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: 320863|回复: 240
go

Retrovirus-Mediated Gene Transfer of Receptor Activator of Nuclear Factor-B-Fc P [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:03 |只看该作者 |倒序浏览 |打印
作者:Dohee Kim, Sun Wook Cho, Sun Ju Her, Jae Yun Yang, Sang Wan Kim, Seong Yeon Kim, Chan Soo Shin作者单位:Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea % q- Z" A$ I' M: g
                  
$ ~+ N) b  j2 b                  
% k- ?3 y& L* r9 h9 B" u1 R          , k3 J, ]- S2 y9 F: }0 n4 l: t
                         5 `8 K1 @' J4 r- M  a- R
            - |& p; h; `' l  e
            
! u7 o1 y7 x6 O6 A8 }            ' U7 U5 Y1 V( a, w9 n. ~$ M
            ) |- u$ G( v$ B% \
                      7 Q/ Q; ?2 ^+ N( b! X: n* @' L0 Y
        ) l5 i( ~+ @8 G" b) R: h3 i
        
# l. `5 r1 W1 c* }+ n. g) T% L; e        
) Y4 x# m  F* z3 }+ w* T2 u          【摘要】
2 {" C. ~$ ?& L8 ]- l      Postmenopausal osteoporosis is characterized by increased bone resorption due to estrogen deficiency. Receptor activator of nuclear factor-B-Fc (RANK-Fc), a fusion protein that specifically blocks receptor activator of nuclear factor ligand binding to RANK, has been known to be efficient and well tolerated in animal models of osteoporosis. Here we show that cell-based gene therapy with RANK-Fc effectively prevented bone loss in ovariectomized (OVX) mice. Thirty-one young adult female C57Bl/6 mice were used, and repeated intraperitoneal injection of mesenchymal stem cells (MSCs) transduced with retrovirus was performed as follows: 1) Sham-operated mice (n = 8); 2) OVX mice treated with phosphate-buffered saline (OVX-PBS; n = 8); 3) OVX mice injected with MSCs transduced with control retrovirus (OVX-green fluorescent protein ; n = 7); and 4) OVX mice injected with MSCs transduced with RANK-Fc (OVX-RANK-Fc; n = 8). Cellular expression of RANK-Fc was confirmed by Western blot analysis of cell lysates and conditioned medium and also by enzyme-linked immunosorbent assay for the mice serum. Measurement of bone mineral density (BMD) by dual-energy x-ray absorptiometry (PIXImus) revealed that the OVX-RANK-Fc group gained significantly higher BMD than either the OVX-PBS group or OVX-GFP group after 8 weeks. The expression of GFP, which is coexpressed with RANK-Fc, was detected by polymerase chain reaction analysis of DNA isolated from femur and intra-abdominal fat, whereas no GFP signal was identified in liver, brain, heart, lung, or bone marrow aspirates. These suggest that expression of RANK-Fc by genetically modified MSCs may be a feasible option for the prevention of bone loss induced by ovariectomy.
/ b5 l7 }- Z( I% `( `. u          【关键词】 Receptor activator of nuclear factor-B Receptor activator of nuclear factor-B-Fc Mesenchymal stem cell Gene therapy Osteoporosis Ovariectomy, C( f$ X$ Q0 H" y1 M* u! X
                  INTRODUCTION
1 I; x) x" J" r( g/ H# ^! h4 U8 Z# B1 {2 h# F
Osteoporosis is defined as a skeletal disorder characterized by compromised bone strength, which predisposes increased risk of fracture, most commonly in the vertebrae, wrist, and hip .4 ], v8 p" a& a+ ^. _5 l

1 l9 l6 H$ x, o3 {! M) ~6 w  ?+ [Osteoclasts, the bone-resorbing cells in the bone marrow environment, are derived from the hematopoietic macrophage system through the interactions between the receptor activator of the nuclear factor-B ligand (RANKL)/RANK system (reviewed in ref. . The RANK-Fc has the potential advantage over OPG of greater specificity for RANKL.
0 ~$ c9 K, @/ Z. ?  [, o' G# j, L5 e2 E/ l  e) h4 t  Y! F
The introduction of RANK-Fc via MSCs could be an option that could in theory overcome a number of conventional problems of recombinant protein technology, including complexity of purification, unstable biological activity, antibody formation, toxicity, and the need for frequent administration due to short half-life. These problems may be solved using gene delivery to express the targeted protein . This study was undertaken to test whether introduction of the MSCs genetically modified with a retrovirus engineered to express mouse RANK-Fc could prevent the bone loss induced by ovariectomy in mice.
8 O2 r" J/ M4 t$ J; t9 Y4 D4 y3 @
MATERIALS AND METHODS9 w6 y; B3 Y  K. v
4 w' ~! L! J- `
Animals( H; T9 b( O& A8 V! Q" m$ r
# T& H0 \' n! t* d# U: |& N! T5 G/ I
Female C57Bl/6 mice (5¨C6 weeks of age) were purchased from Orient Corporation (Kapyoung, Gyeonggi, Korea). All mice used in our experiments were maintained in the animal facility at Seoul National University Hospital Clinical Research Institute, in accordance with the animal care guidelines stipulated by the hospital. Diets and tap water were provided ad libitum throughout the study.
; q2 T" _$ x  q" P: M/ G+ x
' b6 i5 ]: t& Q5 B/ ?Retroviral Vector Construction% {$ [- [2 o: _, S) w

; F& t" ]7 A6 v8 o* oThe pMT-RANK-Fc vector containing a DNA sequence encoding the extracellular domain of mouse RANK (Met1-Pro213) fused via a linker to the Fc region of human immunoglobulin G1 (IgG1) was provided by Dr. Jaerang Rho (Chungnam National University, Daejon, Korea) and has been described previously  downstream of the MSCV long terminal repeat (LTR) and upstream of the IRES-eGFP cassette (Fig. 1), thereby allowing expression of both RANK-Fc and eGFP from a single bicistronic mRNA. The resulting construct was designated pMSCV-RANK-Fc-IRES-eGFP (pMSCV-RANK-Fc-eGFP; Fig. 1). In this system, gene expression is under the transcriptional control of MSCV LTR. The nucleotide sequence of the RANK-Fc coding region was confirmed by sequencing.% }4 D- h+ ?- h2 r; u8 C
( n' q: P# G2 s. J  U: @
Figure 1. Schematic representation of retroviral vectors used. RANK-Fc cDNA is located downstream of MSCV LTR and upstream of IRES-eGFP cassette, allowing the bicistronic expression of both RANK-Fc and eGFP from a single bicistronic mRNA. Abbreviations: eGFP, enhanced green fluorescent protein gene; IRES, internal ribosomal entry site of the encephalomyocarditis virus; MSCV-LTR, Moloney murine leukemia virus long terminal repeat; RANK-Fc, fusion protein of receptor activator of nuclear factor-B (Met1¨CPro213) with Fc region of human immunoglobulin G1.
$ r1 Y4 m- F) G* n6 x5 T$ z' E$ R; }. m& e9 p2 e+ _  Q3 J% @
Isolation and Culture Expansion of MSCs+ J! q5 v2 _+ p! k0 ]9 X

( A# {" {" K( A$ o2 w  i1 aFemale C57Bl/6 mice (6¨C8 weeks old) were sacrificed by cervical dislocation. MSCs were isolated from bone marrow of the femur and tibia by inserting a 27-gauge needle into the shaft of the bone and flushing with 1 ml of -minimum essential medium (MEM; Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com) supplemented with 10% fetal bovine serum (FBS; Cambrex, Walkersville, MD, http://www.cambrex.com), 2 mM L-glutamine, and 100 U/ml penicillin/100 µg/ml streptomycin (Invitrogen, Grand Island, NY, http://www.invitrogen.com). Cells were filtered through a 70-µm nylon filter (Falcon, Franklin Lakes, NJ), centrifuged for 5 minutes at 260g, suspended, and plated out in complete Dulbecco¡¯s modified Eagle¡¯s medium-high glucose (DMEM; Invitrogen) containing 15% horse serum (Invitrogen), 15% fetal calf serum (Invitrogen), 2 mM L-glutamine, 10¨C4 M 2-mercaptoethanol (Sigma-Aldrich), 10¨C6 M hydrocortisone (Sigma-Aldrich), and 100 U/ml penicillin-100 µg/ml streptomycin, at a density of 1 x 106 nucleated cells per ml in 25-cm2 culture flasks (Techno Plastic Products AG, Trasadingen, Switzerland). Cells were grown in complete medium at 37¡ãC at 5% CO2 for 7 days, and the medium was replaced every 2¨C3 days. Adherent cells were grown to 90% confluence, washed with phosphate-buffered saline (PBS), and incubated with 0.25% trypsin/2 mM EDTA (Invitrogen). Nondetached cells were discarded, and the remaining cells were designated passage 1 (P1). Confluent MSCs were passaged further and plated at a dilution of 1:2 to 1:3. P2 to P5 MSCs were transduced with retrovirus.
3 Y- G0 a) Z$ f
: T3 q/ |+ c7 R7 u9 xEstablishment of MSCs Overexpressing RANK-Fc' x, X! S7 B  S4 f  e

2 v5 q. l1 N# {& XTo obtain the viral supernatant, 293T packaging cells were transfected with the retroviral construct using Lipofectamine PLUS (Invitrogen). Briefly, 24 hours prior to transfection, 5 x 106 293T cells were plated in 100-mm dishes containing DMEM supplemented with 10% FBS and 100 U/ml penicillin-100 µg/ml streptomycin. The transfection solution contained DNA (4 µg of pMD-gag-pol, 4 µg of pMD-VSVG, and 4 µg of retroviral vector pMSCV-eGFP or pMSCV-RANK-Fc-eGFP alone), Lipofectamine PLUS, and serum-free DMEM. The medium was replaced after 3 hours. Viral supernatant fractions were collected at 48 hours, filtered through a 0.45-µm syringe filter (Nalgene, Rochester, NY), and stored at ¨C80¡ãC.
1 _3 O! ~+ y4 H  I3 Z6 D; W( f# v( |" \
MSCs were plated on six-well plates at a density of 1.5 x 105 cells per well, 24 hours prior to retroviral transduction. The culture medium was removed, and retroviral supernatant (MSCV-eGFP or MSCV-RANK-Fc-eGFP) with polybrene (Sigma-Aldrich) at a final concentration of 4 µg/ml was added to the culture. Culture plates were centrifuged at 2,800 rpm at 32¡ãC for 90 minutes and incubated at 37¡ãC for 2 hours. A second round of infection was performed as described above, and the retrovirus supernatant was replaced with culture medium for overnight incubation. Following the third and fourth rounds of infection, cells were cultured until near confluency. Transduced cells were directly evaluated for GFP expression using a standard fluorescent microscope (Olympus IX70) and flow cytometry with a fluorescence-activated cell sorter (FACS) (FACS Vantage SE; Becton, Dickinson and Company, Franklin Lakes, NJ, http://www.bd.com) at 530 ¡À 30 nm.
/ T2 \. d9 k$ f+ N5 E4 f+ U5 i4 C4 w) F
Expression of RANK-Fc In Vitro- H+ t6 w% E. T7 w% f! H( o8 O
; i2 o. I; P# _2 o  a! U1 Z- E
To assess the cellular expression of RANK-Fc, Western blot analysis was performed using cell lysates from MSCs transduced with MSCV-RANK-Fc-eGFP or empty MSCV-eGFP retrovirus as described previously . Briefly, cell lysates were prepared as follows: cells were treated with lysis buffer (150 mM NaCl, 50 mM Tris-Cl, pH 7.4, 20 mM EDTA, 1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS, and protease inhibitors), fractionated by SDS-polyacrylamide gel electrophoresis (10% gels), and transferred to nitrocellulose membranes. Blots were probed with goat polyclonal anti-mouse RANK antibody (0.1 µg/ml) or goat polyclonal anti-human IgG Fc fragment-specific antibody (1:1,000; Jackson Laboratory, Bar Harbor, ME, http://www.jax.org), followed by horseradish peroxidase (HRP)-conjugated polyclonal anti-goat IgG (1:3,000; Santa Cruz Biotechnology Inc., Santa Cruz, CA, http://www.scbt.com) and detected by enhanced chemiluminescence using an enhanced chemiluminescence kit (Amersham Biosciences, Piscataway, NJ, http://www.amersham.com), according to the manufacturer¡¯s instructions." X+ ~# B" F! n+ D1 O' F. o% }" o
0 V" i- o* t1 }; o3 N, f, G: v6 y' h
For assaying RANK-Fc secretion in vitro, serum-free DMEM was added to confluent plates of MSCs transduced with MSCV-RANK-Fc-eGFP or MSCV-eGFP. After 24 hours, culture supernatants were incubated at 4¡ãC overnight with goat polyclonal anti-mouse RANK antibody (1 µg/ml; R&D Systems), followed by a 4¡ãC overnight incubation with 20 µl of the agarose conjugate suspension (protein A/G-agarose; Santa Cruz Biotechnology Inc.). Samples were centrifuged at 2,500 rpm for 30 seconds at 4¡ãC, and the supernatant was aspirated. Next, pellets were washed three times and suspended with 40 µl of 2x electrophoresis sample buffer. Samples were boiled for 5 minutes and then subjected to Western blot analysis using anti-mouse RANK antibody or anti-human IgG Fc fragment-specific antibody as above.
9 o# @" ~3 _6 S$ ~
4 x5 d  E1 S9 |" E# qExperimental Design
; X$ Z1 B+ x/ \& p% j# L+ O6 u/ e. O  ]0 L
Mice were randomly divided into four groups as follows: 1) Sham-operated mice (SHAM; n = 8); 2) OVX mice treated with PBS (OVX-PBS; n = 8); 3) OVX mice injected with MSCs transduced with control retrovirus (OVX-GFP; n = 7); and 4) OVX mice injected with MSCs transduced with RANK-Fc (OVX-RANK-Fc; n = 8). Bilateral ovariectomy in mice is a standard method to evaluate agents that ameliorate bone loss associated with estrogen deficiency . On study day ¨C1, mice were anesthetized with ketamine (50 mg/kg) and xylazine (15 mg/kg) to allow exposure of the ovaries. Specifically, the gonads were removed in OVX groups but only manipulated in the SHAM cohorts. To determine whether prophylactic treatment with RANK-Fc prevents ovariectomy-induced bone loss in an osteoporosis model, animals underwent repeated intraperitoneal injection of the indicated doses of transduced cells (OVX-GFP, OVX-RANK-Fc) or PBS (SHAM, OVX-PBS) on days 0, 2, 4, and 6.# {, l( \% M( ^1 |/ Q3 Z' A8 c
2 ^- ?" S  I% l
Transduced MSCs were trypsinized, centrifuged, and suspended at a concentration of 1 x 107 nucleated cells per milliliter of PBS. Suspensions of transduced MSCs were intraperitoneally injected into C57Bl/6 mice in a total volume of 0.2 ml. Intraperitoneal injections were repeated every other day for a total of four doses, based on a previous study . Sera were collected on the indicated days. Blood samples from retro-orbital capillaries were taken under anesthesia with diethyl ether (Duksan Chemical, Duksan, Korea), and serum was recovered by centrifugation at 8,000g for 10 minutes. Samples were stored at ¨C80¡ãC until analysis., y1 o# B7 \: ~% O

- q+ v! h3 K2 c4 n7 DBone Mineral Density Measurement
- V* {; E$ x6 P4 x' C( v
8 `9 d- D' l  o2 W" C0 _/ qBone mineral density (BMD) and body composition were measured using a Lunar PIXImus densitometer (software version 2.0; GE Lunar, Madison, WI) on indicated days. PIXImus scanner is a proven device for bone mineral and body composition measurements of mice and other small animals weighing 10¨C50 g. PIXImus acquires images in less than 5 minutes with dual-energy x-ray absorptiometry (DEXA). Mice were anesthetized with a mixture of ketamine (50 mg/kg) and xylazine (15 mg/kg) in PBS and placed prone on the platform of the PIXImus, and whole body bone mineral content (BMC; mg) and areal BMD (g/cm2) with the exclusion of head was acquired with mouse-specific software (version 1.47). Basically, BMD is calculated by dividing the BMC by projected body area. Body fat content can be also obtained by the same software. All DEXA scans were conducted by the same researcher (D.K.). Percent coefficient of variation of BMC and BMD for the repeated scans was 1%¨C2%.! G5 X2 u6 [2 N, y1 h
8 w- b  S% H" o- ]% O3 q5 z. q4 S
Expression of RANK-Fc In Vivo7 x8 n* g! P- @5 k1 q

; s: s& F* {, l- f' J# f6 A' y# XFor enzyme-linked immunosorbent assay (ELISA), 96-well microplates were coated with goat polyclonal anti-mouse RANK antibody (1 µg/well) overnight at 4¡ãC, washed three times with PBS-Tween (0.5% Tween 20 per liter, pH 7.4), and incubated at room temperature with blocking solution (1.59% Na2CO3, 2.93% NaHCO3, 0.2% NaN3, 1% BSA, 5% sucrose) for 1¨C2 hours. Wells were washed, incubated overnight at 4¡ãC with RANK-Fc protein standard prepared in Sf9 cells (a kind gift from Dr. Jaerang Rho, Chungnam National University, Daejeon, Korea)  and diluted culture supernatant or mouse serum samples (100 µl); rewashed; and treated with HRP-conjugated goat anti-human IgG Fc fragment-specific antibody (1:5,000; Jackson Laboratory) for 2¨C4 hours. Plates were washed and incubated at room temperature with substrate solution (tetramethylbenzidine base; Sigma-Aldrich) for 20 minutes. Following the addition of stop solution (1 M H2SO2), the optical density of each well was determined using a microplate reader set (ThermoMax; Scientific Surplus, Belle Mead, NJ) at 450 nm.* {3 f; j2 r1 r* n. Q  Y9 y( r

9 V6 b# Z* q! Y- c  T' n. cDNA PCR for GFP  ~$ I- o4 }" \6 R8 N
1 c" e2 `  n) B* u6 N+ g9 M, E! E
Femurs from sacrificed mice were dissected free of surrounding tissue and fixed in 4% paraformaldehyde-PBS (pH 7.4 with 10 N NaOH) at 4¡ãC for 3 days. Soft tissues (liver, brain, heart, lung, and intra-abdominal fat) were dissected and placed in paraformaldehyde overnight. Bone marrow cells were isolated from the femur and tibia by inserting a 27-gauge needle into the shaft of the bone and flushing with 1 ml of -minimum essential medium as described above. To detect GFP by polymerase chain reaction (PCR), tissues and bone marrow cells were placed in a lysis buffer containing 100 mM NaCl, 100 mM Tris-Cl (pH 8.0), 25 mM EDTA (pH 8), 0.5% sodium dodecyl sulfate, and 0.1 mg/ml proteinase K. The bones were homogenized, and then lysis buffer was added. DNA was extracted from the resultant fluid by incubating in a 50¡ãC incubator overnight, followed by addition of a phenol:chloroform mixture (1:1) to layer out the DNA from the organic phase. The supernatant of this solution was combined with 10 mM ammonium acetate, and the DNA was precipitated out with 7.3 ml of 100% ethanol per ml of sample, centrifuged, washed in 70% ethanol, and suspended in Tris:EDTA buffer.1 E6 j3 w6 L: t8 Z4 W
1 z4 E- m0 N( {8 \) r4 d
PCR was performed using 2 µl of template DNA, 20 pmol of each primer (synthesized by Bioneer Corp., Chungwon, Korea), 200 µM dNTPs, 1 mM MgCl2, and 1 U of Taq polymerase in a 50-µl reaction volume containing 1x Taq polymerase buffer using a Perkin-Elmer Gene Amp PCR System 2400. The sense and antisense primers, 5'-CACATGAAGCAGCACGACTT-3' and 5'-AGTTCACCTTGATGCCGTTC-3', were used to amplify GFP-producing bands of 265 base pairs (bp).9 c, K  Y" [4 H& Y9 P& z( a

" E8 p8 R% M7 H: s5 w/ k' dStatistical Methods
% W% u4 W6 o/ q" U
0 d3 D9 U  z, [; |9 e) r% J% aData were assessed using Statistical Package for the Social Sciences (SPSS) software (version 11.0) and summarized with descriptive statistics (mean and standard error). The differences between the four treatment groups at each time point were analyzed with the nonparametric Kruskal-Wallis test. All tests were two-sided, and a significance level of 5% was assigned.; E7 v9 Q9 W* i1 e  _

2 l; v/ A$ o4 g5 s7 \4 pRESULTS
! J4 @: s$ |$ p# l  F+ L7 b* J; ?4 R
Establishment of Cells Stably Expressing RANK-Fc. `9 p2 i3 l3 d% ?+ `

$ ]8 N2 u# p1 n* ~To establish cells stably overexpressing RANK-Fc, P2 to P5 MSCs were transduced with MSCV-RANK-Fc-eGFP and directly evaluated for GFP expression by visualizing under a fluorescent microscope. Since transcription of both RANK-Fc and eGFP genes is driven by the same promoter, MSCV LTR, GFP-positive cells should signify RANK-Fc expression. Using FACS analysis, we have confirmed that the transduction efficiencies were 94.1% and 92.5% in MSCs transduced with MSCV-RANK-Fc-eGFP and MSCV-eGFP, respectively.; k( K# `. I9 l
* ~: |; c: F* u1 b$ p' W+ |, T
To confirm the cellular expression of RANK-Fc, protein isolated from lysates of transduced MSCs were subjected to Western blot analysis using antibodies against mouse RANK or human IgG Fc. As shown in Figure 2A and 2B, cells transduced with the pMSCV-RANK-Fc-eGFP vector overexpressed RANK-Fc, compared with those infected with pMSCV-eGFP. Moreover, to confirm the extracellular secretion of RANK-Fc, we analyzed the culture medium of MSCs overexpressing RANK-Fc by immunoprecipitation-Western blot analysis. Immunoblots disclosed RANK-Fc protein secretion in medium from cells transduced with MSCV-RANK-Fc-eGFP, but no immunoreactivity was observed in conditioned medium from cells transduced with MSCV-eGFP (Fig. 2C, 2D).
* @3 H4 q0 ~( c
, I6 ?5 I; _' g: J) \Figure 2. Cellular expression and secretion of RANK-Fc in vitro. (A, B): Western blot analysis of cellular lysates from transduced mesenchymal stem cells (MSCs). Cell lysates from confluent cultures of MSCs transduced with either plasmid murine stem cell virus (pMSCV)-eGFP or pMSCV-RANK-Fc-eGFP were analyzed by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and Western blotting with goat polyclonal anti-mouse RANK antibody (A) or goat polyclonal anti-human IgG Fc fragment-specific antibody (B). (C, D): Protein samples from conditioned medium were immunoprecipitated with goat polyclonal anti-mouse RANK antibody and resolved by SDS-PAGE. Blots were probed with goat polyclonal anti-mouse RANK antibody (C) or goat polyclonal anti-human IgG Fc fragment-specific antibody (D). Abbreviations: GFP, green fluorescent protein; IP, immuno-precipitation; mRANK, mouse receptor activator of nuclear factor-B; RANK, receptor activator of nuclear factor-B; WB, Western blot.
" C8 U9 A, x, m
; W8 M! J/ _7 e. X4 PRANK-Fc Expression In Vivo
2 a% X5 ?! `) Q3 A- I
& S) W, E1 Y) b# E5 @, ]9 e9 tNext, we investigated whether MSCs transduced with RANK-Fc retrovirus continuously secrete biologically active RANK-Fc in vivo after intraperitoneal injection. Blood was collected from retro-orbital capillaries, and serum samples were prepared and analyzed for RANK-Fc activity (Fig. 3). Serum RANK-Fc levels measured by ELISA peaked at 2 weeks after the first injection (9.65 ¡À 2.66 ng/ml, mean ¡À SE), and gradually decreased to 0.60 ¡À 0.21 ng/ml at 8 weeks after the first injection. Serum RANK-Fc levels were much lower (approximately 50-fold) than those from conditioned medium from the cultured MSCs (data not shown). No RANK-Fc was detected in control animals.
& B1 R" R$ e. N! o& F* E9 G! B
4 u6 s/ ~& T% k  y+ |: rFigure 3. Production of RANK-Fc from transplanted mesenchymal stem cells (MSCs) in vivo. Sera from the C57Bl/6 mice that underwent intraperitoneal injection of MSCs transduced with p-murine stem cell virus-RANK-Fc-enhanced green fluorescent protein (pMSCV-RANK-Fc-eGFP) retrovirus were collected on days 5, 14, 28, and 56 after the first injection. The samples were subjected to enzyme-linked immunosorbent assay analysis using anti-Fc antibody. Data are expressed as means and standard errors. n = 7¨C8 per group. Abbreviation: RANK, receptor activator of nuclear factor-B.
6 `4 w$ K; v/ R+ t* n& y" w, J+ v) }0 X' Z8 p" D& e3 ~
RANK-Fc Expression Attenuates Bone Loss in the OVX Model
3 b' A5 e) Y  K; e* K5 ]9 I
7 `5 A5 e5 ^0 P' hAfter ovariectomy, all OVX mice exhibited higher body weight and body fat mass (%) than sham-operated animals, indicating that estrogen deficiency had successfully induced (Fig. 4A, 4B). As a surrogate marker for the functional capacity of the genetically modified cells in vivo, we determined the changes in BMD in the mice that were given MSCs. As shown in Figure 4C and 4D, all OVX mice (OVX-PBS or OVX-GFP group) gained significantly less BMD and BMC, from baseline compared with the sham-operated animals (p
, b% v2 E! B  J' \3 L5 a) J0 c- c' I7 o3 c; }, J
Figure 4. Changes in the body weight, fat mass, BMD, and BMC in the animals. Seven-week-old female C57Bl/6 mice were sham-operated or ovariectomized. OVX-RANK-Fc mice, n = 8; OVX-GFP mice, n = 7; OVX-PBS mice, n = 8). SHAM mice (n = 8) were used as a control. Body weight (g), body fat mass percent (%), BMD (g/cm2), and BMC (g) were measured at baseline and at 4 and 8 weeks after the operation using dual-energy x-ray absorptiometer (PIXimus; GE Healthcare). Shown are percent changes in body weight (A), body fat mass (B), BMD (C), and BMC (D) compared with the baseline. Data are expressed as means and standard errors. *, p * }5 v/ _4 O0 A" X3 F
! x5 @9 ^( s  j: |1 z4 f) x4 O
Table 1. Body weight, BMC, BMD, and body fat changes of the animals
6 g; ^- T% i) f8 P* F! P6 }$ |9 T9 U, H" A3 G1 b( f
It is noteworthy that all the mice in this experiment actually gained BMD because they were still in the growing phase of development . However, the percentage increase in BMD in mice transplanted with MSCs overexpressing RANK-Fc was significantly greater than that of the OVX-PBS group (p
) H8 E9 j( M& _  X" ?  `& J' N$ l/ l: e& u
Detection of GFP Expression by PCR# C; U$ P4 e6 _! q  T, E9 w

- {/ y; }( A- s4 {4 E: `We next tested whether the bone-protective effect of RANK-Fc was a result of engraftment of RANK-Fc producing cells into bone. To address this, PCR analysis for GFP was performed using genomic DNA isolated from various tissues or bone marrow aspirates. As shown in Figure 5A, expected GFP DNA fragment (265 bp) was weakly but clearly amplified from femur and intra-abdominal fat of OVX-RANK-Fc mice, whereas tissues from OVX PBS group did not reveal any positive signal (Fig. 5B). DNA isolated from liver, brain, heart, lung, or bone marrow aspirates did not give a positive band in either of the two groups. Essentially the same pattern of GFP expression (i.e., only from femur and fat tissue) was also observed from OVX GFP mice (data not shown). These results suggest that some degree of engraftment to bone has occurred after systemic injection of retrovirus-transduced MSCs, although a comparable amount of expression was also observed in fat tissue.4 s7 r5 F8 a) {- A

4 \; i$ K8 c) p! wFigure 5. Localization of RANK-Fc-secreting cells in tissue sections of transduced mesenchymal stem cell transplants. Tissues and bone marrow aspirates from the C57Bl/6 mice that underwent intraperitoneal injection of the transduced mesenchymal stem cells or PBS were harvested 56 days after the first injection and prepared for the analysis of green fluorescent protein (GFP) by polymerase chain reaction (PCR). DNA PCR using oligonucleotide primers for the GFP gene was performed using in genomic DNA isolated from various tissues as template. Lane 1, liver; lane 2, brain; lane 3, heart; lane 4, lung; lane 5, intra-abdominal fat; lane 6, femur; lane 7; bone marrow aspirates; lane 8, positive control. Abbreviations: M, 100-base pair DNA ladder; OVX, ovariectomized; PBS, phosphate-buffered saline; RANK-Fc, receptor activator of nuclear factor-B-Fc.
: o, O$ ~5 J! D; A+ q; I
( Y; h8 a$ C, w: J  ^9 T+ o, {4 \  D8 _/ YDISCUSSION
& S8 X( U$ o: {; ^8 z
6 y5 ~2 I# {5 e) pThe objective of this study was to examine the feasibility of using genetically modified MSCs as a platform for sustained systemic delivery of therapeutic proteins into the circulation in an osteoporosis model. Our data show that MSCs are effectively transduced with MSCV-based retrovirus and subsequently are capable of secreting biologically active RANK-Fc in vitro and in vivo. Moreover, RANK blockade by RANK-Fc is an effective method to prevent ovariectomy-induced bone loss.
. z( P8 ^% f$ l7 y& k! Z7 }/ T- L$ D2 F5 N# t
MSCs attract considerable attention in efforts to develop cell and gene therapies .
# c) z+ Q8 E/ }+ W- G+ {5 ?' M# m' q4 u  f9 J! G
Our data demonstrate that mesenchymal cell-based gene therapy with RANK-Fc has clearly protected bone loss associated with ovariectomy in C57Bl/6 mice. Effects on bone density are small, however, possibly due to the rapid skeletal growth of the mice that renders the evaluation of a "bone-protective" effect difficult in this rodent OVX model. In our study, the circulating level of RANK-Fc peaked at 1 week after the last injection, consistent with previous data  and/or loss of transduced cells. Although these results are promising, they highlight the need for careful consideration of ex vivo methods, choice of promoter to direct the desired biological activity, and assessment of the self-maintenance of transduced MSCs upon in vivo transplantation.5 q) E" M' m6 @3 U1 |

9 m# n$ S* T2 ~& q  T' yIt remains a matter of controversy whether donor-derived MSCs are capable of homing and engraftment into the marrow cavity following transplantation. A number of reports indicate that marrow-derived cells engraft as differentiated cells into multiple tissues following infusion into experimental animals . In our experiments, GFP signals were observed in bone and intra-abdominal fat, but not from any other tissues, including bone marrow aspirates by PCR analysis. The presence of GFP signal from intra-abdominal fat seems to be a consequence of intraperitoneal injection of cells resulting in initial accumulation of a large number of cells locally. Our finding of positive PCR bands from femur but not from freshly isolated bone marrow cells is intriguing. One can speculate that the injected cells engrafted, differentiated, or moved, and then resided within the bone tissue, such as bony lacunae or lining cells. However, we have not traced the migration of cells directly, nor were we able to identify the cell types that express GFP using sensitive assays, such as fluorescent in situ hybridization. Therefore, the positive PCR results from femur do not definitely prove the engraftment of donor cells. In addition, the cells we used as a vehicle were not isolated by specific selection method using cell surface markers, and introducing retrovirus by multiple rounds of transduction may have resulted in alteration of stem cell characteristics, including the capacity for homing. Indeed, in our study, the therapeutic effects of RANK-Fc could have been obtained regardless of the location of the transplanted cells once they are connected to the systemic circulation. To confirm whether the systemically administered cells have engrafted, in situ immunological analysis will be required in our model.- `' r( B, e4 j$ e$ k

. d, ~, K/ I' tMSCs are easily isolated from iliac crest bone marrow aspirates. The systemic infusion of autologous MSCs appears to be well tolerated in a Phase I clinical trial . Accordingly, it may be possible to use gene-engineered MSCs from patients for therapy of common diseases, such as osteoporosis, in which marrow ablation cannot be justified. However, notwithstanding the potential benefits of MSC as a cellular vehicle to convey therapeutic gene product, strategies to overcome the risks imposed by retroviral transduction, that is, uncontrolled proliferation and even tumor formation, which can occur depending on the integration of the virus, should be developed before any clinical application could be tried.
7 q5 A$ [# B2 O, Q* U! \: B0 T) X/ Q* X& @6 T
The main limitation of our study is that by using mice in a growing phase, it is difficult to directly extrapolate our results to postmenopausal or involutional model of osteoporosis. Indeed, the role of the RANK/OPG system in the growing mice has not been well established. Moreover, since BMD measurement using DEXA is significantly affected by the size of the bones, interpreting the changes in BMD in animals of different size is very complicated ., I; M; F$ \& H  H7 t

* J- l* u$ e, g' E4 R; k& u  P4 yIn summary, genetically modified MSCs are capable of sustaining to secrete RANK-Fc up to 8 weeks after intraperitoneal injection, and RANK blockade by RANK-Fc may be an effective method to prevent ovariectomy-induced bone loss. These data support the use of bone marrow-derived MSCs as cellular vehicles for gene therapy and pave the way for a future therapeutic approach for osteoporosis using genetically modified MSCs.
, F% O  C! f1 A# l; `& V* ?: g4 I3 Z" r5 R
DISCLOSURES
& H1 F" V8 Z0 i, T* b! d! F5 g1 y  _7 ]' H  X
The authors indicate no potential conflicts of interest.& r4 A6 z* e' Z5 p% Y
( `9 a) c/ X' u* z8 [/ z
ACKNOWLEDGMENTS
2 w1 z; I- m1 ^: e+ O$ k* w' C  x- z9 v4 P5 a: @
This work was supported by grant 06-2005-053-0 from Seoul National University Hospital. D.K. is currently affiliated with the Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Korea.
& m- N* C, D* U3 q( z- w          【参考文献】9 m! N" W& o+ |

# L2 [! l9 l" [* i, g3 y$ j
  c( V5 [" N  J9 o5 {! _* O+ t4 \NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA 2001;285:785¨C795.
" a9 O( ~5 C( h7 I, ]5 S2 Y' z- W) _: T$ D2 R) J
Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997;276:71¨C74.* t( \( d/ R) I) D

9 j9 s& g! U$ {" FBianco P, Riminucci M, Gronthos S et al. Bone marrow stromal stem cells: Nature, biology, and potential applications. STEM CELLS 2001;19:180¨C192.: B. N7 F+ f& l" K, l# W# M

6 k1 {, ^/ U; D# lBartholomew A, Patil S, Mackay A et al. Baboon mesenchymal stem cells can be genetically modified to secrete human erythropoietin in vivo. Hum Gene Ther 2001;12:1527¨C1541.$ \8 n9 e) i1 A

0 d" ~0 u' e* @# u* j7 x* KHofbauer LC, Heufelder AE. Role of receptor activator of nuclear factor-kappaB ligand and osteoprotegerin in bone cell biology. J Mol Med 2001;79:243¨C253.8 x/ q, }. y  z. n) F1 Q) q
- n2 {' K6 V( b% s5 T4 B) J( v* {& r; }
Simonet WS, Lacey DL, Dunstan CR et al. Osteoprotegerin: A novel secreted protein involved in the regulation of bone density. Cell 1997;89:309¨C319.
/ f5 L. d( d  O6 j! K6 C
0 d; E" x4 W/ S0 D6 XHsu H, Lacey DL, Dunstan CR et al. Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc Natl Acad Sci U S A 1999;96:3540¨C3545.4 J# s; F8 e+ r& |

" I1 m$ [# A/ M, `7 _Lacey DL, Timms E, Tan HL et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998;93:165¨C176.4 U1 h  t+ K8 m4 C1 f! N9 v
. x# e& ~; ?# v6 C
Bucay N, Sarosi I, Dunstan CR et al. Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 1998;12:1260¨C1268.
; X! q% j; l$ N3 A' B  r8 ?3 X$ T7 q5 T' c/ f; v9 A. A
Kong YY, Yoshida H, Sarosi I et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 1999;397:315¨C323.
- W" K/ }4 |0 x! G8 {# [* _, }! [5 A+ u
Dougall WC, Glaccum M, Charrier K et al. RANK is essential for osteoclast and lymph node development. Genes Dev 1999;13:2412¨C2424.. J$ e. K7 Z+ L6 w

/ R9 ?% r2 `, \6 R. _Nakashima T, Wada T, Penninger JM. RANKL and RANK as novel therapeutic targets for arthritis. Curr Opin Rheumatol 2003;15:280¨C287.3 \; ]0 e+ Y# y( r& ?+ }
& Q+ F1 U' B8 J& n; _( d
Honore P, Luger NM, Sabino MA et al. Osteoprotegerin blocks bone cancer-induced skeletal destruction, skeletal pain and pain-related neurochemical reorganization of the spinal cord. Nat Med 2000;6:521¨C528.
# A3 e4 H: e- I7 Q  t8 W9 @( ?. @+ a. N4 s! x) M4 `
Bekker PJ, Holloway D, Nakanishi A et al. The effect of a single dose of osteoprotegerin in postmenopausal women. J Bone Miner Res 2001;16:348¨C360.& }% d  Y, _  I  t+ I

2 z: r. m- K" X& IBody JJ, Greipp P, Coleman RE et al. A phase I study of AMGN-0007, a recombinant osteoprotegerin construct, in patients with multiple myeloma or breast carcinoma related bone metastases. Cancer 2003;97:887¨C892.
% z% G4 Q+ Y! \4 S$ y8 r
- t& F- h5 {* D; {/ X; uSordillo EM. Pearse RN RANK-Fc: A therapeutic antagonist for RANK-L in myeloma. Cancer 2003;97:802¨C812.
; f) q  B8 ^; ~( O" U6 Y2 z
7 @" X, ]0 O& e- ePearse RN, Sordillo EM, Yaccoby S et al. Multiple myeloma disrupts the TRANCE/osteoprotegerin cytokine axis to trigger bone destruction and promote tumor progression. Proc Natl Acad Sci U S A 2001;98:11581¨C11586." g8 Q5 a8 V  N/ _

6 e6 w4 e* ]7 ^8 }% lBolon B, Carter C, Daris M et al. Adenoviral delivery of osteoprotegerin ameliorates bone resorption in a mouse ovariectomy model of osteoporosis. Mol Ther 2001;3:197¨C205.
8 [, K6 G6 V) M/ U2 g
, h& ~1 d8 ^" E9 n& EBailey EC, Milenkovic L, Scott MP et al. Several PATCHED1 missense mutations display activity in patched1-deficient fibroblasts. J Biol Chem 2002;277:33632¨C33640.
7 Z3 ^1 d! z+ r/ |8 r9 {
- S7 S% Q8 D2 H# JJeon MJ, Kim JA, Kwon SH et al. Activation of peroxisome proliferator-activated receptor-gamma inhibits the Runx2-mediated transcription of osteocalcin in osteoblasts. J Biol Chem 2003;278:23270¨C23277.( a  j8 W1 v) z& L$ F( o7 @
6 I1 _1 S" q2 Z5 l3 ^% a+ P9 Y
Bellino FL. Nonprimate animal models of menopause: Workshop report. Menopause 2000;7:14¨C24.. _  e& l  v) s# ?
2 Z. m5 Q& S# s* q6 E- q! k6 I
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¨C1147.3 N) e4 z% j/ K1 O7 n$ I' @+ Z

! }3 h2 K3 B% a0 ZAzizi SA, Stokes D, Augelli BJ et al. Engraftment and migration of human bone marrow stromal cells implanted in the brains of albino rats-similarities to astrocyte grafts. Proc Natl Acad Sci U S A 1998;95:3908¨C3913.
% S) }" k; e  U! Q$ e6 l$ Z6 |; P5 e* g
Prockop DJ, Gregory CA, Spees JL. One strategy for cell and gene therapy: Harnessing the power of adult stem cells to repair tissues. Proc Natl Acad Sci U S A 2003;100 (suppl):11917¨C11923.3 Z- D" S9 i% I+ ]1 f
; g7 G6 F7 S5 \# V8 w7 Y
Schwarz EJ, Alexander GM, Prockop DJ et al. Multipotential marrow stromal cells transduced to produce L-DOPA: Engraftment in a rat model of Parkinson disease. Hum Gene Ther 1999;10:2539¨C2549.& q5 v: E* q+ |
" `3 O' P  x/ @, l3 V
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.
2 S, m# q; }- v3 D
0 h  w. q/ U, gLi Y, Chen J, Chen XG et al. Human marrow stromal cell therapy for stroke in rat: Neurotrophins and functional recovery. Neurology 2002;59:514¨C523.
5 _6 M% t- g# p6 J6 N
  Z. X& ^0 J9 ?Jin HK, Carter JE, Huntley GW et al. Intracerebral transplantation of mesenchymal stem cells into acid sphingomyelinase-deficient mice delays the onset of neurological abnormalities and extends their life span. J Clin Invest 2002;109:1183¨C1191.
1 ?& `( I1 q1 k  c% [. [! x
+ m: s& x7 v# K1 {Toma 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¨C98.
, \4 L' O6 n& A, a
' y) R3 V+ Q( ]5 DOrtiz 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./ E4 W9 z9 e$ b7 A

& J& ]2 ~/ F) e5 m/ Q( {0 z0 K" w/ |Krebsbach PH, Zhang K, Malik AK et al. Bone marrow stromal cells as a genetic platform for systemic delivery of therapeutic proteins in vivo: Human factor IX model. J Gene Med 2003;5:11¨C17.
6 B; Y' {% }+ I( }) x, h  Z2 t9 `+ O9 B
Ding LM, Saylors R, Munshi NC. The stromal cell as a vehicle for ex vivo gene transfer. Blood 1997;89:446¨C456.
7 o8 n; w% d: q
8 D& _5 S2 h, E9 x( XChuah MK, Van Damme A, Zwinnen H et al. Long-term persistence of human bone marrow stromal cells transduced with factor VIII-retroviral vectors and transient production of therapeutic levels of human factor VIII in nonmyeloablated immunodeficient mice. Hum Gene Ther 2000;11:729¨C738.0 S9 E. k" W2 z- h* a: e. g1 a1 T
. J  d3 g. @8 ?$ H* |9 P# _
Murry CE, Soonpaa MH, Reinecke H et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 2004;428:664¨C668.
7 K' m3 S9 l6 e  V5 a" S6 |* R6 Q+ S$ O
Balsam LB, Wagers AJ, Christensen JL et al. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 2004;428:668¨C673./ R" E! a3 K5 i

' q$ c. o! |# c5 B: ^2 j! QLazarus HM, Haynesworth SE, Gerson SL et al. Ex vivo expansion and subsequent infusion of human bone marrow-derived stromal progenitor cells (mesenchymal progenitor cells): Implications for therapeutic use. Bone Marrow Transplant 1995;16:557¨C564." R: e4 P9 i$ J2 R+ _6 B9 W( s: N

- ], @0 Y/ R; L9 P% rStewart FM, Crittenden RB, Lowry PA et al. Long-term engraftment of normal and post-5-fluorouracil murine marrow into normal nonmyeloablated mice. Blood 1993;81:2566¨C2571.
6 L4 ?' q! a7 W9 Y8 G
9 [* T4 n9 \& e4 L' Y2 u5 oBienzle D, Abrams-Ogg AC, Kruth SA et al. Gene transfer into hematopoietic stem cells: Long-term maintenance of in vitro activated progenitors without marrow ablation. Proc Natl Acad Sci U S A 1994;91:350¨C354.* `# V0 ^* e) w# ?0 \

( |3 ]4 R: k$ Y. [' a' {% ]Dickson GR, Luczak M, Wlodarski KH. The limitation of DEXA analysis for bone mass determination in mice. Folia Biol (Krakow) 2004;52:125¨C129.
7 {: t: D0 j3 y0 S$ m5 x
$ }/ O8 W: a- U% ?6 PHoldsworth DW, Thornton MM, Drost D et al. Rapid small-animal dual-energy X-ray absorptiometry using digital radiography. J Bone Miner Res 2000;15:2451¨C2457.

Rank: 2

积分
76 
威望
76  
包包
1772  
沙发
发表于 2015-5-25 08:43 |只看该作者
羊水干细胞

Rank: 2

积分
64 
威望
64  
包包
1782  
藤椅
发表于 2015-6-6 19:42 |只看该作者
呵呵,明白了  

Rank: 2

积分
72 
威望
72  
包包
1859  
板凳
发表于 2015-6-14 06:10 |只看该作者
干细胞之家微信公众号
不早了 各位晚安~~~~  

Rank: 2

积分
75 
威望
75  
包包
2193  
报纸
发表于 2015-6-17 19:42 |只看该作者
抢座位来了  

Rank: 2

积分
64 
威望
64  
包包
1769  
地板
发表于 2015-7-5 17:10 |只看该作者
真是天底下好事多多  

Rank: 2

积分
136 
威望
136  
包包
1877  
7
发表于 2015-7-6 20:00 |只看该作者
知道了 不错~~~  

Rank: 2

积分
89 
威望
89  
包包
1794  
8
发表于 2015-8-2 19:42 |只看该作者
干细胞研究还要面向临床

Rank: 2

积分
118 
威望
118  
包包
1769  
9
发表于 2015-8-10 16:53 |只看该作者
风物长宜放眼量  

Rank: 2

积分
75 
威望
75  
包包
2193  
10
发表于 2015-9-8 12:39 |只看该作者
终于看完了~~~  
‹ 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, 2024-4-27 08:13

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.