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: 345164|回复: 240
go

Bone Marrow Transplantation Attenuates the Myopathic Phenotype of a Muscular Mou [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-4 23:58 |只看该作者 |倒序浏览 |打印
作者:Nouzha Salah-Mohellibia, Gaelle Milleta, Isabelle Andr-Schmutzb, Bndicte Desforgesa, Robert Olasoa, Natacha Roblota, Sabrina Courageota, Gilbert Bensimonc, Marina Cavazzana-Calvob, Judith Melkia作者单位:aMolecular Neurogenetics Laboratory, Institut National de la Sant et de la Recherche Mdicale, Inserm, U, Evry, F- France; University of Evry, Evry, F- France;
3 m- r$ M5 C( j' c" t2 }4 r, O3 g. K5 {                  
' k, m2 x9 B* a  c3 o7 {* H                  8 f6 T7 o; b; d3 O+ ~) U
         
) n; E; {4 c6 ]2 e3 b9 ?                        
! `' e$ T# l& K) {9 n+ O: Q' C- e            
; j; @# S  }# I            
5 _1 g4 s5 K8 b            
- q5 N( Q8 S4 T: g            
) u6 |& c* `7 }                      1 [/ r; R" C( T% f5 `
        , @8 v# T! Z1 _% Q
        ( O- }  Y, S5 i  d( |! k1 X) b( A
        
! D9 a$ Z: u; \          【摘要】+ b7 A4 T# a  b: r+ s# i2 e
      Bone marrow (BM) transplantation was performed on a muscular mouse model of spinal muscular atrophy that had been created by mutating the survival of motor neuron gene (Smn) in myofibers only. This model is characterized by a severe myopathy and progressive loss of muscle fibers leading to paralysis. Transplantation of wild-type BM cells following irradiation at a low dose (6 Gy) improved motor capacity ( 85%). This correlated with a normalization of myofiber number associated with a higher number of regenerating myofibers (1.6-fold increase) and an activation of CD34 and Pax7 satellite cells. However, BM cells had a very limited capacity to replace or fuse to mutant myofibers (2%). These data suggest that BM transplantation was able to attenuate the myopathic phenotype through an improvement of skeletal muscle regeneration of recipient mutant mice, a process likely mediated by a biological activity of BM-derived cells. This hypothesis was further supported by the capacity of muscle protein extracts from transplanted mutant mice to promote myoblast proliferation in vitro (1.6-fold increase). In addition, a tremendous upregulation of hepatocyte growth factor (HGF), which activates quiescent satellite cells, was found in skeletal muscle of transplanted mutants compared with nontransplanted mutants. Eventually, thanks to the Cre-loxP system, we show that BM-derived muscle cells were strong candidates harboring this biological activity. Taken together, our data suggest that a biological activity is likely involved in muscle regeneration improvement mediated by BM transplantation. HGF may represent an attractive paracrine mechanism to support this activity. $ h' C# p) g' {: B5 O/ ?; _
          【关键词】 Spinal muscular atrophy Survival of motor neuron Bone marrow Transplantation Skeletal muscle Hepatocyte growth factor
& t$ ^# e1 L& A                  INTRODUCTION
3 S- D0 S: K$ J' ~* b$ C& h. T3 }
Spinal muscular atrophy (SMA) is one of the most frequent genetic causes of death in childhood. SMA is a recessive autosomal disorder caused by mutations of the survival of motor neuron gene (SMN1) and characterized by degeneration of motor neurons associated with muscle paralysis .
1 U5 b  v' H9 w* x+ @
, q' s0 C. P* I' TThe most frequent mutation found in SMA patients is a homozygous deletion of SMN1 exon 7 .* V1 N  }( M- X  h8 R5 A$ w/ D

; M  @. @; r2 J" ~8 a+ RSeveral reports clearly demonstrate that bone marrow-derived cells (BMDCs) contribute to skeletal muscle fibers. Bone marrow transplantations (BMTs) using unfractionated marrow have been performed in mice and showed for the first time that bone marrow contains cells able to be recruited to a damaged muscle . These data have shown a limited capacity of BMDCs to replace damaged myofibers.
% r4 V" K/ m5 n7 i+ t' h
# a( A) D, ?' L8 KMultiple variables are involved in the incorporation of marrow cells to muscle fibers, including irradiation, donor-cell dose, and timing of muscle injury relative to the transplantation . To determine whether BMT might have any beneficial effects on a model exhibiting a severe myopathic phenotype, unfractionated bone marrow cells were transplanted into mild muscular mutant mice (HSA-Cre, SmnF7/F7).% p8 D6 [" o6 t3 ~' r% Z4 a, i" V; F
8 u5 M4 H. \+ l2 Y; H/ ]  l) m" s
MATERIALS AND METHODS& a1 F: L- w/ k$ I& j- @

! ~( l, T& H, g% R+ r! I( oMice
( ^3 l4 z# ^6 l9 b8 h! Q5 V- y+ J$ d
(HSA-Cre, SmnF7/F7) mutant mice were generated by crossing homozygous (SmnF7/F7) mice with those carrying (HSA-Cre, SmnF7/ ) genotype . GFP transgene was also placed on (HSA-Cre, SmnF7/F7) background, and (GFP, HSA-Cre, SmnF7/F7) mice were selected as donors in a second set of experiments (described in Results). All BMT experiments were performed on non-GFP transgenic mice. Animal procedures were performed in accordance with institutional guidelines (agreements A91-228-2 and 3429).% H+ M+ d. E8 J, T
; j9 C5 x; V+ X* H
Bone Marrow Transplantation
7 ~) _. L3 U6 d7 B4 Q% x9 ~2 S' ?1 f+ T! m9 ]  ~/ S7 d
Bone marrow was sterilely isolated from 6¨C10-week-old isogenic wild-type (Smn / ) or (HSA-Cre, SmnF7/F7) transgenic mice (H2b) that ubiquitously expressed enhanced GFP . Femur and tibia were surgically removed and placed in Dulbecco's modified Eagle's medium (DMEM) culture medium with 10% fetal calf serum. Marrow was collected, and red blood cells were lysed in 0.75% NH4Cl in 20 mM Tris, pH 7.2. To determine the rate of bone marrow (BM) cells expressing GFP, BM cells from three Smn /  GFP transgenic mice were labeled with anti-mouse monoclonal antibody CD45.2 (clone 106, 1:200; BD Biosciences, Palo Alto, CA, http://www.bdbiosciences.com), Gr-1 (clone RB6-8C5, 1:500; BD Biosciences), and Sca-1 (clone D7, 1:500; BD Pharmingen, San Diego, http://www.bdbiosciences.com/pharmingen), and labeled cells were analyzed together with GFP fluorescence using a FACSCalibur and CellQuest Software (BD Immunocytometry Systems, San Jose, CA, http://www.bdbiosciences.com). The rate of GFP  CD45 , GFP  Gr-1 , and GFP  Sca-1  was 44 ¡À 0.8%, 39 ¡À 0.3%, and 77 ¡À 1.5%, respectively. In bone marrow cells of GFP mutant mice (GFP, HSA-Cre, SmnF7/F7; n = 3), the rate of GFP  CD45 , GFP  Gr-1 , and GFP  Sca-1  was 34 ¡À 7%, 27 ¡À 6.5%, and 65 ¡À 8%, respectively. No statistically significant difference was observed between wild-type (Smn / ) and mutant GFP mice (HSA-Cre, SmnF7/F7; p > .15 for each comparison). For bone marrow transplantation, the cell pellet was resuspended in culture medium without fetal bovine serum and counted. Unfractionated BM cells (5 x 106) were intravenously injected in the retro-orbital plexus of anesthetized 2-month-old recipient mice within 6 hours following irradiation. Recipient mice were subjected to irradiation at 6 Gy with an x-ray source and maintained in pathogen-free conditions. That dose prevented irradiation-related death of control or mutant mice within the first month, indicating that this dose was not sublethal. One and 3 months after irradiation and BMT, peripheral blood was collected and tested for chimerism by measuring GFP  cells with a flow cytometer. Cells were labeled with biotin-conjugated anti-mouse monoclonal antibody CD45.2 followed by streptavidin-allophycocyanin (1:100; BD Biosciences) and R-phycoerythrin-conjugated rat anti-mouse anti-Gr-1. GFP fluorescence was measured at excitation and emission wavelengths of 489 and 508, respectively. Labeled cells were analyzed together with GFP fluorescence, and the rate of GFP  CD45  and GFP  Gr1  cells was determined.
( m# B' v2 K3 \2 @% E0 v5 z5 B0 }2 \+ O" ]0 Y- t4 k5 L
Histological and Immunofluorescence Experiments on Muscle Tissue* A6 X' t' l+ ]  B+ o9 K0 D
$ [) O! u; A1 E# k
Transverse sections (8¨C10 µm) of isopentane-frozen skeletal muscles including gastrocnemius and soleus of 8-month-old mice were stained with hematoxylin and eosin. To evaluate the total number of muscle fibers with or without central nuclei, serial 400-µm sections of the entire soleus were prepared. The highest number of myofibers per soleus muscle section was retained for statistical analysis. Double immunostaining of GFP and laminin was performed by using rabbit anti-GFP polyclonal (1:1,000; Molecular Probes Inc., Eugene, OR, http://probes.invitrogen.com) and rat monoclonal anti-laminin two antibodies (1:1,000; Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com). A mean of 1,000 myofibers per muscle was examined from each mouse group. Sections were mounted with Vectashield and 4,6-diamidino-2-phenylindole (DAPI) (Vector Laboratories, Burlingame, CA, http://www.vectorlabs.com) and observed under a Zeiss Axiophot fluorescence microscope (Carl Zeiss, Jena, Germany, http://www.zeiss.com).
! [! o5 m6 [0 l
1 T8 K$ r# h3 TSingle Muscle Fiber Isolation+ y9 K8 I8 s6 P4 o7 V  t+ ~
% r# d6 ~/ y% P  F2 M
Single muscle fibers were isolated from the extensor digitorum longus (EDL) muscle of 8-month-old mice according to Rosenblatt et al. . Antibodies were incubated in the following concentrations: monoclonal rat anti-CD34 antibody (1:200; BD Biosciences) or monoclonal mouse anti-Pax 7 (Developmental Studies Hybridoma Bank, Iowa City, IA, http://www.uiowa.edu/dshbwww). The nuclei were stained with DAPI. A total of 25 myofibers (representing around 1,000 nuclei) were examined per animal after CD34 or Pax7 immunolabeling. For GFP detection, 100 myofibers were examined per animal.2 A6 T, ?4 c3 @

- g; r1 g' U, vRotarod Test6 k4 \2 j( e+ o
. C, G0 G6 k4 R& p0 F/ x
Tests started at 2.5 months of age and were performed every 2 days. The protocol consisted in placing mice on a rod placed 20 cm above the floor of apparatus and rotating at 5 rpm (Bioseb, Chaville, France, http://www.bioseb.com). The test was stopped after an arbitrary limit of 7 minutes. Mice were scored either positive (able to maintain their balance for 7 minutes) or negative (fell before 7 minutes). In the case of a positive test at 5 rpm, rotating speed was increased to 10 rpm for 20 minutes. For each session, two trials were performed, with a resting interval of 1 minute. Results are given as time mice were able to maintain their balance at 5 rpm (factor 1) and 10 rpm (factor 2).+ b" c+ a& h1 s; f4 W

( ?# [* D* w! L2 _  X# f7 d* `Determination of Cre Recombinase Activity by DNA Analysis
* M8 ~  e. V8 S0 e9 Q, D; C( Y5 D
5 P6 E! g$ J2 G, I" ^Blood was collected by retro-orbital plexus puncture of anesthetized mice. Red blood cells were lysed in 0.75% NH4Cl. Cells were then incubated in extraction buffer supplemented with 0.1 mg/ml of proteinase K. Detection of the Smn7, wild-type and SmnF7 alleles, and Cre recombinase transgene was performed by PCR amplification as previously described .
2 }. q" h( Q. F6 M
+ ]0 o7 I; y, |5 t) F: hRNA Analysis& _" s% @- Q8 N  ^
' c9 `/ `: K, [  W; W
Total RNA was extracted from skeletal muscle using Trizol reagent (Invitrogen, Carlsbad, CA, http://www.invitrogen.com). cDNA was synthesized from total RNA by reverse transcriptase reaction. For Notch targets, primer sequences were as follows: HES-1, HES1F, 5'-CAA CAC GAC ACC GGA CAA AC-3'; HES1R, 5'-TCT TCT CCA TGA TAG GCT TTG ATG-3'; HEY-1, HEY1F, 5'-CTT GAG TTC GGC GCT GTG TTC C-3'; HEY1R, 5'-GAT GCC TCT CCG TCT TTT CCT-3'; HEY-L, HEYLF, 5'-CCC CTC ACC CTA CTC ACC A-3'; HEYLR, 5'-GCT TCA ACC CAG ACC CAA G-3'. For vascular endothelial growth factor a (VEGFa) transcripts, primer sequences were as follows: VEGFa-FOR1, 5'-AAG GAG AGC AGA AGT CCC ATG A-3'; VEGFa-REV1, 5'-AGC TTC GCT GGT AGA CAT CCA T-3'. For Cre recombinase transcripts, primer sequences were 5'-GCG GTC TGG CAG TAA AAA CTA TC-3' and 5'-GTG AAA CAG CAT TGC TGT CAC TT-3'. For hepatocyte growth factor (HGF) transcripts, primer sequences were as follows: HGF-FOR, 5'-GAG GTA CGC TAC GAA GTC TGT GA-3'; HGF-REV, 5'-GAT TCT GTG TGA TCC ATG GGA-3'. Real-time quantitative PCR was carried out using the SYBR-green master mix (Applied BioSystems, Foster City, CA, http://www.appliedbiosystems.com) and processed on an ABI Prism 7000 (Applied BioSystems). For each set of primers, first optimal primer concentration was determined and then a standard curve using increased dilution of cDNA was established in control tissues. Each reaction was performed in triplicate. Transcript level was normalized to aldolase (forward, 5'-TGAAGCGCTGCCAGTATGTTA-3'; reverse, 5'-GGTC-GCTCAGAGCCTTGTAGA-3') since there is no difference in aldolase transcript level between control and mutant skeletal muscle.3 {" {; I/ a$ p$ K2 ]

+ _  l' f2 j' d5 v& z3 m% C) eImmunoblotting Experiments
" ?% w( K3 y+ j1 {% H; V- l, {5 Z- n% _0 P' U) I
Frozen skeletal muscle (quadriceps) of 8-month-old mutant mice transplanted with wild-type bone marrow (wtBM) (n = 5) and nontransplanted mutants (n = 5) were crushed in liquid nitrogen, transferred into a buffer containing 25 mM sodium phosphate (pH 7.2), 5 mM EDTA, and 1% SDS supplemented with protease inhibitor cocktail (Sigma-Aldrich) then boiled. For HGF and actin immunodetection, anti-HGF (1:100; mouse anti-rat HGF antibody; Institute of Immunology, Tokyo, http://www.tokumen.co.jp/english/index.html) and monoclonal anti-actin antibodies (1:50,000; clone AC-40; Sigma-Aldrich) were used.
# V% Y* z. I: t6 Q9 E$ q7 y: M8 l) v2 T0 _7 @2 s
Myoblast Proliferation Assay8 K( L  \0 P$ Z$ \& }) h

& r" Q- y( D; E6 CPrimary muscle cultures were prepared from newborn wild-type mice (Smn / , 3¨C5 days old) as previously described . Quadriceps muscles of 8-month-old mice were isolated and crushed using a mortar and pestle. The muscles were incubated in cold PBS (500 µl/100 mg of tissue) for 2 hours with gentle shaking at  4¡ãC. The crushed muscle extracts were centrifuged at 10,000g for 10 minutes, and the supernatant was collected and filtered through a 0.2-µm filter. Proteins were extracted from skeletal muscle of mutant mice irradiated and transplanted with wild-type (n = 6) or mutant BM cells (n = 3) or irradiated but not transplanted (n = 4). The myoblast culture was exposed to 0 (PBS as mock), 2, 5, and 10 µg of protein extracts for 1 day. At least three independent experiments were performed. In each experiment, muscle protein extracts of mutant mice transplanted with wild-type BM were run in parallel with those of mutant mice irradiated and transplanted with mutant BM or nontransplanted.- `' J# c; Y/ H, K. n
: Y' G& U- P; M+ R
Statistical Analysis  d2 \) I( b3 `! a! r5 d. ^* {/ _4 v
3 K9 T( q6 ?& U; \
Statistical comparisons were performed using the t test for all experiments except for comparing the number of GFP-positive and GFP-negative muscle fibers in transplanted mutant and control mice (2; Statview, Alsyd, France, http://www.statview.com). Data are presented as mean ¡À SE values.
6 q: O7 k5 `' l# E2 o4 a; V$ `. A# P7 J
RESULTS
0 Q1 A( g& a0 ]: V6 e  }! I* k2 I0 M" {! v( q8 ]1 K
Transplantation of Wild-Type Bone Marrow Leads to Marked Improvement of Muscle Phenotype of Mutant Mice
3 H. m% Z; s' h* t: e* x; f7 L- K
+ g' b( t2 N, D1 X7 LMice receiving irradiation followed by transplantation were designated transplanted, those receiving irradiation without transplantation were designated nontransplanted, and those receiving neither irradiation nor transplantation were designated untreated (Table 1). Depending on whether BM cells were isolated from wild-type (Smn / ) or mutant mice (HSA-Cre, SmnF7/F7), BM was designated wtBM or mutant bone marrow (mutBM), respectively (Table 1). In the first set of experiments, wtBM was used as donor cells. Two-month-old mutant mice were transplanted with wtBM cells (5 x 106 cells; n = 39) from Smn /  GFP mice or nontransplanted (n = 15). Control mice of the same age (n = 10; SmnF7/ ) were transplanted using the same protocol. Hematopoietic chimerism was examined from blood samples collected 1 and 3 months after BMT. Fluorescence-activated cell sorting (FACS) analysis of GFP-positive (GFP ) cells, hematopoietic CD45  cells, or Gr-1  granulocytes was performed on transplanted mutant (n = 39) and control (n = 10) mice. One month after BMT, a mean of 55 ¡À 2.4% and 55.3 ¡À 5% of CD45  cells were GFP  in mutant and control mice, respectively. FACS analysis demonstrated that 3 months after BMT, long-term hematopoietic reconstitution was similar in mutant and control mice (61 ¡À 3% and 64 ¡À 6.5% of GFP  CD45  cells, respectively). Similar results were obtained for GFP  Gr-1  cells (data not shown).
0 |* s! Q2 M6 ~  D- _& h" d' Q0 U. N+ g
Table 1. Characteristics of mice used as donors and recipients& T$ D, |3 R- e6 E' e# L

! T; X; x2 V6 kGastrocnemius and soleus muscles from 8-month-old mutant and control mice transplanted with wtBM were examined for the presence of GFP-positive muscle fibers. Double immunolabeling experiments of GFP and laminin, a specific marker of the basal lamina, revealed 1.95 ¡À 0.1% of GFP  muscle fibers in the transplanted mutant mice (146 GFP  of 7,596 myofibers; six mice; Fig. 1). This proportion was higher in the mutant than in control mice (0.1 ¡À 0.01%; 5 GFP  of 5,039 myofibers; five mice; 2; p
+ v: a( B. s7 ]. ?4 t6 Q$ u4 H. S7 N' F" {% o0 U8 i
Figure 1. Green fluorescent protein (GFP) muscle fibers derived from bone marrow are present in transplanted mutant mice. Double immunofluorescent staining of GFP (red) and basal lamina (laminin, green) on transverse sections of gastrocnemius of 8-month-old control (A) and mutant (B) mice transplanted with wild-type bone marrow (wtBM) revealed GFP  myofibers (arrow). Nontransplanted mutant mouse was used as negative control (D). GFP  muscle fibers are also present in mutant mice transplanted with mutant bone marrow (mutBM) (C) (details in Results). Scale bar = 100 µm.$ D+ O7 N# T/ i" \0 f+ X
9 x! e7 ^5 A- h/ d3 s1 g
Figure 2. Skeletal muscle regeneration is improved in mutant mice transplanted with wild-type bone marrow. At low magnification (A¨CD), soleus muscle from 8-month-old control mice (A), nontransplanted mutant mice (B), and mutant mice transplanted with wild-type bone marrow (wtBM) (C) is framed by a dotted line. Note the larger size of the entire soleus of wtBM transplanted (C) compared with nontransplanted mutant mouse (B). At higher magnification (A'¨CD'), note the increased proportion of regenerating myofibers (with central nuclei, arrow) in mutant mouse transplanted with wtBM (C') compared with nontransplanted (B'). In mutant mouse transplanted with mutant bone marrow (mutBM) (D, D'), note the reduction in size of the entire soleus (D) and the lower proportion of regenerating myofibers (D') compared with mouse transplanted with wtBM (C'). Scale bar = 300 µm (A¨CD), 50 µm (A'¨CD').2 L( n* Z0 S! D3 O% B

! `; f: u' E: _Figure 3. Transplantation of wild-type but not mutant bone marrow cells improves the myopathic phenotype of Smn mutant mice. (A): Total number of regenerating myofibers in the entire soleus. Asterisk indicates statistically significant higher number of regenerating myofibers in mutant mice transplanted with wtBM (n = 7) compared with nontransplanted mutants (n = 5; p = .01). (B): Total number of mature myofibers in the entire soleus. Asterisk indicates statistically significant higher number of mature myofibers in control mice (p 2 _0 \1 g, N2 K- O
* V- C' k$ {% ~; w! f! j' p
Satellite cells were examined to determine whether the gain in muscle fibers derives solely from the activation of satellite cells of recipient mice or occurs in association with the differentiation of BMDCs into satellite cells. Immunolabeling of CD34 and Pax7, two specific markers of satellite cells . The proportion of satellite cells expressing CD34 or Pax7 was evaluated per 100 nuclei of myofibers (including nuclei of myofibers and satellite cells). The number of nuclei per myofiber surface unit was similar in wtBM-transplanted and nontransplanted mutant mice (data not shown). A significant increased proportion of CD34  and Pax7  satellite cells was observed in the transplanted mutant mice (4.5 ¡À 0.17% and 4.7 ¡À 0.24%, respectively) compared with the nontransplanted mutant mice (3 ¡À 0.3% and 3.3 ¡À 0.3%, respectively; p = .003 and p = .01, respectively; Figs. 3C, 4). When only isolated myofibers with central nuclei were analyzed, similar results were obtained (data not shown). No GFP  satellite cells were observed in a total of 100 muscle fibers per EDL from five transplanted mutant or control mice. These data strongly suggest that BMDCs do not differentiate into satellite cells in our model and demonstrate that the improvement in the muscle regeneration process correlated with an increased proportion of CD34  (1.5-fold increase) or Pax7  (1.4-fold increase) satellite cells, a process mediated by BMT.
3 n! G; \# K4 T* p( E/ M- y: E* z
Figure 4. Increased proportions of Pax7 and CD34 satellite cells on freshly isolated muscle fibers of mutant mice transplanted with wild-type but not mutant bone marrow. Immunolabeling experiments of CD34 (A¨CC) and Pax7 (D¨CF) were performed on isolated muscle fibers from extensor digitorum longus of nontransplanted mutant mice (A, A', D, D'), mutant mice transplanted with wild-type bone marrow (wtBM) (B, B', E, E'), and mutant mice transplanted with mutant bone marrow (mutBM) (C, C', F, F'). An increased proportion of CD34 or Pax7  satellite cells (arrows) was observed in mutant transplanted with wtBM compared with nontransplanted mutant mice or mutant mice transplanted with mutBM (see Fig. 3 for quantification). Satellite cell nuclei and myonuclei were stained with 4,6-diamidino-2-phenylindole (A', B', C', D', E', F'). Scale bar = 50 µm.% g9 z, x% A  @9 S& ]

8 @+ W, M: l; R: d. f  G/ R. BTo determine whether the increase in myofibers found in transplanted mutant mice has a beneficial effect on motor behavior, motor performance was evaluated using the rotarod test. From 3 to 5 months of age, similar motor activity profiles were observed in the wtBM transplanted mutant mice (n = 16), untreated mutant mice (n = 10), and wtBM transplanted control mice (n = 4; Fig. 3D). However, from 6 to 9 months of age, a dramatic decline of motor capacity was observed in the untreated mutant mice (from 46% to 77% reduction; n = 10), whereas transplanted mutants showed sustained activity (n = 15; p = .017; Fig. 3D). These results indicate that the motor defect was significantly attenuated following transplantation. In addition, these data suggest that the high proportion of regenerating myofibers is capable of sustaining motor function recovery.
/ w0 v$ }$ p, M5 L5 Q' s/ T. I' a4 B
Skeletal muscle improvement of transplanted mutant mice was not caused by a change in Cre-mediated deletion of Smn in skeletal muscle. Indeed, the quantification of transcripts of the Cre recombinase transgene in skeletal muscle did not reveal any differences among transplanted, nontransplanted, and untreated mutant mice. These data indicate that the improvement of the muscular phenotype cannot be ascribed to changes in Cre-mediated deletion of Smn (supplemental material).
, y9 j5 j# ~  C; V8 x: H6 K3 p; L4 o8 Q
Activation of Skeletal Muscle Regeneration Is Mediated by a Biological Activity of Bone Marrow-Derived Cells
( o. w8 E/ J7 R( E  @5 E/ E8 X! v2 A+ o" d. z/ I+ Q& v
Surprisingly, wild-type bone marrow transplantation of mutant mice revealed a low proportion of GFP  muscle fibers (2%), which contrasted with a significant increase in the number of regenerating myofibers (1.6-fold increase). These data indicate that BM cells had a very limited capacity to replace or fuse to mutant myofibers (2%), in agreement with previous reports. Nevertheless, wtBM transplantation was able to attenuate the myopathic phenotype of mutant recipient mice through an improvement of skeletal muscle regeneration. These results suggested an activation of muscle regeneration mediated by a biological activity of the BMDCs.
0 s) c' p% ]  ^9 \1 u0 F* T! h4 D+ k, Q2 Q4 @& S
To test this hypothesis, a myoblast proliferation assay was performed. Proteins were extracted from quadriceps muscle of 8-month-old mutant mice transplanted with wtBM (six mice) and nontransplanted mutant mice (four mice) and added to the medium of primary cultures of wild-type myoblasts (Smn / ) 2 days after purification. Myoblast proliferation was evaluated by comparing the number of myoblasts before (day 2) and after a 1-day incubation with protein extracts (day 3). We found a 5.3-fold increase in the number of myoblasts in the presence of 10 µg of protein extracts from mutant mice transplanted with wtBM compared with a 3.5- or 2.7-fold change using nontransplanted muscle protein extracts or mock, respectively (p
0 ?/ I. N; g" w$ j. S- o) B+ b' y5 V1 u  j1 P" T; _( ?  ]
Figure 5. Evidence for a biological activity of bone marrow-derived cells on myoblast proliferation. Myoblast proliferation was evaluated as the ratio of wild-type myoblast number at day 3 (1 day after incubation with the protein extracts or mock) to that at day 2 (before incubation). The same fields were examined at both stages. Zero corresponds to the mock (phosphate-buffered saline without protein extract). Student's t test revealed statistically significant higher myoblast proliferation (indicated by *) in the presence of muscle extracts from mutant mice transplanted with wtBM (six mice) compared with nontransplanted (four mice) or mutant mice transplanted with mutBM (three mice). Note the dose effect of protein extracts from mutant mice transplanted with wtBM (0¨C10 µg) on myoblast proliferation ratio. Asterisks indicate p value : Z6 P0 g! R+ a$ q

& h& L) N1 {) u8 G" J  i* A8 WWild-Type BM-Derived Muscle Cells Are Strong Candidate Cells to Activate Skeletal Muscle Regeneration+ d$ `/ n  F; p
* U0 B8 j  r4 }
In the above experiments, we compared wtBM transplanted and nontransplanted mutant mice and showed a beneficial effect of transplantation. This effect could come from either circulating blood cells or muscle cells derived from bone marrow. Since the Cre recombinase expression was restricted to differentiated myotubes only , no genotype difference at the Smn locus was expected between circulating blood cells of untreated and wtBM transplanted mutant mice (Table 1). To exclude the possibility of an ectopic expression of Cre recombinase in the blood cells of untreated mutant mice, deletion of Smn exon 7 was examined in genomic DNA extracted from the circulating blood cells. No deletion of Smn exon 7 was observed, indicating that the benefit of transplantation on mutant phenotype could not be mediated by the circulating blood cells (data not shown). These data suggested that muscle cells derived from wild-type bone marrow might be regarded as candidate cells.& ]' s+ F* {* G- @6 C

3 K4 _0 K4 n& gTo evaluate the role of BM-derived muscle cells in the muscle improvement of recipient mutant mice, mutant mice were transplanted with mutant bone marrow. Mutant bone marrow cells carry the (HSA-Cre, SmnF7/F7) genotype, and Cre-mediated deletion of Smn exon 7 will only occur in differentiated muscle fibers. Therefore, in BM-derived circulating blood cells, the Smn locus (SmnF7/F7) remains intact since the Cre recombinase is not expressed. In contrast, in BM-derived muscle cells, the Cre recombinase is expressed, leading to the deletion of Smn (Smn7/7; Table 1). The GFP transgene was transferred into a (HSA-Cre, SmnF7/F7) background through successive backcrosses, and (GFP, HSA-Cre, SmnF7/F7) mice were selected as donors. Transplantation was performed on irradiated 2-month-old non-GFP mutant mice (HSA-Cre, SmnF7/F7; n = 11) using the same protocol. Hematopoietic reconstitution of the mutant mice transplanted with mutBM (65 ¡À 8% and 61 ¡À 6% of GFP CD45  cells, 1 and 3 months after transplantation, respectively) was similar to mutant mice transplanted with wtBM. Immunofluorescent labeling of GFP on transverse muscle sections revealed 1 ¡À 0.1% of GFP  muscle fibers in 8-month-old mutant mice transplanted with mutBM (47 GFP  of 4,579; five mice; Fig. 1). Importantly, the total number of regenerating muscle fibers in the entire soleus (246 ¡À 27) as well as the proportion of CD34  (2.4 ¡À 0.1%) and Pax7  (2.6 ¡À 0.2%) satellite cells per 100 myofiber nuclei were markedly reduced in 8-month-old mutant mice transplanted with mutBM (five mice) compared with mutant mice transplanted with wtBM (p
5 r" g) x- f7 c/ c: \) L0 i: k3 S0 ], z& _5 I
Moreover, myoblast proliferation assay was performed and did not reveal any significant increased myoblast proliferation after a 1-day incubation with 10 µg of protein extracts from mutant mice transplanted with mutBM (3.5-fold change) compared with nontransplanted mutant mice or mock (3.4- and 2.7-fold change, respectively; Fig. 5)./ x0 z7 z# S. J) i# M/ Y
( X/ p4 n$ e" ]! k3 b; K
These data strongly suggest that activation of myoblast proliferation was mediated by a biological activity of skeletal muscle cells from mutant mice transplanted with wild-type but not mutant bone marrow. Several candidate pathways could account for this activation and were tested using real-time PCR analysis of transcripts normalized to aldolase. The Notch signaling pathway has been shown previously to mediate activation of muscle regeneration , revealed dramatic changes. A tremendously increased expression of HGF was observed in skeletal muscle of 8-month-old mutant mice transplanted with wtBM (1.5 ¡À 0.08; n = 5) compared with mutBM (0.45 ¡À 0.09; n = 5) or nontransplanted mutants (0.045 ¡À 0.007; n = 5, p
' Q3 j: Q' ?: }2 t/ ?3 k
+ ^1 j: ^- u& g/ ZFigure 6. Expression analysis of HGF, Notch targets, and vascular endothelial growth factor a (VEGFa). (A): Real-time polymerase chain reaction (PCR) amplification analysis of transcripts encoding HGF in skeletal muscle of nontransplanted mutant mice (n = 5), mutant mice transplanted with wild-type bone marrow (wtBM) (n = 5), mutant mice transplanted with mutant bone marrow (mutBM) (n = 5), and untreated mutant (n = 5). Asterisks indicate statistically significant changes (p
8 U; X. h% P7 z' C0 U, S7 V- r. W* c% O, K5 ~$ u' _. q$ K# J) f0 g
DISCUSSION
! V5 C1 K/ Z/ @1 l: o% S
5 J$ X: u* S5 l+ E2 w: X8 u/ ?Here, we provide strong evidence that the transplantation of wild-type bone marrow cells improves important aspects of the myopathic phenotype of Smn muscular mutant mice, including normalization of myofiber number and motor performance. These beneficial effects tightly correlate with the activation of skeletal muscle regeneration and a higher proportion of CD34 and Pax7 satellite cells. The remarkable attenuation of mutant phenotype contrasts with low recruitment of BMDCs into myofibers, as previously reported in other models .
; K* D9 j. x, D* y% `! K
5 b0 t; N( o" Y) U/ ]4 x$ }The biological activity of BM-derived cells was further suggested by the in vitro myoblast proliferation assay showing an enhanced activation of myoblast proliferation in the presence of proteins extracts from skeletal muscle of mutant mice transplanted with wtBM but not with mutBM. Importantly, a tremendous upregulation of the HGF gene was found in skeletal muscle of mutant mice transplanted with wtBM compared with mice transplanted with mutBM or nontransplanted mutant mice. HGF, a ligand for the c-met protooncogene product, has emerged as an important candidate molecule in muscle regeneration and has been shown to act in a paracrine manner. The c-met receptor is present on quiescent satellite cells in skeletal muscle, and HGF has the ability to activate satellite cells .
3 y$ ~& F& l! B7 o' Y6 O3 w2 U5 r/ b& u5 y- h$ d
Thanks to the Cre-loxP system, we were able to exclude the BM-derived circulating blood cells as candidate to harbor this biological activity. Therefore, no difference exists at the Smn locus in circulating blood cells of wild-type or (HSA-Cre, SmnF7/F7) mice. In contrast, the transplantation of BM cells derived from wild-type (Smn / ) but not mutant (HSA-Cre, SmnF7/F7) donors provides wild-type Smn alleles to deficient pre-existing myofibers or is able to form new wild-type myofibers. Even at a low percentage, wtBM-derived myofibers can be regarded as candidate cells able to activate muscle regeneration through a biological activity. We cannot exclude other candidates, such as cells of muscle lineage, homing in skeletal muscle and coming from bone marrow. Additional experiments should contribute to determining the fraction of bone marrow-derived cells homing in skeletal muscle and participating in the upregulation of HGF. Although the nature of this biological activity still remains to be characterized, these results provide strong evidence that this activity may attenuate a myopathic phenotype by delaying or preventing myofiber loss.( r$ j( W5 \8 Q: T" n7 J
" o$ `% \. h9 e5 `
It has been shown that the proportion of muscle fibers incorporating adult BMDCs depends on the type of skeletal muscles or physiological stress applied to muscles  should be investigated to determine whether the recruitment of BMDCs into skeletal muscle may be correlated with the severity of myofiber loss of recipient mice. Higher contribution of BMDCs to myofibers using physiological stress might allow stronger beneficial effects. Elucidating the nature of BMDCs able to fuse to or to differentiate into myofibers on one hand and the pathway underlying HGF gene upregulation, which may represent an attractive paracrine mechanism to support this biological activity, on the other hand should be helpful in identifying new means to improve skeletal muscle regeneration.
5 @' |0 I( ?4 D
+ _' f- l# {0 ODISCLOSURES
" h4 ]9 V* D- I
' h. e  c/ ?* T& bThe authors indicate no potential conflicts of interest.
9 @* J4 e5 N) _$ E- O* m2 y. R. i6 X$ |. X" M
ACKNOWLEDGMENTS
4 ?5 _! ?* y0 V3 j1 ]0 t9 ?0 t- |# h! i
This work was supported by Institut National de la Sant¨¦ et de la Recherche M¨¦dicale, the Association Française contre les Myopathies, Families of Spinal Muscular Atrophy (USA), the Conseil R¨¦gional d'Ile de France, and the Fondation Bettencourt Schueller. We are very grateful to C. Collins for helpful suggestions in isolating myofibers, A. Joutel for advice on investigation of the Notch pathway, and E. Tournier-Lasserve and A. Fischer for fruitful comments. We thank P. Ardouin and A. Rouches (Institut Gustave Roussy) for assistance in the irradiation process and M. Okabe for providing us with GFP transgenic mice.
) \& C( ?# [* |5 i          【参考文献】
3 {4 y+ B2 _% C: {2 W  y) V
) f3 `4 Y+ C; ], I: l& ]+ b& `+ M8 g! E
Lefebvre S, Burglen L, Reboullet S et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 1995;80:155¨C165.6 F; q% T  |; @: b  K

+ }% n7 F' W& |2 H1 |, xFrugier T, Nicole S, Cifuentes-Diaz C et al. The molecular bases of spinal muscular atrophy. Curr Opin Genet Dev 2002;12:294¨C298.
8 @4 V. ~; y) F. Q0 r
# G' D$ S: J9 P. l# s* ULiu Q, Dreyfuss G. A novel nuclear structure containing the survival of motor neurons protein. EMBO J 1996;15:3555¨C3565.
2 B+ @0 c  u" |- V' q) h% O% f5 _4 _* g9 A6 d
Yong J, Wan L, Dreyfuss G. Why do cells need an assembly machine for RNA-protein complexes? Trends Cell Biol 2004;14:226¨C232." L2 h0 N& B  f# Z- x
, U7 I9 W# F  x( P: T5 D) Y4 b
Jones KW, Gorzynski K, Hales CM et al. Direct interaction of the spinal muscular atrophy disease protein SMN with the small nucleolar RNA-associated protein fibrillarin. J Biol Chem 2001;276:38645¨C38651.; \. q  O1 n0 @$ V/ i
3 B& K( V8 F7 y, c6 A! Z
Pellizzoni L, Kataoka N, Charroux B et al. A novel function for SMN, the spinal muscular atrophy disease gene product, in pre-mRNA splicing. Cell 1998;95:615¨C624.3 |$ t' {5 o# G8 O: }

; e6 R$ h" i9 }  S( Z2 CPellizzoni L, Charroux B, Rappsilber J et al. A functional interaction between the survival motor neuron complex and RNA polymerase II. J Cell Biol 2001;152:75¨C85.
& \( o1 l' Q# ?. ~" W
  N$ ^% L  u* j7 B1 W) E; r( ^Cifuentes-Diaz C, Frugier T, Tiziano FD et al. Deletion of murine SMN exon 7 directed to skeletal muscle leads to severe muscular dystrophy. J Cell Biol 2001;152:1107¨C1114.
5 i0 |, r9 G7 f- V; I
' L3 q0 C( A. p$ SCifuentes-Diaz C, Nicole S, Velasco ME et al. Neurofilament accumulation at the motor endplate and lack of axonal sprouting in a spinal muscular atrophy mouse model. Hum Mol Genet 2002;11:1439¨C1447.
; h: ^6 Z; a: n. Q! U; a. z0 Y; ~' Y( ?3 V
Frugier T, Tiziano FD, Cifuentes-Diaz C et al. Nuclear targeting defect of SMN lacking the C-terminus in a mouse model of spinal muscular atrophy. Hum Mol Genet 2000;9:849¨C858.
' {7 i5 z0 T" F) I
" N- J' h4 H3 ]/ n5 uNicole S, Desforges B, Millet G et al. Intact satellite cells lead to remarkable protection against Smn gene defect in differentiated skeletal muscle. J Cell Biol 2003;161:571¨C582.
: F& L6 w* x5 e9 c+ y4 p0 H/ E/ ]4 G+ u3 o
Miniou P, Tiziano D, Frugier T et al. Gene targeting restricted to mouse striated muscle lineage. Nucleic Acids Res 1999;27:e27.5 d5 w4 h! @, i
1 f$ K1 M( g! K/ f; _  e# ?
Vitte JM, Davoult B, Roblot N et al. Deletion of murine Smn exon 7 directed to liver leads to severe defect of liver development associated with iron overload. Am J Pathol 2004;165:1731¨C1741.( ]  S$ ~4 S! q2 O  H  P! B" u: C8 M

, G) d. A/ I+ _1 z- j1 pFerrari G, Cusella-De Angelis G, Coletta M et al. Muscle regeneration by bone marrow-derived myogenic progenitors. Science 1998;279:1528¨C1530.( x7 V/ t! t# c8 N( T7 s4 |0 K& q

5 c) q% X/ _6 F% @6 W7 x; nGussoni E, Soneoka Y, Strickland CD et al. Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 1999;401:390¨C394.  F8 _$ H& g* V" {% G4 Y8 O3 |

7 ^6 i9 e; W: X) R3 p/ HFerrari G, Stornaiuolo A, Mavilio F. Failure to correct murine muscular dystrophy. Nature 2001;411:1014¨C1015.
: O# s5 R* _) o$ H$ Z3 N3 [6 I
9 R: Z' ?$ H: B- n  V" WCamargo FD, Green R, Capetanaki Y et al. Single hematopoietic stem cells generate skeletal muscle through myeloid intermediates. Nat Med 2000;9:1520¨C1527.: b  g9 Z9 B1 ~, O- V
( y5 Z, G! B: c* S
Wang X, Willenbring H, Akkari Y et al. Cell fusion is the principal source of bone-marrow-derived hepatocytes. Nature 2003;422:897¨C901.
0 ]* G4 Z( q4 Y$ T2 B: p, o. Z4 y
* e3 a3 {6 v4 f/ Y4 K5 B; T( NWeimann JM, Johansson CB, Trejo A et al. Stable reprogrammed heterokaryons form spontaneously in Purkinje neurons after bone marrow transplant. Nat Cell Biol 2003;5:959¨C966.
! N& a' N5 r6 v, X# Y+ q$ P% `- W  J1 d, N1 ^1 i! \# w
Doyonnas R, LaBarge MA, Sacco A et al. Hematopoietic contribution to skeletal muscle regeneration by myelomonocytic precursors. Proc Natl Acad Sci USA 2004;101:13507¨C13512.1 N3 A5 b' C% T; h. @3 A3 d# N
% Y' K6 Y0 Q, m
Abedi M, Greer DA, Foster BM et al. Critical variables in the conversion of marrow cells to skeletal muscle. Blood 2005;106:1488¨C1494.
: n/ i$ V( ^$ X0 y. p- z' q% a3 J
+ |* k: W0 h* c) o& QBrazelton TR, Nystrom M, Blau HM. Significant differences among skeletal muscles in the incorporation of bone marrow-derived cells. Dev Biol 2003;262:64¨C74.
* u7 @2 R( T  j( \/ F$ @% w6 t1 j4 d8 x' G( o5 z* @
Palermo AT, Labarge MA, Doyonnas R et al. Bone marrow contribution to skeletal muscle: A physiological response to stress. Dev Biol 2005;279:336¨C344.' F) g  \0 A9 C' C( X! s1 z( x  L

& f2 S& S9 t( K! @Carnwath JW, Shotton DM. Muscular dystrophy in the mdx mouse: Histopathology of the soleus and extensor digitorum longus muscles. J Neurol Sci 1987;80:39¨C54.. ~! m" h# R7 C4 Q( j9 r$ W- l
4 J+ ?% @0 V# h4 _: @# ]
Okabe M, Ikawa M, Kominami K et al. ¡®Green mice¡¯ as a source of ubiquitous green cells. FEBS Lett 1997;407:313¨C319.2 N( Q) N4 N- `4 ^
( V3 K* A; k6 Z. u' T; c+ @  d
Rosenblatt JD, Lunt AI, Parry DJ et al. Culturing satellite cells from living single muscle fiber explants. In Vitro Cell Dev Biol Anim 1995;31:773¨C779.
  N; N3 k! ^5 V+ U5 X* M8 i- n# ~+ @) v3 P% y9 o% g7 t
Beauchamp JR, Heslop L, Yu DS et al. Expression of CD34 and Myf5 defines the majority of quiescent adult skeletal muscle satellite cells. J Cell Biol 2000;151:1221¨C1234.
  Z* z3 d# |: Y* p$ h% V. B" l- c4 M( x! ?3 u- }* w
Conboy IM, Conboy MJ, Smythe GM et al. Notch-mediated restoration of regenerative potential to aged muscle. Science 2003;302:1575¨C1577.
& v9 ~  _# ^" V$ C* X8 |' Q' h) z+ `; ~; C
Iso T, Kedes L, Hamamori Y. HES and HERP families: Multiple effectors of the Notch signaling pathway. J Cell Physiol 2003;194:237¨C255.. {7 B6 o$ M4 i. j3 F2 L0 j, x; c

% y6 ^5 W/ o6 M2 |& z$ s9 JAllen RE, Sheehan SM, Taylor RG et al. Hepatocyte growth factor activates quiescent skeletal muscle satellite cells in vitro. J Cell Physiol 1995;165:307¨C312.
; ^4 _/ l2 x7 k% w: c' W9 V0 P/ u" n* w; m' l7 {* K+ V5 T7 \: b
Bischoff R. Chemotaxis of skeletal muscle satellite cells. Dev Dyn 1997;208:505¨C515.3 u; V- h; }( H- T% {+ w

' Q+ ^* ~, L# n3 Y& [Tatsumi R, Anderson JE, Nevoret CJ et al. HGF/SF is present in normal adult skeletal muscle and is capable of activating satellite cells. Dev Biol 1998;194:114¨C128.
9 F: D6 s+ b  C+ x( j% N; R) A$ f" {1 P
Wagers AJ, Weissman IL. Plasticity of adult stem cells. Cell 2004;116:639¨C648.! Z* V  I5 a% g/ A  _0 ^$ q* t5 k
6 @+ p2 o6 ^# p
Izumida Y, Aoki T, Yasuda D et al. Hepatocyte growth factor is constitutively produced by donor-derived bone marrow cells and promotes regeneration of pancreatic beta-cells. Biochem Biophys Res Commun 2005;333:273¨C282.& T9 v8 X: v+ O# J5 x% L
# g' ?/ U: l' ^# L$ k" o
Ebens A, Brose K, Leonardo ED et al. Hepatocyte growth factor/scatter factor is an axonal chemoattractant and a neurotrophic factor for spinal motor neurons. Neuron 1996;17:1157¨C1172.

Rank: 2

积分
64 
威望
64  
包包
1769  
沙发
发表于 2015-5-31 14:32 |只看该作者
给我一个女人,我可以创造一个民族;给我一瓶酒,我可以带领他们征服全世界 。。。。。。。。。  

Rank: 2

积分
72 
威望
72  
包包
1942  
藤椅
发表于 2015-6-15 19:34 |只看该作者
生殖干细胞

Rank: 2

积分
73 
威望
73  
包包
1833  
板凳
发表于 2015-7-7 14:42 |只看该作者
干细胞之家微信公众号
必须顶  

Rank: 2

积分
161 
威望
161  
包包
1862  
报纸
发表于 2015-7-31 17:12 |只看该作者
围观来了哦  

Rank: 2

积分
72 
威望
72  
包包
1859  
地板
发表于 2015-8-4 14:43 |只看该作者
先看看怎么样!  

Rank: 2

积分
64 
威望
64  
包包
1782  
7
发表于 2015-8-7 10:18 |只看该作者
昨晚多几分钟的准备,今天少几小时的麻烦。  

Rank: 2

积分
56 
威望
56  
包包
1853  
8
发表于 2015-8-22 19:29 |只看该作者
免疫细胞治疗  

Rank: 2

积分
66 
威望
66  
包包
1790  
9
发表于 2015-8-25 21:14 |只看该作者
看贴回复是好习惯  

Rank: 2

积分
69 
威望
69  
包包
1788  
10
发表于 2015-9-3 12:35 |只看该作者
顶你一下,好贴要顶!  
‹ 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, 2024-5-5 22:26

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.