 2, for 15 minutes). Biotinylated secondary antibodies (1:250) were used, and immunoreactivity was detected using the ABC peroxidase-based system in combination with 3,3'-diaminobenzidine, the Vector Blue alkaline phosphatase-based system, or a combination of both (Vector Laboratories, Burlingame, CA, http://www.vectorlabs.com), following the manufacturer's protocol. Absent and/or nonspecific primary negative controls were included. For secondary antibodies conjugated with fluorochromes, the sections were incubated with Sudan Black (0.1% in 70% ethanol for 30 minutes) to lower autofluorescence.$ f; |) ~8 }5 }. o
: f( F: h9 s# e+ s  n7 X7 X
For the Alizarin Red S stain for calcium, sections were dewaxed and taken to 95% alcohol. The sections were then air-dried before being placed in Alizarin solution for 1¨C5 minutes (1% aqueous red S , pH 6.3) until the desired intensity was seen, and they were then counterstained with methyl green (Vector Laboratories) for 2 minutes, rinsed in water, blotted, and then rinsed in acetone for 30 seconds.2 l3 P& P0 C. D, h( N
6 j8 y& v0 s, x. D: T
All microscopy was performed on a Nikon Eclipse E1000 microscope, and all images were captured using a Nikon DXM1200F digital camera (Tokyo, http://www.nikon.com).
% V9 }$ c+ d; _$ ^3 N9 V3 z" N
) f2 K+ W) L, @% N2 K9 rKaryotype Analysis
9 m: L; H; w2 @% c1 ^8 y
1 F' Y1 Z% V2 [7 @4 @Metaphase analysis was performed by using 80% confluent flasks of MSCs that were subsequently incubated with 0.1 µg/ml colcemid (Roche Diagnostics, Basel, Switzerland, http://www.roche-applied-science.com) overnight for passage 1 and for 5 hours for later passages and human fetal MSCs. Mitotic cells were washed in PBS and trypsinized, and the cells were incubated in hypotonic solution (75 mM KCl) for 15 minutes at 37¡ãC and fixed in methanol/acetic acid (3:1). Fixed chromosomes spreads were stained with 200 ng/ml 4,6-diamidino-2-phenylindole in 2x standard saline citrate and examined by immunofluorescence microscope. Forty metaphases of each passage and cell type were counted.
/ v. P7 D. k+ |# c& ?$ {$ s; E8 |4 s! `9 Z! ^; l, r# U
RESULTS  M6 C0 `6 \9 M4 Z4 G6 c) d

5 O. E! G* U* L1 x4 r0 T3 n) h9 _Isolation and Characterization of Murine MSCs
* @) B6 E. w( ^/ t; N8 ^* j; B: n* u; P5 L# ~! w% L/ T
The murine MSCs used in these experiments have been extensively characterized, as previously described ).
; Q+ h) D1 R! {6 V8 u1 F3 L/ i2 x  ?, {' c
MSC Engraftment Occurs Immediately Post-Transplantation by Embolization
& e. f! v/ ~7 m  `
' R. u+ Y: N% \) N& b; zWe performed systemic injections of NOD/SCID-derived MSCs into sublethally irradiated NOD/SCID mice. Two mice per time point were killed on days 1, 2, 7, 14, and 28 in three separate experiments. A fourth experiment consisted of nine mice, all sacrificed at day 28. Only two mice (both killed on day 2, from the second experiment) showed no eGFP-positive cell engraftment. All other mice demonstrated engraftment. On day 1 (six mice), engraftment of eGFP-positive cells was between 0.45% and 3.2% of the total cells counted per section (Fig. 1A, 1B). Histopathological examination showed that murine MSCs embolize in pulmonary capillaries. Figure 1C shows an ectatic capillary with embolized MSCs. After impaction, MSCs transmigrate into air spaces (Fig. 1E), where they proliferate or cluster to form aggregates (Fig. 1F).
) D8 i  w6 u* L0 G: P# z+ c
4 K) _% `: D. ~$ |) o1 H* mFigure 1. NOD/SCID enhanced green fluorescent protein (eGFP)-tagged MSCs embolize into NOD/SCID mice lung parenchyma and migrate into air spaces after 24 hours. (A, B): Low-power (x10) views of MSCs engrafted into lung parenchyma (, anti-eGFP). (C¨CF): Ectatic capillaries with embolized MSCs (solid arrows) (anti-eGFP; x100). (E): An MSC transmigrating into the alveolar air space (x100). (F): A cluster of MSCs within an alveolus (x100).6 z  ?- C# |! A
  m4 e# U% l7 E
MSC Plasticity Is Rare in the Lung Despite Expansion of Engrafted Donor-Derived Cells0 D# s& H! U( x1 j) o* s

  K1 j  C, l% R" ?We and others have previously reported that bone marrow-derived cells have the ability to differentiate into lung epithelial cells . However, the majority of eGFP-positive tissue derived from donor MSCs did not costain with either a lung epithelial cell nuclear marker (TTF-1) or an epithelial cell membrane marker (AE1/AE3) (Fig. 2E, 2F). Indeed, the lung epithelial architecture within the eGFP-positive tissue was grossly distorted by proliferating eGFP-positive cells.) ~1 ~( J! x0 L  r  S0 V
& y) N! o4 ?5 `; z/ y
Figure 2. NOD/SCID-derived MSCs at 14 days post-transplantation showing rare enhanced green fluorescent protein (eGFP)-positive cells costaining with epithelial markers in the lung. (A¨CD): Costaining of anti-eGFP (A), anti-TTF-1 (B), and 4,6-diamidino-2-phenylindole (DAPI) (C); (D): Merged image with double-stained cell marked with a white arrowhead. (E, F): Shown is a large lesion containing eGFP-positive tissue but no costaining with epithelial markers. (E): Green, anti-TTF-1; red, anti-eGFP; blue, DAPI. (F): Green, anti-AE1/AE3; red, anti-eGFP; blue, DAPI. Magnification, x100 (A¨CD), x20 (E), x40 (F).  ?2 M$ T! r8 z% |9 x. K$ B' |

( v4 L4 |  A) B/ I6 ^0 @Donor-Derived Murine MSCs Form Tumors Within the Lung Consisting of Bone and Cartilage
  J% d8 c- c* h8 n- w9 ?* e
- [- n' ?* d, N6 G$ |7 CPostengraftment, donor-derived NOD/SCID MSCs rapidly expanded, forming large areas of tissue within the lungs of NOD/SCID mice by day 7 (Fig. 3A, 3B). The majority of this tissue had the morphological appearance of cartilage or bone, and by day 14, areas of lung stained clearly for cartilage-specific marker collagen 2 (Fig. 3C) and bone markers Alizarin Red (Fig. 3F) and osteocalcin (Fig. 3G, 3H). Large areas had the characteristic collagen deposition of bone on polarized light microscopy (Fig. 3I, 3J). The tumors continued to expand obliterating normal lung tissue until between 40% and 60% of the lung parenchyma contained eGFP-positive cells in 6 of 12 mice sacrificed at 28 days (six of six mice from the first three experiments). The other nine mice (five transduced with lentivirus-eGFP and four untransduced) had multiple nodules, but they were of smaller size, occupying
/ U! e, w( G7 I
& K! e' i! K, R7 iFigure 3. Postengraftment, the donor-derived NOD/SCID MSCs rapidly expand, forming large areas of tissue within the lungs of NOD/SCID mice by day 7 (, polarized light; magnification, x10). These tumors continued to expand, obliterating normal lung tissue until between 40% and 60% of the lung tissue was formed of eGFP-positive cells by day 28 and the mice were sacrificed because of breathlessness. Detailed histopathological examination revealed tumors resembling exuberant fracture callus or well-differentiated osteosarcoma. Magnification, x40 (K), x10 (L).' H6 [1 o: L" W2 z" A& u
( m, q( u- N4 H3 |% z+ F
Although other organs were analyzed in the first three experiments, no tumor formation was detected in kidney, liver, or heart (data not shown).
" O/ h+ W# F, c/ J$ I0 d' n
5 t1 @+ p9 C" a* |Murine MSCs Grew Within Vessel Lumen, Infiltrating and Tracking Along Vessel Walls, but Did Not Costain with Endothelial Markers! ]5 N1 A7 x2 r" @1 j$ Y

+ p; P. [$ S* d8 _& f8 m6 xReports of MSC differentiation into endothelial cells led us to search for donor-derived cells that costained with endothelial markers. MSCs were frequently seen to expand within vessels differentiating into bone or cartilage (Fig. 4A). eGFP-positive cells also invaded the wall of the vessel in an apparently organized manner (Fig. 4B, 4C) but did not costain for the endothelial markers CD31 or endomucin. Within the vessel wall, cells positive for endothelial markers were not donor-derived (Fig. 4C). eGFP-positive cells "pagetoid spread" along the vascular basement membrane, undermining and displacing the endothelium (Fig. 4E, 4F), but again, these donor-derived cells did not stain for endothelial markers.' ^! K5 j) M8 {3 y* z; ]1 z) Z0 D

# O* W7 j6 }; a6 g! |1 x4 aFigure 4. NOD/SCID MSCs formed tumors within vessels that infiltrated vessel walls. (A): Histopathological analysis shows cartilage tissue growing within a vessel at 14 days (magnification, x10). (B, C): Shown are enhanced green fluorescent protein (eGFP)-positive cells infiltrating the vessel wall but not expressing the endothelial marker endomucin (). Magnification, x40 (D), x100 (E, F). Engrafted MSCs were highly proliferative from day 1 postdelivery. (G): MSCs on day 1 postinjection. (H): a day 14 lesion. Both slides (G, H) costained with anti-eGFP (blue) and anti-Ki67 (brown). Magnification, x100 (G), x20 (H).- |1 O- N/ s% C5 @! d1 ]) H
& j4 M1 }2 B$ Q+ L& E
Murine MSCs Rapidly Proliferated Following Systemic Injection( g( N) P2 w) S

# v8 c2 z* Y; Y& hLung epithelium divides irregularly in the steady state, with rare cells staining positively for proliferative markers. Using immunohistochemistry for Ki67, a marker of cellular proliferation, we determined that injected MSCs were proliferating following embolization within lung parenchymal capillaries within 24 hours (Fig. 4G) and continued to proliferate within the lung, explaining the rapid growth of the lesions formed (Fig. 4H).
) X0 o! d+ E  {; L. W$ ^& ~+ K/ ]) U8 d" ?- Z$ o
Lentiviral Transduction of Donor Cells Does Not Cause the Tumor Formation# s7 k5 c  t5 D

4 I$ X  _4 ]( H- t, a# L" W  W* bThe cells injected in the first three experiments had been transduced with a lentivirus expressing eGFP, allowing easy identification of donor cells. To examine whether the lentiviral transduction led to the tumor formation, we performed a fourth experiment, in which we injected four NOD/SCID mice with nontransduced NOD/SCID cells and five NOD/SCID mice with cells transduced with the lentivirus. All nine mice developed equivalent tumors (Fig. 5A¨C5C). As noted above, the tumors were smaller than those seen in the first three experiments but had the same morphological and differentiation characteristics, indicating that the formation of tumor was not related to virus integration.6 o4 W. h" o0 o

- q' g3 W: E1 ]# J6 z  E9 Q* EFigure 5. Tumor formation analysis of mouse and human MSCs. MSCs from NOD/SCID mice form tumors with or without lentiviral transduction. Magnification, x20 (A¨CC). (A): Enhanced green fluorescent protein (eGFP)-positive donor cells in the tumors formed after injection of NOD/SCID MSCs that were transduced with lentivirus-eGFP; (B): Serial section stained with an isotype control. MSCs from NOD/SCID mice without lentiviral transduction were also capable of forming tumors at the same level and, as expected, were negative for eGFP (C). (D): Shown is tumor formed after LacZ MSCs were injected into NOD/SCID mice (arrow; magnification, x40; H&E), whereas no tumors were found in the other eight mice analyzed (; magnification, x10: H&E).+ a: D! |- F( _0 h

* e8 ]5 U0 [6 K; i  M% s& [Murine MSCs from a Nonimmunocompromised Mouse Strain Show Less Tumor Formation Following Systemic Administration
' x+ e8 L- `' S' n* a; i
( W% M5 q, k4 r4 f; ^$ `To examine whether the formation of tumors only occurred in immunodeficient mice, we repeated our experiments in immunocompetent Bl6/129 mice. Donor cells from Rosa26-LacZ (Bl6/129) mice were cultured under the same conditions and infused into 13 sublethally irradiated Bl6/129 mice using two different cell doses (eight mice with 2 x 106 cells; five mice with 0.35 x 106 cells). The mice were sacrificed 28 days after infusion. The lungs were sectioned at different levels separated by 50 µm. Between 8 and 12 H&E sections (through at least two lobes each) per mouse were examined for evidence of tumor formation. None of the 13 mice analyzed had tumors (Table 1), suggesting that tumor formation may be dependent on mouse strain. To examine whether the absence of tumors in the Bl6/129 mice can be explained by recipient strain differences, we performed the same experiments injecting Rosa26-LacZ-derived donor cells into sublethally irradiated NOD/SCID recipient mice. Histopathological analysis showed a single tumor in one mouse (Fig. 5D; Table 1) but no tumors in the other eight mice (Fig. 5E; Table 1). This low frequency of tumor formation suggests that strain differences in the donor cells are important.; s1 E: U$ M  ^4 m  J8 I
( }& y1 r) T/ y) h/ E, [/ O
Table 1. Summary of tumor nodule formation
7 m8 g3 U+ J! {0 X5 e# c7 ?) N- x
Lentivirally Transduced Human Adult and Fetal MSCs Did Not Form Tumors
" i! o9 i: Z+ D, C9 |. u2 y9 d" F2 @
We next wished to determine whether human MSCs cultured in similar circumstances formed tumors after transplantation into mice. We used both human adult and fetal blood MSCs (passages 6 and 7, respectively). Human fetal MSCs have been extensively characterized by us previously and demonstrated in vitro differentiation into adipocytes, chondrocytes, and osteocytes . NOD/SCID mice were infused intravenously with 2 million fetal MSCs or 1 or 2 million adult MSCs after identical sublethal irradiation of 375 cGy to the murine experiments. For fetal MSC experiments, two mice were killed on day 1, two on day 14, two on day 21, six on day 28, and four at 8 weeks. No tumors were detected at any time point (Fig. 5F; Table 2). Indeed, the number of cells detected with anti-green fluorescent protein (GFP) staining reduced from approximately 1 in 350 at day 1 to
0 P5 L3 h8 j' |3 l# X4 K$ t2 C7 S8 i) x8 C% C( M
Table 2. Summary of lung engraftment and tumor formation in bone marrow transplantation of human fetal enhanced green fluorescent protein-transduced MSCs by histology% s0 a" |" ]* `8 J4 e% ~
% ]  r: R" A' U9 ?9 ?6 x6 I' x- K5 }
Table 3. Summary of lung engraftment and tumor formation in bone marrow transplantation of human adult enhanced green fluorescent protein-transduced MSCs by histology8 h2 t. `4 `! i% e% y( s# ]
& q, C) g% _0 f! K
Murine MSCs Have an Abnormal Karyotype, but Human MSCs Do Not" [2 R" C. ~7 }& E% k& g
+ I( H; u/ e6 _* [+ E9 G: x
Chromosomal abnormalities are thought to be critical in tumor formation, and recent studies have identified abnormalities in murine MSCs after high passage numbers . To check whether the donor MSCs used in our study had normal chromosomes, we performed a karyotype analysis on all the cell types injected, counting at least 40 metaphases per cell type and passage. We found that in both NOD/SCID- and Rosa26-LacZ (Bl6/129)-derived MSCs, the majority of metaphases had the expected number of chromosomes (n = 40) at passage 1 (data not shown). Unexpectedly, at the time of injection (passage 4 or 5), both strains of MSCs showed numerical chromosomal abnormalities, with a modal chromosome number of 69 for NOD/SCID-derived MSCs and 76 for LacZ-derived cells (Fig. 6A, 6B). These changes in cell karyotype may be responsible, in part, for the tumor formation observed. However, almost all the human fetal MSCs (35 of 40 cells) showed the normal number of chromosomes (n = 46) at passage 7 (Fig. 6A, 6B).
2 Q/ p0 m; K( V5 ~# {8 a- e; t( V% u7 r+ S5 R! u& k7 N, S, t, U
Figure 6. Murine MSCs but not human MSCs show abnormal karyotype. The analysis of 40 metaphases of murine and human MSCs at the time of injection (P4, P5, or P7) showed numerical chromosomal abnormalities in both NOD/SCID and LacZ MSCs but not in the human MSCs (; magnification, x100; 4,6-diamidino-2-phenylindole staining). Although the majority of the human cells had an expected number of chromosomes (n = 46), both strains of murine MSCs showed changes in the number of chromosomes having the majority of metaphases within the chromosome range 41¨C79 (B). Abbreviation: P, passage.
) y! q9 D. E8 C% o9 b( o, q4 I# C0 g, h9 f7 t$ l' v1 U" i+ q7 w
DISCUSSION
- O3 E0 W; H# |4 Y, @' P+ Z& ?; @# ]6 F* @7 M+ a
We systemically injected an enriched population of murine MSCs into mice following low-dose irradiation. The aim of our study was to evaluate the extent of lung parenchyma engraftment as epithelial cells under these conditions. Although we saw rare evidence of differentiation of MSCs into cells with staining characteristics of lung epithelium, we saw widespread engraftment and expansion of MSCs into large tumors, showing differentiation into bone and cartilage cells with destruction of normal lung architecture.) d5 o! P5 w/ |2 [- d/ @3 y

$ \( |1 B! x' n4 p! ^# WWe have demonstrated that not only do murine MSCs cultured under our conditions have high in vitro capacity for osteogenic and chondrogenic differentiation .8 I" O5 P% s+ M9 T6 D/ p
& Y" }/ X( E% K7 i6 J0 V( b
Additional experiments using a different mouse strain, Bl6/129, revealed no tumor formation (0 of 13 mice analyzed). Hence, there does appear to be some strain dependence. To establish whether this was due to the genetic background of the recipient, we administered Rosa26-LacZ (Bl6/129)-derived MSCs into NOD/SCID mice and found that only one of nine mice displayed a small tumor. These results suggest that tumor formation is dependent on both recipient strain and the strain from which donor cells are derived. Although both donor murine strains had numerical abnormalities in karyotype, it is possible that differences in the cytogenetic events occurring between the two strains could explain the differences in the tumorigenesis capacity. This hypothesis is supported by a report that shows a variety of cytogenetic abnormalities in MSCs derived from C57/Bl6 mice, some capable of forming sarcomas in C57/Bl6 mice but others not . Moreover, another possible explanation could be differences in the expression of cell adhesion molecules that may favor the higher engraftment in the lung parenchyma of NOD/SCID-derived MSCs. Of note, all four separate cultures of NOD/SCID MSCs produced lung tumors upon injection in our experiments.$ U0 N* m3 y# o5 n/ d$ ~+ A  j) ~, I

& _. b; V: ]) m4 C; i; VMurine MSCs, although less well studied than human MSCs, have similar in vitro differentiation potential but differ in a number of other attributes, including cell surface molecule expression . This may alter cell homing and engraftment after systemic injection. To assess whether human MSCs would also form tumors in our model, we injected both human fetal and adult MSCs carrying a lentivirally expressed eGFP marker. Human cells engrafted into the lung parenchyma, but at a lower level. Importantly, we found no evidence of tumor formation after 2 months, and the levels of GFP-positive cells decreased over time, with no evidence of proliferation.
$ c2 Y' Z8 f1 y: d* G9 f; F/ {6 t2 h0 a
Few studies have examined the effects of systemic delivery of MSCs. Ortiz et al. reported an attenuation of bleomycin-induced inflammation and fibrosis after transplanting murine MSCs, resulting in engraftment of donor-derived type 2 pneumocytes, although at lower levels than those of HSC transplants , indicating that the nature of the chromosomal aberrations and/or other factors might influence the capacity of MSCs to form tumors. Importantly, the culture systems in these different studies and our own used standard culture conditions and thus are unlikely to be responsible for the tumor formation.- V$ C8 ?) ]. |8 q& t' \' H
+ k! @. p2 ?3 b1 g  j* m
Why these enriched MSCs form tumors is intriguing. We have found that our highly selected murine MSCs rapidly accrue genetic changes, as demonstrated by their abnormal karyotype. It is likely that this has led to abnormal expression of genes or genetic/epigenetic changes responsible for tumor formation. It would be interesting to examine this short-term-cultured MSC population for the genetic abnormalities frequently found in osteosarcoma tumors and cell lines. The irradiation dose we administrated in our experiments was not likely critical for the tumor formation, as previous reports showed that this sublethal dose is not harmful to the lung . Supporting these data, the transplanted human MSCs were also given a sublethal dose of irradiation and no tumors were seen.
0 d! C+ t# u! j2 o! r. f# r2 F$ h" o
Our data provide further evidence for the rapid acquisition of chromosomal abnormalities and potential adverse effects of systemic injection of murine MSCs after a minimal in vitro culture necessary to obtain a quantity of cells required for cell therapy studies. These results stress the importance of careful analysis of cells used for cell therapy both before and after transplantation.
4 J7 o1 o0 c! y  s6 g$ f( r& C2 }6 i! w; @8 `7 }
DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
. |# G" F: N- L8 z- C( V6 W1 b2 y) [; N( n' L
The authors indicate no potential conflicts of interest.
  W+ O$ q3 v+ |# Q$ B! p
: _' u( J5 s- Y; v, }  K% QACKNOWLEDGMENTS
/ X7 b' O4 j' F. p9 I+ V( \7 F' `
/ X& I# u# c8 z3 ^  vWe thank Fernando Anjos-Afonso for help with these experiments, George Elias and his team in the Cancer Research UK histopathology laboratories, Karen Groot for critical reading of the manuscript, and Denise Sheer for help with karyotyping analysis. This work was supported by Cancer Research UK. S.M.J. is a Medical Research Council Clinician Scientist. D.B. and S.M.J. are joint last authors.
0 Y; v( [6 C8 O& i) U# [          【参考文献】" N: }. `. C" `  |% ?' q% g  \

$ m8 S8 m' n. N6 s: f  f  e
. N/ ]$ {! l4 `4 \% YPittenger MF, Mackay AM, Beck SC et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143¨C147.) j- _; c. N/ }6 z( h  {
( ]/ G: K- H% _2 R+ m
Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997;276:71¨C74.: e& f3 g% N- E4 c$ B- i

9 m! v, [9 b5 b+ Z; U# qWoodbury D, Schwarz EJ, Prockop DJ et al. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res 2000;61:364¨C370.
7 _. z- a2 M0 T/ u* }  o7 a2 [) D+ @- M! J' S' P( O
Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve 1995;18:1417¨C1426.) ^, H  C3 ^. k( y( Y! r
# R' k, o& A- L: b
Anjos-Afonso F, Siapati EK, Bonnet D. In vivo contribution of murine mesenchymal stem cells into multiple cell-types under minimal damage conditions. J Cell Sci 2004;117:5655¨C5664.# @+ b/ J' t) w2 a
0 B: }" @% U/ m5 D9 _. O' M# o9 h. B
Wang G, Bunnell BA, Painter RG et al. Adult stem cells from bone marrow stroma differentiate into airway epithelial cells: Potential therapy for cystic fibrosis. Proc Natl Acad Sci U S A 2005;102:186¨C191.
3 r' p2 X' f- Z4 Z; p- e8 z1 f9 ?" f/ `+ v& q1 h9 ?- n* z  [, F/ l
Hofstetter CP, Schwarz EJ, Hess D et al. Marrow stromal cells form guiding strands in the injured spinal cord and promote recovery. Proc Natl Acad Sci U S A 2002;99:2199¨C2204.  D) u. o/ h; s

$ o8 G* P- J  d/ P: J) QKopen GC, Prockop DJ, Phinney DG. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci U S A 1999;96:10711¨C10716.
! X: v! k6 M& {; k# s) W3 }  ?9 k
Pereira RF, O'Hara MD, Laptev AV et al. Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta. Proc Natl Acad Sci U S A 1998;95:1142¨C1147.5 R- M) Q! g# }" E/ V8 V' ?
# [2 |8 Z( w) d4 G8 W
Liechty KW, MacKenzie TC, Shaaban AF et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med 2000;6:1282¨C1286.
( b4 ^) ~6 Z+ L2 C6 ~/ O1 [  l2 v5 q6 b$ B
Devine 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¨C3001.
( H' g0 }5 x: ^3 i
$ R  ]  G/ s: H7 c4 IKrause DS, Theise ND, Collector MI et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 2001;105:369¨C377.
& h' x8 e+ A  c; F; Y
9 c2 u0 K+ F, ]" ]Kotton DN, Ma BY, Cardoso WV et al. Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development 2001;128:5181¨C5188.
, L( r% G6 P) `
5 L9 |9 k. u4 }$ c; l; t0 @4 L5 o( NYamada M, Kubo H, Kobayashi S et al. Bone marrow-derived progenitor cells are important for lung repair after lipopolysaccharide-induced lung injury. J Immunol 2004;172:1266¨C1272.
: W5 u7 q+ J* O6 }! J
7 u% c1 b8 ^4 ^; ^0 }Ishizawa K, Kubo H, Yamada M et al. Bone marrow-derived cells contribute to lung regeneration after elastase-induced pulmonary emphysema. FEBS Lett 2004;556:249¨C252.: l, `& w  v! M& |
" I  V! X$ M* d" I) D! a
Ortiz LA, Gambelli F, McBride C et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A 2003;100:8407¨C8411.; J2 b6 N6 R( a
. }) Z2 p6 \& [/ E4 X! z
Hashimoto N, Jin H, Liu T et al. Bone marrow-derived progenitor cells in pulmonary fibrosis. J Clin Invest 2004;113:243¨C252.
  ~$ g# J. H9 s; V3 O. k  q# e
2 M% B( |/ F: y# OChan J, O'Donoghue K, de la Fuente J et al. Human fetal mesenchymal stem cells as vehicles for gene delivery. STEM CELLS 2005;23:93¨C102.
6 w3 {% c( @6 w& f
: r  O$ F" j2 x; R. {$ K/ n/ GPhinney DG, Kopen G, Isaacson RL et al. Plastic adherent stromal cells from the bone marrow of commonly used strains of inbred mice: Variations in yield, growth, and differentiation. J Cell Biochem 1999;72:570¨C585.+ R& k0 T7 f9 d4 `4 X

2 \" [6 k4 z( p& b1 D8 OPeister A, Mellad JA, Larson BL et al. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood 2004;103:1662¨C1668.$ v& T( [- |4 R. P9 K& ^
9 W  F5 L# T0 p/ u. k
Kotton DN, Fabian AJ, Mulligan RC. Failure of bone marrow to reconstitute lung epithelium. Am J Respir Cell Mol Biol 2005;33:328¨C334.. H: n) v9 C! p. d" \% R
$ W9 W; K: V- w* X
Chang JC, Summer R, Sun X et al. Evidence that bone marrow cells do not contribute to the alveolar epithelium. Am J Respir Cell Mol Biol 2005;33:335¨C342./ P9 N. w* H, ?2 J# j2 p
' ]" D2 g1 P! G! i# o! s
Chan J, O'Donoghue K, Gavina M et al. Galectin-1 induces skeletal muscle differentiation in human fetal mesenchymal stem cells and increases muscle regeneration. STEM CELLS 2006;24:1879¨C1891.$ ?- q, d4 l* v! c

5 h+ ~* @0 U( R8 D0 N$ LMiura M, Miura Y, Padilla-Nash HM et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. STEM CELLS 2006;24:1095¨C1103., C$ {' ~( h- M7 h, x
* P) F& e0 G5 D- @. [5 k4 z
Hackett JA, Feldser DM, Greider CW. Telomere dysfunction increases mutation rate and genomic instability. Cell 2001;106:275¨C286.: v1 _9 \# M% m1 k! f. u1 g6 l& W

% X0 g: ?( `+ n4 K# LFeldser DM, Hackett JA, Greider CW. Telomere dysfunction and the initiation of genome instability. Nat Rev Cancer 2003;3:623¨C627.) Q) F! y, y( g4 m4 l1 O+ c* B! R

; U  K. l& b# u! fRam¨ªrez N, Vilella FE, Colon M et al. Osteogenesis imperfecta and hyperplastic callus formation in a family: A report of three cases and a review of the literature. J Pediatr Orthop B 2003;12:88¨C96.
0 I6 W2 D/ w) t
% k1 r4 L* X9 x. a; P5 uTolar J, Nauta AJ, Osborn MJ et al. Sarcoma derived from cultured mesenchymal stem cells. STEM CELLS 2007;25:371¨C379.% R& C  J: f' y0 }$ H( Y7 E( n
- s3 B  V2 d* {+ \9 w. I' l! G
Deans RJ, Moseley AB. Mesenchymal stem cells: Biology and potential clinical uses. Exp Hematol 2000;28:875¨C884.
' m; i* F' ~! E" f! d7 g
$ o! R! k1 V' J3 @9 i% pRubio D, Garcia-Castro J, Martin MC et al. Spontaneous human adult stem cell transformation. Cancer Res 2005;65:3035¨C3039.
$ F- G% U/ r! F# _' P+ r7 g7 P) y5 S3 I, J$ ~
Burns JS, Abdallah BM, Guldberg P et al. Tumorigenic heterogeneity in cancer stem cells evolved from long-term cultures of telomerase-immortalized human mesenchymal stem cells. Cancer Res 2005;65:3126¨C3135.
8 A1 Q% c2 d* B/ f# H
6 q9 V, V$ p# O; N1 v1 O2 M0 h, CTheise ND, Henegariu O, Grove J et al. Radiation pneumonitis in mice: A severe injury model for pneumocyte engraftment from bone marrow. Exp Hematol 2002;30:1333¨C1338.. g9 q. g% \9 e1 c7 a
7 ^6 w/ V, o& k  e
Herzog EL, Van Arnam J, Hu B et al. Threshold of lung injury required for the appearance of marrow-derived lung epithelia. STEM CELLS 2006;24:1986¨C1992.

Rank: 2

积分
136 
威望
136  
包包
1877  
沙发
发表于 2015-6-10 17:18 |只看该作者
不错,支持下  

Rank: 2

积分
118 
威望
118  
包包
1769  
藤椅
发表于 2015-6-19 20:07 |只看该作者
这样的贴子,不顶说不过去啊  

Rank: 2

积分
101 
威望
101  
包包
1951  
板凳
发表于 2015-8-31 16:55 |只看该作者
干细胞之家微信公众号
不错 不错  比我强多了  

Rank: 2

积分
163 
威望
163  
包包
1852  
报纸
发表于 2015-9-11 20:10 |只看该作者
肌源性干细胞

Rank: 2

积分
61 
威望
61  
包包
1757  
地板
发表于 2015-9-13 21:14 |只看该作者
楼主福如东海,万寿无疆!  

Rank: 2

积分
122 
威望
122  
包包
1876  
7
发表于 2015-10-11 18:58 |只看该作者
ding   支持  

Rank: 2

积分
77 
威望
77  
包包
1730  
8
发表于 2015-10-12 11:35 |只看该作者
希望可以用些时间了~````  

Rank: 2

积分
97 
威望
97  
包包
1738  
9
发表于 2015-10-12 18:53 |只看该作者
一个有信念者所开发出的力量,大于99个只有兴趣者。  

Rank: 2

积分
72 
威望
72  
包包
1942  
10
发表于 2015-10-27 11:34 |只看该作者
病毒转染干细胞
‹ 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, 2024-5-19 16:20

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.