骨髓间充质干细胞移植对扩张型心肌病大鼠模型促修复作用的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
扩张型心肌病(dilated cardiomyopathy,DCM)是一种临床常见的心肌病,该病预后差,5-10年生存率较低(30%-40%)。DCM的治疗目前尚无特效药物及方法。干细胞移植是治疗DCM的一种新方法,已经成为国内外研究的热点。骨髓间充质干细胞(Bone marrow mesenchymal stem cells, BMSCs)是存在于骨髓中的非造血实质的基质干细胞,具有来源广泛、取材方便、在体外容易扩增、具有较强的自我复制和多向分化能力、免疫源性弱、可以自体移植、不存在伦理方面的问题、可以进行基因修饰、能分化为心肌细胞和内皮细胞等优势,BMSCs已经成为心血管疾病细胞移植治疗的理想种子细胞。本研究旨在探讨在体外不同诱导条件下BMSCs分化为心肌细胞的作用;建立稳定可靠的DCM动物模型,以供基础实验研究;探讨BMSCs移植治疗DCM模型大鼠的作用及机制。
     本研究采用密度梯度离心法结合贴壁培养法分离培养大鼠BMSCs,在体外分别用5-氮胞苷(5-azacytidine, 5–aza)、DCM模型大鼠血清+5–aza、DCM模型大鼠心肌细胞裂解液等作为条件培养液培养,观察BMSCs形态变化、用免疫组化和RT-PCR法检测心肌肌钙蛋白T(cardiac troponin T;CTnT)的表达情况。选用雄性Wistar大鼠,接受腹腔注射阿霉素的方法建立DCM模型。然后用5-溴脱氧尿嘧啶核苷(5-bromodeoxyuridine ,Brdu)标记BMSCs、DCM模型大鼠血清+5–aza培养诱导的细胞进行细胞移植,采用单次静脉注射和重复静脉注射的方法,观察各组大鼠的状态及死亡率、超声心动图、脑利钠肽(brain natriuretic peptide,BNP)水平、病理改变、Masson染色结果、用激光共聚焦显微镜追踪移植细胞在心脏中的分布情况和分化情况。用免疫组化和RT-PCR法检测血管内皮生长因子(vascular endothelial growth factor,VEGF)表达情况。
     本研究得出的结论是:密度梯度离心法结合贴壁培养法可以分离培养到大量、高活性、纯化的BMSCs;在体外用5–aza、DCM大鼠模型的心肌细胞裂解液、DCM大鼠模型血清+5–aza三种方法均可诱导BMSCs向心肌细胞分化,其中DCM模型大鼠血清+5–aza(5μmol/L)诱导组细胞生长状态、向心肌细胞分化的结果最佳;用阿霉素2.5mg/(kg·week),连续6周腹腔注射给药,建立DCM大鼠模型的方法成功率高,造模周期较短,结果可靠,可以做为典型、稳定的供基础研究的动物模型;经静脉移植是一种有效的移植途径,重复静脉治疗提高了移植细胞的数量、延长了补充供体细胞的时间,治疗效果优于单次静脉移植治疗;直接用BMSCs移植治疗和用先经体外诱导分化后的BMSCs移植治疗均能改善心功能,两者无显著差异;BMSCs在体内可以分化为心肌细胞,并表达心肌特异性cTnT;BMSCs移植治疗可以提高DCM大鼠的存活率、改善心功能、抑制心肌纤维化、增加VEGF的表达,改善心室重构。
Dilated cardiomyopathy (DCM) is a clinical common cardiac disease. Its etiology may be related to infection, autoimmune, metabolic abnormalities and family history. It is mainly characterized by dilatation and systolic dysfunction of left or bilateral ventricular. It is histologically marked by myocardial cell hypertrophy, degeneration, mitochondrial function defect, and different levels of fibrosis. The prognosis is poor, and 5-10-year survival rate is low (30% to 40%). Up to now, there is no specific drug treatment and methods. The current therapy is to control heart failure, lessen symptom and prevent ventricular remodeling, including the use of medicines, such as angiotensin converting enzyme inhibitor (ACEI), diuretics, digoxin,βblockers, calcium antagonists and other drugs. As the poor prognosis of the DCM patients, heart transplantation is the most effective treatment of patients with DCM in advanced stage, but many problems have been encountered in clinical practice, such as donor source, immunological rejection and costs. So cardiologists have been looking for a new treatment method. Stem cell transplantation as a new treatment of DCM has became a hot research area in recent years. Bone marrow mesenchymal stem cells (BMSCs) are non-hematopoietic stem cells in the bone marrow with a variety of sources, drawing convenience, easy expansion in vitro, pluripotency, and possess a weak immune rejection, and can be autologous transplantation with no ethical problems, and can be genetically modified, and can differentiate into myocardial cells and endothelial cells and other advantages. So BMSCs have become ideal seed cells for cell transplantation to treat cardiovascular disease. The treatment of acute myocardial infarction using stem cell transplantation has unparalleled advantages compared with conventional treatment, while the researches about the therapy of non-ischemic cardiovascular diseases with the BMSCs are less. Meanwhile many problems still remains, such as the choice of seed cells, the quantity of transplanted cells, the path of transplantation, treatment time, the duration of BMSCs proliferation and survival in the recipient, and how to improve the differentiation capabilities of BMSCs, when to follow-up after treatment, security in clinical application, which are need to be solved.
     Objective
     1. To explore the effect of differentiation of BMSCs into myocardial cells in different inducing conditions in vitro.
     2. To establish a stable and reliable animal model with DCM for basic experimental research.
     3. To explore the effect of BMSCs on the therapy of DCM with different transplantation methods and its mechanism
     Methods
     1. BMSCs were isolated with the density gradient centrifugation combined with adherent method and cultured. The growth curve, cell cycle, phenotype of BMSCs were determined. BMSCs were induced to differentiate into osteoblasts and adipocytes. In vitro BMSCs were cultured in the medium supplemented with 5–azacytidine(5–aza) or serum of rat with DCM combined with 5–aza or cadiocyte lysate of rat with DCM. Then the morphological changes were observed and the expression of cardiac troponin was detected using the methods of immunocytochemistry and RT-PCR.
     2. Rats DCM models were established by the following method. Male Wistar rats were administered doxorubicin 2.5 mg / (kg ? week) by intraperitoneal injection for 6 weeks. Then rats were observed for 2 weeks after the withdrawal. While rats in the control group were given the same volume normal saline with the same exposure route. The body weight changes and mortality were recorded, and echocardiography was performed. In addition to, plasma brain natriuretic peptide (BNP) level was detected, left ventricular internal diameter was measured, and pathological observation, electron microscopy, Masson staining of myocardium were performed.
     3. BMSCs labeled with Brdu and differentiated cells derived from the BMSCs in the medium supplemented with the serum of rat with DCM combined 5–aza were transplanted using a single intravenous injection and repeated intravenous injection method. The rats were randomly divided into normal control group (12 rats), model control group (17 rats), treatment group 1 (12 rats) which represents the single intravenous injection with BMSCs , treatment group 2 (12 rats) which represents the repeat intravenous injection with BMSCs, treatment group 3 (12 rats) which represents the single intravenous injection with differentiated cells cultured with the serum of rat with DCM combined 5–aza, treatment group 4 (12 rats) which represents the repeat intravenous injection with the same cells as in the treatment group 3. The state and mortality of the rats were observed, echocardiography, BNP levels, pathological observation and Masson staining were performed. The distribution and differentiation of transplanted cells in the heart were traced with laser scanning confocal microscope. The expression of VEGF was detected using immunohistochemistry and RT-PCR.
     Results
     1. Most of BMSCs isolated with the density gradient centrifugation combined adherent method and cultured adhered to the wall with the colony growth. Passaged cells quickly adhered, proliferated rapidly, and almost no colonies were formed. About 6 - 7d later, the cells crowded the bottom of the flask with the spindle-shaped uniform distribution. After many passages, cells have a more uniform and orderly distribution of fibroblast-like. After passaged for consecutive 9 times, cells had no significant changes in morphology, no signs of aging. The passage cycle is 6-7d, but after the 9 passage, the proliferation of cells was slow and the passage cycle was extended. The Passage1, 3, and 5 cells were high proliferation, the growth curve shape of which were similar. The majority of the cells were relatively quiescent. BMSCs could differentiate into osteoblasts and to fat cells under particular induced conditions. After the phenotypes of BMSCs were determined by flow cytometry, they were CD44-positive andCD34-negative. All the three methods could induce BMSCs to differentiate into cardiocytes-like cells with the expression of troponin, among which the growth state of cells in 5–aza group was poor; the cells in the other two groups grew better. The positive-expression rate of cardiac troponin of differentiated cells in the serum of rat with DCM combined 5–aza group was highest.
     2.The bodyweight was significantly different between normal group and model group on the 8th weeks. Rat mortality rate in the model group was 16.7%. The echocardiography findings, plasma BNP, left ventricular internal diameter were significantly different between normal group and model group. In the normal group, the results of HE showed that cardiac muscle fibers arranged regularly, no myocardial fiber breakage, uniform and rich cytoplasm, normal cell space and no edema. The results of electron microscopy displayed myocardial fibers arranged regularly, the uniform size of mitochondria, no edema, intact membrane and normal density of crest. The results of myocardial Masson staining showed collagen distributing sparse and complete collagen fibers nets of the neighbor cells and pale staining. In the model group, the results of HE staining showed myocardial fibers disorganized, myocardial fiber breakage, widened cell gap, edema and vacuole degeneration. The results of electron microscopy showed myocardial fiber fracture, swelling mitochondria, vacuole degeneration, and crest depletion. The results of myocardial Masson staining showed increased collagen in myocardium, breakage and derangement of collagen fibers surrounding the myocardial cells.The CVF compared with the normal group had significantly different.
     3. In model group without cell transplantation, the state of rats were poor, and body weight did not increase, seven rats died, and the mortality rate was 41.2%. While the state of rats in cell transplantation groups were improved, the body weight increased, the mortality of rats in all the cell transplantation groups was lower than that in the model group. The mortality rate of rats in repeat intravenous injection group was lower than that in single intravenous injection group. The cardiac function with echocardiography 2 weeks after cell transplantation had no significantly difference (p> 0.05) between treatment groups and model group. The results of cardiac function with echocardiography and plasma BNP 4 weeks after cell transplantation were: the results of treatment group 1, 2, 3, 4 were significantly different compared with that in the control group (p <0.05); the results in treatment group 2 and 4- were significantly different from that in the model group (p<0.05); there were significant difference between the treatment group 1 and treatment group 2 (p <0.05); there were significant difference between treatment group 3 and treatment group 4 (p <0.05); there was no significant difference between the treatment group 1, 3 and model group (p>0.05); there was no significant difference between treatment group 2 and treatment 4 (p> 0.05). The distribution and cTnT expression of transplant cells in myocardial tissue using the laser confocal microscope showed that transplanted cells labeled with Brdu could be found in the treatment group 1, 2, 3, 4, which distributed disperse, fused with myocardial cell, and expressed cTnT. However, the numbers of the transplanted cells labeled with Brdu in repeated intravenous injection treatment group 2, 4 were more than that in the single intravenous injection treatment group 1, 3, and the expression of cTnT in cells was significantly increased. There was no significant difference between treatment group 1 and treatment group 3 and between treatment group 2 and treatment group 4. The results of HE staining showed that myocardial cells still arranged disordered, myocardial fiber breakage was alleviated in treatment group 1 and treatment group 3 compared with model group. However, some changes still remained, such as widened cell gap, cell edema and vacuole degeneration in treatment group 1 and treatment group 3.The result of HE staining displayed that myocardial cells arrangement were no obvious disorder, no significant cardiac fiber broke, cell gap widened, edema and degeneration occurred, which significantly were alleviated compared with that in the model group in treatment group 2 and treatment group 4. Masson staining results showed there was still more myocardial collagen, and collagen fiber was still broken and disordered in treatment group 1 and treatment group 3. While there was less myocardial collagen in treatment group 2 and treatment group 4 than that in model group. In treatment group 2 and treatment group 4, there was no significant collagen fiber breakage and disorderly arrangement, and expression of VEGF was higher in treatment group 2 and treatment group 4 than that in other groups by immunohistochemistry and RT-PCR.
     Conclusion
     1. A number of highly active purified BMSCs were isolated by density gradient centrifugation combined with adherent method.
     2. In vitro the three methods can induce BMSCs to differentiate into cardiocyte-like cells, which include 5–aza, the serum of rat with DCM model combined with 5–aza, the myocardium lysate of rat with DCM.
     3. The induced effects were better in the medium supplemented with the serum of rat with DCM combined with 5–aza, the myocardium lysate of rat with DCM than that with 5–aza. The cells growth state and differentiation into cardiocyte-like cells were the best in the medium supplemented with serum of rat with DCM combined with 5–aza (5μmol / L).
     4. The method with doxorubicin 2.5mg/(kg ? week) for 6 weeks by intraperitoneal injection to establish rat model with DCM was high success rate, shorter cycle of establishing models and reliable, which can be used as preparing animal models for basic research.
     5. The vein transplantation was an effective way, repeated intravenous treatment can increase the number of transplanted cells, extend the time of add donor cells, and its effect was better than that in single intravenous treatment.
     6. There were no significant difference between BMSCs transplantation treatment and transplantation treatment of cardiocyte-like cells differentiated from BMSCs.
     7.In vivo BMSCs can differentiate into myocardial cells, which expressed cardiac-specific cTnT.
     8. BMSCs transplantation for treatment of DCM in rats can reduce mortality and improve cardiac function, inhibit myocardial fibrosis, increase expression of VEGF and improve ventricular remodeling.
引文
[1] RICHARDSON P,MCKENNA W,BRISTOW M,et a1.Report of the 1995 world health organization/in—ternational society and federation of cardiology task force on the definition and classification of cardiomyopathies[J].Circulation,1996,93(5):841--842.
    [2] MARON B J,TOWBIN J A,THIENE G,et a1.Contemporary definitions and classification of the cardiomyopathies: an American Heart Association Scientific Statement from the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention[J].Circulation,2006,113(14):1807—1816.
    [3]中华医学会心血管病分会、中华心血管病杂志编委会、中国心肌病诊断与治疗建议工作组。心肌病诊断与治疗建议[J].中华心血管病杂志,2007,35(1):3--16.
    [4] ELLIOTT P,ANDERSSON B,ARBUSTINI E,et al. Classification of the cardiomyopathies:a position staement from the european society of cardiology working group on myocardial and pericardial diseases[J].Eur Heart J,2008,29(2):270-276.
    [5] Luk A,Ahn E,Soor GS,et al. Dilated cardiomyopathy:a review[J]. J Clin Pathol,2009,62(3):219-225.
    [6] Cihakova D,Rose NR. Pathogenesis of myocarditis and dilated cardiomyopathy [J]. Adv Immunol,2008,99:95-114.
    [7] Mazzone M,La Sala M,Portale G,et al. Review of dilated cardiomyopathies. Dilated cardiomyopathies and altered prothrombotic state: a point of view of the literature[J]. Panminerva Med,2005,47(3):157-167.
    [8]杨英珍,陈瑞珍.扩张型心肌病发病机制和治疗的研究新动向[J].中华心血管病杂志,2006,3(34):196-197.
    [9]吕安林,张晶,杜娟娟.扩张型心肌病发病机制新论[J].临床医学工程,2009,3(16):99-100.
    [10]吕凤英,宋来凤,刘蕾,等.心肌扩张型心肌病的病理形态特征及其临床意义[J].中华病理学杂志,2007,12(36):796-800.
    [11] Thiene G,Basso C,Calabrese F,et al. Twenty years of progress and beckoning frontiers in cardiovascular pathology: cardiomyopathies[J]. Cardiovasc Pathol, 2005,14(4):165-169.
    [12] Thompson BS. Sudden cardiac death and heart failure[J]. AACN Adv Crit Care, 2009,20(4):356-365.
    [13] Olson TM,Michels VV,Ballew TD,et al. Sodium channel mutations and susceptibility to heart failure and atrial fibrillation[J]. JAMA,2005,293(4):447-454.
    [14] Towbin JA, Bowles NE. The failing heart[J]. Nature,2002,415(6868):227-233.
    [15]王玉亭,王庭槐.扩张型心肌病发病机理及治疗进展[J].中国心血管杂志,2005,10(3):230-231.
    [16] Acquatella H. Echocardiography in Chagas heart disease[J]. Circulation,2007,115(9):1124-1131.
    [17] Thomas DE,Wheeler R,Yousef ZR,et al. The role of echocardiography in guiding management in dilated cardiomyopathy[J]. Eur J Echocardiogr,2009,10(8):15-21.
    [18] Abrams D,Derrick G,Penn DJ,et al. Cardiol complications in children following infection with varicella zoster virus[J ]. CardiolYoung,2001,11(6):647-652.
    [19] Tam PE. Coxsackievirus myocarditis: interplay between virus and host in the pathogenesis of heart disease[J]. Viral Immunol,2006,19(2):133-146.
    [20] Comar M,D'Agaro P,Campello C,et al. Human herpes virus 6 in archival cardiac tissues from children with idiopathic dilated cardiomyopathy or congenital heart disease[J]. J Clin Pathol,2009,62(1):80-83.
    [21 ]Reis FJ,Viana M,Oliveira M,et al. Prevalence of hepatitis C and B virus infection in patients with idiopathic dilated cardiomyopathy in Brazil:a pilot study[J]. Braz J Infect Dis,2007,11(3):318-321.
    [22] Sani MU. Myocardial disease in human immunodeficiency virus (HIV) infection: a review[J]. Wien Klin Wochenschr, 2008, 120(3-4): 77-87.
    [23] PatanèS, Marte F, Sturiale M, et al. Myocarditis and cardiomyopathy HIV associated. Int J Cardiol. 2009 Jan 29. [Epub ahead of print]
    [24] Chuichi Kawai MD. From myocarditis to cardiomyopathy :mechanisms of inflammation and cell death[ J ]. Circulation, 1999, 99(8): 1091-1100.
    [25] Chapman NM, Kim KS.Persistent coxsackievirus infection: enterovirus persistence in chronic myocarditis and dilated cardiomyopathy[J]. Curr Top Microbiol Immunol, 2008, 323: 275-292.
    [26] Jahns R, Boivin V, Schwarzbach V, et al. Pathological autoantibodies in cardiomyopathy[J]. Autoimmunity, 2008, 41(6): 454-461.
    [27] Caforio AL, Tona F, Bottaro S, et al. Clinical implications of anti-heart autoantibodies in myocarditis and dilated cardiomyopathy[J]. Autoimmunity, 2008, 41(1): 35-45.
    [28]张晓伟,蔡文锋.扩张型心肌病免疫病理学发病机制研究进展[J].心血管病学进展, 2009, 5(30): 814-816.
    [29] Burkett EL, Hershberger RE. Clinical and genetic issues in familial dilated cardiomyopathy. J Am Coll Curdiol[J], 2005, 45(7): 969-981.
    [30] Frey N, Katus HA. Dilated cardiomyopathy as a genetic disease: molecular and clinical aspects. Internist (Berl), 2008, 49(1): 43-50.
    [31] Lushnikova EL, Nepomniashchikh LM. Genetic factors of the development of dilated cardiomyopathy[J]. Vestn Ross Akad Med Nauk, 2006, (7): 43-52.
    [32]徐磊,孙爱军,葛均波.特发性扩张型心肌病致病基因研究进展[J].中华心血管病杂志,2008,7(36):664-666.
    [33] Malhotra R, Mason PK. Lamin A/C deficiency as a cause of familial dilated cardiomyopathy[J]. Curr Opin Cardiol, 2009, 24(3): 203-208.
    [34] Modjtahedi N, Giordanetto F, Madeo F,et al. Apoptosis-inducing factor: vital and lethal[J]. Trends Cell Biol, 2006, 16(5): 264-272.
    [35]冯震博,伍伟锋,刘唐威,等.扩张型心肌病的心肌损伤与细胞凋亡.医学研究杂志[J],2006,35(3):34.
    [36] Jahns R, Boivin V, Lohse MJ. beta(1)-Adrenergic receptor function, autoimmunity, and pathogenesis of dilated cardiomyopathy[J]. Trends Cardiovasc Med, 2006, 16(1): 20-24.
    [37] Poole-Wilson PA, Swedberg K, et al. Comparison of carvedilol and metoprolol on clinical outcomes in patients with chronic heart failure in the Carvedilol Or Metoprolol European Trial (COMET): randomised controlled trial[J]. Lancet, 2003, 362(9377): 7 - 13.
    [38] Braun M, Edelmann F, Knapp M, et al. The calcium channel blocker felodipine attenuates the positive hemodynamic effects of the beta-blocker metoprolol in severe dilated cardiomyopathy--a prospective, randomized, double-blind and placebo-controlled study with invasive hemodynamic assessment[J]. Int J Cardiol, 2009, 132(2): 248-256.
    [39] Zhao P, Sharma AC, Ren J. Pathogenesis and therapy of autoimmunity-induced dilated cardiomyopathy[J]. Front Biosci,. 2009, 14: 1708-1715.
    [40] Felix SB, Staudt A. Immunoadsorption as treatment option in dilated cardiomyopathy [J]. Autoimmunity, 2008, 41(6): 484-489.
    [41] Kishimoto C, Shioji K, Kinoshita M, et al. Treatment of acute inflammatory cardiomyopathywith intravenous immunoglobulin ameliorates left ventricular function associated with suppression of inflammatory cytokines and decreased oxidative stress[ J ]. Int J Cardiol, 2003, 91 (2 /3): 173-178.
    [42] Hessel FP,Wegner C,Muller J, et al. Economic evaluation and survival analysis of immunoglobulin adsorp tion in patients with idiopathic dilated cardiomyopathy [J]. Eur J Health Econ, 2004, 5(1): 58-63.
    [43] Cleland JG,Daubert JC, Erdmann E, et al. The effect of cardiac resynchronization on morbidity and mortality in heart failure [ J ]. NEngl J Med, 2005, 352 (15):1539-1549.
    [44] Komamura K, Tatsumi R, Miyazaki J, et al. Treatment of dilated cardiomyopathy with electroporation of hepatocyte growth factor gene into skeletal muscle [ J ]. Hypertension, 2004, 44 ( 3 ): 365-371.
    [45] Sleeper MM, Bish LT, Sweeney HL. Gene therapy in large animal models of human cardiovascular genetic disease[J]. ILAR J, 2009, 50(2): 199-205.
    [46] Thekkudan J, Rogers CA, Thomas HL, et al. Trends in adult heart transplantation: a national survey from the United Kingdom Cardiothoracic Transplant Audit 1995-2007[J]. Eur J Cardiothorac Surg, 2010, 37(1): 80-86.
    [47] Matsumiya G. Surgical ventricular restoration[J]. Nippon Rinsho,2006, 64(5): 975-978. [48古丽帕日·克力.扩张型心肌病的病因与治疗研究进展[J].心血管病学进展, 2009, 3 (30): 508-512.
    [49] Loeffier M, Roeder I. Tissue stem cell:definition,plasticity,heterogeneity, self-organization and models- a conceptual approach[J].Cells Tissue Organs, 2002, 171(1): 8-26.
    [50] Neef TA.Why embryonic stem cell research should be banned forever[J]. Del Med J, 2005, 77(6): 233-234.
    [51] Verfaillie CM.Adult stem cell:assessing the case for pluripotency[J].Trends Cell Biol, 2002, l2(11):502-508.
    [52] Korbling M, Estrov Z. Adult stem cells for tissue repair– a new therapeutic concept[J]? N Engl J Med, 2003, 349(6): 570–582.
    [53] Friendenstein A, Chailakhyan R, Gerasimov U. Bone marrow osteogenic stem cells:in vitro cultivation and transplantion in diffusion chambers[J]. Cell Tissue Kinet, 1987, 20(3): 263-272.
    [54]裴雪涛.干细胞生物学[M].北京:科学出版社,2003.
    [55] Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells[J]. Science, 1999, 284(5411): 143-147.
    [56]胡资兵,曾荣,郭伟韬,林颢.骨髓间充质干细胞诱导分化特征[J].中国组织工程研究与临床康复,2008,12(43):8561-8566.
    [57] Lee OK, Kuo TK, Chen WM, et al. Isolation of multipotent mesenchymal stem cells from umbilical cord blood[J]. Blood, 2004, 103(5): 1669-1675.
    [58] Carlo-Stella C, Gianni MA.Biology and clinical applications of marrow mesenchymal stem cells[J]. Pathol Biol , 2005, 53(3): 162-164.
    [59] Luc Sensebe′, Philippe Bourin.Mesenchymal Stem Cells for Therapeutic Purposes[J]. Transplantation, 2009, 87(15), 49-53.
    [60] Dvorakova J, Hruba A, Velebny V,et al.Isolation and characterization of mesenchymal stem cell population entrapped in bone marrow collection sets[J]. Cell Biol Int, 2008, 32(9): 1116-1125.
    [61] Jorgensen C, Djouad F, Apparailly F,et al. Engineering mesenchymalstem cells for immunotherapy[J]. Gene Ther, 2003, 10(10): 928–931.
    [62] Barry FP, Murphy JM. Mesenchymal stem cells: clinical applications and biological characterization[J]. Int J Biochem Cell Biol, 2004, 36(4): 568–584.
    [63] Le Blanc K, Ringden O. Immunobiology of human mesenchymal stem cellsand future use in hematopoietic stem cell transplantation[J]. Biol Blood Marrow Transplant ,2005, 11(5): 321–334.
    [64] Le Blanc K, Pittenger M. Mesenchymal stem cells: progress toward promise[J]. Cytotherapy ,2005, 7(1): 36–45.
    [65] Satija NK, Gurudutta GU, Sharma S,et al. Mesenchymal stem cells: molecular targets for tissue engineering[J]. Stem Cells Dev, 2007, 16(1): 7-23.
    [66] Shin M,Yoshimoto H,Vacanti JP. In vivo bone tissue engineering using mesenchymal stem cells on a novel electrospun nanofibrous scaffold. [J].Tissue Eng, 2004, 10(1-2): 33-41.
    [67] Woodbury D,Schwarz EJ,Prockop DJ,et al. Adultrat and human bone marrow stromal cells differentiate into neurons[J]. J Neurosci Res, 2000, 61(4): 364-370.
    [68] Waller EK,Huang S,Terstappen L,et al. Changes in the Growth Properties of CD34 +, CD38 -, Bone Marrow Progenitors during Human Fetal Development[J]. Blood, 1995, 86(2): 710-718.
    [69] Bruder SP, Jaiswal N, Haynesworth SE. Growth kinetics, self-renewal, and the osteogenic potential of purified human mesenchymal stem cells during extensive subcultivation and following cryopreservation[J]. J Cell Biochem, 1997, 64(2): 278–294.
    [70] Bruder SP, Kurth AA, Shea M, et al. Bone regeneration by implantation of purified, culture-expanded human mesenchymal stem cells[J]. J Orthop Res , 1998, 16(2): 155–162.
    [71] Kadiyala S, Young RG, Thiede MA, et al. Culture expanded canine mesenchymal stem cells possess osteochondrogenic potential in vivo and in vitro[J]. Cell Transplant , 1997, 6(2): 125–134.
    [72] Young RG, Butler DL, Weber W, et al. Use of mesenchymal stem cells in a collagen matrix for Achilles tendon repair[J]. J Orthop Res, 1998,16(4): 406–413.
    [73] Awad HA, Butler DL, Boivin GP, et al. Autologous mesenchymal stem cell-mediated repair of tendon[J].Tissue Eng, 1999,5(3): 267–277.
    [74] Ferrari G, Cusella-De Angelis G, Coletta M, et al.Muscle regeneration by bone marrow-derived myogenic progenitors[J]. Science, 1998, 279(5356):1528–1530.
    [75] Galmiche MC, Koteliansky VE, Briere J, et al. Stromal cells from human long-term marrow cultures are mesenchymal cells that differentiate following a vascular smooth muscle differentiation pathway[J]. Blood, 1993, 82(1): 66–76.
    [76] Dennis JE, Merriam A, Awadallah A, et al. A quadripotential mesenchymal progenitor cell isolated from the marrow of an adult mouse[J]. J Bone Miner Res, 1999, 14(5): 700–709.
    [77] Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues[J]. Science, 1997, 276(5309): 71–74.
    [78] Woodbury D, Schwarz EJ, Prockop DJ, et al. Adult rat and human bone marrow stromal cells differentiate into neurons[J]. J Neurosci Res , 2000, 61(4): 364-370.
    [79] Chagraoui J, Lepage-Noll A, Anjo AA, et al. Fetal liver stroma consists of cells in epithelial-to-mesenchymal transition[J]. Blood ,2003,101(8): 2973-2982.
    [80] Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine[J]. Muscle Nerve, 1995, 18(12): 1417-1426.
    [81] Makino S, Fukuda K, Miyoshi S, et al. Cardiomyocytes can be generated from marrow stromal cells in vitro[J]. J Clin Invest, 1999, 103(5): 697-705.
    [82] Planat-Be′nard V, Menard C, Andre′M, et al. Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells[J]. Circ Res, 2004, 94(2): 223-229.
    [83] Oswald J, Boxberger S, Jorgensen B, et al. Mesenchymal stem cells can be differentiated into endothelial cells in vitro[J]. Stem Cells, 2004, 22(3): 377-384.
    [84]李焕萍,刘春蓉,张永亮.骨髓间充质干细胞在医学领域中的研究与应用[J].中国组织工程研究与临床康复,2007,11(28):5622—5625.
    [85]胡资兵,曾荣,郭伟韬,林颢.骨髓间充质干细胞诱导分化特征[J].中国组织工程研究与临床康复,2008,12(43):8561-8566.
    [86] Pittenger MF ,Martin BJ . Mesenchymal stem cell s and t heir potential as cardiac t herapeutics[J ] . Circ Res, 2004, 95 (1): 9-20.
    [87] Soonpaa MH, Koh GY, Klug MG et al. Formation of nascent intercalated disks between grafted fetal cardiomyocytes and host myocardium[J]. Science, 1994, 1264(5155): 98-101.
    [88] Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis[J]. Science Feb, 1997, 275(5302):964–967.
    [89] Beltrami AP, Barlucchi L, Torella D, et al.Adult cardiac stem cells are multipotent and support myocardial regeneration[J]. Cell Sep, 2003, 114(6): 763–776.
    [90] Behfar A, Zingman LV, Hodgson DM,et al. Stem cell differentiation requires a paracrine pathway in the heart[J]. FASEB J, 2002, 16(12): 1558–1566.
    [91] Dai W, Field LJ, Rubart M, et al. Survival and maturation of human embryonic stem cell-derived cardiomyocytes in rat hearts[J]. J Mol Cell Cardiol, 2007, 43(4): 504–516.
    [92] Ballard VL, Edelberg JM. Stem cells for cardiovascular repair - The challenges of the aging heart[J]. J Mol Cell Cardiol, 2008, 45(4): 582-592.
    [93] Marsboom G, Janssens S. Endothelial progenitor cells: new perspectives and applications in cardiovascular therapies[J]. Expert Rev Cardiovasc Ther, 2008, 6(5): 687-701.
    [94] Matthew J, Callaghan, Daniel J, et al. Hyperglycemiainduced reactive oxygen species and impaired endothelial progenitor cell function[J]. Antioxid Redox Signal,2005,7(1112):1476-1482.
    [95] Rubart M, Soonpaa MH, Nakajima H, et al. Spontaneous and evoked intracellular calcium transients in donor-derived myocytes following intracardiac myoblast transplantation[J]. J Clin Invest, 2004, 114(6): 775–783.
    [96] Dib N, Michler RE, Pagani FD, et al. Safety and feasibility of autologous myoblast transplantation in patients with ischemic cardiomyopathy: four-year follow-up[J]. Circulation , 2005, 112(12): 1748–1755.
    [97] Shmelkov SV, Meeus S, Moussazadeh N, et al. Cytokine preconditioning promotes codifferentiation of human fetal liver CD133+ stem cells into angiomyogenic tissue[J]. Circulation , 2005, 111(9): 1175–1783.
    [98] Herrera MB, Bruno S, Buttiglieri S, et al. Isolation and characterization of a stem cell population from adult humanliver[J]. Stem Cells Dec, 2006, 24(12): 2840–2850.
    [99] Anderson PA, Muller-Borer BJ, Esch GL, et al. Calcium signals induce liver stem cells to acquire a cardiac phenotype[J]. Cell Cycle Jul, 2007, 6(13): 1565–1569.
    [100]Wakitani S,Saito T,Capian AI.Myogenic cells derived from rat bone marow mesenchymal stem cells exposed to 5-azacytidine[J].Muscle Nerve,1995, 18(12):1417-1426.
    [101]Makino S, Fukuda K, Miyoshi S, et al. Cardiomyocytes can be generated from marrow stromal cells in vitro[J]. J Clin Invest Mar, 1999, 103(5): 697–705.
    [102]Wang J, Shum-Tim D, Galipeau J, et al. Marrow stromal cells for cellularcardiomyoplasty:feasibility and potential clinical advantages[J]. J Thorac Cardiovasc Surg, 120(5): 999-1006
    [103]Strauer BE,Brehm M,Zeus T,et a1.Intracoronary, human autologousstem celI transplantation for myocardial regeneration following myocardialinfarction [J].Dtsch Med Wochenschr, 2001, 126(34—35):932-938.
    [104]Assmus B, Fischer-Rasokat U, Honold J, et al. Transcoronary transplantation of functionally competent bmcs is associated with a decrease in natriuretic peptide serum levels and improved survival of patients with chronic postinfarction heart failure: results of the topcare-chd registry[J]. Circ Res, 2007, 100(8): 1234–1241.
    [105]Assmus B, Honold J, Schachinger V, et al. Transcoronary transplantation of progenitor cells after myocardial infarction[J]. N Engl J Med, 2006, 355(12): 1222–1232.
    [106]Agbulut O,Menot ML,Li Z,et al. Temporal patterns of bone marrow cell differentiation following transplantation in doxorubicin induced cardiomyopathy [J]. Cardiovasc Res, 2003, 58(2): 451-459.
    [107]Nagaya N,Fujii T,1wase T,et at.Intravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesis[J]. Am J Physiol Heart Circ Physiol, 2004, 287(6): 2670-2676.
    [108]Noriteshi Nagaya,Kenji Kngawa,el a1.Transplantation of Mesenchymal Stem Cells lmpmves Cardiac Function in a Rat Model of Dilated Cardiomyopathy[J].Circulation,2005,112(8):1128-1135.
    [109]陈蓉,张国辉,蒋世忠,等.骨髓间充质干细胞静脉移植对扩张型心肌病大鼠毛细血管生成的影响[J].中国微循环,2009,13(3):161-165.
    [110]王建安,谢小洁,何红,等.骨髓间质干细胞移植治疗原发性扩张型心肌病的疗效与安全性[J].中华心血管病杂志,2006,34(2):107-110.
    [111]余更生,沈兴,田杰,等.同种异体骨髓间充质干细胞移植对扩张型心肌病模型兔心功能与结构及电生理的影响[J].中国组织工程研究与临床康复,2008,12(34):6776-6780.
    [112]Ip JE,Wu Y, Huang J, et al. Mesenchymal stem cells use integrinbetal not CXC chemokine receptor 4 for myocardial migration and engraftment[J]. Mol Biol Cell, 2007, 18(8): 2873–2882.
    [113]Wang Y, Haider HK, Ahmad N, et al. Combining pharmacological mobilization with intramyocardial delivery of bone marrow cells over-expressing VEGF is more effective for cardiac repair[J]. J Mol Cell Cardiol , 2006, 40(5): 736–745.
    [114]蔡继锐,刘仁光.骨髓干细胞动员治疗急性心肌梗死研究进展[J].中国心血管病研究杂志,2006,4(3):225-228.
    [115]Nathan M Novotny, Rinki Ray, Troy A Markel, et al.Stem Cell Therapy in Myocardial Repair and Remodeling[J]. Stem Cell Therapy, 2008,207( 3): 423-434.
    [116]Ebelt H, Jungblut M, Zhang Y, et al. Cellular cardiomyoplasty: improvement of left ventricular function correlates with the release of cardioactive cytokines[J]. Stem Cells , 2007, 25(1): 236–244.
    [117]Crisostomo PR, Wang M, Markel TA, et al. Stem cell mechanisms and paracrine effects: potential in cardiac surgery[J]. Shock, 2007, 28(4): 375–383.
    [118]Reffelmann T, Dow JS, Dai W, et al. Transplantation of neonatal cardiomyocytes after permanent coronary artery occlusion increases regional blood flow of infarcted myocardium[J]. JMol Cell Cardiol, 2003, 35(6): 607–613.
    [119]Kocher AA, Schuster MD, Szabolcs MJ, et al. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function[J]. Nat Med , 2001, 7(4): 430–436.
    [120]Orlic D, Kajstura J, Chimenti S, et al.Bone marrow stem cells regenerate infarcted myocardium[J]. Pediatr Transplant, 2003, Suppl(3): 86-88.
    [121]Badorff C, Brandes RP, Popp R, et al. Transdifferentiation of blood-derived human adult endothelial progenitor cells into functionally active cardiomyocytes [J]. Circulation , 2003, 107(7): 1024–1032.
    [122]Assmus B, Fischer-Rasokat U, Honold J, et al. Transcoronary transplantation of functionally competent BMCs is associated with a decrease in natriuretic peptide serum levels and improved survival of patients with chronic postinfarction heart failure: results of the TOPCARE-CHD Registry[J]. Circ Res, 2007, 100(8): 1234–1241.
    [123]Dohmann HF, Perin EC, Takiya CM, et al. Transendocardial autologous bone marrow mononuclear cell injection in ischemic heart failure: postmortem anatomicopathologic and immunohistochemical findings[J]. Circulation, 2005, 112(4): 521–526.
    [124]Huttmann A, Duhrsen U, Stypmann J, et al. Granulocyte colony-stimulating factor-induced blood stem cell mobilization in patients with chronic heart failure–feasibility, safety and effects on exercise tolerance and cardiac function [J]. Basic Res Cardiol , 2006, 101(1): 78–86.
    [125]Menasche P, Hagege AA, Vilquin JT, et al. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction[J]. J Am Coll Cardiol , 2003, 41(7): 1078–1083.
    [126]Siminiak T, Kalawski R, Fiszer D, et al. Autologous skeletal myoblast transplantation for the treatment of postinfarction myocardial injury: phase I clinical study with 12 months of follow-up[J]. Am Heart J, 2004, 148(3): 531–537.
    [127]Kawasuji M. Cell transplantation for cardiac repair[J]. Ann Thorac Cardiovasc Surg, 2006, 12(2): 1–3.
    [128]黄裕立,麦炜颐.骨髓间充质干细胞移植在心脏再生医学中的分子机制[J].国际内科学杂志,2008,35(9):531-535.
    [129]Guo J,Lin GS,Bao CY,et aL Anti-Intlammation Role for MesenchymM Stem Cells Transplantation in Myocardial Infarction[J].Inflammation,2007,30(34):97-104.
    [130] Ohnishi S,Yarmgawa B,Tanaka K,et a1.Transplantation of mesenchymal stem cells attenuates myocardial injury anddysfunction in a rat model of acutemyocarditis.J Mol Cell Cardiol,2007,42(1):88—97.
    [131]Badorff C, Brandes RP, Popp R, et al. Transdifferentiation of blood-derived human adult endothelial progenitor cells into functionally active cardiomyocytes[J]. Circulation, 2003, 107(7): 1024–1032.
    [132]Fukuhara S, Tomitas S, Yamashivo S, et al. Direct cell-cell interactior of cardiomyocytes is key for bone marrow stromal cells to go into cardial lineage in vitro[J]. Thorac Cardiovasc Surg, 2003, 125(6): 1470-1480.
    [133]Park JS, Huang NF, Kurpinski KT, et al. Mechanobiology of mesenchymal stem cells and their use in cardiovascular repair[J].Front Biosci, 2007, 12: 5098–5116.
    [134]陈沅.骨髓间充质干细胞分化为心肌细胞的分子机制及其微环境[J].实用儿科临床杂志,2008,23(1):4-6.
    [135]Liu XN, Yin Q, Zhang H,et al.Tissue extracts from infarcted myocardium of rats in promoting the differentiation of bone marrow stromal cells into cardiomyocyte-like cells[J]. Biomed Environ Sci, 2008, 21(2): 110-117.
    [136]袁岩,陈连凤,张抒扬,等.心肌细胞裂解液对骨髓间充质干细胞向心肌细胞分化诱导作用的研究[J].中华心血管病杂志,2005,33(2):170-173.
    [137]刘建华,洪亮,钟雪云,等.心肌梗死大鼠血清对大鼠骨髓间充质干细胞分化为心肌细胞的影响[J].中国组织工程临床研究与临床康复,2009,13(14):2797-2800.
    [138]Piccini JP,Hranitzky P. Diagnostic monitoring strategies in heart failure management[J]. Am Heart J, 2007, 153(4 Suppl):12-17.
    [139]Nassiri SM,Khaki Z,Soleimani M,et al. The similar effect of transplantation of marrow-derived mesenchymal stem cells with or without prior differentiation induction in experimental myocardial infarction[J]. J Biomed Sci, 2007, 14(6): 745-755.
    [140]Nagaya N,Kangawa K,Itoh T,et al. Transplantation of mesenchymal stem cells improves cardiac function in a rat model of dilated cardiomyopathy[J]. Circulation, 2005, 112(8):1128-1135.
    [141]Wang TZ,Ma AQ,Xu ZY,et al. Differentiation of mesenchymal stem cells intocardiomyocytes induced by cardiomyocytes[J]. Zhong Nan Da Xue Xue Bao Yi Xue Ban, 2005, 30(3): 270-275.
    [142]Zhou H,Jin Z,Liu J,et al. Mesenchymal stem cells might be used to induce tolerance in heart transplantation[J]. Med Hypotheses, 2008, 70(4): 785-787.
    [143]Iop L,Chiavegato A,Callegari A,et al. Different cardiovascular potential of adult- and fetal-type mesenchymal stem cells in a rat model of heart cryoinjury[J]. Cell Transplant, 2008, 17(6): 679-694.
    [144].Silva GV,Litovsky S,Assad JA,et al. Mesenchymal stem cells differentiate into an endothelial phenotype,enhance vascular density,and improve heart function in a canine chronic ischemia model[J]. Circulation, 2005, 111(2): 150-156.
    [145]Iwase T,Nagaya N,Fujii T,et al. Comparison of angiogenic potency between mesenchymal stem cells and mononuclear cells in a rat model of hindlimb ischemia[J]. Cardiovasc Res, 2005, 66(3): 543-551.
    [146]Moscoso I,Centeno A,Lopez E,et al. Differentiation "in vitro" of primary and immortalized porcine mesenchymal stem cells into cardiomyocytes for cell transplantation[J]. Transplant Proc, 2005, 37(1): 481-482.
    [147]Xu W,Zhang X,Qian H,et al. Mesenchymal stem cells from adult human bone marrow differentiate into a cardiomyocyte phenotype in vitro[J]. Exp Biol Med (Maywood), 2004, 229(7): 623-631.
    [148]Rangappa S,Entwistle JW,Wechsler AS,et al. Cardiomyocyte-mediated contact programs human mesenchymal stem cells to express cardiogenic phenotype[J]. J ThoracCardiovasc Surg, 2003, 126(1): 124-132.
    [149]Miyahara Y,Nagaya N,Kataoka M,et al. Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction[J]. Nat Med, 2006, 12(4): 459-465.
    [150]Chang SA,Lee EJ,Kang HJ,et al. Impact of myocardial infarct proteins and oscillating pressure on the differentiation of mesenchymal stem cells:effect of acute myocardial infarction on stem cell differentiation[J]. Stem Cells,2008;26(7): 1901-1912.
    [151]Muscari C,BonaféF,Carboni M,et al. Difluoromethylornithine stimulates early cardiac commitment of mesenchymal stem cells in a model of mixed culture with cardiomyocytes[J]. J Cell Biochem, 2008, 103(4): 1046-1052.
    [152]Yamada Y,Yokoyama S,Wang XD,et al. Cardiac stem cells in brown adipose tissue express CD133 and induce bone marrow nonhematopoietic cells to differentiate into cardiomyocytes[J]. Stem Cells, 2007, 25(5):1326-1333.
    [153]Wang T,Xu Z,Jiang W,et al. Cell-to-cell contact induces mesenchymal stem cell to differentiate into cardiomyocyte and smooth muscle cell[J]. Int J Cardiol, 2006, 109(1):74-81.
    [154].赵鹏,李玉明,庞伟,等. 5-氮胞苷不同时点对骨髓间质干细胞的诱导分化作用[J].武警医学,2006,17(9):670-672.
    [155]Tomita S, Li RK, Weisel RD et al. Autologous transplantation of bone marrow cells improves damaged heart function.Circulation, 1999, 100(19 suppl): 247-256.
    [156]吴铁军,蔡振杰,俞世强等.不同浓度5 -氮胞苷对骨髓间质干细胞体外诱导分化的作用[J].第四军医大学学报,2003,24(15):1373-1375.
    [157]方志成,王玮,刘志祥,等. 5-氮胞苷诱导对骨髓间充质干细胞凋亡影响的实验研究[J].心血管康复医学杂志,2006,15(2):111-113.
    [158]Panteghini M. Present is sues in the determination of troponins and other markers of cardiac damage[J]. Clin Biochem, 2000, 33(3): 161-166.
    [159]Moscoso I, centrno A, lopez E, et al. Differentiation "in vitro" of primary and immortalized porcine mesenchymal stem cells into cardiomyocytes for cell transplantation[J]. Transplant Proc, 2005, 37(1): 481-482.
    [160].付国伟,赵文增,文冰,等.不同浓度的5-氮胞苷对人骨髓间充质干细胞向心肌细胞分化的影响[J].心脏杂志(Chin Heart J ),2008,20 (5):545-548.
    [161]Shengwen Calvin Li, Lang Wang, Hong Jiang, et al. Stem cell engineering for treatment of heart diseases: Potentials and challenges[J]. Cell Biology International, 2009, 33 (3): 255-267.
    [162] Pankaj Kumar Mishra. Bone marrow-derived mesenchymal stem cells for treatment of heart failure: is it all paracrine actions and immunomodulation? [J]Journal of Cardiovascular Medicine,2008, 9(2): 122–128.
    [163]Massimo F. Piepoli. Transplantation of progenitor cells and regeneration of damaged myocardium: more facts or doubts? Insights from experimental and clinical studies[J]. Journal of Cardiovascular Medicine, 2009, 10(8): 624–634.
    [164]Recchia FA, Lionetti V. Animal models of dilated cardiomyopathy for translational research[J]. Vet Res Commun, 2007, 31(Suppl 1): 35-41.
    [165]黄荣杰,刘唐威,伍伟锋,等.呋喃唑酮制备大鼠扩张型心肌病模型[J].中国病理生理杂志,2005,21 (11) :2147-2150.
    [166]邓海,龚云,曾晶晶.小剂量阿霉素间断静注诱导兔扩张型心肌病模型的实验研究[J].今日药学,2008,18 (3):80-82.
    [167]Nithipongvanitch R, Ittarat W, Cole MP, et al. Mitochondrial and nuclear p53 localization in cardiomyocytes: redox modulation by doxorubicin (Adriamycin)? [J]. Antioxid Redox Signal, 2007, 9(7): 1001-1008.
    [168]Berthiaume JM, Wallace KB. Adriamycin-induced oxidative mitochondrial cardiotoxicity[J]. Cell Biol Toxicol, 2007, 23(1): 15-25.
    [169]Viale PH, Yamamoto DS.Cardiovascular toxicity associated with cancer treatment[J]. Clin J Oncol Nurs, 2008, 12(4): 627-638.
    [170]Hayward R, Hydock DS.Doxorubicin cardiotoxicity in the rat: an in vivo characterization[J].J Am Assoc Lab Anim Sci, 2007, 46(4): 20-32.
    [171]Bekeredjian R, DeFilippis NJ, Siddiquee Z, et al.Transcriptional analysis of doxorubicin-induced cardiotoxicity[J]. Yi X Am J Physiol Heart Circ Physiol[J], 2006, 290(3): 1098-1102.
    [172]Hou XW, Son J, Wang Y, et al. Granulocyte colony-stimulating factor reduces cardiomyocyte apoptosis and improves cardiac function in adriamycin-induced cardiomyopathy in rats[J]. Cardiovasc Drugs Ther, 2006 , 20(2): 85-91.
    [173]钟国强,马国添,刘唐威,等.腹腔注射阿霉素建立扩张型心肌病大鼠模型的方法学改进.基础医学与临床[J],2007,27(11):1289-1292.
    [174]Stamm C, et al. Cell Therapy for Heart Disease: Great Expectations, As Yet Unmet[J]. Heart Lung Circulation, 2009, 18(4): 245-256.
    [175]Dongxia Ge, Xiaojing Liu, Liang Li, et al. Chemical and physical stimuli induce cardiomyocyte differentiation from stem cells[J]. Biochemical and Biophysical Research Communications, 2009, 381(3): 317–321.
    [176]蔡继锐,刘仁光.骨髓干细胞动员治疗急性心肌梗死研究进展[J].中国心血管病研究杂志,2006,4(3):225-228.
    [177]Koc ON, Day J, Nieder M, et al. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy(MLD) and Hurler syndrome (MPS2IH) [J]. Bone Marrow Transplant, 2002, 30 (4): 215-222.
    [178]郭牧,宋昱.脑利钠肽的临床应用[J]中国心血管病研究,2008, 6(5):386-389.
    [179]冯善伟,姚晓黎,李中,等. Brdu体外标记大鼠骨髓间充质干细胞的研究[J].第一军医大学学报,2005,25(2):184-186.
    [180]Edelberg JM, Tang L, Hattori K, et al. Young adult bone marrow-derived endothelial precursor cells restore aging-impaired cardiac angiogenic function[J]. Circ Res, 2002, 90(10): E89–93.
    [181]V.L.T. Ballard, J.M. Edelberg. Stem cells for cardiovascular repair—The challenges of the aging heart[J]. Journal of Molecular and Cellular Cardiology , 2008, 45(4): 582–592.
    [182]Counter CM. The roles of telomeres and telomerase in cell life span[J]. Mutat Res, 1996, 366(1): 45–63.
    [183]Noritoshi Nagaya, Kenji Kangawa, et al. Intravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesis[J]. Am J Physiol Heart Circ Physiol, 2004, 287(6): 2670-2676.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700