低能量激光治疗对成骨细胞及骨愈合的影响及相关机制的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
激光是一种新型光源,具有方向性好、亮度高、单色性好、相干性好等优异的物理特性,可以在医学的诊断和治疗方面发挥巨大作用。低能量激光治疗(Low-Level-Laser- Therapy,LLLT)具有生物刺激效应,学者们发现它可以减轻疼痛、炎症、水肿,促进伤口、深层组织和神经的愈合。已有大量的体外和动物实验的研究证实LLLT可以促进成骨细胞增殖、分化和骨组织愈合,但应用于口腔种植领域的研究较少。LLLT的有效照射剂量和照射方式尚无统一标准,现仍有一些负面报道认为它对骨组织愈合没有作用,因此还不是一个被广泛接受的治疗手段。本研究尝试将LLLT应用于口腔种植领域,以验证LLLT是否能够有效的促进成骨细胞增殖、分化和骨形成,并探讨不同照射方式的LLLT是否有区别,为其应用于临床提供理论依据。
     本研究采用MTT、流式细胞术、细胞化学钙钴法检测人成骨样细胞MG-63的增殖、细胞周期、细胞凋亡和碱性磷酸酶(alkaline phosphatase,ALP)的表达,观察了LLLT对MG-63生物学特性的影响。采用免疫组化、RT-PCR和Western Blot的方法检测骨桥蛋白(osteopontin,OPN)、I型胶原(collagen I,coll I)的mRNA的表达量和蛋白表达量,观察了LLLT对MG-63细胞外基质蛋白的影响。此外,本研究观察了LLLT对大鼠股骨实验性骨缺损的刺激作用。结果表明,3.75 J·cm-2照射剂量的LLLT可以促进体外人成骨样细胞MG-63的增殖,改变细胞周期时相,抑制细胞凋亡,促进MG-63分泌成骨细胞早期标志物ALP,促进细胞外基质蛋白OPN、coll I的表达,30 J·cm-2照射剂量的LLLT可以促进大鼠股骨实验性骨缺损的骨愈合。此外,实验组中多次照射和单次照射比较并无不同。
     综上所述,本研究证实了LLLT可以促进成骨细胞增殖、分化,促进动物模型的骨形成,LLLT可能对种植术后的骨愈合有促进作用,具有临床意义,为LLLT应用于口腔种植领域打下坚实的实验基础。
As a new light source, a laser possesses many excellent physical characteristics such as tiny divergence angle, brightness, monochromaticity and coherence. The era of laser medicine came when the American scientist Maiman invented the first laser in 1960. Lasers have played an important role in medical diagnosis and therapy. According to the power of the laser utilized, there are two areas of laser therapy: one is for high power lasers which can be used in ablation, cutting, solidification and hemostasis; the other is for low power lasers which can be used for irradiating human tissue, without causing damage, and then promoting its healing.
     Low level laser(LLL),also called low intensity/power laser, cold laser, soft laser, weak laser and so on, is one kind of visible light and infrared light, which possesses satisfactory monochromatism, between 600~950nm of the wavelength, below 250mW of the output power. At the same time, instead of heat effect, laser has biochemical effects which could not result in irreversible damage to bio-tissue, but rather a similar effect obtained by physical factors such as ultrasound, apuncture and moxibustion. That is called“biostimulation”, and now“photo- biomodulation”.The corresponding therapy is called low-level-laser-therapy (LLLT).For medical workers, there are three areas where LLLT could play an important role: 1) triggering immunological chain reaction, promoting wound healing and tissue repair; 2) reduction of inflammatory process and its associated pain and edema; 3) relief of neurogenic pain influence. Many in vitro and in vivo studies have offered theoretical founation for the clinical application of LLLT. Bone defect is common in oral implantology, which can influence the bone mass and therapeutic schemes. There are several ways of bone repair, for instance, bone graft which includs autogenous bone graft, allogenic bone graft,artificial bone graft, physical stimulus which includes electromagnetic stimulation, recompression therapy, ultrasound. LLLT is a physical treatment which has been demonstrated to improve the proliferation and differen- tiation of osteoblast and bone healing in animal models.
     In spite of many convicing study of the positive effects of LLLT on bone healing, there are several studies confirmed that LLLT has no effect on osteoblast and bone healing. LLLT is not a widely accepted therapy. Therefore, more experiments are needed to verify the effective dose and irradiation method of LLLT. Meanwhile, few studies were applied to oral implantology. No research is found in domestic area. If LLLT can be applied to bone healing in oral implatology, LLLT may help improve the success ratio of implant.
     In this study, the effect of LLLT on proliferation,differentiation of MG-63 and bone healing in an animal model was detected. The different methods of LLLT was investigated too. This study can provide a theoretical foundation for the application of LLLT in oral implantology.
     1 Effect of LLLT on biological characteristics of MG-63
     OBJECTIVE: To elavuate the biological characteriscs of human osteoblast-like cell MG-63 which was used as an osteoblastic model.
     METHOD: MG-63 was seeded into H-DMEM with 10% fetal bovine serum. There were 3 groups randomly divided: laser group 1 (L1), laser group 2 (L2), control group(C). The laser was 808 nm, outpower 66 mW, power density 33 mW·cm-2. L1 was multi-irradiated group which was irradiated 3 consecutive days for 40 s per day, that was 1.25 J·cm-2 per day. L2 was single-irradiated group which was irradiated for 120 s on the 1st day, that was 3.75 J·cm-2 and was not irradiated on 2nd and 3rd days.The total dose of both of laser groups was 3.75 J·cm-2 . C was not irradiated. The proliferation, cell cycle, apoptosis, ALP of MG63 were measured by MTT, flow cytometry(FCM) and cytochemical calcium-cobalt method.
     RESULTS: On the 4th day after single irradiation, MTT of laser groups was higher than that of control group(P<0.05). Compared to control group, laser groups presented more S-stage cells, less G1-stage cells and higher proliferation index(PI,PI=S + G2/ G1 +S + G2)(P<0.05). The apoptosis rates in laser groups were lower than that in control group with significant difference (P<0.05). The expression of ALP in laser groups were higher than that in control group (P<0.05). However, for these 4 indexes, there was no significant difference between laser group 1 and laser group 2 (P>0.05).
     2 Effect of LLLT on extracellular matrix protein of MG-63
     OBJECTIVE: To investigated the effect of LLLT on mRNA and protein expression of OPN,coll I of MG-63, and to detect the effect of LLLT on ECMP and the mechanism of LLLT.
     METHOD: MG-63 was seeded into H-DMEM with 10% fetal bovine serum. There were 3 groups randomly designed: laser group 1(L1), laser group 2(L2), control group(C). The laser was 808 nm, outpower 66 mW, power density 33 mW·cm-2 . L1 was multi-irradiated group which was irradiated 3 consecutive days for 40s per day, that was 1.25 J·cm-2 per day. L2 was single-irradiated group which was irradiated for 120s on the 1st day, that was 3.75 J·cm-2 and was not irradiated on the 2nd and 3rd days.The total dose of both of laser groups was 3.75 J·cm-2. C was not irradiated. The mRNA and protein expression of OPN,coll I of MG63 were measured by immunohistochemical method , RT-PCR and Western Blot.
     RESULTS: On the 4th day after single irradiation, with immunohistochemical method, the expression of OPN,coll I in laser groups were higher than that in control group (P<0.05). With RT-PCR method, the expression of OPN,coll I in laser groups were higher than that in control group (P<0.05). With western blot, the expression of OPN,coll I in laser groups were higher than that in control group (P<0.05).However, for these 3 indexes, there was no significant difference between laser group 1 and laser group 2 (P>0.05).
     3 Stimulatory effect of LLLT on bone defect created surgically in rat femur
     OBJECTIVE:To observe the stimulatory effect of LLLT on bone defect sites created surgically in rat femur, and to evaluate the histological changes including inflammation and bone formation around bone defect.
     METHOD: Thirty rabbits were randomly divided into 3 groups: laser group 1, laser gourp 2 and control group. Each group was divided into 2 sub-groups according to different sacrificed time: 15d sub-group and 30d sub-group. The laser was 808nm, output 2.6 mW,power density 83 mW cm-2 .L1 was multi-irradiated group which was irradiated 3 consecutive days for 2 min per day, that was 10 J·cm-2 per day. L2 was single-irradiated group which was irradiated for 6 min on the 1st day, that was 30 J·cm-2 and was not irradiated on the 2nd and 3rd days.The total dose of both of laser groups was 30 J·cm-2, C was not irradiated. After an general anesthesia, the femur was perforated with a surgical bone drill 2 mm diameter×1 mm depth. Wound closure was done in layers. The rabbits were killed on 15 d and 30 d after the surgery. The femur was removed, then fixed in 4% paraformaldehyde, decalcified, dehydrated, embedded in paraffin and sectioned for HE staining. The inflammation and bone formation of each group was analyzed under a light micorscope.
     RESULTS: Control group: At 15 days, the presence of disorganized connective tissue with moderate or severe inflammation. was observed. At 30 days, the wounds were partially filled by newly formed bone tissue composed of numerous osteoblasts differentiating into osteocytes with mild inflammation. Laser group: the presence of organized connective tissue with mild inflammation. was observed. At 30 days, no inflammation was observed. All specimens presented new bone formation and had the defect almost completely filled with bone tissue with characteristics of secondary bone.A large number of blood capillaries were clearly observed in this period. The inflammation score of laser groups were lower than that of control group with significant difference (P<0.05), meanwhile, the bone formation score of laser groups were higher than that of control group with significant difference (P<0.05). However, there was no significant difference between laser group 1 and laser group 2 (P>0.05).
     In conclusion, LLLT with proper dose may have a positive effect on in vitro or in vivo experiments on early stage, for instance, promoting proliferation and differentiation, improving bone healing of bone defect created in animal model. In this study, there was no different between multi-irradiated group and single-irradiated group. More research on the difference of their biological effect is needed in future.
引文
[1]罗湘杭,廖二元,周后德,等.人成骨肉瘤MG263细胞分化特性及分化过程中的基因表达[J].湖南医科大学学报,2001,26(2):701-011.
    [2]Kim SJ, Moon SU, Kang SG,et al. Effects of low-level laser therapy after Corticision on tooth movement and paradental remodeling[J]. Lasers Surg Med,2009,41(7):524-533.
    [3]Frigo L, Fávero GM, Campos Lima HJ, et al. Low-Level Laser Irradiation (InGaAlP-660 nm) Increases Fibroblast Cell Proliferation and Reduces Cell Death in a Dose-Dependent Manner[J]. Photomed Laser Surg,2009 Sep 21.
    [4]Tuby H, Maltz L, Oron U. Implantation of low-level laser irradiated mesenchymal stem cells into the infarcted rat heart is associated with reduction in infarct size and enhanced angiogenesis[J]. Photomed Laser Surg,2009,27(2):227-233.
    [5]Saracino S, Mozzati M, Martinasso G, et al. Superpulsed laser irradiation increases osteo- blast activity via modulation of bone morphogenetic factors[J]. Lasers Surg Med,2009, 41(4):298-304.
    [6]Martinasso G, Mozzati M, Pol R, et al. Effect of superpulsed laser irradiation on bone formation in a human osteoblast-like cell line[J]. Minerva Stomatol,2007,56(1-2):27-30.
    [7]王学金,刘慧颖,郝晓宁.牙龈卟啉单胞菌对纯钛表面成骨细胞表达OPG和RANKL mRNA的影响[J].口腔医学研究,2008,24(4):483-783.
    [8]韩雪,宋九余,张力,等.阳极氧化处理后的新型钛合金表面成骨细胞OPG、RANKL基因表达研究[J].临床口腔医学杂志,2008,24(4):203-206.
    [9]刘慧颖,王学金,伊哲,等.氟离子注入钛表面改性对成骨细胞黏着斑形成的影响[J].华西口腔医学杂志,2008,26(2):137-143.
    [10]赵震锦,张翀,张扬,等.表面粗糙度对种植体钛片表面成骨细胞的影响[J].解剖科学进展,2008,14(2):178-182.
    [11]马红梅,吴琳,艾红军,等.纳米羟基磷灰石/胶原/聚乳酸支架材料对人成骨细胞早期附着影响的实验研究[J].生物医学工程与临床,2007,11(3):161-163.
    [12]狄升蒙,骞爱荣,田宗成,等.强磁重力环境对人成骨细胞发动蛋白-2表达和定位的影响[J].细胞生物学杂志,2008,30(6):766-770.
    [13]Hou JF, Zhang H, Yuan X, et al. In vitro effects of low-level laser irradiation for bonemarrow mesenchymal stem cells: proliferation, growth factors secretion and myogenic differentiation[J]. Lasers Surg Med, 2008,40(10):726-733.
    [14]Eduardo Fde P, Bueno DF, de Freitas PM,et al. Stem cell proliferation under low intensity laser irradiation: a preliminary study[J]. Lasers Surg Med,2008,40(6):433-438.
    [15]Mirzaei M, Bayat M, Mosafa N,et al. Effect of low-level laser therapy on skin fibroblasts of streptozotocin-diabetic rats[J]. Photomed Laser Surg,2007,25(6):519-525.
    [16]Pires Oliveira DA, de Oliveira RF, Zangaro RA,et al. Evaluation of low-level laser therapy of osteoblastic cells[J]. Photomed Laser Surg,2008,26(4):401-404.
    [17]Arisu HD, Türk?z E, Bala O. Effects of Nd:Yag laser irradiation on osteoblast cell cultures[J]. Lasers Med Sci,2006,21(3):175-180.
    [18]Xu M, Deng T, Mo F,et al. Low-intensity pulsed laser irradiation affects RANKL and OPG mRNA expression in rat calvarial cells[J]. Photomed Laser Surg, 2009,27(2):309-315.
    [19]Fukuhara E, Goto T, Matayoshi T,et al. Optimal low-energy laser irradiation causes temporal G2/M arrest on rat calvarial osteoblasts[J].Calcif Tissue Int, 2006,79 (6):443-450.
    [20]Shefer G, Partridge TA, Heslop L,et al. Low-energy laser irradiation promotes the survival and cell cycle entry of skeletal muscle satellite cells[J]. J Cell Sci,2002,115:1461-1469.
    [21]Carnevalli CM, Soares CP, Zangaro RA,et al. Laser light prevents apoptosis in Cho K-1 cell line[J]. J Clin Laser Med Surg,2003,21(4):193-196.
    [22]Zhang L, Xing D, Zhu D,et al. Low-power laser irradiation inhibiting Abeta25-35-induced PC12 cell apoptosis via PKC activation[J]. Cell Physiol Biochem,2008,22(1-4):215-222.
    [23]Wu S, Xing D, Wang F,et al.Mechanistic study of apoptosis induced by high-fluence low-power laser irradiation using fluorescence imaging techniques[J]. J Biomed Opt,2007, 12(6):064015.
    [24]Haxsen V, Schikora D, Sommer U,et al. Relevance of laser irradiance threshold in the induction of alkaline phosphatase in human osteoblast cultures[J]. Lasers Med Sci,2008, 23(4):381-384.
    [25]Karu T. The science of low-power laser therapy[M]. Amsterdam:Gord Brea Sci Pub, 1998, 53-94.
    [26]Pinheiro AL, Cavalcanti ET, Pinheiro TI,et al. Low-Level Laser Therapy in management of disorders of the maxillofacial region[J]. J Clin Laser Med Surg,1997,15:181–183.
    [27]Powell K, Low P, McDonnell PA,et al. The Effect of Laser irradiation on Proliferation of Human Breast Carcinoma, Melanoma, and Immortalized Mammary Epithelial Cells[J]. Photomed Laser Surg, 2010 ,28(1):115-123.
    [28]Hawkins D, Abrahamse H. Effect of multiple exposures of low-level laser therapy on the cellular responses of wounded human skin fibroblasts[J]. Photomed Laser Surg,2006, 24(6):705-714.
    [29]Karu TI, Afanas'eva NI. Cytochrome c oxidase as the primary photoacceptor upon laser exposure of cultured cells to visible and near IR-range light[J].Dokl Akad Nauk,1995,342 (5):693-695.
    [30]Eells JT, Wong-Riley MT, VerHoeve J,et al. Mitochondrial signal transduction in accel- erated wound and retinal healing by near-infrared light therapy[J]. Mitochondrion, 2004,4(5-6):559-567.
    [31]Karu TI, Kolyakov SF. Exact action spectra for cellular responses relevant to photo- therapy[J]. Photomed Laser Surg,2005,23:355-361.
    [32]Yu W, Naim JO, McGowan M,et al. Photomodulation of oxidative metabolism and electron chain enzymes in rat liver mitochondria[J]. Photochem Photobiol,1997, 66:866-871.
    [33]Passarella S. He-Ne laser irradiation of isolated mitochondria[J]. J Photochem Photobiol B,1989,3:642-643.
    [34]Liu H, Colavitti R, Rovira II,et al. Redox-dependent transcriptional regulation[J].Circ Res,2005,97:967-974.
    [35]Saygun I, Karacay S, Serdar M,et al. Effects of laser irradiation on the release of basic fibroblast growth factor (bFGF), insulin like growth factor-1 (IGF-1), and receptor of IGF-1 (IGFBP3) from gingival fibroblasts[J]. Lasers Med Sci,2008,23(2):211-215.
    [36]Safavi SM, Kazemi B, Esmaeili M,et al. Effects of low-level He-Ne laser irradiation on the gene expression of IL-1beta, TNF-alpha, IFN-gamma, TGF-beta, bFGF, and PDGF in rat's gingiva[J]. Lasers Med Sci,2008,23(3):331-335.
    [37]Gavish L, Perez LS, Reissman P,et al.Irradiation with 780 nm diode laser attenuates inflammatory cytokines but upregulates nitric oxide in lipopolysaccharide-stimulated macrophages: Implications for the prevention of aneurysm progression[J]. Lasers SurgMed,2008,40(5):371-378.
    [38]Nissan J, Assif D, Gross MD,et al.Effect of low intensity laser irradiation on surgically created bony defects in rats[J]. J Oral Rehabil,2006,33(8):619-924.
    [39]Miyata H, Genma T, Ohshima M,et al.Mitogen-activated protein kinase/extracellular signal-regulated protein kinase activation of cultured human dental pulp cells by low-power gallium-aluminium- arsenic laser irradiation[J]. Int Endod J,2006,39(3):238-244.
    [40]Aimbire F, Bjordal JM, Iversen VV,et al. Low level laser therapy partially restores trachea muscle relaxation response in rats with tumor necrosis factor alpha-mediated smooth airway muscle dysfunction[J]. Lasers Surg Med,2006,38(8):773-778.
    [41]Kipshidze N, Petersen JR, Vossoughi J,et al. Low-power laser irradiation increases cyclic GMP synthesis in penile smooth muscle cells in vitro[J].J Clin Laser Med Surg, 2000, 18(6): 291-294.
    [42]Alaluf S, Muir-Howie H, Hu HL,et al. Atmospheric oxygen accelerates the induction of a post-mitotic phenotype in human dermal fibroblasts: the key protective role of glutathione [J]. Differentiation, 2000,66:147-155.
    [43]Yamamoto M, Tamura K, Hiratsuka K,et al. Stimulation of MCM3 gene expression in osteoblast by low level laser irradiation[J].Lasers Med Sci,2001,16(3):213-217.
    [44]Hamajima S, Hiratsuka K, Kiyama-Kishikawa M,et al. Effect of low-level laser irradiation on osteoglycin gene expression in osteoblasts[J].Lasers Med Sci,2003,18(2):78-82.
    [45]Stein A, Benayahu D, Maltz L,et al.Low-level laser irradiation promotes proliferation and differentiation of human osteoblasts in vitro[J]. Photomed Laser Surg, 2005, 23(2): 161-166.
    [46]Stein E, Koehn J, Sutter W,et al. Initial effects of low-level laser therapy on growth and differentiation of human osteoblast-like cells[J].Wien Klin Wochenschr, 2008,120 (3-4): 112-117.
    [47]Chen CH, Tsai JL, Wang YH,et al. Low-level laser irradiation promotes cell proliferation and mRNA expression of type I collagen and decorin in porcine Achilles tendon fibroblasts in vitro[J].J Orthop Res,2009,27(5):646-650.
    [48]吴佳奇,张晓刚,王清.骨缺损的基因治疗[J].泸州医学院学报,2004,27(3): 268-270.
    [49]Miloro M, Miller JJ, Stoner JA. Low-level laser effect on mandibular distractionosteogenesis[J]. J Oral Maxillofac Surg,2007,65(2):168–176.
    [50]Trelles MA, Mayayo E. Bone fracture consolidates faster with low-power laser[J].Lasers Surg Med,1987,7(1):36-45.
    [51]Hedner E, Linde A. Efficacy of bone morphogenetic protein (BMP) with osteopromotive membranes–an experimental study in rat mandibular defects[J]. Eur J Oral Sci,1995,103(4): 236–241.
    [52]Dahlin C, Linde A, Gottlow J,et al. Healing of bone defects by guided tissue regen- eration[J]. Plast Reconstr Surg,1988,81(5):672–676.
    [53]Bell R, Beirne OR. Effect of hydroxylapatite, tricalcium phosphate, and collagen on the healing of defects in the rat mandible[J]. J Oral Maxillofac Surg,1988,46(7):589–594.
    [54]Ray RD, Holloway JA. Bone implants:preliminary report of an experimental study[J]. J Bone Joint Surg Am,1957,39:1119–1128.
    [55]Nicola RA, Jorgetti V, Rigau J,et al.Effect of low-power GaAlAs laser (660 nm) on bone structure and cell activity:an experimental animal study[J].Lasers Med Sci, 2003,18(2): 89-94.
    [56]Khadra M, Kasem N, Haanaes HR,et al. Enhancement of bone formation in rat calvarial bone defects using low-level laser therapy[J]. Oral Surg Oral Med Oral Pathol Oral Radiol Endod,2004,97(6):693-700.
    [57]Ruhe PQ, Kroese-Deutman HC, Wolke JG,et al. Bone inductive properties of rhBMP-2 loaded porous calcium phosphate cement implants in cranial defects in rabbits[J]. Biomaterials,2004,25(11):2123–2132.
    [58]Spector JA, Luchs JS, Mehrara BJ, et al. Expression of bone morphogenetic proteins during membranous bone healing[J]. Plast Reconstr Surg,2001,107(1):124–134.
    [59]Yasko AW, Lane JM, Fellinger EJ,et al. The healing of segmental bone defects, induced by recombinant human bone morphogenetic protein (rhBMP-2). A radiographic, histological, and biomechanical study in rats[J]. J Bone Joint Surg Am,1992,74(5):659–670.
    [60]Garavello-Freitas I, Baranauskas V, Joazeiro PP,et al. Low-power laser irradiation improves histomorphometrical parameters and bone matrix organization during tibia wound healing in rats[J].J Photochem Photobiol B,2003,70(2):81–89.
    [61] Bell R, Beirne OR. Effect of hydroxylapatite, tricalcium phosphate, and collagen on thehealing of defects in the rat mandible[J]. J Oral Maxillofac Surg,1988,46(7):589–594.
    [62]Nagahara K, Mouri K, Kanematsu N,et al. An in vivo evaluation of an osteoinductive implantable material produced by osteoblastic cells in vitro[J].Int J Oral Maxillofac Implants,1994,9(1):41–48.
    [63]Ono I, Ohura T, Murata M,et al. A study on bone induction in hydroxyapatite combined with bone morphogenetic protein[J].Plast Reconstr Surg,1992,90(5):870–879.
    [64]Hedner E, Linde A. Efficacy of bone morphogenetic protein (BMP) with osteopromotive membranes–an experimental study in rat mandibular defects[J]. Eur J Oral Sci,1995,103(4): 236–241.
    [65]Guzzardella GA, Fini M, Torricelli P, Giavaresi G, Giardino R. Laser stimulation on bone defect healing: An in vitro study[J]. Lasers Med Sci, 2002,17(3):216–220.
    [66]Pinheiro AL, Limeira Junior Fde A, Gerbi ME,et al. Effect of 830-nm laser light on the repair of bone defects grafted with inorganic bovine bone and decalcified cortical osseus membrane[J].J Clin Laser Med Surg,2003,21(5):301–306.
    [67]Dahlin C, Sandberg E, Alberius P,et al. Restoration of mandibular nonunion bone defects. An experimental study in rats using an osteopromotive membrane method[J].Int J Oral Maxillofac Surg,1994,23(4):237–242.
    [68]Aukhil I. Biology of wound healing[J]. Periodontol 2000,2000,22: 44–50.
    [69]Sodek J, McKee MD. Molecular and cellular biology of alveolar bone[J]. Periodontol 2000,2000,24:99–126.
    [70]Lozano AJ, Cestero HJ Jr, Salyer KE. The early vascularization of onlay bone grafts[J]. Plast Reconstr Surg,1976,58(3):302–305.
    [71]Garavello-Freitas I, Baranauskas V, Joazeiro PP,et al.Low-power laser irradiation improves histomorphometrical parameters and bone matrix organization during tibia wound healing in rats[J].J Photochem Photobiol B,2003,70(2):81–89.
    [72]Freitas AC, Pinheiro AL, Miranda P,et al. Assessment of anti-inflammatory effect of 830nm laser light using C-reactive protein levels[J]. Braz Dent J,2001,12(3):187–190.
    [73]Silva Junior AN, Pinheiro AL, Oliveira MG,et al. Computerized morphometric assessment of the effect of low-level laser therapy on bone repair: An experimental animal study[J]. J Clin Laser Med Surg,2002,20(2):83–87.
    [74]Khadra M, R?nold HJ, Lyngstadaas SP,et al. Low level laser therapy stimulates bone-implant interaction: An experimental study in rabbits[J].Clin Oral Impl Res, 2004, 15(3):325-332.
    [75]Kazem Shakouri S, Soleimanpour J, Salekzamani Y,et al. Effect of low-level laser therapy on the fracture healing process[J]. Lasers Med Sci,2010,25(1):73-77.
    [76]Pires-Oliveira DA, Oliveira RF, Amadei SU,et al. Laser 904 nm action on bone repair in rats with osteoporosis[J]. Osteoporos Int,2010 Mar 4.
    [77]Javadieh F, Bayat M, Abdi S,et al. The effects of infrared low-level laser therapy on healing of partial osteotomy of tibia in streptozotocin-induced diabetic rats[J]. Photomed Laser Surg,2009,27(4):641-646.
    [78]Blaya DS, Guimar?es MB, Pozza DH,et al. Histologic study of the effect of laser therapy on bone repair[J].J Contemp Dent Pract,2008,9(6):41-48.
    [79]Dortbudak O, Haas R, Mallath-Pokorny G. Biostimulation of bone marrow cells with a diode soft laser[J]. Clin Oral Implants Res,2000,11(6):540–545.
    [80]Wilder-Smith P. The soft laser: Therapeutic tool or popular placebo ? [J] Oral Surg Oral Med Oral Pathol,1988,66(6):654–658.
    [81]Yamamoto M, Tamura K, Hiratsuka K,et al. Stimulation of MCM3 gene expression in osteoblast by low level laser irradiation[J]. Lasers Med Sci,2001,16(3):213–217.
    [82]Sato S, Ogura M, Ishihara M,et al. Nanosecond, high-intensity pulsed laser ablation of myocardium tissue at the ultraviolet, visible, and near-infrared wavelengths: in-vitro study[J]. Lasers Surg Med,2001,29:464–473.
    [83]Gerbi ME, Pinheiro AL, Marzola C,et al. Assessment of bone repair associated with the use of organic bovine bone and membrane irradiated at 830 nm[J].Photomed Laser Surg, 2005,23(4):382-388.
    [84]Carvalho Pde T, Silva IS, Reis FA,et al. Effect of 650 nm low-power laser on bone morphogenetic protein in bone defects induced in rat femors[J]. Acta Cir Bras,2006, 21,Suppl 4:63-68.
    [85]Gerbi ME, Marques AM, Ramalho LM, et al. Infrared laser light further improves bone healing when associated with bone morphogenic proteins: an in vivo study in a rodent model[J]. Photomed Laser Surg,2008,26(1):55-60.
    [86]da Silva RV, Camilli JA. Repair of bone defects treated with autogenous bone graft and low-power laser[J].J Craniofac Surg,2006,17(2):297-301.
    [87]Weber JB, Pinheiro AL, de Oliveira MG,et al. Laser therapy improves healing of bone defects submitted to autologous bone graft[J].Photomed Laser Surg,2006,24(1):38-44.
    [88]Ng GY, Fung DT, Leung MC,et al. Ultrastructural comparison of medial collateral ligament repair after single or multiple applications of GaAlAs laser in rats[J]. Lasers Surg Med, 2004,35(4):317–323.
    [89]Schneider NP,Soudry M,Remusat M,et al. Modifications of growth dynamics and ultrastr- ucture after helium-neon laser treatment of human gingival fibroblasts[J]. Quintessence Int,1989,20:887-893.
    [90]Rochkind S, Rousso M, Nissan M,et al. Systemic effects of low-power laser irradiation on the peripheral and central nervous system, cutaneous wound and burns[J]. Lasers Surg Med,1989,9:174-182.
    [91]Airaksinen O, Kolari PJ, Hietanen M,et al. Low power lasers in physical therapy: measurement of optical output power of devices[J]. Acupunc Electrother Res,1993,18:9-16.
    [92]Inoue K, Nishioka J, Hukuda S. Suppressed tuberculine reaction in guinea pigs following laser irradiation[J].Lasers Surg Med,1989,9:271-275.
    [93]Schindl A, Heinze G, Schindl M,et al. Systemic effects of low-intensity laser irradiation on skin microcirculation in patients with diabetic microangiopathy[J]. Microvasc Res, 2002, 64:240-246.
    [94]Braverman B, McCarthy RJ, Ivankovich AD,et al. Effect of helium-neon and infrared laser irradiation on wound healing in rabbits[J]. Lasers Surg Med,1989,9:50-58.
    [95]Gordjestani M, Dermaut L, Thierens H. Infrared laser and bone metabolism: a pilot study[J]. Int J Oral Maxillofac Surg,1994,23: 54-56.

© 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