Join Us | Latest Articles | Contact

Journal Home


Editorial Board


Archive


Submit to this journal


Current issue

Journal of Musculoskeletal Disorders and Treatment




Perspectives on Improving the Efficacy of PRP Treatment for Tendinopathy

James H-C. Wang* and Xavier Nirmala


MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, USA


*Corresponding author: James H-C. Wang, MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, USA, Tel: 412-648-9102, E-mail: wanghc@pitt.edu
J Musculoskelet Disord Treat, JMDT-2-015, (Volume 2, Issue 2), Commentary
Received: March 28, 2016; Accepted: June 29, 2016; Published: June 30, 2016
Citation: Wang JH-C, Nirmala X (2016) Perspectives on Improving the Efficacy of PRP Treatment for Tendinopathy. J Musculoskelet Disord Treat 2:015.
Copyright: © 2016 Wang JH-C, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.



Keywords

Tendinopathy, Inflammation, Degeneration, PRP, Efficacy


Introduction

Platelet-rich plasma (PRP), popularly called PRP, is almost like a "house-hold" name these days because of its wide recognition by physicians and patients due to its prevalent use. The popularity of PRP is due to its efficacy in treating chronic tendon injury (or tendinopathy) in some clinical studies. It is particularly preferred by elite athletes because PRP injections were reported to accelerate healing and enable quicker return to sport activities [1,2]. However, there is no doubt that the popularity of PRP usage is also driven by financial gain. Typically, PRP injections are performed in out-patient clinics in a relatively short time period and a decrease in tendon pain and improvement in tendon function have been reported in patients with chronic plantar fasciitis [3], elbow tendinopathy [4-8], chronic Achilles tendinopathy [9-11] and patellar tendinopathy [12,13]. It has been estimated that more than 86,000 athletes receive PRP treatment every year in the United States and Europe [14]. In fact, the market value of PRP is estimated to grow to $126 million this year [15]. Despite such reports on the high efficacy of PRP treatment on tendon injuries, not all PRP treatments in clinics have been reported to be successful. For example, in some studies, PRP treatment did not improve tendon function and associated pain in patients with Achilles tendinopathy [16-18]. These conflicting clinical trial findings have raised skepticism in the use of PRP for the treatment of tendinopathy. However, despite critics opposing its use there is no decline in the use of PRP to treat tendon injuries in clinics, likely because PRP is autologous and has not been reported to harm the body by inducing immunogenic reactions, hyperplasia, carcinogenesis or tumor growth [19]. It is indeed made from a persons’ own blood by one or two rounds of centrifugation processes such that it contains highly concentrated platelets in a small volume of plasma [20]. Therefore, the reported benefits of PRP and the lack of side effects thereof continue to fuel its use in clinics to treat tendon injuries. In this commentary, we attempt to discuss the scientific basis for the use of PRP to treat tendon injuries in clinics through a close analysis of the existing scientific data. We also provide suggestions to improve the PRP efficacy in clinics.


Growth Factors in PRP

A critical question to be answered about the use of PRP treatment in clinics is 'what confers the healing effects on PRP?' The consensus answer is growth factors (GFs), which both supporters and critics of PRP agree on. As the name suggests, PRP is the plasma portion of the blood containing platelets in high concentrations [21]. Platelets are the natural healing cells recruited to the wound area soon after a tissue injury; in addition to containing antibacterial and fungicidal proteins that prevent infections, platelets are reservoirs of numerous GFs such as platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), insulin-like growth factor-I (IGF-I), fibroblastic growth factor (FGF) and hepatocyte growth factor (HGF) [22-24]. GFs are in fact, the effectors that enhance the healing of tendon injuries by PRP due to their specialized functions [25,26]. For example, animal tendons treated with IGF-I [27], PDGF [28,29] or FGF [30] showed increased cell proliferation and collagen production. TGF-β also stimulated cell migration and proliferation in rat Achilles tendons [31] and increased collagen type I synthesis in tendon cells in vitro [32,33]. Further, our studies showed that HGF exerts anti-inflammatory effects on tendon cells in vitro and injured tendons in vivo [34]. Besides the benefits of GFs, PRP also contributes to tendon healing by forming a fibrin matrix when activated thus providing a conductive bio-scaffold for cell migration and new matrix formation [35,36]. Lastly, we can presume that the presence of physiological proportions of GFs in PRP may better benefit tendon healing than administering arbitrary proportions of GFs individually or in combinations [37]. Thus, the presence of GFs, fibrin matrix and "physiological" composition of GFs in PRP preparations may effectively drive the healing process of injured tendons after PRP treatment.

Recent studies on the PRP-induced cellular effects revealed that PRP exerts its effects on tendon stem/progenitor cells (TSCs), which are tendon specific adult stem cells discovered in the tendons of humans, rabbits, rats and mice [38-41]. In vitro treatment of TSCs with PRP in the form of platelet-rich clot releasate (PRCR) induced the differentiation of TSCs into active tenocytes, which are the dominant resident cells in tendons. These active tenocytes produced abundant collagen [25,42]. Similar effects were observed after the in vivo treatment of rat Achilles tendons with PRP and TSCs together [43]. From these studies, it may be inferred that PRP is safe to use for treating tendon injuries because PRCR, mainly containing releasates (GFs, cytokines, etc.) from activated platelets, did not induce non-tenocyte differentiation of TSCs [25,42], which may lead to the formation of non-tendinous tissues such as fatty-, cartilage-, or bone-like tissues, thus compromising tendon structure and therefore causing more damage to the treated tendons. Based on these previous findings, it can be concluded that there are basic science data that support the use of PRP to treat tissue injuries in general and tendinopathy in particular because GFs and possibly other factors in PRP can reduce tendon inflammation and hence pain, and enhance tendon wound healing by exerting their anti-inflammatory and anabolic effects on tendon cells [34].


PRP Treatment Variations

Scientifically speaking, if PRP treatment for tendon injuries is reasonable, then why do clinical trials report many conflicting results? We believe that multiple factors normally practiced in clinical PRP treatments may critically influence the PRP treatment outcome. These include patient-related and PRP-related factors.


Patient-Related Factors

First is a list of variables related to patients; examples of these include variations in age, gender, recommendation or not of rehabilitation along with PRP treatment, disease history, treatment history, etc. Existing data suggest that age may be a critical factor in determining the efficacy of PRP treatment because aging affects stem cells in two ways; it decreases their number as well as reduces their quality. For example, the number of MSCs in human bone marrow [44] and the number of TSCs in mice [45] and rat tendons [40] declined with age. Similarly, stem cell qualities such as stemness, proliferation and differentiation capacities also decreased with age in humans and animals. Specifically, adipose-derived stem cells (ADSCs) [46], bone marrow stem cells (BMSCs) [46], TSCs [40,45] and ACL-derived cells [47] from young animals proliferated at a higher rate than those from older animals and, the "fitness" of human MSCs [44] reduced with age. Moreover, the fate of stem cells were also altered in TSCs with more cells from aging rats and mice differentiating into non-tenocyte lineages such as adipocytes, chondrocytes and osteocytes [40,45]. Aging not only diminishes TSC properties, but also has its effects on GFs; increased numbers of GF receptors were observed in young than older porcine ACL fibroblasts [48] suggesting that GF receptors could be more in young tendons than older tendons although evidence for this is warranted. In addition, PDGF concentrations were higher in P-PRG (pure-platelet rich gel) derived from young horses than older horses [49]. Thus, the increased number of GF receptors and higher amounts of GFs in young individuals may trigger a better cellular response to PRP treatment in young than older individuals. These findings shed some insights on why PRP treatment may be less effective or even ineffective in older patients who may have fewer numbers and "poor quality" of stem cells [20]. Therefore, the efficacy of PRP treatment in healing tendon wounds in aged patients is expected to be low. Also, current clinical trials mostly include patients of various ages ranging from 18 - 70 years old [17,50-52] that may likely skew the results due to variable PRP treatment outcomes because PRP treatment in older individuals may be poor or non-effective. Therefore, clinical trials should have age as an inclusion criterion for PRP treatment; separating young and old cohorts may improve the PRP treatment outcome: a smaller number of young patients is needed to demonstrate the efficacy of PRP treatment for tendinopathy, whereas a larger number of older patients is required for the same efficacy simply because of large variations in treatment effects in older patient populations. The age may be an important factor that causes inconsistent results in clinical trials.

Gender could be another main determinant of PRP efficacy because of known gender differences in the viscoelastic properties of tendons [53], incidences of Achilles tendon ruptures [54,55] and wound healing [56]. These reports indicate that sex may influence the PRP treatment outcomes due to inherent differences in the biological properties of tendons between males and females that may impact their response to PRP treatment differently. However, in most clinical trials, males and females are grouped together with significantly larger number of males than females, which may confound the PRP treatment efficacy. In a recent study, the levels of all GFs were reported to be higher in human females than males with significant differences in the levels of EGF, HGF, IGF-I and PDGF [57]. Similarly, the concentration of PDGF in P-PRG derived from female horses was significantly higher than from male horses [49]. Lastly, the PRP treatment in aged or post-menopausal females should be given particular attention because reduced estrogen levels have been linked to decreased tensile strength [58] and diminished wound healing response of dermal fibroblasts [59,60], and exogenous estrogen has been shown to improve tendon morphology and biomechanical properties post-menopause [61], and regulate VEGF [62] and IGF-I expression [63]. To effectively evaluate the issue of gender in PRP treatment, clinical studies should separate patients based on gender prior to treatment. Moreover, to demonstrate the efficacy of PRP treatment on tendon injuries, male and female patients should be separated or only one sex should be included in clinical trials to reduce variations due to gender and hence increase statistical power of a clinical trial. On the other hand, if both male and female subjects are included in a clinical trial for PRP treatment, a much larger sample size (the number of patients in both control and treatment groups) must be adopted to increase statistical power to detect the effects of PRP treatment.

Another patient-related factor that should be addressed is whether or not to recommend rehabilitation with PRP treatment. A study investigating the effects of PRP with or without mechanical loading on rat Achilles tendons found that nullifying loading with Botox injections negated the PRP treatment effects while in normal tendons (not treated with Botox) both PRP and mechanical loads independently increased tendon repair without synergistic effects [64]. A randomized control trial (RCT) also studied the effect of eccentric exercise after PRP treatment on patients with lateral elbow epicondylitis [8]. This study showed that the VAS scores were improved in patients in the exercise group in comparison with the control group that remained sedentary [8]. In general, most studies suggest that strengthening exercises may benefit tendon structure and function although the timing of applying rehabilitation seems questionable with some suggesting resuming activity immediately along with any repair treatment [65] and others arguing that a slightly delayed loading may improve mechanical and biological parameters to a greater extent than immediate loading or prolonged post-operative immobilization [66,67].

Another recent addition to the patient-related factors is the likely presence of single nucleotide polymorphism (SNPs) in the genes transcribing GFs and other PRP-related factors [68]. Although direct evidence for the presence of SNPs in GFs and how they affect PRP treatment efficacy has not been established yet, the IGF2 GC genotype was found to reduce the degree of muscle injuries in elite soccer players when compared to those with IGF2 CC or GG genotypes [69]. Similarly, a SNP in elastin gene (AA genotype) was found to predispose patients to severe muscle injuries and prolong recovery times compared to the AG or GG genotypes [70]. It remains to be seen whether such SNP occurrences influence the PRP efficacy treatment in clinical trials.

Considering the above discussions together, we suggest that clinical trials of PRP treatment on tendon injuries should be large, randomized and selective in patient enrollment so that the statistical power of the PRP treatment outcomes in trials can be enhanced. On the other hand, more basic science studies are also needed to obtain a comprehensive understanding of the age, gender and rehabilitation effects on the PRP treatment efficacy. Other factors such as genetics, disease history and prior treatment history after PRP treatment should be considered as variables in clinical trials. Finally, in clinical trials of PRP treatment, appropriate selection of controls is critical in assessing the efficacy of PRP treatment. These controls may include needle penetration and saline injection. We believe that platelet-poor plasma (PPP) injection may be the best control because it includes needle penetration, and plasma contained in most PRP preparations [34].


PRP-Related Factors

The second major factor that may cause inconsistent results in PRP treatment efficacy in clinics is related to PRP itself. This includes differences in the platelet concentration, PRP composition and activation method. In clinical studies, PRP is prepared using commercial kits that do not yield "one-type" of PRP with the same composition. The main difference in PRP preparations appears to be in the platelet concentrations. Indeed, manufacturers describe that in current commercial preparations platelet yield could increase by 1-fold (Autolo Gel System, Secquire), 3-5-fold (Biomet GPS, Cell Saver Based Systems, Sorin Angel, Harvest Smart Pre BMC, Depuy Symphony, Arteriocyte Medical Magellan) or even up to 10-fold (GenesisCS) higher than in whole blood depending on the kit used. While the rationale to use PRP is to deliver concentrated GF containing platelets at the site of injury, 'more' does not always translate to 'better'. No improvement in tendon function (ATRS score) was observed when PRP containing 10-fold more platelets than in whole blood was used to treat acute Achilles tendon ruptures in patients [18]. Therefore, most clinical and basic science studies only use PRP with platelet concentrations 3-5-fold higher than in whole blood due to apparent beneficial effects; decreased pain, increased tendon cell proliferation, improved collagen synthesis and organization, etc. [42,71-73].

Besides platelet concentration, PRP composition may differ. Specifically, leukocyte levels in PRP preparations may vary and can affect PRP treatment outcomes. While leukocytes promote chemotaxis, cell proliferation and differentiation [74] they also release inflammatory cytokines such as interleukin-1 β (IL-1β), tumor necrosis factor-α (TNF-α) and reactive oxygen species that can harm the already injured tissues [75]. Indeed, leukocyte containing PRP (L-PRP) inflicted catabolic effects on human chondrocytes [76] and rabbit TSCs [73]. More importantly, pure PRP (P-PRP) or leukocyte free PRP promoted anabolic effects by increasing collagen II and aggrecan expression in chondrocytes [76] and upregulating alpha-smooth muscle actin (α-SMA), collagen types I and III in rabbit TSCs [73]. These and other studies suggest that to promote tendon healing it may be beneficial to use PRP with low concentrations of leukocytes.

Although the platelet and leukocyte concentrations in PRP can be easily optimized, thus far, a standard PRP composition to treat tendon injuries has not been worked out. The lack of guidelines and inconsistencies in the PRP preparations made from kits skew the PRP efficacy reported in clinical trials. A comparison between MTF Cascade, Arteriocyte Magellan and Biomet GPS III PRP preparation systems using the same blood sample showed no significant differences in platelet concentration but huge variations in the concentrations of leukocytes, PDGF and VEGF [77]. Similarly, the platelet concentration was similar in aphaeresis-derived platelets, Buffy coat-derived platelets and tube method-derived platelets. However, the leukocyte concentration differed significantly between the samples [78]. If any of these leukocyte containing PRP preparations are used to treat tendon injuries, the end result would be the induction of catabolic effects that may skew PRP treatment efficacy.

Another less frequently discussed PRP-related factor is platelet activation that releases the GFs stored in alpha granules [78]. Traditionally, PRP is activated using an external source such as thrombin, calcium chloride or collagen [11,64,79]; however, some authors inject non-activated PRP because platelets become activated in vivo when in contact with collagen [8,37].

Due to these variations in PRP-related factors, some authors have suggested a universal PRP labeling system to be followed with description of PRP components. Some examples of classification systems are the use of PAW, which indicates the Platelet concentration, Activation method and presence of White blood cells or leukocytes [80] or DEPA, which indicates the Dose of platelets, Efficiency of production, Purity of the PRP and Activation of PRP [81]. While the various technologies and in-house preparation methods provide flexibility to choose a kit/method for clinical use, careful consideration should be given to the resulting PRP preparation for the intended treatments. For instance, tendon injuries in the early phase of healing are mainly inflammatory [82-84], and may be treated with P-PRP because of its anabolic and anti-inflammatory effects. In the early phase, tendons are not degenerative and P-PRP is likely to induce TSCs to proliferate and differentiate into tenocytes and as a result, enhance tendon healing. However, advanced stage tendinopathy may not be treatable with P-PRP because this stage is characterized by degenerative conditions with extensive formation of lipids, proteoglycan accumulation, and calcification in tendon lesions, either alone or in combination [85]. With only few TSCs remaining in the degenerative tendons, P-PRP may not be able to enhance sufficient TSC proliferation to repopulate the degenerated tendon, promote collagen production or reverse the already differentiated non-tenocyte cells; therefore, an effective repair of advanced stage tendon injury by P-PRP is unlikely. However, surgical removal of tendon lesions (wound debridement) has been advocated in clinics to induce healing in a non-tenogenic environment in advanced stage tendon injuries [35,86]. This debridement procedure may be followed by L-PRP treatment, which due to the presence of leukocytes, may kick-off a healing process in injured tendons. Finally, platelets in PRP are known to contain pro-angiogenic (e.g. VEGF) or anti-angiogenic (e.g. endostatin) factors [87] that may have differential effects on the healing of tendon injuries. These factors are also selectively released after platelet activation by specific protease activated receptors, PAR1 or PAR4. Therefore, future studies should investigate the effect of selective platelet activation and its influence on the healing of tendon injuries.


Conclusion

Considering the above discussion, it seems reasonable to use PRP to treat tendon injuries effectively because it can reduce tendon inflammation and hence improve tendon function and accelerate tendon healing, mainly due to the abundant GFs released from the platelets in PRP once activated. It is apparent that the current "one size-fits-all" approach to treat "any" tendon injury using "any" PRP preparation would almost certainly lead to contradicting results or would diminish the efficacy of PRP treatment. Thus, instead of the "one-size-fits-all" approach, a "tailored" approach with considerations on the patients' age, gender, disease history, rehabilitation, etc., and an appropriate type of PRP, must be applied to treat tendon injuries with PRP so that the maximum efficacy of PRP treatment can be achieved in clinics.


Acknowledgements

Funding support from NIH/NIAMS (AR065949, AR061395, and AR060920) is gratefully acknowledged (JHW).


References
  1. Rowan TL, Drouin JL (2013) A multidisciplinary approach including the use of platelet-rich plasma to treat an elite athlete with patellar tendinopathy - a case report. J Can Chiropr Assoc 57: 301-309.

  2. Schwarz A (2009) A Promising Treatment for Athletes, in the Blood. The New York Times 1-3.

  3. Wilson JJ, Lee KS, Miller AT, Wang S (2014) Platelet-rich plasma for the treatment of chronic plantar fasciopathy in adults: a case series. Foot Ankle Spec 7: 61-67.

  4. Peerbooms JC, Sluimer J, Bruijn DJ, Gosens T (2010) Positive effect of an autologous platelet concentrate in lateral epicondylitis in a double-blind randomized controlled trial: platelet-rich plasma versus corticosteroid injection with a 1-year follow-up. Am J Sports Med 38: 255-262.

  5. Mishra A, Pavelko T (2006) Treatment of chronic elbow tendinosis with buffered platelet-rich plasma. Am J Sports Med 34: 1774-1778.

  6. Mishra A, Woodall J Jr, Vieira A (2009) Treatment of tendon and muscle using platelet-rich plasma. Clin Sports Med 28: 113-125.

  7. Hechtman KS, Uribe JW, Botto-vanDemden A, Kiebzak GM (2011) Platelet-rich plasma injection reduces pain in patients with recalcitrant epicondylitis. Orthopedics 34: 92.

  8. Thanasas C, Papadimitriou G, Charalambidis C, Paraskevopoulos I, Papanikolaou A (2011) Platelet-rich plasma versus autologous whole blood for the treatment of chronic lateral elbow epicondylitis: a randomized controlled clinical trial. Am J Sports Med 39: 2130-2134.

  9. Rabago D, Wilson J, Zgierska A (2010) Platelet-rich plasma for treatment of Achilles tendinopathy. JAMA 303: 1696-1697.

  10. Gaweda K, Tarczynska M, Krzyzanowski W (2010) Treatment of Achilles tendinopathy with platelet-rich plasma. Int J Sports Med 31: 577-583.

  11. Filardo G, Kon E, Di Matteo B, Di Martino A, Tesei G, et al. (2014) Platelet-rich plasma injections for the treatment of refractory Achilles tendinopathy: results at 4 years. Blood Transfus 12: 533-540.

  12. Kon E, Filardo G, Delcogliano M, Presti ML, Russo A, et al. (2009) Platelet-rich plasma: new clinical application: a pilot study for treatment of jumper's knee. Injury 40: 598-603.

  13. Filardo G, Kon E, Della Villa S, Vincentelli F, Fornasari PM, et al. (2010) Use of platelet-rich plasma for the treatment of refractory jumper's knee. Int Orthop 34: 909-915.

  14. Wasterlain AS, Braun HJ, Harris AH, Kim HJ, Dragoo JL (2013) The systemic effects of platelet-rich plasma injection. Am J Sports Med 41: 186-193.

  15. Matava MJ (2012) Platelet-rich plasma: the next big thing?: commentary on an article by Ujash Sheth, BHSc, et al.: "Efficacy of autologous platelet-rich plasma use for orthopaedic indications: a meta-analysis". J Bone Joint Surg Am 94: e25.

  16. de Jonge S, de Vos RJ, Weir A, van Schie HT, Bierma-Zeinstra SM, et al. (2011) One-year follow-up of platelet-rich plasma treatment in chronic Achilles tendinopathy: a double-blind randomized placebo-controlled trial. Am J Sports Med 39:1623-1629.

  17. de Vos RJ, Weir A, van Schie HT, Bierma-Zeinstra SM, Verhaar JA, et al. (2010) Platelet-rich plasma injection for chronic Achilles tendinopathy: a randomized controlled trial. JAMA 303: 144-149.

  18. Schepull T, Kvist J, Norrman H, Trinks M, Berlin G, et al. (2011) Autologous platelets have no effect on the healing of human achilles tendon ruptures: a randomized single-blind study. Am J Sports Med 39: 38-47.

  19. Sampson S, Gerhardt M, Mandelbaum B (2008) Platelet rich plasma injection grafts for musculoskeletal injuries: a review. Curr Rev Musculoskelet Med 1: 165-174.

  20. Wang JH (2014) Can PRP effectively treat injured tendons? Muscles Ligaments Tendons J 4: 35-37.

  21. Webb CA (2012) Platelet-rich plasma update: clinical use in musculoskeletal care: the therapy has seen increased demand from the general patient population. The Journal of Musculoskeletal Medicine 29: 96.

  22. Anitua E, Andia I, Sanchez M, Azofra J, del Mar Zalduendo M, et al. (2005) Autologous preparations rich in growth factors promote proliferation and induce VEGF and HGF production by human tendon cells in culture. J Orthop Res 23: 281-286.

  23. Alsousou J, Ali A, Willett K, Harrison P (2013) The role of platelet-rich plasma in tissue regeneration. Platelets 24: 173-182.

  24. Yuan T, Guo SC, Han P, Zhang CQ, Zeng BF (2012) Applications of leukocyte- and platelet-rich plasma (L-PRP) in trauma surgery. Curr Pharm Biotechnol 13: 1173-1184.

  25. Chen L, Dong SW, Tao X, Liu JP, Tang KL, et al. (2012) Autologous platelet-rich clot releasate stimulates proliferation and inhibits differentiation of adult rat tendon stem cells towards nontenocyte lineages. J Int Med Res 40: 1399-1409.

  26. Parafioriti A, Armiraglio E, Del Bianco S, Tibalt E, Oliva F, et al. (2011) Single injection of platelet-rich plasma in a rat Achilles tendon tear model. Muscles Ligaments Tendons J 1: 41-47.

  27. Dahlgren LA, van der Meulen MC, Bertram JE, Starrak GS, Nixon AJ (2002) Insulin-like growth factor-I improves cellular and molecular aspects of healing in a collagenase- induced model of flexor tendinitis. J Orthop Res 20: 910-919.

  28. Yoshikawa Y, Abrahamsson SO (2001) Dose-related cellular effects of platelet-derived growth factor-BB differ in various types of rabbit tendons in vitro. Acta Orthop Scand 72: 287-292.

  29. Wang XT, Liu PY, Tang JB (2004) Tendon healing in vitro: genetic modification of tenocytes with exogenous PDGF gene and promotion of collagen gene expression. J Hand Surg Am 29: 884-890.

  30. Chan BP, Fu S, Qin L, Lee K, Rolf CG, et al. (2000) Effects of basic fibroblast growth factor (bFGF) on early stages of tendon healing: a rat patellar tendon model. Acta Orthop Scand 71: 513-518.

  31. Kashiwagi K, Mochizuki Y, Yasunaga Y, Ishida O, Deie M, et al. (2004) Effects of transforming growth factor-beta 1 on the early stages of healing of the Achilles tendon in a rat model. Scand J Plast Reconstr Surg Hand Surg 38: 193-197.

  32. Heinemeier K, Langberg H, Olesen JL, Kjaer M (2003) Role of TGF-beta1 in relation to exercise-induced type I collagen synthesis in human tendinous tissue. J Appl Physiol (1985) 95: 2390-2397.

  33. Klein MB, Yalamanchi N, Pham H, Longaker MT, Chang J (2002) Flexor tendon healing in vitro: effects of TGF-beta on tendon cell collagen production. J Hand Surg Am 27: 615-620.

  34. Zhang J, Middleton KK, Fu FH, Im HJ, Wang JH (2013) HGF mediates the anti-inflammatory effects of PRP on injured tendons. PLoS One 8: e67303.

  35. Anitua E, Sánchez M, Nurden AT, Nurden P, Orive G, et al. (2006) New insights into and novel applications for platelet-rich fibrin therapies. Trends Biotechnol 24: 227-234.

  36. Xie X, Wang Y, Zhao C, Guo S, Liu S, et al. (2012) Comparative evaluation of MSCs from bone marrow and adipose tissue seeded in PRP-derived scaffold for cartilage regeneration. Biomaterials 33:7008-7018.

  37. Marx RE (2004) Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg 62: 489-496.

  38. Bi Y, Ehirchiou D, Kilts TM, Inkson CA, Embree MC, et al. (2007) Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nat Med 13: 1219-1227.

  39. Zhang J, Wang JH (2010) Characterization of differential properties of rabbit tendon stem cells and tenocytes. BMC Musculoskelet Disord 11: 10.

  40. Zhou Z, Akinbiyi T, Xu L, Ramcharan M, Leong DJ, et al. (2010) Tendon-derived stem/progenitor cell aging: defective self-renewal and altered fate. Aging Cell 9: 911-915.

  41. Rui YF, Lui PP, Li G, Fu SC, Lee YW, et al. (2010) Isolation and characterization of multipotent rat tendon-derived stem cells. Tissue Eng Part A 16: 1549-1558.

  42. Zhang J, Wang JH (2010) Platelet-rich plasma releasate promotes differentiation of tendon stem cells into active tenocytes. Am J Sports Med 38: 2477-2486.

  43. Chen L, Dong SW, Liu JP, Tao X, Tang KL, et al. (2012) Synergy of tendon stem cells and platelet-rich plasma in tendon healing. J Orthop Res 30: 991-997.

  44. Stolzing A, Jones E, McGonagle D, Scutt A (2008) Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev 129: 163-173.

  45. Zhang J, Wang JH (2015) Moderate Exercise Mitigates the Detrimental Effects of Aging on Tendon Stem Cells. PLoS One 10: e0130454.

  46. Liu HY, Huang CF, Lin TC, Tsai CY,Tina Chen SY, et al. (2014) Delayed animal aging through the recovery of stem cell senescence by platelet rich plasma. Biomaterials 35: 9767-9776.

  47. Mastrangelo AN, Haus BM, Vavken P, Palmer MP, Machan JT, et al. (2010) Immature animals have higher cellular density in the healing anterior cruciate ligament than adolescent or adult animals. J Orthop Res 28: 1100-1106.

  48. Vavken P, Saad FA, Murray MM (2010) Age dependence of expression of growth factor receptors in porcine ACL fibroblasts. J Orthop Res 28: 1107-1112.

  49. Giraldo CE, López C, Álvarez ME, Samudio IJ, Prades M, et al. (2013) Effects of the breed, sex and age on cellular content and growth factor release from equine pure- platelet rich plasma and pure-platelet rich gel. BMC Veterinary Research 9: 1-10.

  50. Ferrero G, Fabbro E, Orlandi D, Martini C, Lacelli F, et al. (2012) Ultrasound-guided injection of platelet-rich plasma in chronic Achilles and patellar tendinopathy. J Ultrasound 15: 260-266.

  51. Seijas R, Ares O, Catala J, Alvarez-Diaz P, Cusco X, et al. (2013) Magnetic resonance imaging evaluation of patellar tendon graft remodelling after anterior cruciate ligament reconstruction with or without platelet-rich plasma. J Orthop Surg (Hong Kong) 21: 10-14.

  52. Kesikburun S, Tan AK, Yilmaz B, Yasar E, Yazicioglu K (2013) Platelet-rich plasma injections in the treatment of chronic rotator cuff tendinopathy: a randomized controlled trial with 1-year follow-up. Am J Sports Med 41: 2609-2616.

  53. Kubo K, Kanehisa H, Fukunaga T (2003) Gender differences in the viscoelastic properties of tendon structures. Eur J Appl Physiol 88: 520-526.

  54. Nillius SA, Nilsson BE, Westlin NE (1976) The incidence of Achilles tendon rupture. Acta Orthop Scand 47: 118-121.

  55. Nyyssönen T, Lüthje P, Kröger H (2008) The increasing incidence and difference in sex distribution of Achilles tendon rupture in Finland in 1987-1999. Scand J Surg 97: 272-275.

  56. Guo S, Dipietro LA (2010) Factors affecting wound healing. J Dent Res 89: 219-229.

  57. Evanson JR, Guyton MK, Oliver DL, Hire JM, Topolski RL, et al. (2014) Gender and age differences in growth factor concentrations from platelet-rich plasma in adults. Mil Med 179: 799-805.

  58. Slauterbeck J, Clevenger C, Lundberg W, Burchfield DM (1999) Estrogen level alters the failure load of the rabbit anterior cruciate ligament. J Orthop Res 17: 405-408.

  59. Ashcroft GS, Dodsworth J, van Boxtel E, Tarnuzzer RW, Horan MA, et al. (1997) Estrogen accelerates cutaneous wound healing associated with an increase in TGF-beta1 levels. Nat Med 3: 1209-1215.

  60. Gilliver SC, Ashworth JJ, Ashcroft GS (2007) The hormonal regulation of cutaneous wound healing. Clin Dermatol 25: 56-62.

  61. Hansen M, Kjaer M (2014) Influence of sex and estrogen on musculotendinous protein turnover at rest and after exercise. Exerc Sport Sci Rev 42: 183-192.

  62. Mueller MD, Vigne JL, Minchenko A, Lebovic DI, Leitman DC, et al. (2000) Regulation of vascular endothelial growth factor (VEGF) gene transcription by estrogen receptors alpha and beta. Proc Natl Acad Sci U S A 97: 10972-10977.

  63. Denger S, Bahr-Ivacevic T, Brand H, Reid G, Blake J, et al. (2008) Transcriptome profiling of estrogen-regulated genes in human primary osteoblasts reveals an osteoblast-specific regulation of the insulin-like growth factor binding protein 4 gene. Mol Endocrinol 22: 361-379.

  64. Virchenko O, Aspenberg P (2006) How can one platelet injection after tendon injury lead to a stronger tendon after 4 weeks? Interplay between early regeneration and mechanical stimulation. Acta orthopaedica 77: 806-812.

  65. Rajabi H, Shahin HS, Norouzian M, Mehrabani D, Nazhvani SD (2015) The Healing Effects of Aquatic Activities and Allogenic Injection of Platelet-Rich Plasma (PRP) on Injuries of Achilles Tendon in Experimental Rat. World J Plast Surg 4: 66-73.

  66. Thomopoulos S, Williams GR, Soslowsky LJ (2003) Tendon to bone healing: differences in biomechanical, structural, and compositional properties due to a range of activity levels. J Biomech Eng 125: 106-113.

  67. Killian ML, Cavinatto L, Galatz LM, Thomopoulos S (2012) The role of mechanobiology in tendon healing. J Shoulder Elbow Surg 21: 228-237.

  68. Pruna R, Til L, Artells R (2014) Could single nucleotide polymorphisms influence on the efficacy of platelet-rich plasma in the treatment of sport injuries? Muscles Ligaments Tendons J 4: 63-65.

  69. Pruna R, Artells R, Ribas J, Montoro B, Cos F, et al. (2013) Single nucleotide polymorphisms associated with non-contact soft tissue injuries in elite professional soccer players: influence on degree of injury and recovery time. BMC Musculoskeletal Disord 14: 1-7.

  70. Pruna R, Artells R (2015) New Tendencies in Risk Factors for Soft Tissue Injuries: Genetic Biomakers. International Journal of Orthopaedics 2: 307-311.

  71. Marx RE (2001) Platelet-rich plasma (PRP): what is PRP and what is not PRP? Implant Dent 10: 225-228.

  72. Zhang J, Wang JH (2014) PRP treatment effects on degenerative tendinopathy - an in vitro model study. Muscles Ligaments Tendons J 4: 10-17.

  73. Zhou Y, Zhang J, Wu H, Hogan MV, Wang JHC (2015) The differential effects of leukocyte- containing and pure platelet-rich plasma (PRP) on tendon stem/progenitor cells - implications of PRP application for the clinical treatment of tendon injuries. Stem Cell Res Ther 6: 173.

  74. Wrotniak M, Bielecki T, Gazdzik TS (2007) Current opinion about using the platelet-rich gel in orthopaedics and trauma surgery. Ortop Traumatol Rehabil 9: 227-238.

  75. McCarrel TM, Minas T, Fortier LA (2012) Optimization of leukocyte concentration in platelet-rich plasma for the treatment of tendinopathy. J Bone Joint Surg Am 94.

  76. Cavallo C, Filardo G, Mariani E, Kon E, Marcacci M, et al. (2014) Comparison of platelet-rich plasma formulations for cartilage healing: an in vitro study. J Bone Joint Surg Am 96: 423-429.

  77. Castillo TN, Pouliot MA, Kim HJ, Dragoo JL (2011) Comparison of growth factor and platelet concentration from commercial platelet-rich plasma separation systems. Am J Sports Med 39: 266-271.

  78. Zimmermann R, Jakubietz R, Jakubietz M, Strasser E, Schlegel A, et al. (2001) Different preparation methods to obtain platelet components as a source of growth factors for local application. Transfusion 41: 1217-1224.

  79. Eppley BL, Woodell JE, Higgins J (2004) Platelet quantification and growth factor analysis from platelet-rich plasma: implications for wound healing. Plast Reconstr Surg 114: 1502-1508.

  80. DeLong JM, Russell RP, Mazzocca AD (2012) Platelet-rich plasma: the PAW classification system. Arthroscopy 28: 998-1009.

  81. Dohan Ehrenfest DM, Andia I, Zumstein MA, Zhang C-Q, Pinto NR, et al. (2014) Classification of platelet concentrates (Platelet-Rich Plasma-PRP, Platelet-Rich Fibrin-PRF) for topical and infiltrative use in orthopedic and sports medicine: current consensus, clinical implications and perspectives. Muscles Ligaments Tendons J 4: 3-9.

  82. Fung L, Wong B, Ravichandiran K, Agur A, Rindlisbacher T, et al. (2009) Three-dimensional study of pectoralis major muscle and tendon architecture. Clin Anat 22: 500-508.

  83. Nakama LH, King KB, Abrahamsson S, Rempel DM (2005) Evidence of tendon microtears due to cyclical loading in an in vivo tendinopathy model. J Orthop Res 23: 1199-1205.

  84. Andarawis-Puri N, Kuntz AF, Ramsey ML, Soslowsky LJ (2011) Effect of supraspinatus tendon repair technique on the infraspinatus tendon. J Biomech Eng 133: 031008.

  85. Kannus P, Jozsa L (1991) Histopathological changes preceding spontaneous rupture of a tendon. A controlled study of 891 patients. J Bone Joint Surg Am 73: 1507-1525.

  86. Kang YH, Jeon SH, Park JY, Chung JH, Choung YH, et al. (2010) Platelet-rich fibrin is a Bioscaffold and reservoir of growth factors for tissue regeneration. Tissue Eng Part A 17: 349-359.

  87. Ma L, Perini R, McKnight W, Dicay M, Klein A, et al. (2005) Proteinase-activated receptors 1 and 4 counter-regulate endostatin and VEGF release from human platelets. Proc Natl Acad Sci USA 102: 216-220.

International Journal of Anesthetics and Anesthesiology (ISSN: 2377-4630)
International Journal of Blood Research and Disorders   (ISSN: 2469-5696)
International Journal of Brain Disorders and Treatment (ISSN: 2469-5866)
International Journal of Cancer and Clinical Research (ISSN: 2378-3419)
International Journal of Clinical Cardiology (ISSN: 2469-5696)
Journal of Clinical Gastroenterology and Treatment (ISSN: 2469-584X)
Clinical Medical Reviews and Case Reports (ISSN: 2378-3656)
Journal of Dermatology Research and Therapy (ISSN: 2469-5750)
International Journal of Diabetes and Clinical Research (ISSN: 2377-3634)
Journal of Family Medicine and Disease Prevention (ISSN: 2469-5793)
Journal of Genetics and Genome Research (ISSN: 2378-3648)
Journal of Geriatric Medicine and Gerontology (ISSN: 2469-5858)
International Journal of Immunology and Immunotherapy (ISSN: 2378-3672)
International Journal of Medical Nano Research (ISSN: 2378-3664)
International Journal of Neurology and Neurotherapy (ISSN: 2378-3001)
International Archives of Nursing and Health Care (ISSN: 2469-5823)
International Journal of Ophthalmology and Clinical Research (ISSN: 2378-346X)
International Journal of Oral and Dental Health (ISSN: 2469-5734)
International Journal of Pathology and Clinical Research (ISSN: 2469-5807)
International Journal of Pediatric Research (ISSN: 2469-5769)
International Journal of Respiratory and Pulmonary Medicine (ISSN: 2378-3516)
Journal of Rheumatic Diseases and Treatment (ISSN: 2469-5726)
International Journal of Sports and Exercise Medicine (ISSN: 2469-5718)
International Journal of Stem Cell Research & Therapy (ISSN: 2469-570X)
International Journal of Surgery Research and Practice (ISSN: 2378-3397)
Trauma Cases and Reviews (ISSN: 2469-5777)
International Archives of Urology and Complications (ISSN: 2469-5742)
International Journal of Virology and AIDS (ISSN: 2469-567X)
More Journals

Contact Us

ClinMed International Library | Science Resource Online LLC
3511 Silverside Road, Suite 105, Wilmington, DE 19810, USA
Email: contact@clinmedlib.org
 

Feedback

Get Email alerts
 
Creative Commons License
Open Access
by ClinMed International Library is licensed under a Creative Commons Attribution 4.0 International License based on a work at https://clinmedjournals.org/.
Copyright © 2017 ClinMed International Library. All Rights Reserved.