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: 410457|回复: 234
go

Cartilage-Derived Stromal Cells: Is It a Novel Cell Resource for Cell Therapy to [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 00:11 |只看该作者 |倒序浏览 |打印
作者:Wenjun Sua, Hao Zhanga, Zhuqing Jiab, Chunyan Zhoub, Yingjie Weia, Shengshou Hua作者单位:a Research Center for Cardiovascular Regenerative Medicine, Ministry of Health, and Department of Cardiovascular Surgery, Cardiovascular Institute and Fu-Wai Hospital, CAMS and PUMC;b Stem Cell Center, Beijing University, Beijing, Peoples Republic of China
* @* {" A, p, }4 Y* @                  
7 D1 ?! I8 K( r9 R& N! @                  1 ^; J7 g: U3 t- b: t
         
& C! r: B# p' `" h                         ; d: F5 x  _, T7 D9 }" A4 X! ^
            7 Y' D# i0 S3 P1 a- o1 x  _
            
# B- R- r, ^4 J) t; D            
: d0 Z+ b$ [1 E" V) L. K            ' @( ?0 ]! D7 `- f
                      3 N1 Z) q# Q% B: |, u/ P. W
        7 X. e3 T2 w! C
        3 |. y5 I( Y. `. Z+ O, Y
        8 x! P! i2 c+ B  w2 l, G
          【摘要】6 |% y( _# s0 d# S& s9 G
      Human cartilage is reported to contain multipotent stromal cells. We evaluated the effect of human cartilage-derived stromal cells (CDSCs) on heart function when transplanted into the infarcted myocardium of rats. CDSCs were isolated and cultured from human articular cartilage and subjected to fluorescence-activated cell sorting (FACS) analysis. The CDSCs were consistently negative for CD14, CD34, CD38, CD45, CD49f, CD104, CD105, CD106, CD117, HLA-DR, and ABCG-2, and positive for CD10, CD44, CD71, CD73, CD90, CD147, and HLA-A, -B, and -C by FACS analysis. Myocardial infarction (MI) was created in rats by ligation of the left anterior descending artery. Three weeks after MI, the CDSCs labeled with Hoechst stain were injected into the infarct and border zone. Echocardiography, histological examination, and reverse transcription-polymerase chain reaction (RT-PCR) were performed 4 weeks after cell transplantation. Echocardiography indicated that CDSC transplantation could improve heart function. The number of capillaries increased in the injection regions in the transplantation group. Histological examination showed that Hoechst-labeled CDSCs in islands within the infarcted region were stained positively for desmin and smooth muscle actin but negatively for alpha-sarcomeric actin and troponin-I. RT-PCR results indicated the expression level of collagen I, collagen III, tissue inhibitor of metalloproteinase-1, transforming growth factor-ß1, and vascular endothelia growth factor were much higher in the scar tissue in the transplantation group than in the medium and control groups. Our findings suggested that CDSCs might promote angiogenesis, prevent left ventricular remodeling, and improve the heart function when transplanted into injured heart in the rat model of myocardial infarction. 0 g% Q9 J( [2 R$ ]  O! F& M5 p; A; F
          【关键词】 Myocardial infarction Cartilage Stromal cells Cell transplantation" I7 ~6 a" Y$ H: ?% y$ y
                  INTRODUCTION
5 d* o+ D9 F3 C, f* ~8 h
8 e" `+ F" `; M* P& l$ J- BThe adult heart lacks the potential of effective regeneration. Permanent loss of cardiomyocytes and nonfunctional fibrous scar formation after myocardial infarction (MI) result in irreversible damage of cardiac function, and the subsequent remodeling process leads to expansion of the initial infarct area and dilation of the left ventricular lumen . It has not yet been proved that human cartilage-derived stromal cells (CD-SCs) could improve the heart function when transplanted into the injured heart. We transplanted CDSCs into the infarct and border zone of injured heart in rat animal model to evaluate the possibility of cell differentiation and the influence on angiogenesis, left ventricle (LV) remodeling, and heart function.
2 `$ d" ^7 |' U) D% c' I) i; o# r" W( ^
MATERIALS AND METHODS7 e  ~2 S. {0 ^, s' Y

; Z7 H0 j$ Z% t( H7 PIsolation and Culture of Human CDSCs
! d: V. O8 S( B' @) h* `" [6 k  U" T6 ?% P. o3 a
Cells were isolated from articular cartilage of 12- to 20-week-old spontaneously aborted fetuses as previously described . The Ethical Committee of Peking University Health Science Center and CAMS and PUMC granted permissible use of human tissue. Articular cartilage from glenohumeral, humeroulnar, and knee joints was excised and thoroughly minced. CDSCs were separated by incubation with 0.2% collagenase II for 2 hours with stirring and cultured in Iscove¡¯s modified Dulbecco¡¯s medium supplemented with 100 U/ml penicillin, 100 µg/ml streptomycin, 1.25 µg/ml amphorectin B, 2 mM L-glutamine, and 10% fetal calf serum. Cultured cells were seeded at a density of 1 x 105 cells/cm2 and incubated at 37¡ãC with 5% humidified carbon dioxide for 24 hours, nonadherent debris was removed, and adherent cells were cultured at a density of 5 x 103/cm2, with replacement of the medium every 3 to 4 days. When the cells had grown to 80% confluence, they were harvested by trypsinization (0.25% trypsin-EDTA) for 5 minutes at 37¡ãC. Passaging of the cultures (1:3 split) was carried out.
9 y4 r& X  t3 s3 e" P! o1 |" Y' u! y6 q+ ?
Fluorescence-Activated Cell Sorting
" P: D! k/ K% d) z! I8 O$ \$ A1 k- p1 Q% M' H! R6 D# `
Fluorescence-activated cell sorting (FACS) detection of CDSCs (at passages 3 through 10) was performed according to the procedure of FACS staining described previously . Briefly, cells were detached from the cell culture dish with 0.25% trypsin-EDTA and stained with the following mouse anti-human monoclonal antibodies: fluorescein isothiocyanate (FITC)-labeled antibodies against CD14, CD34, CD45, CD49f, CD90, CD105, CD106, CD 147, HLA-DR, HLA-A, -B, and -C, and phycoerythrin (PE)-labeled antibodies against CD10, CD38, CD44, CD71, CD73, CD104, CD117, and ABCG-2. After staining, cells were washed using 1x phosphate-buffered saline (PBS) and then resuspended in 1 ml of PBS for FACS flow cytometry (Becton, Dickinson and Company, Franklin Lakes, NJ, http://www.bd.com) analysis.
) E4 F6 {. a# M  R: f
% c3 s4 V, h9 ~% L( l, j" o# VAnimals and Myocardial Infarction
8 p: G# j" n3 h1 Z7 o* {
% I( P) c; ^- ~Experiments were performed in 8-week-old male Sprague-Dawley rats with an initial body weight of approximately 280 g (n = 90). All procedures were approved by the Animal Care Committee of the Cardiovascular Institute and Fu-Wai Hospital and performed according to the Guide for the Care and Use of Laboratory Animals prepared by the Institutes of Laboratory Animal Resources, National Research Council, and published by the National Academy Press, revised 1996 (NIH publication No. 85-23). Under general anesthesia induced with a ventilation mask with 4%¨C5% isoflurane and oxygen at 2 to 3 L/min, adult rats were oral-tracheal intubated with an 18-gauge intravenous catheter, and positive pressure ventilation (200 ml/min) was maintained with 2%¨C3% isoflurane in oxygen using a Harvard ventilator (model 683, Harvard Apparatus, Holliston, MA, http://www.harvardapparatus.com) . Successful performance of coronary occlusion is verified by observation of the development of pale color in the distal myocardium and the ventricular arrhythmia in the electrocardiographic monitor after ligation. The muscle layer and skin incision were closed with 3¨C0 silk sutures, and penicillin G procaine (150,000 U/ml) was given intramuscularly (0.4 ml per rat). Three weeks after MI, the survived rats with left ventricular ejection fraction (LVEF) less than 60% in ultrasonic assessment were randomized into three groups¡ªCDSC group (n = 23), medium group (n = 22), and control group (n = 23).
/ Z* t  H( m3 A: ^% I$ E
. Q( H# y* W! [  R9 H6 p: U: HCDSC Preparation and Transplantation
* E3 {! Z; c1 X- J) g
; f" d! A" }; j9 S$ }! LBefore cell transplantation, CDSCs at the fifth passage were harvested and stained with 5 µg/ml Hoechst 33342 (Sigma-Aldrich, St. Louis, http://www.sigmaaldrich.com) for 90 minutes at 37¡ãC as described previously . Control rats only underwent the procedure of thoracotomy with the injection of 50 µl of saline (n = 23, control group). After cell transplantation, cyclosporine was administered subcutaneously (10 mg/kg) every day in both CDSC and medium groups for immunosuppression.
  P) ^! g9 d% q8 L0 n" X! D. A6 F, q2 B! \' J2 \" G. b9 h' t
Evaluation of Left Ventricular Function+ B+ H. k, ^# Q- D: U

8 T+ {. B) _; ~Transthoracic echocardiography was performed 3 weeks after MI (as baseline, before cell transplantation) and 4 weeks after cell transplantation according to the protocols described previously . The animals were anesthetized with a ventilation mask with 4%¨C5% isoflurane during the procedure. Under anesthesia, the chest was shaved and a layer of acoustic coupling gel was applied. A commercially available echocardiographic system equipped with a 12-MHZ probe (Philip 5500; Philip Corp., Beijing, http://www.philips.com.cn) was used in the measurement. Initially, a two-dimensional short-axis view of the LV was obtained at the level of the papillary muscles. After optimizing gain settings and ensuring that the image was on axis, M-mode tracings were recorded through the anterior and posterior LV walls at a paper speed of 100 mm/sec. This orientation was chosen to allow delineation of wall thickness and motion in infarct and noninfarct territories. The results were recorded on optical discs, and the M-mode tracings were analyzed. Relative posterior wall thickness and LV internal dimensions were measured from at least three consecutive cardiac cycles. We also used posterior wall thickening, endocardial fractional shortening (FS), and LVEF as indices to estimate LV systolic function.
( P/ d9 z; z+ l/ I6 C- k
, i, }3 {* V7 q* o/ u  ~) EIdentification of Transplanted Cells and Histological Analysis# D; l4 u2 ?) C

" p& T( v- D5 `2 {1 xWithin 3 days after follow-up echocardiography, all animals were killed with an overdose of ketamine and pentobarbital 4 weeks after cell transplantation. The hearts were quickly removed and the ventricles were cross-sectioned into three specimens from apex to base, and each of these was cut into two pieces. The first one was fixed in 4% paraformaldehyde, embedded in paraffin, and sectioned to yield 8-µm slices. The sections were stained with hematoxylin and eosin as described in the manufacturer¡¯s specifications (Sigma-Aldrich). The second piece of each specimen was embedded in OCT compound (Tissue-Tech; Miles Inc., Elkhart, IN, http://www.bayerus.com) and frozen in isopentane cooled in liquid nitrogen. Eight-micrometer cryosections were fixed in 4% paraformaldehyde. The transplanted CDSCs in frozen sections were identified by fluorescent microscopy. Endogenous peroxidase activity was quenched with 0.3% H2O2 for 15 minutes. After rinsing with PBS three times, the slices were incubated with antibodies against desmin, alpha-sarcomeric actin, smooth muscle actin, troponin-I, collagen types I and II, and von Willebrand factor for 16 hours at 21¡ãC. Negative control samples were incubated in PBS without the primary antibodies. After being rinsed three times for 15 minutes each, the slices were incubated with goat anti-rabbit immunoglobulin G conjugated to FITC and Rhodamine (tetramethylrhodamine isothiocyanate) at 37¡ãC for 45 minutes . The specimens were observed immediately under fluorescent microscope.& T% N3 D( U$ I$ ^$ N$ {4 ]

# d1 K  B! G8 ~Von Kossa Staining* b! \8 J+ W+ v

. V) O+ i( h3 S" z) |  z0 nVon Kossa staining was performed to detect abnormal deposits of calcium within the myocardium. The heart tissue slices were incubated in the dark with a 2% (wt/vol) silver nitrate solution. The slices were then washed with distilled water and exposed to bright light (while covered with water) for 15 minutes and then dehydrated with ethanol . The calcium is reduced by a strong light and replaced with silver deposits, visualized as metallic silver. By von Kossa staining, calcium mineral appears black." |: T% ]3 p' I5 w% ~

/ w, q5 s4 j5 `  ?3 w! vMeasurement of Capillary Density in the Scar
: h7 s# ?7 o$ \- K" A8 r
0 _/ ~. s$ F: R; sAfter staining with antibody of von Willebrand factor, the number of capillary vessels was counted in the scar tissue of all groups using a standard light microscope at a x 400 magnification . Five high-power fields within the scar of each section were randomly selected, and the number of capillaries was averaged and expressed as the number of capillary vessels per high-power field (0.2 mm2).+ _. ?* }1 I# N9 n4 l3 {4 ^
  e( v3 Q. g& b% ^0 x  O
Reverse Transcription-Polymerase Chain Reaction
& ]/ D$ P( [7 N( W
4 d! f. i6 `, q  A  zTotal RNA was extracted from the fresh heart scar tissue using TRI Reagent (Invitrogen, Carlsbad, CA, http://www.invitrogen.com) according to the manufacturer¡¯s instructions. After the extraction of total RNA, a reverse transcription-polymerase chain reaction (RT-PCR) analysis was performed as described previously . The cDNA was generated from 2 µg of total RNA using Moloney murine leukemia virus reverse transcription (Promega, Madison, WI, http://www.promega.com) and random primers during 2-hour incubation at 42¡ãC. PCR was performed with 2 µl of cDNA using Taq DNA polymerase (Promega). Preliminary experiments were carried out for each gene to select the optimal number of cycles to enable the amplification reaction to proceed in a linear range. PCR of a constitutively expressed gene, GAPDH, was used as an internal control for the amount of input RNA. RNA samples from each group were assayed using primers for the following genes: collagen I, collagen III, matrix metalloproteinase-1 (MMP-1), tissue inhibitor of metalloproteinase-1 (TIMP-1), transforming growth factor-ß1 (TGF-ß1), and vascular endothelia growth factor (VEGF). Ten microliters of each PCR product was run on 1.5% agarose gels, and bands were visualized with a UV transilluminator (Bio-Rad, Hercules, CA, http://www.bio-rad.com).
8 v5 [1 X* c/ l5 g6 o
' v5 w0 c$ n1 @! m2 bData Analysis; I& ]. v! B6 e# L: {* ^

% ~; O0 E9 \3 v8 |' Q; Y2 EAll data are expressed as mean ¡À standard deviation. SPSS (SPSS Inc., Chicago, http://www.spss.com) for Windows 11.0 was used for all analyses. Comparisons of continuous variables among animal groups were studied by a one-way analysis of variance. Longitudinal studies comparing data within each group were achieved by the use of paired t-tests. Statistical significance was assumed at p 6 G5 L1 H6 e7 W) N4 Q, R& C& g/ m
) Z& s8 A6 J( Y6 z: w; d
RESULTS
" O5 L1 K; ?5 ]) H( K4 _
! l) x2 ~' u; M* T( {/ {* N$ C6 }! {Cell Morphology, FACS Analysis, and Immunohistochemistry/ Q  Y7 }8 t/ {: b7 `4 N9 l
3 Z5 U- @# |* p6 n1 V
Cells derived from human articular cartilage exhibited spindle-shaped morphology, attached to the culture plate tightly, proliferated in the culture medium, appeared morphologically to be a homogeneous population, and maintained similar morphology with passages (Fig. 1A). We were able to passage the cells more than 20 times without detecting any signs of senescence. FACS analysis showed that the cells were consistently negative for CD14, CD34, CD38, CD45, CD49f, CD104, CD105, CD106, CD117, HLA-DR, and ABCG-2 and positive for CD10, CD44, CD71, CD73, CD90, CD147, and HLA-A, -B, -C (Fig. 1B). In immunohistochemical analysis, the cultured CDSCs stained negatively for desmin, smooth muscle actin, alpha-sarcomeric actin, and cardiac specific troponin-I (data not shown). Before cell transplantation, CDSCs were detached and stained with Hoechst 33342 (Fig. 1C).
5 Z+ u$ A0 j7 R0 l2 n  {# y" C* `) a; s7 L
Figure 1. (A): Cartilage-derived stromal cells (CDSCs) were spindle-shaped and attached to the culture tightly (original magnification x 400). CDSCs were detached, labeled with fluorescein isothiocyanate-conjugated or phycoerythrin-conjugated monoclonal antibodies, and detected by flow cytometry. Values represent the mean percentage of all assessed cells positively stained by the respective antibodies in the flow cytometry analyses. The results showed that CDSCs were consistently negative for CD14, CD34, CD38, CD45, CD49f, CD104, CD105, CD106, CD117, HLA-DR, and ABCG-2 and positive for CD10, CD44, CD71, CD73, CD90, CD147, and HLA-A, -B, and -C (B). CDSCs were detached with trypsin/EDTA and stained with Hoechst 33342 before transplantation (C).
6 ~7 h. Z4 @+ N: x( u# y
$ E5 [% n% Q* }* G: iSurgical Outcome of Animal Model5 D8 K5 L9 K) F/ H1 \1 N8 T

9 Y5 f9 {7 d% LMortality over the 3-week period after myocardial infarction was 17.8% (16 of 90). Most deaths occurred within 48 hours of MI. Of the survived rats with LVEF less than 60% in ultrasonic assessment, 91.9% (68 of 74) were randomized into three groups¡ªCDSC group (n = 23), medium group (n = 22), and control group (n = 23). The mortality over the 4-week period after the second surgical procedure was 13.0% (3 of 23), 18.2% (4 of 22), and 17.4% (4 of 23) for CDSC, medium, and control groups, respectively. Most deaths also occurred within 48 hours after the second surgical procedure. The survived rats had no apparent signs of abnormalities throughout the experiments.1 R; s8 R# [2 B2 {
) \$ t8 r# k. p
Evaluation of Ventricular Function
. r! W2 d% l) {' o  d. z0 U4 J  @/ _6 I! Y
Two-dimensional and M-mode transthoracic echocardiographic images were obtained. Baseline echocardiography was performed 3 weeks after myocardial infarction, the day before cell transplantation, and there was no significant difference in heart function between the three groups. At the 4-week time point after cell transplantation, the heart function stayed unchanged in the CDSC group except the improved left ventricle posterior wall thickening (54.7% ¡À 17.7% vs. 40.3% ¡À 9.7%, p = .016) compared with the corresponding values of baseline echocardiography. Four weeks after cell transplantation, the echocardiographic parameter of the CDSC-treated group showed higher LVEF (55.2% ¡À 6.1% vs. 47.9% ¡À 8.7% and 50.2% ¡À 8.1%), FS (25.6% ¡À 3.6% vs. 21.5% ¡À 3.7% and 21.6% ¡À 4.1%), and posterior wall thickening (54.7% ¡À 17.7% vs. 33.4% ¡À 15.3% and 31.5% ¡À 16.2%) compared with medium and control groups (p 7 k* ]8 I. F4 g3 ]

7 G7 M0 D9 ?/ t9 j& zFigure 2. Effect of CDSC transplantation on cardiac function. CDSCs (CDSC group, n = 20), medium alone (medium group, n = 18), and saline (control group, n = 19) were injected into ischemic myocardium. LVEF, FS, LVEDd, LVEds, LVPWd, LVPWs, and LVPW thickening were calculated 3 weeks (baseline) after myocardial infarction and 4 weeks after cell transplantation (*p
( L8 O& C7 }  m0 J. T- K+ a
8 h: O  }; w4 j8 ]( jHistological Examination
, D/ G- q' f% c3 S0 {2 [7 G
) Z2 F# A* [, F# VThe LAD ligation led to a transmural infarction in the anterior wall of all examined rats, and fibrous scar tissue developed in the infarction area (Figs. 3A, 3B). Hoechst-labeled CDSCs were detected in islands within the infarct region under fluorescent microscopy (Fig. 3C) and formed vessel lumen¨Clike structure in the scar tissue of cell transplantation rats (Figs. 4A, 4B), but they could not be found in the hearts of medium and control groups. Von Willebrand factor staining indicated that the transplanted CDSCs were involved in the formation of microvessels, and endothelial cells that stained positively were found at the lumen side of the vessel-like structure formed by CDSCs (Figs. 4C¨C4E). By immunofluorescent examination, the CDSCs that transplanted into the scar and border zone were stained negatively for alpha-sarcomeric actin and cardiac-specific troponin-I (data not shown) and positively for desmin and smooth muscle actin (Figs. 4F, 4G). There are neither calcium deposits nor chondroplasia in the heart tissue 4 weeks after CDSC transplantation by von Kossa staining and immunohistochemical staining of collagen II (data not shown).
8 m: l1 \% R$ j/ i
, d6 T$ s. z* Y* c( x. i% r+ XFigure 3. The morphology of the heart 3 weeks after myocardial infarction. The left anterior descending ligation led to a transmural infarction in anterior wall of all examined rats, and fibrous scar tissue developed in the infarction area (A, B). Hoechst-labeled cartilage-derived stromal cells (CDSCs) were detected in islands within the infarct region under fluorescent microscopy 4 weeks after transplantation (C) (original magnification x 200). Hematoxylineosin staining in the tissue slice adjacent to slice C (D).
) n/ n+ M$ g7 O/ _& n7 H
2 S: M) h) P: t/ ^0 CFigure 4. Grafted Hoechst-labeled CDSCs were detected within the infarct region and formed vessel lumen-like structure 4 weeks after cell transplantation (original magnification x 200 ). Von Willebrand factor staining in the tissue slice adjacent to slice A indicated that the transplanted CDSCs were involved in the formation of microvessels (B) (original magnification x 200). Immunohistofluorescent staining indicated that grafted cartilage-derived stromal cells (CDSCs) were negative for von Willebrand factor. Endothelial cells stained positively for von Willebrand factor were found at the lumen side of the vessel-like structure formed by CDSCs (C, Hoechst fluorescent of grafted CDSCs; D, immunohistofluorescent staining of von Willebrand factor in the same tissue slice; E, merged picture of C and D; original magnification x 400). The transplanted CDSCs were stained positively for desmin (F) and smooth muscle actin (G). Arrows indicated the positive staining.
3 b* S5 ^. Z9 W( G0 I- q( u8 m- C) D/ F
Capillary Density in the Scar$ v- u1 f1 n) ^- F) |9 W( [9 K

, D  U) D3 P% g, g9 W) dTo examine whether transplanted CDSCs induced an angiogenetic effect in injured myocardium, the density of blood vessels was calculated in the infarcted area with von Willebrand factor staining. The number of capillaries increased significantly after 4 weeks in infarcted myocardium in rats with CDSC transplantation compared with the medium and control groups (5.58 ¡À 1.12 vs. 3.14 ¡À 0.91 and 2.03 ¡À 0.82 vessels per 0.2 mm2; p
$ q0 H0 y8 i8 N3 C; N% D" ?  f/ W+ a2 i. B
Figure 5. Effects of cartilage-derived stromal cell (CDSC) transplantation on neovascularization. Endothelial cells were stained with antibody of von Willebrand factor, and vessel numbers within infarcted myocardium of rat hearts were counted. Total numbers of vessels in five areas were evaluated in each animal (A, CDSC group, n = 20; B, medium group, n = 18; C, control group, n = 19; original magnification x 200). The number of capillaries increased significantly after 4 weeks in infarcted myocardium in rats with CDSC transplantation compared with the medium and control groups (D) (*p
. E: A6 f* Y% X% R2 a* @  j# j2 T2 H, W# g% a5 G  p( m! q
Reverse Transcription-Polymerase Chain Reaction
5 Q! _% k* }. W) K8 g. x5 y; D4 Y# ~$ P# x/ R  h7 p# }) @
RT-PCR results indicated that the expression levels of collagen I, collagen III, TIMP-1, TGF-ß1, and VEGF were much higher (p 8 ^! j- q. E8 E) D0 X

) Y, V6 U4 m' k. qFigure 6. The mRNA expression levels semiquantified by reverse transcription¨Cpolymerase chain reaction. Amounts of cDNAs from different RNA samples were normalized using the levels of GAPDH as an internal control (A). Relative intensities of polymerase chain reaction bands relative to those GAPDH bands amplified in the experiment were determined and summarized (B). There was a clear increase in the expression levels of collagen I, collagen III, TIMP-1, TGF-ß1, and VEGF 4 weeks after CDSC transplantation compared with medium and control groups (*p + z8 L; C1 |& g) c- B6 t

+ @" G& y  ]# W' I( v* J: KDISCUSSION
8 ?  _, ?  B1 R" I9 U' L
( X+ g3 G7 J4 q! _Adult cardiac myocytes are thought to be terminally differentiated and to therefore lose their proliferative capacity after birth. Recent studies have shown the presence of cardiac stem cells residing in the heart . Resident cardiac primitive cells seem to be the most promising candidate for novel, cell-based treatment of patients with heart disease. However, their number is very limited, and how to activate the resident stem cells to regenerate damaged myocardium and restore the heart function after myocardial infarction remains a question. Therefore, multipotent stem cell transplantation could be an alternative treatment modality for cardiac regeneration at present.* H% b, \- n7 G, `
1 e* L% Z: e: t+ M
There is a threshold of the number of stem cells needed to generate adequate heart muscle to contribute to cardiac function. Adult stem cells are limited in supply in each patient and therefore are difficult to isolate and purify. On the other hand, the stem cells derived from patients must be isolated and expanded in culture to obtain a sufficient amount of stem cells for transplantation. However, it is not practical to wait a long time for the manipulation of stem cells in vitro before transplantation in clinical use .) f! t  [! S  C3 J# X& ]7 u

* C: s" `& [; v9 F  wIn our experiment, human CDSCs were spindle-shaped, proliferated in the culture medium, appeared morphologically to be a homogeneous population, and maintained similar morphology with passages. The CDSCs could be passaged more than 20 times without detecting any signs of senescence. Most of the markers expressed by CDSCs coincided with mesenchymal stem cells . When transplanted into infarcted rat hearts, the CDSCs could promote angiogenesis, prevent LV remodeling, and then improve the heart function. Our study results indicated that transplantation of the CDSCs might provide a novel and promising approach to improve the injured heart function following myocardial infarction.+ X1 W! V8 t) A: ]$ C/ ?
' L" Z$ V, x: _( H
The cellular and molecular mechanisms that mediate the functional benefits observed in the failing heart after cell transplantation are not clear. First, an active process whereby the engrafted cells replace lost contractile elements and contract synchronously with the host myocardium may occur. Second, it is possible that some of the beneficial effects of cell transplantation may be the result of a reorganization of the structural elements surrounding the engrafted cells. In our experiment, there is no evidence indicating that CDSCs transplanted into the infarcted rat hearts differentiated into myocardium and were involved in myocardial contraction, whereas the CDSCs were positive for desmin and smooth muscle actin. The implanted CDSCs participated in the formation of microvessels and significantly elevated VEGF expression levels accompanied by increased vascular density and regional blood flow in the infarct zone.. \) x& [* `& \3 [0 Z

) L# D4 w3 b2 B- U3 ^  b6 LIt has been well documented that BM stromal cell transplantation can improve infarcted heart function of rat, porcine, and human . It is interesting that single injection of cell-free medium could also induce the angiogenesis in the heart compared with the control group, although the angiogenic effect induced by cell-free medium is not as strong as that induced by CDSC transplantation. Growth factors and/or cytokines supplemented in the medium may be responsible for the angiogenic effect induced by cell-free medium.
8 S8 u0 b6 I+ F6 z9 Z+ j' K3 r. `4 A% z; W
The interstitial or extracellular collagen matrix (ECCM) provides the structural framework for coordinated muscle cell contraction and sequesters growth factors and cytokines that interact with local cell receptors to influence cellular behavior and survival. Remodeling of the ECCM after MI plays a major role in LV remodeling, whereby decrease, disruption, and/or defective composition of the ECCM promote LV dilation and rupture  reported that although twofold to threefold increase in myocardial collagen above the normal level results in increased LV stiffness and mild dysfunction, a very small decrease in collagen below normal can lead to drastic consequences, including LV dilation and rupture. It thus seems that ECCM is a double-edged sword for the heart, as both ECCM deficiency and overproduction may lead to the damage of heart.
' o0 E3 i, `( c  C( ?# c1 ~/ d; {0 r6 D. X0 I. c; S
Collagen types I and III composed most of the macromolecules of ECCM. The data presented in this study showed that the expression levels of collagen types I and III in infarcted zone were much higher in the transplantation group than those in the medium and control groups. Fedak et al.  reported that cell transplantation might alter the matrix components of the injured heart region as well as the normal myocardium to prevent the thinning and dilatation that constitutes remodeling after myocardial infarction. The increase in expression levels of collagen types I and III in infarcted zone of rats may protect the heart from LV remodeling and dilation. In view of the fact that ECCM overproduction may lead to damage of the heart, in our long-term study project, we will pay more attention to the negative effects induced by ECCM accumulation and search for a new strategy to recover the normal level of collagen in the damaged heart.7 ?$ Q7 f$ w  `5 F0 ~+ O8 @; v  R& A

+ D' z2 j: U( V6 n' y0 ^$ iA fine balance between MMPs that degrade the ECCM and the endogenous TIMPs maintains normal remodeling and function, and an imbalance can result in adverse remodeling . The functional improvement of rat hearts could also be explained by the ability of the engrafted CDSCs to protect ECCM and limit infarct expansion.( k: ?; a, `2 r( z- R2 w. w
) L; e" G2 T) S# Y7 U
There have been concerns about the potential for adverse effects or unregulated differentiation after stem cell transplantation in vivo . It is therefore necessary to verify the safety of CDCS transplantation. In our study, there was no evidence of calcium deposits or chondroplasia in the heart tissue 4 weeks after CDCS transplantation. Further studies are still required to evaluate the long-term safety." K) d9 Q( ?: j) R

; J( ?4 [: B$ g1 t: K7 S& SRecent reports have provided substantial new insights into stem cell populations in a variety of adult tissues. In addition to BM, other sources of stem cells with mesenchymal potential include adipose tissue . The possible therapeutic mechanisms involved myogenic and/or angiogenic and paracrine effects of the stem cells. In our study, mesenchymal stem cells could be isolated from cartilage tissue. The cells might differentiate into smooth muscle and secrete cytokines to repair the damaged heart. The cells can be used as a new cell source to be applied in cell-based therapy. In view of the fact that the mesenchymal stem cells from cartilage tissue can be easily cultured and amplified in vitro, we can establish a cell bank for allotransplantation and use the mesenchymal stem cells from cartilage tissue for the treatment of patients with heart disease but not suitable for BM harvesting procedure.
" G! n$ s; ~* {$ _! `! f3 ~2 d7 A, Y
To investigate the influence of CDSC transplantation on heart function, angiogenesis, and ventricular remodeling after myocardial infarction, the rats were usually killed and examined 4 weeks after the transplantation of CDSCs in our experiment. Indeed, long-term observation is necessary to track the survival of the rats with repaired heart function to evaluate the long-term efficacy and adverse events. We do have data about the survival of the rats (data not shown). In our experiment, six rats with repaired heart function survived for 12 weeks with CDSC transplantation before euthanasia (four rats received daily inoculation of cyclosporine for 4 weeks, and two rats received daily inoculation for 12 weeks). The general status of the rats was similar to the healthy rats during the 12 weeks when they were fed in the cages.
" c0 |5 j( r; c) E! H* n# _, a# x, Q; N0 a- N7 _
Although statistical analysis showed cardiac function improved in the stem cell transplantation group, the difference was not impressive. We used a single dose (5 x 106 cells) for cell transplantation. Previous studies have demonstrated that cell dose has a significant influence on the beneficial effects of cell transplantation on ventricular function . We believe that more improved functional effects could be achieved with an optimal dose of cells and optimal time point for transplantation after myocardial infarction. To determine the optimal number of cells and optimal time point for transplantation, a dose-response and a time-response curve would need to be conducted. However, it is likely that attempts to enhance cell survival after implantation may be more effective in increasing the number of viable engrafted cells than increasing the number of cells implanted by means of combination of cell transplantation with gene therapy of growth factor and cytokines that have functions to enhance survival of the implanted cells.
5 \- B2 Z1 V5 |
$ l% B" ]! }1 C6 x+ G: `0 _3 RIn conclusion, the present study showed that stromal cells from human articular cartilage may promote angiogenesis, prevent LV remodeling, limit infarct expansion, and improve the heart function when transplanted into infarcted rat hearts. The CDSCs might become a new candidate cell source for cell transplantation in the treatment of myocardial infarction.
2 C3 Y6 d- {" `8 n# I. @
# S6 y4 G: q/ P8 kACKNOWLEDGMENTS  h5 S* R- m/ R4 f4 j( ?. P6 g3 ~3 j

1 x+ q% g' ]6 ], W0 x- p4 Q. IW.S. and H.Z. contributed equally to this work. This research was supported by grants from National Outstanding Young Foundation (30125039), Beijing Science Technology Committee (H020220010490), and National 863 Program. We would like to thank Dr. Xi Chen and Dr. Weiquan Zhu for their valuable advice and technical assistance in the experiments of molecular biology. We also thank Dr. Ping Chen and Yinan Liu for cell culture and fluorescence-activated cell sorting analysis., U" t) K; @( C9 j* ^" s

5 o1 n$ U4 x9 Q0 ]DISCLOSURES
9 G/ @: M- j7 I2 j5 w2 [' _
+ X1 s5 C3 d4 C$ GS.H. has performed contract work within the last 2 years for National Outstanding Young Foundation, Beijing Science Technology Committee, and National 863 Program.
6 f" K9 f( l0 ^5 z% q, J          【参考文献】
  S- {) k8 v; c5 U9 y 7 |7 i. n+ X$ G& J7 _6 v  ~
5 E2 a" ~- z+ M2 `6 T; N$ R
Pfeffer JM, Pfeffer MA, Fletcher PF et al. Progressive ventricular remodeling in rat with myocardial infarction. Am J Physiol 1991;260: 1406¨C1414.
6 s- Q& L. S, N7 n# D) H/ b3 U) l6 Y' d; B
Yang F, Liu YH, Yang XP et al. Myocardial infarction and cardiac remodeling in mice. Exp Physiol 2002;87:547¨C555.
/ V# X& s/ }8 Z/ B( D8 {% _/ f( C3 d5 i0 v; ?: \" ~( }
Litwin SE, Katz SE, Morgan JP et al. Serial echocardiographic assessment of left ventricular geometry and function after large myocardial infarction in the rat. Circulation 1994;89:345¨C354.' _9 X: D; ?0 O' y( A5 }! V& o

1 j6 W! ~4 A$ |Taylor DA, Atkins BZ, Hungspreugs P et al. Regenerating functional myocardium: Improved performance after skeletal myoblast transplantation. Nat Med 1998;4:929¨C933.
5 k) p+ x& {5 a4 Z. r# c
8 f/ y5 |' Y! z. [- F0 q' b1 CTomita S, Li R-K, Weisel RD et al. Autologous transplantation of bone marrow cells improves damaged heart function. Circulation 1999; 100(suppl II):247¨C256.
' U* r* c2 o0 _( Y2 H5 s  n4 F9 s
. Y" l3 n. U1 _- P2 qGalinanes M, Loubani M, Davies J et al. Autotransplantation of unmanipulated bone marrow into scarred myocardium is safe and enhances cardiac function in humans. Cell Transplant 2004;13:7¨C13.
* `$ G" u* a* M1 V* V- r/ a3 t- \5 [
Fujii T, Yau TM, Weisel RD et al. Cell transplantation to prevent heart failure: A comparison of cell types. Ann Thorac Surg 2003;76:2062¨C2070.
# o0 k2 W" B6 H5 ^# `) \$ T* L% v( m, |& D) P- D
Rangappa S, Fen CH, Lee EH et al. Transplantation of adult mesenchymal stem cells isolated from the fatty tissue into cardiomyocytes. Ann Thorac Surg 2003;75:775¨C779.
! X( D  P1 S1 `* X# ~9 A
$ {* z$ Y: X% \  c* @! V& aBarbero A, Ploegert S, Heberer M et al. Plasticity of clonal populations of dedifferentiated adult human articular chondrocytes. Arthritis Rheum 2003;48:1315¨C1325.5 x2 z- w/ Z- C1 p/ d6 n6 e4 P: R
( ~4 ]/ ?: C  O7 C8 n5 j
Cui Y, Wang H, Xu TR et al. Stem cell characteristics of human fetal articular cartilage-derived cells2 ]; w- e: r; z, t% C6 ?* t$ q
: L" _" m$ W1 S" L
Tallheden T, Dennis JE, Lennon DP et al. Phenotypic plasticity of human articular chondrocytes. J Bone Joint Surg Am 2003;85:93¨C100.$ l0 i  B2 f; c

5 ~, z, m' D( C) f% y5 OMiranville A, Heeschen C, Sengenes C et al. Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation 2004;110:349¨C355.
- a( {+ u9 M5 y8 Z  s: p* t& T" b4 h/ h. s& [. G, b7 P
Li RK, Jia ZQ, Weisel RD et al. Cardiomyocyte transplantation improves heart function. Ann Thorac Surg 1996;62:654¨C661.* k+ S# c  u' G* S1 S4 J

* U4 V; b, N7 o; ZOtt HC, Marksteiner BR, Wolf D et al. Combined transplantation of skeletal myoblasts and bone marrow stem cells for myocardial repair in rats. Eur J Cardiothorac Surg 2004;25:627¨C634.3 b, x5 O2 n) j

# l6 }3 N4 `- W6 iTang TL, Zhao Q, Zhang YC et al. Autologous mesenchymal stem cell transplantation induce VEGF and neovascularization in ischemic myocardium. Regul Pept 2004;117:3¨C10.0 I0 P- p8 ?+ ^' H# Z  ^1 K

* `$ ^: p" F" ]Goodell M, Brose K, Paradis G et al. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 1996;183:1797¨C1806.
* ^6 y9 j! w- q7 h1 E$ D) p+ k
3 _  Q, R8 U+ F& }1 w8 n7 XSjaastad I, Sejersted OM, Ilebekk A et al. Echocardiographic criteria for detection of postinfarction congestive heart failure in rats. J Appl Physiol 2000;89:1445¨C1454.$ C  H- c  N- M: S
% d" X) V% q# R5 _3 @2 I4 E1 v- i
Bacou F, Andalousi RB, Daussin PA et al. Transplantation of adipose tissue-derived stromal cells increases mass and functional capacity of damaged skeletal muscle. Cell Transplant 2004;13:103¨C111.- N2 O: P0 x) k/ x$ _; `' F

' _" {6 y# c+ d, J" n  n8 a* N& I" ^' }Jaiswal N, Haynesworth SE, Caplan AI et al. Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vivo. J Cell Biochem 1997;64:295¨C312.( O4 O- B0 ]: C4 \6 ]
; Y8 \& i  n5 b% z) P! ^
Murtuza B, Suzuki K, Bou-Gharios G et al. Transplantation of skeletal myoblasts secreting an IL-1 inhibitor modulates adverse remodeling in infarcted murine myocardium. Proc Natl Acad Sci U S A 2004;101: 4217¨C4221.
8 J7 C. v! W3 E- n' T2 Z
5 q8 v1 W( ?" ]. wDawn B, Stein AB, Urbanek K et al. Cardiac stem cells delivered intravascularly traverse the vessel barrier, regenerate infarcted myocardium, and improve cardiac function. Proc Natl Acad Sci U S A 2005; 102:3766¨C3771.
+ v+ v9 z6 _4 G8 X
; I; o8 \) R* x( A8 G2 m2 z) g) xBeltrami AP, Urbanek K, Kajstura J et al. Evidence that human cardiac myocytes divide after myocardial infarction. N Engl J Med 2001;344: 1750¨C1757.
# u& O( u0 F" w% Q4 u+ m5 C; t1 s" \
, S# n/ F- Q/ p. m5 ]Urbanek K, Torella D, Sheikh F et al. Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure. Proc Natl Acad Sci U S A 2005;102:8692¨C8697.% T1 g2 b; k5 u" \/ X: p" X* L

. T7 A* K( U- |8 O+ vBeltrami AP, Barlucchi L, Torella D et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763¨C766.! g. h9 U2 j. _3 X& C. [. v* p) ~
- M9 f% ^% I: ?3 W% i
Matsuura K, Nagai T, Nishigaki N et al. Adult cardiac Sca-1-positive cells differentiate into beating cardiomyocytes. J Biol Chem 2004;279: 11384¨C11391.; Y" Y- Y& J3 U+ ?4 l

" |, j7 e$ c$ j" FRafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 2003;9:702¨C712.! e8 m" X' y# N" M: u. Y8 c1 S9 n
# v8 r2 U8 N2 S" M" ]1 P1 y
Aicher AM, Erenner W, Zuhayra M et al. Assessment of the tissue distribution of transplanted human endothelial progenitor cells by radioactive labeling. Circulation 2003;107:2134¨C2139.$ d( M; |7 W  j, w
% o3 e1 ]& a) i! C
Yoon YS, Park JS, Tkebuchava T et al. Unexpected severe calcification after transplantation of bone marrow cells in acute myocardial infarction. Circulation 2004;109:3154¨C3157.  ?1 i! w0 F# c9 ^7 w

$ M3 W- c) D6 M1 H# P: lPittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res 2004;95:9¨C20.
- G; r% z9 |, e0 t; p9 Y3 N$ o7 k9 T, G4 a
Roberts L. Mesenchymal stem cells. Vox Sang 2004;87(suppl 2):s38¨Cs41.
& d, v% {- M5 @+ Z; G
% q( T- g! k8 b6 J' kTomita S, Mickle DAG, Weisel RD et al. Improved heart function with myogenesis and angiogenesis after autologous porcine bone marrow stromal cell transplantation. J Thorac Cardiovasc Surg 2002;123:1132¨C1140." W6 G- @! Z. l' G7 ^" f* n2 k
8 e4 r1 p* I4 [6 q4 i1 a( E
Jugdutt BI. Remodeling of the myocardium and potential targets in the collagen degradation and synthesis pathways. Curr Drug Targets Cardiovasc Haematol Disord 2003;3:1¨C30.
" L- w$ `. k% e7 a/ A9 @1 S3 B% a9 G7 L: m; l5 N% T
Covell JW. Factors influencing diastolic function: Possible role of the extracellular matrix. Circulation 1990;81(suppl III):115¨C158.
% Y" ?& e9 N, y0 ~
4 y* R( c# y& Q/ fCleutjens JPM, Kandala JC, Guarda E et al. Regulation of collagen degradation in the rat myocardium after infarction. J Mol Cell Cardiol 1995;27:1281¨C1292.4 A1 j- f5 j! x/ Y* B$ D
/ [7 p$ D& g" ]) E" W5 H- D
Fedak PW, Altamentova SM, Weisel RD. Matrix remodeling in experimental and human heart failure: A possible regulatory role for TIMP-3. Am J Physiol 2003;284:626¨C634.3 e+ I/ G8 K# }9 E3 v) Z
/ B+ \% i* Q: o* R" k: D
Jugdutt BI. Ventricular remodeling after infarction and the extracellular collagen matrix: When is enough enough? Circulation 2003;108:1395¨C1343.
9 N* E! L( ?6 A) ]1 M$ _. W$ {3 I' f7 |7 l! v! P& S+ k4 M4 c( [. k
Weber KT. Extracellular matrix remodeling in heart failure: A role for de novo angiotensin II generation. Circulation 1997;96:4065¨C4082.
2 Q6 B" f, D. J7 `7 s6 R# Q5 V1 `' K, C$ F  V8 h5 s' n! K! _
De Ugarte DA, Morizono K, Elbarbary A et al. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 2003;174:101¨C109.
: C  v+ T) G- [8 v# x6 [. s" \1 Y- w& Y! p4 K
Jankowski RJ, Deasy BM, Huard J. Muscle-derived stem cells. Gene Ther 2002;9:642¨C647.* ?- x$ }5 y( C3 q
. a& @6 A1 k; ], t) H1 q% X
Zvaifler NJ, Marinova-Mutafchieva L, Adams G et al. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res 2000;2: 477¨C488.
/ @: U0 }# ^& e7 c2 @4 o7 x
' L1 g3 l5 h4 Z4 z( a3 KTuli R, Seghatoleslami MR, Tuli S et al. A simple, high-yield method for obtaining multipotential mesenchymal progenitor cells from trabecular bone. Mol Biotechnol 2003;23:37¨C49.0 Z) v" `4 B+ t
' E1 `' r  N& B7 {) B6 y
Noth U, Osyczka AM, Tuli R et al. Multilineage mesenchymal differentiation potential of human trabecular bone-derived cells. J Orthop Res 2002;20:1060¨C1069., Q) l8 P8 h$ q/ d
$ f( J: q9 k  p5 p0 U$ Y3 N
De Bari C, Dell¡¯Accio F, Tylzanowski P et al. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum 2001; 44:1928¨C1942.
- ?# o9 E1 E4 g0 }/ I
$ m: M5 o7 t" I% a& t) }6 w' HPouzet B, Vilquin JT, Hagege AA et al. Factors affecting functional outcome after autologous skeletal myoblast transplantation. Ann Thorac Surg 2001;71:844¨C850.

Rank: 1

积分
威望
0  
包包
32  
沙发
发表于 2009-10-17 20:38 |只看该作者
thanks

Rank: 1

积分
37 
威望
37  
包包
241  
藤椅
发表于 2009-10-20 17:49 |只看该作者
附件: 你需要登录才可以下载或查看附件。没有帐号?注册
已有 1 人评分威望 包包 收起 理由
细胞海洋 + 10 + 10 极好资料

总评分: 威望 + 10  包包 + 10   查看全部评分

Rank: 2

积分
74 
威望
74  
包包
945  
板凳
发表于 2009-12-12 18:04 |只看该作者
干细胞之家微信公众号
ding

Rank: 3Rank: 3

积分
304 
威望
304  
包包
1983  

优秀会员 帅哥研究员

报纸
发表于 2010-6-5 16:51 |只看该作者
很感兴趣 谢谢分享

Rank: 2

积分
136 
威望
136  
包包
1877  
地板
发表于 2015-7-19 22:33 |只看该作者
好啊,谢楼主

Rank: 2

积分
122 
威望
122  
包包
1876  
7
发表于 2015-7-23 19:37 |只看该作者
想都不想,就支持一下  

Rank: 2

积分
72 
威望
72  
包包
1730  
8
发表于 2015-8-6 12:55 |只看该作者
围观来了哦  

Rank: 2

积分
162 
威望
162  
包包
1746  
9
发表于 2015-8-9 17:51 |只看该作者
21世纪,什么最重要——我!  

Rank: 2

积分
64 
威望
64  
包包
1782  
10
发表于 2015-8-31 06:53 |只看该作者
厉害!强~~~~没的说了!  
‹ 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, 2024-6-17 20:10

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.