Journal articles on the topic 'Intra-osseous defect'

To see the other types of publications on this topic, follow the link: Intra-osseous defect.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 38 journal articles for your research on the topic 'Intra-osseous defect.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

HASEGAWA, T., and Y. YAMANO. "Arthroplasty of the Proximal Interphalangeal Joint Using Costal Cartilage Grafts." Journal of Hand Surgery 17, no. 5 (October 1992): 583–85. http://dx.doi.org/10.1016/s0266-7681(05)80248-7.

Full text
Abstract:
Seven intra-articular fractures in five patients with partial bone loss at the PIP joint were reconstructed using a graft of costal cartilage. In all cases there were total or partial cartilagenous defects of the proximal phalangeal side of the joint. Early treatment of two joints, using only costal cartilage grafts, resulted in bony ankylosis due to necrosis of the grafted cartilage. In five joints the grafted cartilage included osseous portions using the costo-osteochondral junction, leading to an average range of movement of 64° with satisfactory clinical results. The technique is a useful alternative to other forms of arthroplasty or arthrodesis, and can provide satisfactory functional results when there is a partial defect of the head of the proximal phalanx.
APA, Harvard, Vancouver, ISO, and other styles
2

El-dien, Asma M. Sarag, Shereen Fathy, and Yasmine Alaa El-din. "Potential Bone Regenerative Effects of DFDBA, Simvastatin and Platelet Rich Fibrin, Radiographically and Histologically of Intra-Bony Periodontal Defects in White New Zealand Rabbits." Open Access Macedonian Journal of Medical Sciences 9, no. D (April 11, 2021): 72–80. http://dx.doi.org/10.3889/oamjms.2021.5848.

Full text
Abstract:
AIM: This study aimed to evaluate and to compare the regenerative power of simvastatin, Demineralized Freeze-Dried Bone Allograft (DFDBA) allograft, platelets rich Fibrin (PRF), and a combination of these materials radiographically and histologically in the intra-bony periodontal defects in white New Zealand rabbits. MATERIALS AND METHODS: This study was conducted on 54 defects in 27 adult male rabbits (n = 27) which were divided into three groups according to the follow-up preplanned scheduled for 1, 2, and 3 weeks. The selected materials were induced as following: A=DFDBA, B=Simvastatin, C= PRF, D=A+C, E=B+C, and F=negative (control group). The intra-bony periodontal defects were induced as the form of one osseous wall defect of 10 mm height, 4 mm depth between the first and the second molars. Then, samples were prepared for histological evaluation. Radiographic assessment was done using computed tomography radiography which was carried at different time intervals as the following baseline, 1, 2, and 3 weeks later. Statistical analysis was performed using ANOVA. RESULTS: After evaluating the results, macroanatomy, radiographically, and histologically, it is thus confirmed that DFDBA allograft combined with PRF create the best bone regenerative results, followed by DFDBA, Simvastatin, simvastatin+ PRF, control group, and finally PRF. CONCLUSION: All of the materials examined in this study showed different percentage in terms of bone density and bone regenerative effects. However, the best results for bone density of the DFDBA + PRF group were recorded after 3 weeks. Thus, the study concludes that a combination of DFDBA + PRF reflects the best properties of both materials in terms of bone density results of the defect. Such results are particularly significant for the selection of bone regeneration materials, and generally, for periodontal regeneration.
APA, Harvard, Vancouver, ISO, and other styles
3

Raveendran, Dr Shruthi, Dr Shruthi S, Dr Nisha K J, Dr Sanjeela Guru, Dr Parichaya Batra, and Dr Anjana Suresh U. "NovaboneTM dental putty as a potential regenerative material for treating horizontal defects in chronic periodontitis patients." RGUHS Journal of Dental Sciences 11, no. 2 (2019): 2–10. http://dx.doi.org/10.26715/rjds.11_2_2.

Full text
Abstract:
ntroduction: Periodontitis is a multifactorial disease, which, when not adequately treated, is followed with progressive attachment loss which leads to tooth mobility and eventually tooth loss. Periodontal regenerative surgery aims to regenerate and reconstruct the lost periodontal tissue. Regeneration with novabone putty has shown to be effective in reducing probing pocket depth, gain in clinical attachment level and increase in horizontal bone level. Aim: This interventional clinical trial was to evaluate the osseous regenerative potential of a calcium phosphosilicate bioactive glass NOVABONE TM in the treatment of horizontal bone defects. Materials and method: A total of 20 sites with horizontal bone defect was treated with open flap debridement with intra marrow penetration and novabone putty. Statistical analysis: Plaque index, gingival index and radiographic determination were analysed by paired t test. Probing pocket depth and clinical attachment levels were analysed by Wilcoxon Signed Ranks Test. Result: There was significant reduction seen in plaque index, gingival index, probing pocket depth and radiographic crestal level when compared between baseline and 12 months respectively (2.48 ± 0.44, 2.92 ± 0.39, 6.80 ± 0.89, 8.29 ± 0.87) and (1.89 ± 0.42, 1.92± 0.51, 3.00± 0.67, 5.48 ± 0.89) and significant gain in clinical attachment level from 4.40 ± 0.96 to 1.20 ± 0.91. Conclusion: The present study showed novabone putty significantly improved the clinical parameters in horizontal bone defects.
APA, Harvard, Vancouver, ISO, and other styles
4

Herten, M., M. Sager, L. Benga, J. C. Fischer, M. Jäger, M. Betsch, M. Wild, M. Hakimi, P. Jungbluth, and J. P. Grassmann. "Bone marrow concentrate for autologous transplantation in minipigs." Veterinary and Comparative Orthopaedics and Traumatology 26, no. 01 (2013): 34–41. http://dx.doi.org/10.3415/vcot-11-11-0165.

Full text
Abstract:
SummaryAutologous bone marrow plays an increasing role in the treatment of bone, cartilage and tendon healing disorders. Cell-based therapies display promising results in the support of local regeneration, especially therapies using intra-operative one-step treatments with autologous progenitor cells. In the present study, bone marrow-derived cells were concentrated in a point-of-care device and investigated for their mesenchymal stem cell (MSC) characteristics and their osteogenic potential.Bone marrow was harvested from the iliac crest of 16 minipigs. The mononucleated cells (MNC) were concentrated by gradient density centrifugation, cultivated, characterized by flow cytometry and stimulated into osteoblasts, adipocytes, and chondrocytes. Cell differentiation was investigated by histological and immunohistological staining of relevant lineage markers. The proliferation capacity was determined via colony forming units of fibroblast and of osteogenic alkaline-phosphatase-positive-cells.The MNC could be enriched 3.5-fold in nucleated cell concentrate in comparison to bone marrow. Flow cytometry analysis revealed a positive signal for the MSC markers. Cells could be differentiated into the three lines confirming the MSC character. The cellular osteogenic potential correlated significantly with the percentage of newly formed bone in vivo in a porcine metaphyseal long-bone defect model.This study demonstrates that bone marrow concentrate from minipigs display cells with MSC character and their osteogenic differentiation potential can be used for osseous defect repair in autologous transplantations.
APA, Harvard, Vancouver, ISO, and other styles
5

Shimon, V. M., A. A. Shereghii, S. P. Alfeldii, and M. V. Shimon. "OUR EXPERIENCE IN THE TREATMENT OF CALCANEUS FRACTURES." Актуальні проблеми сучасної медицини: Вісник Української медичної стоматологічної академії 19, no. 3 (November 8, 2019): 91–95. http://dx.doi.org/10.31718/2077-1096.19.3.91.

Full text
Abstract:
Treatment of Calcaneus fractures needs improvement of modern methods. The purpose of the work is to reveal the structural and functional advantages of the results of surgical treatment of patients with intra-articular fractures of the calcaneus with the use of bone plastic. Material and methods. Case histories of 51 patients treated surgically at the clinical bases of the Department of General Surgery between 2014–2019, retrospectively studied for impressionable intraatricular fractures of 53 calcaneus bones with displacement. The average age of trauma victims was 34.7 ± 1.6 years (26 to 51 years), women - 9 (17.64%), men - 42 (82.35%). All patients operated by open reduction and osteosynthesis (group A; n = 10; 19.6%), and in combination with an osteoplasty by autograft from the iliac crest, biocermic implant (group B; n = 37; 80.4%). Results. At 18 months, excellent results (100–90 points for AOFAS) were achieved in 14 (41.6% - group A; 58.4% - group B) patients, good (89–80 points for AOFAS) in 23 (42, 8% - group A; 57.2% - group B) patients, satisfactory (79-70 points for AOFAS) - in 10 (60.0% - group A; 40.0% - group B) patients, unsatisfactory (less from 70 AOFAS points) - in 4 (75.0% - group A; 25.0% - group B) patients. Conclusions. The use of a bio-ceramic bone implant has proven to be a promising direction for osteoplastic interventions with good deformation resistance in the long term. Replacement of bone defect with an auto-osseous graft at the reference osteoplasty in separate terms showed higher bone density compared to none, but less than when using a bioceramic filler Post-operative deformity of the calcaneus testifies to the difficulties of treatment of this pathology, which requires further search for the most optimal materials for replacement of the bone cavity in order to achieve the possibility of early mobilization, preservation of the anatomic structure of the calcaneus, congruence of joint surfaces, prevention of complications.
APA, Harvard, Vancouver, ISO, and other styles
6

Chun, Yong-Min. "Remplissage Procedure: When and How?" Orthopaedic Journal of Sports Medicine 7, no. 11_suppl6 (November 1, 2019): 2325967119S0045. http://dx.doi.org/10.1177/2325967119s00457.

Full text
Abstract:
Hill-Sachs lesions were first described in 1940 as grooved defect in the posterior aspect of the humeral head associated with traumatic anterior glenohumeral dislocation. The reported incidence of Hill-Sachs lesions following traumatic anterior instability events ranges from 60% to 90%. Despite recognition of the Hill-Sachs lesion for more than 75 years, most shoulder stabilization procedures have focused on repairing or augmenting the torn or deficient anterior soft tissues and glenoid bone in hopes of preventing engagement of the posterior humeral head defect. To help identify lesions that are important causes of instability, both Palmer and Widen and Burkhart and De Beer described the ‘‘engaging’’ Hill-Sachs lesion, which refers to one that engages the rim of the glenoid when the shoulder is physiologically abducted and externally rotated. Engaging Hill-Sachs lesions lead to recurrent instability, and a high rate of failure when treated with arthroscopic Bankart repair alone. There are two methods to assess the Hill-Sachs lesion related with surgical decision making for concomitant remplissage procedure. One method is dynamic examination. During arthroscopic surgery, the relative relationship between the Hill-Sachs lesion and the glenoid can be assessed. The important thing is that this dynamic examination should be performed after the Bankart repair. The disadvantage of this method is that there is a risk of damaging the repair during the dynamic examination. The second method is to use the ‘glenoid track’ concept. The width of the glenoid track, defined as the distance between the medial margin of the glenoid track and the medial margin of the footprint of the rotator cuff was 83% of the glenoid width when the arm was at 90° of abduction in live shoulders. Our institution use en face views of both glenoids and the posterior view of the involved humeral head on 3D CT. First, we measure the width of the intact glenoid and calculate 83% of the glenoid width(0.83D). Then, this 83% value (0.83D) is applied to the involved glenoid en face view. If there is a bony defect of the glenoid, the defect width ‘d’ needs to be subtracted from the 83% value (0.83D) to obtain the true width of the glenoid track (0.83D - d). We apply this width (0.83D - d) to the posterior view of the humeral head. If the medial margin of the Hill-Sachs le stays within the glenoid track, there is no risk that this Hill-Sachs lesion engages with the anterior rim of the glenoid. If the Hill-Sachs lesion extends more medially over the medial margin of the glenoid track, there is a risk of engagement and dislocation. The former used to be called ‘on-track HSL’ and the latter ‘off-track Hill-Sachs lesion’. Based on the on-track/off-track concept, treatment strategy is as follows. For shoulders with on-track Hill-Sachs lesion and glenoid bone loss of < 25%, Bankart repair alone is sufficient. With on-track Hill-Sachs lesion and glenoid bone loss of ≥ 25%, the glenoid bone loss needs to be fixed, for example by the Latarjet procedure. With off-track Hill-Sachs lesion and the glenoid bone loss of < 25%, Bankart repair plus remplissage is needed. In addressing recurrent anterior shoulder instability, surgical decision making for additional remplissage procedure is inevitable. There are two methods: one is glenoid track method which can be employed in preoperative evaluation. The other is assessment of engaged Hill-Sachs lesion during arthroscopic evaluation. Which one do you prefer? References Hill, H.A. and M.D. Sachs, The grooved defect of the humeral head: a frequently unrecognized complication of dislocations of the shoulder joint. Radiology, 1940. 35(6): p. 690-700. Burkhart, S.S. and J.F. De Beer, Traumatic glenohumeral bone defects and their relationship to failure of arthroscopic Bankart repairs: significance of the inverted-pear glenoid and the humeral engaging Hill-Sachs lesion. Arthroscopy: The Journal of Arthroscopic & Related Surgery, 2000. 16(7): p. 677-694. Flatow, E.L. and J.J. Warner, Instability of the shoulder: Complex problems and failed repairs: Part 1. Relevant biomechanics, multidirectional instability, and severe loss of glenoid and humeral bone. Journal of Bone and Joint Surgery, 1998. 80(1): p. 122. Lynch, J.R., et al., Treatment of osseous defects associated with anterior shoulder instability. Journal of shoulder and elbow surgery, 2009. 18(2): p. 317-328. Buza, J.A., 3 rd, et al., Arthroscopic Hill-Sachs remplissage: a systematic review. J Bone Joint Surg Am, 2014. 96(7): p. 549-55. Spatschil, A., et al., Posttraumatic anterior-inferior instability of the shoulder: arthroscopic findings and clinical correlations. Archives of orthopaedic and trauma surgery, 2006. 126(4): p. 217-222. Yiannakopoulos, C.K., E. Mataragas, and E. Antonogiannakis, A comparison of the spectrum of intra-articular lesions in acute and chronic anterior shoulder instability. Arthroscopy: The Journal of Arthroscopic & Related Surgery, 2007. 23(9): p. 985-990. Itoi, E., ‘On-track’ and ‘off-track’ shoulder lesions. EFORT Open Rev, 2017. 2(8): p. 343-351. Parke, C., et al. Arthroscopic remplissage for humeral defect in anterior shoulder instability: is it needed. in 39th annual meeting of Japan Shoulder Society, Tokyo. 2012. Yamamoto, N., et al., Contact between the glenoid and the humeral head in abduction, external rotation, and horizontal extension: a new concept of glenoid track. Journal of shoulder and elbow surgery, 2007. 16(5): p. 649-656. Omori, Y., et al., Measurement of the glenoid track in vivo as investigated by 3-dimensional motion analysis using open MRI. The American journal of sports medicine, 2014. 42(6): p. 1290-1295. Di Giacomo, G., E. Itoi, and S.S. Burkhart, Evolving concept of bipolar bone loss and the Hill-Sachs lesion: from “engaging/non-engaging” lesion to ”on-track/off-track” lesion. Arthroscopy: The Journal of Arthroscopic & Related Surgery, 2014. 30(1): p. 90-98. Locher, J., et al., Hill-Sachs off-track lesions as risk factor for recurrence of instability after arthroscopic Bankart repair. Arthroscopy: The Journal of Arthroscopic & Related Surgery, 2016. 32(10): p. 1993-1999. Shaha, J.S., et al., Clinical validation of the glenoid track concept in anterior glenohumeral instability. JBJS, 2016. 98(22): p. 1918-1923. Yamamoto, N., et al., The stabilizing mechanism of the Latarjet procedure: a cadaveric study. JBJS, 2013. 95(15): p. 1390-1397. Connolly, J., Humeral head defects associated with shoulder dislocation: their diagnostic and surgical significance. Instr. Course Lect., 1972. 2: p. 210-218. Purchase, R.J., et al., Hill-Sachs “remplissage”: an arthroscopic solution for the engaging Hill-Sachs lesion. Arthroscopy: The Journal of Arthroscopic & Related Surgery, 2008. 24(6): p. 723-726. Boileau, P., et al., Arthroscopic Hill-Sachs Remplissage with Bankart Repair: Strategy and Technique. JBJS Essent Surg Tech, 2014. 4(1): p. e4.
APA, Harvard, Vancouver, ISO, and other styles
7

Fernando, Adrian F., and Joselito F. David. "Combination of Autologous Protein-Rich Fibrin and Bone Graft: An Invaluable Option for Reconstruction of Segmental Mandibular Defects." Philippine Journal of Otolaryngology-Head and Neck Surgery 28, no. 1 (June 18, 2013): 38–42. http://dx.doi.org/10.32412/pjohns.v28i1.509.

Full text
Abstract:
Dear Editor, Reconstruction of mandibular defects resulting from ablative surgery for benign and malignant tumors remains a reconstructive challenge. For the past decade, the fibular free flap has been the workhorse for large mandibular defects because of its length, versatility, and ability to be harvested with a skin paddle for soft tissue closure. Although its success rate has continuously improved to almost 95%, donor site morbidity remains a matter of concern.1,2 Bone grafts are already widely used in dental surgery but only as fillers for chipped or marginal defects and not for large segmental mandibular defects. We present a new technique of reconstructing segmental mandibular defects using bone grafts combined with autologous platelet-rich fibrin (PRF), a biomaterial derived intra-operatively from the patient that incorporates leukocytes, platelets, growth factors, and a wide range of glycoproteins in a dense fibrin matrix. Moreover, we describe the essential role of PRF in bone healing and regeneration that offers an invaluable reconstructive option that is free of donor site morbidity without sacrificing the main goal of reconstruction in restoring both form and function. Keywords: Mandibular reconstruction; segmental mandibular defect; bone graft; autologous Platelet-Rich Fibrin (PRF) MATERIALS AND METHODS Subject and Indications A 23-year-old male underwent reconstruction with allogenic bone graft in combination with autologous platelet-rich fibrin (PRF) for a large segmental angle to parasymphyseal mandibular defect. (Figure 1) As in this example, the authors’ technique for segmental mandibular defect reconstruction using bone grafts with PRF was best performed as a second stage procedure following tumor ablation to prevent contamination from oral cavity secretions. As with any elective procedure, a thorough review of the medical history, control of systemic disease and informed consent were necessary. Patients with contraindications for fibular free flaps, such as history of peripheral vascular disease, unfavorable imaging of the lower extremity, venous insufficiency, and anomalous lower extremity vasculature may benefit from this technique.3 Patients who had failed mandibular reconstruction with other methods such as those reconstructed with single alloplastic material, titanium plate, non-vascularized autologous bone graft, or free flaps are likewise candidates for this option. Procedure A submandibular incision was made and a sub-platysmal flap was raised to expose the entire length of the mandibular defect. Apart from the preservation of vital structures in the area, it was important not to violate the oral mucosa to prevent contamination of the reconstruction site. A cortico-cancellous allogenic bone graft (Maxgraft®, Botiss Medical AG , Berlin, Germany) was fitted to the mandibular defect and anchored with bicortical screws (2.5; 2.8 mm, lengths 14-20 mm) to a pre-bended 2.5 mm reconstruction titanium plate (Modus Reco 2.5, Medartis, Hochbergerstrasse Basel Switzerland). The remaining gaps between the inlaid grafts were filled with the remaining cancellous bone and biomimetic composite materials. (Figure 2) Although allogenic bone grafts with cortical and cancellous components are recommended for mandibular ramus and condylar reconstruction as these regions are composed of nearly 100% cortical bone, xenogenic (Cerabone®, Botiss Medical AG , Berlin, Germany) or combined alloplastic material- (Maxresorb®, Botiss Medical AG , Berlin, Germany) bone grafts may be used in other regions. (Figure 3) Meanwhile, venous blood was simultaneously drawn from the patient and placed in a 10 cc glass collecting tube for single centrifuge processing using a PC-O2 centrifuge (PC-O2, Process, Nice, France). The specific centrifuge processes 8 uncoated tubes using a standard protocol specially manufactured for processing PRF using 33° tube angulation at 2700 RPM’s, soft spin for 12 minutes.4 (Figure 4) At the end of the centrifugation process, three distinct fractions of blood components were produced where the intermediate fraction composed of dense PRF clot was used. The other blood components separated by the centrifugation process- serum or platelet-poor plasma (PPP) and red blood cell concentrates, were respectively situated in the superficial and bottom layers of the collecting tube. The PRF clots were then transferred to a PRF processing box (PRF Box®, Process, Nice, France) to prepare standardized membranes and harvest the PRF exudates in a sterile environment.5 (Figure 5) Collagen material of native pericardium (GTR/GBR) membrane (Jason® membrane, Botiss Medical AG, Berlin, Germany) was placed underneath the graft recipient site and the processed PRF membranes were layered over the graft recipient site to stimulate osteoblastic differentiation and neoangiogenesis.6 The entire recipient site was then enveloped with the collagen material, mechanically securing the autologous PRF in contact with the bone grafts. (Figure 6) This established a membrane barrier for guided bone regeneration (GBR) and guided tissue regeneration (GTR) by preventing growth of undesired cells inside the neomandible and allowing osteogenesis and angiogenesis.7 Commercially available collagen biomaterials vary from native collagen membrane to enhanced Ca/P collagen composite materials such as the 3D-stable collagen graft (Mucoderm®, Botiss Medical AG, Berlin, Germany) with larger available sizes for long mandibular defect coverage. The skin was closed in the usual manner and the patient was initially maintained on a liquid diet, progressing to a soft diet over 2-4 weeks. Plain panoramic radiographs after a week confirmed proper alignment of the bone grafts and monthly radiographic series was recommended for the first 6 months after reconstruction. RESULTS Monthly panoramic radiographs for the first 6 months after reconstruction showed absence of bone resorption. A 3D reconstruction CT imaging of the mandible was done after 10 months for placement of three osteo-integrated dental implants. (Figure 7) Bone biopsies were also taken in conjunction with placement of dental implants, and sent to the University of Bonn, Germany for histologic evaluation. (Figure 8) The trichrome-stained specimens showed new mineralized tissues consisting of woven bone characterized by high numbers of distributed osteocytes and irregularly arranged fiber bundles within the new bone matrix confirming bone regeneration. (Figure 9) The latest panoramic radiograph of the patient at 26 months after surgery showed absence of gaps between the bone grafts and their junction with the normal mandible, evincing complete bone regeneration and a successful mandibular reconstruction. (Figure 10) A total of 14 cases of segmental mandibular defects have been reconstructed by the authors using the particular technique from January 2011 to February 2013 with 100% success rate and will be reported as a series in the near future. DISCUSSION Advancements in mandibular reconstruction have continued to develop over the past decades. The use of alloplastic materials like titanium plating implants for repairing mandibular defects provided patients with rapid rigid mandibular restoration but was limited by numerous complications such as infection, plate extrusion, and subsequent failure. Recently, the concept of distraction osteogenesis, involving bone distraction with an external mechanical device and progressive lengthening of the bone to allow a gap of new bone during the consolidation phase has also been used for mandibular reconstruction but has been limited by poor scar formation, delayed return to function, and inadequate formation of desired bone length.8 The advent of microvascular reconstructive surgery enabled the transfer of vascularized osseous flaps with the most commonly used fibular free flap showing superior results over non-vascularized bone transfer and better quality of life outcome. However, it did not remain free of donor-site morbidities.9 Tissue engineering led to the development and use of bone grafts that hold promise for the future of head and neck repair.10 Numerous clinical studies demonstrated the utility of tissue engineering in developing bone grafts for mandibular defect reconstruction.11 Such have already been widely used over the past decades in oro-maxillary and dental reconstruction, including the recombinant bone morphogenetic protein (rhBMP-2) that is now used with great success in cleft palate repair, alveolar ridge augmentation, and sinus lift procedures.12 Autogenous bone grafts derived from the patient work through osteogenesis, osteoinduction and osteoconduction. However, such are not recommended because apart from enabling a donor site morbidity-free technique, they are best harvested as microvascular flaps. Allogenic bone grafts on the other hand are cadaveric processed grafts that may be cortical, trabecular, or combined in composition and have both osteoconductive and osteinductive properties. Xenografts or processed animal bone graft are a subgroup of the synthetically manufactured alloplasts known to form new bones from their osteoconductive activities. The use of autologous PRF is already widely used in combination with bone grafts for dental surgeries but not for large mandibular defects.13 Our reconstructive technique using bone grafts for large segmental mandibular defects emphasizes the important role of PRF with its intrinsic factors and leukocyte contents that release high amounts of growth factors such as TGBß1, PDGF-AB, VEGF and matrix glycoproteins.14 Collagen membrane used to envelop the entire recipient site creates a membrane barrier to prevent the growth of soft tissues and allow angiogenesis within the neomandible.15 Fascia lata may be used as a membrane barrier but defeats the authors’ goal of an absolute donor site morbidity-free procedure. Overall, this particular technique along with gentle tissue handling and avoidance of oral cavity contamination for reconstructing large mandibular defects has been found to enhance bone regeneration capable for osteo-integrated dental implantation. Generally, the harvesting and processing of autologous PRF is simple and inexpensive. Its use with bone grafts is a good substitute for segmental mandibular reconstruction in patients with contraindications to free flap procedures or in cases where patients simply wish to be free from any donor-site morbidity. However, this technique is limited to defects secondary to trauma and ablation of benign conditions as bone regeneration is expected in approximately 6 to 9 months. Mandibular defects following resection of malignant oral neoplasms are still best reconstructed with fibular free flaps as radiation therapy is warranted at the soonest possible time. Histologic validation of bone regeneration and osteoblastic activity index for the 13 other cases performed by the authors using this particular technique necessitates bone research centers that are capable of advanced bone analysis, and none are locally-available at this time. Meanwhile, this technique of combining autologous PRF in bone grafting remains an innovative and invaluable option for mandibular reconstruction today.
APA, Harvard, Vancouver, ISO, and other styles
8

Lukovic, Natalija, Obrad Zelic, Sasa Cakic, and Vanja Petrovic. "The use of beta-tricalcium phosphate and bovine bone matrix in the guided tissue regeneration treatment of deep infra-bony defects." Srpski arhiv za celokupno lekarstvo 137, no. 11-12 (2009): 607–12. http://dx.doi.org/10.2298/sarh0912607l.

Full text
Abstract:
Introduction. The primary goal of bone regeneration procedures with application of various regenerative biologic agents and biomaterials is to facilitate the formation of periodontal tissues lost as a result of periodontitis. Objective. The aim of the study was to compare clinical outcome of the guided tissue regeneration (GTR) treatment with the use of ?-tricalcium phosphate and with bovine bone matrix in human deep intra-osseous defects. Methods. Twenty-one systemically healthy subjects with moderate to advanced periodontitis, between 30 and 56 years of age, 11 females and 10 males, were selected. Patients having two similar inter-proximal defects with pocket probing depths following initial therapy greater than 5 mm were recruited for the study. Experimental sites were grafted with pure ?-tricalcium phosphate biomaterial (Cerasorb?) and a biomembrane, while control sites were treated with bovine-bone hydroxiapatite xenograft (Bio-oss?) and a biomembrane. Immediately before surgery and 12 months after surgery, pocket probing depth (PPD), epithelial attachment level (EAL) and gingival recession (GR) were evaluated. Results. In the experimental group PPD amounted to 6.76?0.83 mm before surgery, and decreased significantly to 2.67?0.48 mm 12 months following surgery, while in the control group PPD significantly decreased from 7.14?0.65 mm presurgically to 2.85?0.57 mm postsurgically. After one year, EAL gain was 2.76?0.99 mm in the experimental group, and 3.24?0.16 mm in the control group. After twelve months postoperatively GR amounted to 1.33?0.79 mm in the experimental group and to 1.05?0.80 mm in the control group. No statistically significant differences for PPD reduction, EAL gain and GR increase were detected between the groups. Conclusion. Results from the present study indicate that GTR treatment of deep intra-osseous defects with Bio-oss? and Cerasorb? resulted in clinically and statistically significant improvement of EAL gain and PPD reduction. A GR was slightly increased, with no statistical significance.
APA, Harvard, Vancouver, ISO, and other styles
9

Md. Jalaluddin, DhirendraK Singh, Ipsita Jayanti, Prasad Kulkarni, Mohamed Faizuddin, and Fouzia Tarannum. "Use of platelet rich plasma in the management of periodontal intra-osseous defects: A clinical study." Journal of International Society of Preventive and Community Dentistry 7, no. 2 (2017): 105. http://dx.doi.org/10.4103/jispcd.jispcd_28_17.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Chhabra, Dr Anukriti, Dr AshitaS Uppoor, Dr Swati Pralhad, Dr Neelesh Singh, and Dr DilipG Nayak. "QUANTIFICATION OF INTRA OSSEOUS BONE DEFECTS BY CONVENTIONAL VERSUS 3DIMENSIONAL IMAGING TECHNIQUES - AN IN VIVO STUDY." International Journal of Advanced Research 4, no. 5 (May 31, 2016): 1377–85. http://dx.doi.org/10.21474/ijar01/460.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Parashari, Umesh Chandra, Sachin Khanduri, Samarjit Bhadury, and Fareena Akbar Qayyum. "Radiographic diagnosis of a rare case of oculo-dento-digital dysplasia." South African Journal of Radiology 15, no. 4 (December 7, 2011): 134. http://dx.doi.org/10.4102/sajr.v15i4.359.

Full text
Abstract:
Oculodentodigital dysplasia (ODDD), also known as oculodento-osseous dysplasia, is an extremely rare autosomal dominant disorder with high penetrance, intra- and interfamilial phenotypic variability, and advanced paternal age in sporadic cases. The incidence of this disease is not precisely known, with only 243 cases reported in the scientific literature, suggesting an incidence of around 1 in 10 million people. It is marked mainly by eye abnormalities, craniofacial dysmorphism, dental anomalies, hand and foot malformations, various skeletal defects, and mildly delayed mental development. Neurological changes may appear earlier in each subsequent generation. This case report describes a radiological diagnosis of ODDD based on physical appearance, clinical features and radiographic findings in a 16-year-old girl.
APA, Harvard, Vancouver, ISO, and other styles
12

Hagen, Mia, William Hannay, Quinn Saluan, T. Sean Lynch, Robert Westermann, and James Rosneck. "How Well Does MRI Predict Chondral Lesions in Patients with Femoroacetabular Impingement? An Analysis of 545 Cases (137)." Orthopaedic Journal of Sports Medicine 9, no. 10_suppl5 (October 1, 2021): 2325967121S0027. http://dx.doi.org/10.1177/2325967121s00276.

Full text
Abstract:
Objectives: Articular cartilage injury has been identified as a risk factor for poor outcomes following hip arthroscopy for femoroacetabular impingement syndrome (FAI). The purpose of this study was to evaluate the efficacy of magnetic resonance imaging (MRI) in detecting cartilage defects, and to identify specific MRI findings associated with cartilage injury. Methods: All patients undergoing hip arthroscopy between February 2015 and May 2017 at one institution were enrolled in a prospective cohort. Intra-articular findings were documented at the time of surgery. MRI reports were retrospectively reviewed for radiologist-reported articular cartilage, osseous or synovial abnormalities. Sensitivity and specificity of MRI findings were calculated; multivariate logistic regression analysis determined which findings were associated with high-grade chondral damage at time of arthroscopy and used to create an online risk calculator, https://orthop.washington.edu/hiprisk/. Results: Out of a total of 598 patients who underwent hip arthroscopy, 550 had MRI reports available for review (92%). Grade III and IV cartilage injuries were reported on arthroscopy in 70 patients (13%) of average age 33 ± 13 years. On univariate analyses, individual MRI findings were not found to be sensitive in detection of articular cartilage injury (mean 22%, range 1.4% – 46%), but positive findings were highly specific (mean 90%, range 76% – 99%). Multivariate analysis revealed that older age (OR 1.09 [1.06-1.11], p < .001) and osseous findings such as subchondral cyst or edema (OR 4.77 [2.51-9.05], p <.001) were most predictive of grade III and IV defects (p < .001). An example of use of the online calculator is in Figure 1. Conclusions: MRI was a specific but not sensitive tool in diagnosing articular cartilage injury. Surgeons should be aware that osseous findings such as cysts or edema are highly predictive of full-thickness cartilage loss in FAI.
APA, Harvard, Vancouver, ISO, and other styles
13

Grob, Karl, Chan-Hi Kim, and Tobias Dietrich. "Postoperative Imaging after Impingement Surgery." Seminars in Musculoskeletal Radiology 21, no. 05 (October 12, 2017): 529–38. http://dx.doi.org/10.1055/s-0037-1606131.

Full text
Abstract:
AbstractIn addition to the patient's medical history and clinical evaluation, conventional radiographs and magnetic resonance imaging (MRI) are important tools to indicate appropriate conservative treatment or even revision surgery in patients with symptoms after surgical management of femoroacetabular impingement (FAI). We present an overview of current evidence in postoperative imaging after impingement surgery. Undercorrection of the underlying osseous FAI configuration is the most frequent indication for revision surgery within the first 2 years after index FAI surgery. Femoral neck fractures, iatrogenic chondral injuries, early conversion to total hip arthroplasty, loose bodies, and heterotopic ossifications are rare but typical early complications after surgical treatment of FAI. Abnormal MRI findings after FAI surgery such as intra-articular adhesions, labral tears, cartilage defects, and anterior capsular defects are common findings in both asymptomatic and symptomatic postoperative patients. Avascular necrosis of the femoral head is an extremely rare complication after surgical treatment of FAI.
APA, Harvard, Vancouver, ISO, and other styles
14

Vougioukas, Vassilios I., Ulrich Hubbe, Vera van Velthoven, Thomas M. Freiman, Alexander Schramm, and Uwe Spetzger. "Neuronavigation-assisted Cranial Reconstruction." Neurosurgery 55, no. 1 (July 1, 2004): 162–67. http://dx.doi.org/10.1227/01.neu.0000126940.20441.e7.

Full text
Abstract:
Abstract OBJECTIVE: Cranial defects resulting from congenital deformities, ablative resection of osseous tumors, traumatic injury, and destructive infectious lesions are often severe enough to warrant surgical reconstruction. In particular cases, satisfactory cosmetic results may be difficult to achieve because of the extent and location of the lesion. METHODS: We evaluated the role of neuronavigation for reconstruction of large cranial defects with prefabricated titanium and intraoperatively constructed neuronavigation-assisted polymethylmethacrylate implants. RESULTS: Neuronavigation-assisted cranial reconstruction was performed in 14 patients. Surgical procedure, illustrative cases, postoperative results, and apparent benefits of the technique are presented and discussed. In all patients, excellent cosmetic results were achieved. CONCLUSION: In cranial reconstruction, neuronavigation is of value not only for intraoperative determination of resection margins but also for preoperative assessment and planning. The combination of navigation techniques with prefabricated or intra-operatively constructed implants enables achievement of excellent cosmetic results.
APA, Harvard, Vancouver, ISO, and other styles
15

Persson, G. Rutger, Hanne Falk, and Lars Laurell. "A retrospective radiographic outcome assessment study of intra-bony defects treated by osseous surgery or by bone graft procedures." Journal of Clinical Periodontology 27, no. 2 (February 2000): 104–8. http://dx.doi.org/10.1034/j.1600-051x.2000.027002104.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
16

abd Al Rahman, ola, hala hazzaa, and mai Attia. "Evaluation of autogenous Bone Graft with or without Intra-Marrow Penetration in Treating One Osseous Wall Defects: An experimental study." Al-Azhar Dental Journal for Girls 8, no. 3 (July 1, 2021): 437–42. http://dx.doi.org/10.21608/adjg.2021.23244.1231.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Minenna, Luigi, Federico Herrero, Mariano Sanz, and Leonardo Trombelli. "Adjunctive effect of a polylactide/polyglycolide copolymer in the treatment of deep periodontal intra-osseous defects: a randomized clinical trial." Journal of Clinical Periodontology 32, no. 5 (May 2005): 456–61. http://dx.doi.org/10.1111/j.1600-051x.2005.00696.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

Masood, Farah, Christos Angelopoulos, and Alan Glaros. "A Comparison Between Two Types of Radiographic Film for Accuracy of Measurements of Approximal Osseous Defects." Journal of Contemporary Dental Practice 7, no. 3 (2006): 22–29. http://dx.doi.org/10.5005/jcdp-7-3-22.

Full text
Abstract:
Abstract Aim The purpose of this study was to compare two types of conventional radiographic film: Ekta-speed plus and Insight (Eastman Kodak Co, Rochester NY, USA) for accuracy of measurements of approximal bone loss. Methods and Materials Four dried human mandibles with complete dentition were selected. Radiographic images were made with a standardized technique. Mesial and distal bone levels on the mandibular premolar and molar teeth were measured on the two types of radiographs (Ekta-speed plus and Insight) by nine observers. The data obtained by the observers were compared with the primary investigators’ corresponding measurements, which served as “the ground truth” for this study. Results The results of the analysis of variance (ANOVA) demonstrated a significant difference for the Ektaspeed plus film (p = .001), but the difference was not clinically significant. The level of intra-rater reliability was high for the observers (0.969 to 0.990). Conclusion Performance of Ekta-speed plus and Insight films was found to be similar. Citation Masood F, Angelopoulos C, Glaros A. A Comparison Between Two Types of Radiographic Film for Accuracy of Measurements of Approximal Osseous Defects. J Contemp Dent Pract 2006 July;(7)3:022-029.
APA, Harvard, Vancouver, ISO, and other styles
19

Jindal, Vikas, AmarjitSingh Gill, Daljit Kapoor, and Harinder Gupta. "The comparative efficacy of decalcified allogenic bone matrix and intra-oral free osseous autografts in the treatment of periodontal intrabony defects." Journal of Indian Society of Periodontology 17, no. 1 (2013): 91. http://dx.doi.org/10.4103/0972-124x.107482.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Sipos, P. M., B. G. Loos, F. Abbas, M. F. Timmerman, and U. van der Velden. "The combined use of enamel matrix proteins and a tetracycline-coated expanded polytetrafluoroethylene barrier membrane in the treatment of intra-osseous defects." Journal of Clinical Periodontology 32, no. 7 (July 2005): 765–72. http://dx.doi.org/10.1111/j.1600-051x.2005.00754.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Listl, Stefan, Yu-Kang Tu, and Clovis Mariano Faggion Jr. "A cost-effectiveness evaluation of enamel matrix derivatives alone or in conjunction with regenerative devices in the treatment of periodontal intra-osseous defects." Journal of Clinical Periodontology 37, no. 10 (August 19, 2010): 920–27. http://dx.doi.org/10.1111/j.1600-051x.2010.01611.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Seoud, Muhammed Abouel, Medhat Kataia, Mouchira Salah El Din, Magdy Ali, and Adel AboelFattouh. "The Usage of Low Level Laser Therapy Bio Stimulation and Bone Grafting in Accelerating the Healing of chronic intra-Osseous Defects: Vivo Study." Indian Journal of Public Health Research & Development 10, no. 9 (2019): 1172. http://dx.doi.org/10.5958/0976-5506.2019.02602.0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Stachel, Daniel, Nora Naumann-Bartsch, Patrick Morhart, Markus Metzler, Thorsten Langer, Klaus Schwarz, and Wolfgang Holter. "TBI Based Conditioning Regimen and Intra Osseous Application of PBSC to Overcome Graft Resistance In a Patient with Autoimmune Lymphoproliferative Syndrome (ALPS)." Blood 116, no. 21 (November 19, 2010): 4696. http://dx.doi.org/10.1182/blood.v116.21.4696.4696.

Full text
Abstract:
Abstract Abstract 4696 Autoimmune lymphoproliferative syndrome (ALPS) is a rare immunologic disorder caused by defects in the Fas-induced programmed cell death pathway. Impaired lymphocyte apoptosis results in gradual lymphocyte accumulation and dysregulation of lymphocyte homeostasis. ALPS patients usually suffer from persistent generalized lymphadenopathy, hepato-splenomegaly, immune-mediated cytopenias, and other autoimmune phenomena. A 15-month-old boy was diagnosed with ALPS when he presented with typical symptoms. Lab exams showed a pancytopenia, elevated serum immunoglobulin levels, a peripheral expansion of double-negative T lymphocytes of up to 40% of TCRab+ T cells and impaired lymphocyte apoptosis. Molecular analysis confirmed the diagnosis of a type Ia ALPS by identifying a heterozygous Fas gene mutation (D260H). Despite treatment with repeated pulses of high-dose methylprednisolone, intravenous immunoglobulins, and mycophenolate mofetil (MMF) to control lymphoproliferation and recurrent pancytopenia his disease progressed. At three years of age he developed numerous arterial aneurysms of the iliac, mesenterial, renal, hepatic, right middle meningeal, brachial and femoral arteries up to 2.6 cm in diameter and lymphoproliferation resulting in paraplegia and right arm paresis. Stem cell transplantation was considered, and BMT from a 9/10 matched unrelated donor (MUD) after a reduced intensity conditioning regimen (CR) with Fludarabin (150 mg/m2), Melphalan (140 mg/m2/d) and ATG (60mg/kg) was performed using 5.22 × 106 CD34+ cells/kg body weight. However, graft failure had to be diagnosed on day +27. 53 days after the first BMT a PBSCT from the same donor after myeloablative CR using Busulfan (19.2 mg/kg/d i.v.), Etoposid (30 mg/kg/d), Cyclophosphamid (120 mg/kg/d) and ATG (60mg/kg) was performed (24.5 × 106 CD34+ cells/kg bw). Again, graft failure was seen. At day +55 a third HSCT using PBSCT from another 9/10 MUD after CR with Fludarabin i.v. (160 mg/m2), Thiotepa (5 mg/kg/d), 4 Gy total body irradiation (TBI) and campath (1 mg/kg) was performed. 10.8 × 106 CD34+ cells/kg bw were given intra osseous, 9.8 × 106 CD34+ cells/kg bw were given i.v. Engraftment was slow (Leukos 980 day +35), but chimerism showed 99 % donor cells. Two years later the patient is alive and well, with persistent engraftment and good hematological and immunological function. Arterial aneurysms stopped growing and some have thrombosed. This case illustrates some interesting points. Atypical and unusually severe manifestations of ALPS forced us to perform a HSCT in this patient. In severe ALPS stem cell engraftment is difficult to achieve as previously reported in the Fas deficient lpr mouse model. One reason might be the reduced ability of cytotoxic drugs to induce apoptosis in the Fas deficient recipient T cells. Thus, recipient T cells could persist and kill donor cells resulting in graft failure or rejection. Additionally, increased FasL expression on recipient cells could induce apoptosis in Fas bearing donor stem cells as shown in lpr mice. Also, trapping of infused stem cells in the extremely enlarged liver and spleen could have played a role. A 3rd attempt was therefore designed to overcome graft resistance and proved finally successful. The use of TBI together with campath possibly induced more T cell apoptosis than chemotherapy alone. The intra osseous application probably increased engraftment efficiency by avoiding trapping of stem cells in liver and spleen and possibly by induction of tolerance by intra osseous application as described before. Disclosures: No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
24

Trombelli, Leonardo, Marco Annunziata, Salvatore Belardo, Roberto Farina, Alessandro Scabbia, and Luigi Guida. "Autogenous bone graft in conjunction with enamel matrix derivative in the treatment of deep periodontal intra-osseous defects: a report of 13 consecutively treated patients." Journal of Clinical Periodontology 33, no. 1 (January 2006): 69–75. http://dx.doi.org/10.1111/j.1600-051x.2005.00865.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
25

Yanke, Adam, Navya Dandu, Blake Bodendorfer, Reem Darwish, Athan Zavras, Brian Forsythe, Brian Cole, and Nicholas Trasolini. "Paper 18: Effect of Bone Marrow Aspirate Concentrate on Osteochondral Allograft Transplantation Incorporation: A Prospective, Randomized, Single Blind Investigation." Orthopaedic Journal of Sports Medicine 10, no. 7_suppl5 (July 1, 2022): 2325967121S0058. http://dx.doi.org/10.1177/2325967121s00582.

Full text
Abstract:
Objectives: Osteochondral allograft transplantation for cartilage defects of the knee has demonstrated excellent long-term clinical outcomes and survival, which largely depends on successful graft osseointegration. Biologics have been suggested as a viable adjunct to enhance successful healing in several musculoskeletal applications. In the context of OCA, early results have suggested that BMAC may improve cellular repopulation and viability within the osseus portion of an implanted graft. However, few clinical studies to date have investigated the impact of BMAC on patient outcomes following OCA. The purpose of this study was to investigate the effect of bone marrow aspirate concentrate (BMAC) on osseointegration and patient-reported outcome metrics (PROMs) after osteochondral allograft transplantation in a prospective, randomized controlled single-blinded trial. Methods: Patients undergoing osteochondral allograft transplantation of the knee were consented and enrolled. Prior to surgery, patients were randomized into either the BMAC or sham incision groups. In the BMAC group, the osteochondral allograft plug was soaked in BMAC for a minimum of 2 minutes prior to implantation. All patients underwent postoperative computed tomography (CT) scanning at 6 months postoperatively and completed PROMs preoperatively, 6 months, and 1 year postoperatively. Two board-eligible orthopaedic surgeons blinded to treatment allocation independently assessed and graded each CT according to the ACTOCA system proposed by Gelber et al. Results: Thirty-six patients enrolled between April 2018 to December 2020 (17 female, 19 male) were included for analysis. There were no significant differences between the BMAC and non-BMAC groups in graft signal density (Grader 1: p=0.283, Grader 2: p=0.467), osseous integration (both graders: p=0.489), surface percentage with discernible cleft (Grader 1: 0.287, Grader 2: 0.469), or intra-articular fragments (Grader 1: p=0.617, Grader 2: p=0.810) (Table 1). A significantly greater number of patients receiving BMAC demonstrating cystic changes <3 mm (Grader 1: p=0.015, Grader 2: p=0.05) (Figure 1). At 1 year, BMAC patients reported significantly better WOMAC Pain (87.82±14.26 vs 75.80±15.56, p=0.043) and trended towards improved PROMIS Pain (54.14±8.31 vs 61.79±5.24, p=0.09). Conclusions: Patients receiving BMAC soaked OCA grafts demonstrated no difference from controls with respect to graft signal intensity, osseous integration, intra-articular fragments, or discernible graft-host clefts at 6-months postoperatively. BMAC patients had a significantly greater occurrence of small (<3 mm) cystic changes. At 1 year, BMAC patients reported significantly less pain than controls on WOMAC Pain, with similar trends on PROMIS Pain Interference. [Table: see text][Figure: see text]
APA, Harvard, Vancouver, ISO, and other styles
26

Jayakumar, Avula, Palaparthi Rajababu, Surabhi Rohini, Kalakonda Butchibabu, Anumala Naveen, Pathakota Krishnajaneya Reddy, Sisinty Vidyasagar, Durvasula Satyanarayana, and Sayini Pavan Kumar. "Multi-centre, randomized clinical trial on the efficacy and safety of recombinant human platelet-derived growth factor with β-tricalcium phosphate in human intra-osseous periodontal defects." Journal of Clinical Periodontology 38, no. 2 (December 6, 2010): 163–72. http://dx.doi.org/10.1111/j.1600-051x.2010.01639.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Torres Torrillas, M., E. Damiá, B. Cuervo, D. Chicharro, A. Del Romero, L. Miguel, M. Rubio, et al. "Intra-osseous application of plasma rich in growth factors to treat acute cartilage defects in rabbits: evaluation of blood biomarkers (hyaluronic acid and collagen type II cleavage epitope)." Osteoarthritis and Cartilage 29 (April 2021): S409—S410. http://dx.doi.org/10.1016/j.joca.2021.02.533.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Scabbia, Alessandro, and Leonardo Trombelli. "A comparative study on the use of a HA/collagen/chondroitin sulphate biomaterial (BiostiteR) and a bovine-derived HA xenograft (Bio-OssR) in the treatment of deep intra-osseous defects." Journal of Clinical Periodontology 31, no. 5 (May 2004): 348–55. http://dx.doi.org/10.1111/j.1600-051x.2004.00483.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
29

Helal, Marwa EL-Desouky, Riham Omar Ibrahim, Mahmoud Ibrahim Fahmy El-Refaie, and Sarah Mohammed Kenawy. "Clinical and radiographic evaluation of non-incised papilla surgical approach (NIPSA) versus modified minimally invasive surgical technique (M-MIST) in treatment of intraosseous defects in patients with stage III periodontitis." International journal of health sciences, July 8, 2022. http://dx.doi.org/10.53730/ijhs.v6ns6.10482.

Full text
Abstract:
Background: Minimally invasive surgical (MIS) procedure" in multiple and isolated intra-osseous defects maintained the entire interdental papilla with significant improvement in clinical and radiographic outcomes compared to traditional periodontal surgeries. Methods: Thirty-two intra-osseous defects in 15 patients were enrolled for this study and were randomly allocated to both groups i.e. (16 defects) per group. Patients in group I (test group) went for Non-Incised Papilla Surgical Approach (NIPSA), while, in group II (control group), patients received Modified Minimally Invasive Surgical Technique (M-MIST). All clinical and radiographic periodontal parameters were measured at baseline, six months and twelve months. Results: Patients in both groups showed significant improvements in clinical and radiographic outcomes after twelve months of follow-up including reductions in CAL, potential effect in post-surgical patients’ satisfaction and intra-osseous defect fill. Absence of significant difference regarding intergroup comparisons. The minimal surgical flap design plays a very important role in the success of the therapy, creating and maintaining a stable space for the clot. Conclusions: NIPSA is viewed as a promising minimally invasive technique even in hopeless teeth with severe periodontal bone loss, clinical attachment loss to the apex and minimal keratinized tissue.
APA, Harvard, Vancouver, ISO, and other styles
30

Abdulrahman, Yasser Ali, Manal Mohamed Hosny, Ahmed Elfana, and Karim Mohamed Fawzy El-Sayed. "Clinical and radiographic evaluation of low-speed platelet-rich fibrin (PRF) for the treatment of intra-osseous defects of stage-III periodontitis patients: a randomized controlled clinical trial." Clinical Oral Investigations, July 25, 2022. http://dx.doi.org/10.1007/s00784-022-04627-2.

Full text
Abstract:
Abstract Aim The current randomized controlled trial assessed for the first time the effect of a low-speed platelet-rich fibrin (PRF) with open flap debridement (OFD) versus OFD alone in the treatment of periodontal intra-osseous defects of stage-III periodontitis patients. Methods Twenty-two periodontitis patients with ≥ 6 mm probing depth (PD) and ≥ 3 mm intra-osseous defects were randomized into test (PRF + OFD; n = 11) and control (OFD; n = 11) groups. Clinical attachment level (CAL)–gain (primary outcome), PD-reduction, gingival recession depth (GRD), full-mouth bleeding scores (FMBS), full-mouth plaque scores (FMPS), radiographic linear defect depth (RLDD), and radiographic bone fill (secondary-outcomes) were examined over 9 months post-surgically. Results Low-speed PRF + OFD and OFD demonstrated significant intra-group CAL-gain and PD- and RLDD-reduction at 3, 6, and 9 months (p < 0.01). Low-speed PRF + OFD exhibited a significant CAL-gain of 3.36 ± 1.12 mm at 6 months (2.36 ± 0.81 mm for the control group; p < 0.05), and a significantly greater PD-reduction of 3.36 ± 1.12 mm at 3 months, of 3.64 ± 1.12 mm at 6 months and of 3.73 ± 1.19 mm at 9 months (2.00 ± 0.89 mm, 2.09 ± 1.04 mm, and 2.18 ± 1.17 mm in the control group respectively; p < 0.05). No significant differences were notable regarding GRD, FMPS, FMBS, RLDD, or bone fill between both groups (p > 0.05). Conclusions Within the current clinical trial’s limitations, the use of low-speed PRF in conjunction with OFD improved CAL and PD post-surgically, and could provide a cost-effective modality to augment surgical periodontal therapy of intra-osseous defects of stage-III periodontitis patients. Clinical relevance Low-speed PRF could provide a cost-effective modality to improve clinical attachment gain and periodontal probing depth reduction with open flap debridement approaches.
APA, Harvard, Vancouver, ISO, and other styles
31

Elbehwashy, Mohamed Talaat, Manal Mohamed Hosny, Ahmed Elfana, Alaa Nawar, and Karim Fawzy El-Sayed. "Clinical and radiographic effects of ascorbic acid-augmented platelet-rich fibrin versus platelet-rich fibrin alone in intra-osseous defects of stage-III periodontitis patients: a randomized controlled clinical trial." Clinical Oral Investigations, April 12, 2021. http://dx.doi.org/10.1007/s00784-021-03929-1.

Full text
Abstract:
Abstract Aim To assess platelet-rich fibrin (PRF) with ascorbic acid (AA) versus PRF in intra-osseous defects of stage-III periodontitis patients. Methodology Twenty stage-III/grade C periodontitis patients, with ≥ 3 mm intra-osseous defects, were randomized into test (open flap debridement (OFD)+AA/PRF; n = 10) and control (OFD+PRF; n = 10). Clinical attachment level (CAL; primary outcome), probing pocket depth (PPD), gingival recession depth (RD), full-mouth bleeding scores (FMBS), full-mouth plaque scores (FMPS), radiographic linear defect depth (RLDD) and radiographic defect bone density (RDBD) (secondary-outcomes) were examined at baseline, 3 and 6 months post-surgically. Results OFD+AA/PRF and OFD+PRF demonstrated significant intragroup CAL gain and PPD reduction at 3 and 6 months (p < 0.001). OFD+AA/PRF and OFD+PRF showed no differences regarding FMBS or FMPS (p > 0.05). OFD+AA/PRF demonstrated significant RD reduction of 0.90 ± 0.50 mm and 0.80 ± 0.71 mm at 3 and 6 months, while OFD+PRF showed RD reduction of 0.10 ± 0.77 mm at 3 months, with an RD-increase of 0.20 ± 0.82 mm at 6 months (p < 0.05). OFD+AA/PRF and OFD+PRF demonstrated significant RLDD reduction (2.29 ± 0.61 mm and 1.63 ± 0.46 mm; p < 0.05) and RDBD-increase (14.61 ± 5.39% and 12.58 ± 5.03%; p > 0.05). Stepwise linear regression analysis showed that baseline RLDD and FMBS at 6 months were significant predictors of CAL reduction (p < 0.001). Conclusions OFD+PRF with/without AA significantly improved periodontal parameters 6 months post-surgically. Augmenting PRF with AA additionally enhanced gingival tissue gain and radiographic defect fill. Clinical relevance PRF, with or without AA, could significantly improve periodontal parameters. Supplementing PRF with AA could additionally augment radiographic linear defect fill and reduce gingival recession depth.
APA, Harvard, Vancouver, ISO, and other styles
32

Bhardwaj, Smiti. "Low Level Laser Therapy in the Treatment of Intra-Osseous Defect- A Case Report." JOURNAL OF CLINICAL AND DIAGNOSTIC RESEARCH, 2016. http://dx.doi.org/10.7860/jcdr/2016/15805.7466.

Full text
APA, Harvard, Vancouver, ISO, and other styles
33

Cotter, Mar, Zeeshan G. Khattak, and Chris Cotter. "A sagittal split osteotomy approach for removal of a large cementoblastoma at the mandibular angle." Journal of the Irish Dental Association, January 11, 2023. http://dx.doi.org/10.58541/001c.67924.

Full text
Abstract:
Benign lesions at the angle of mandible are frequently removed by a conventional intra oral approach to gain access and achieve complete visualisation. This method is quick and effective when dealing with small benign lesions that are superficially located at the angle of mandible. The removal of large and deeply located lesions with a conventional intra oral approach however, bring about a unique set of challenges particularly when the third molar is displaced towards the inferior border of mandible including lack of complete visualisation of the lesion, difficulty in identification and protection of the inferior alveolar nerve and necessitating removal of a considerable amount of osseous structure thus increasing the risk of a mandibular fracture. Alternative techniques for such lesions include an extra oral approach but one which could potentially create a cosmetic defect from cutaneous scarring and can result in facial nerve injury. This case report describes the use of a unilateral sagittal split osteotomy (SSO) in the removal of a mandibular cementoblastoma. This is a safe and effective technique allowing optimal access to the tumor with complete visualisation, identification and protection of the inferior alveolar nerve with minimal bone removal while maintaining mandibular integrity, strength and facial aesthetics.
APA, Harvard, Vancouver, ISO, and other styles
34

Torres-Torrillas, Marta, Elena Damiá, Pau Peláez, Laura Miguel-Pastor, Belén Cuervo, José J. Cerón, José M. Carrillo, Mónica Rubio, and Joaquín J. Sopena. "Intra-osseous infiltration of adipose mesenchymal stromal cells and plasma rich in growth factors to treat acute full depth cartilage defects in a rabbit model: Serum osteoarthritis biomarkers and macroscopical assessment." Frontiers in Veterinary Science 9 (December 20, 2022). http://dx.doi.org/10.3389/fvets.2022.1057079.

Full text
Abstract:
IntroductionIntra-articular infiltration of plasma rich in growth factors (PRGF) and adipose mesenchymal stromal cells (AMSCs) are known to inhibit osteoarthritis progression. However, in severely affected patients, the treatment cannot reach the deeper layers of the articular cartilage; thus, its potential is limited. To overcome this limitation, intra-osseous infiltrations have been suggested. The purpose of this study is to assess the impact of intra-osseous infiltration therapies on serum biomarkers of osteoarthritis and to assess cartilage regeneration macroscopically.Materials and methodsA total of 80 rabbits were divided into four groups based on the intra-osseous treatment administered on the day of surgery: control, PRGF, AMSCs and a combination of PRGF + AMSCs. In addition, all groups received a single intra-articular administration of PRGF on the same day. Serum biomarker levels were measured before infiltration and 28-, 56-, and 84-days post infiltration, and macroscopical assessment was conducted at 56- and 84-days follow-up post infiltration.ResultsIn the PRGF + AMSCs group, significantly lower concentrations of hyaluronic acid and type II collagen cleavage neoepitope were recorded at all time points during the study, followed by PRGF, AMSCs and control groups. Regarding macroscopical assessment, lower scores were obtained in PRGF + AMSCs group at all study times.DiscussionThe results suggest that the combination of intra-articular PRGF with intra-osseous PRGF or AMSCs achieves better results in rabbits with acute chondral defects and that intra-osseous infiltration is a safe procedure.
APA, Harvard, Vancouver, ISO, and other styles
35

"Clinical and Radiographic Evaluation of NIPSA Versus M-MIST in Treatment of Intra-osseous Defects." Case Medical Research, November 4, 2019. http://dx.doi.org/10.31525/ct1-nct04149834.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Naineni, Rohini. "Effect of Alendronate with β – TCP Bone Substitute in Surgical Therapy of Periodontal Intra-Osseous Defects: A Randomized Controlled Clinical Trial." JOURNAL OF CLINICAL AND DIAGNOSTIC RESEARCH, 2016. http://dx.doi.org/10.7860/jcdr/2016/20965.8365.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

DEVI, RENU. "Clinical Evaluation of Insulin like Growth Factor-I and Vascular Endothelial Growth Factor with Alloplastic Bone Graft Material in the Management of Human Two Wall Intra-Osseous Defects." JOURNAL OF CLINICAL AND DIAGNOSTIC RESEARCH, 2016. http://dx.doi.org/10.7860/jcdr/2016/21333.8476.

Full text
APA, Harvard, Vancouver, ISO, and other styles
38

Thi Thanh, Pham, Nguyen Thi Mai, Nguyen Thi Ngoc Ha, Mai Thi Hien, and Nguyen Dac Tu. "Platelet-rich Plasma and Its Application in Clinical Trial." VNU Journal of Science: Medical and Pharmaceutical Sciences 37, no. 3 (September 21, 2021). http://dx.doi.org/10.25073/2588-1132/vnumps.4346.

Full text
Abstract:
Platelet-rich plasma (PRP) is an autologous physiological product that has a platelet concentration above baseline. The PRP contains a high level of important growth factors and cytokines such as PDGF, EGF, VEGF, TGF-β,... which play important roles in healing and tissue regeneration. The PRP is currently used in various medical fields, including orthopedic, plastic surgery, dermatology, dentistry, musculoskeletal and obstetrics-gynecology. To date, the results from in vitro, in vivo studies to clinical trials have proved the effectiveness of PRP in the treating of many diseases. Altogether, PRP is considered to be a very promising futuristic therapy by dint of its simple and highly potential clinical application. Keywords: Platelet-rich plasma, PRP, growth factors, applications of platelet-rich plasma. References [1] J. S. F. Moure, J. L. V. Eps, F. J. Cabrera, Z. Barbosa, G. M. D. Rosal, B. K. Weiner, et al., Platelet-rich Plasma: A Biomimetic Approach to Enhancement of Surgical Wound Healing, Journal of Surgical Research, Vol. 207, No., 2017, pp. 33-44, https://doi.org/10.1016/j.jss.2016.08.063.[2] A. Zarbock, R. K. P. Grabowska, K. Ley, Platelet-neutrophil-interactions: Linking Hemostasis and Inflammation, Blood Reviews, Vol. 21, No. 2, 2007, pp. 99-111, https://doi.org/10.1016/j.blre.2006.06.001.[3] R. E. Marx, Platelet-rich Plasma (PRP): what is PRP and what is not PRP?, Implant Dentistry, Vol. 10, No. 4, 2001, pp. 225-228, https://doi.org/10.1097/00008505-200110000-00002.[4] R. Alves, R. Grimalt, A Review of Platelet-Rich Plasma: History, Biology, Mechanism of Action, and Classification, Skin Appendage Disord, Vol. 4, No. 1, 2018, pp. 18-24, https://doi.org/10.1159/000477353.[5] P. A. Everts, J. T. Knape, G. Weibrich, J. P. Schonberger, J. Hoffmann, E. P. Overdevestet al., Platelet-rich Plasma and Platelet Gel: A Review, The Journal of ExtraCorporeal Technology, Vol. 38, No. 2, 2006, pp. 174-187.[6] A. Mishra, J. W. Jr, A. Vieira, Treatment of Tendon and Muscle Using Platelet-rich Plasma, Clinics in Sports Medicine, Vol. 28, No. 1, 2009, pp. 113-125, https://doi.org/10.1016/j.csm.2008.08.007.[7] R. Dhurat, M. Sukesh, Principles and Methods of Preparation of Platelet-rich Plasma: A Review and Author's Perspective, Journal of Cutaneous Aesthetic Surgery, Vol. 7, No. 4, 2014, pp. 189-197, https://doi.org/10.4103/0974-2077.150734.[8] B. L. Eppley, W. S. Pietrzak, M. Blanton, Platelet-rich Plasma: A Review of Biology and Applications in Plastic Surgery, Plastic and Reconstructive Surgery, Vol. 118, No. 6, 2006, pp. 147e-159e, https://doi.org/10.1097/01.prs.0000239606.92676.cf.[9] K. Mautner, G. Malanga, R. Colberg, Optimization of Ingredients, Procedures and Rehabilitation for Platelet-rich Plasma Injections for Chronic Tendinopathy, Pain Management, Vol. 1, No. 6, 2011, pp. 523-532, https://doi.org/10.2217/pmt.11.56.[10] C. Kia, J. Baldino, R. Bell, A. Ramji, C. Uyeki, A. Mazzocca, Platelet-rich Plasma: Review of Current Literature on its Use for Tendon and Ligament Pathology, Current Reviews in Musculoskeletal Medicine, Vol. 11, No. 4, 2018, pp. 566-572, https://doi.org/10.1007/s12178-018-9515-y.[11] J. Alsousou, M. Thompson, P. Hulley, A. Noble, K. Willett, The Biology of Platelet-rich Plasma and its Application in Trauma and Orthopaedic Surgery: A Review of The Literature, The Journal of Bone and Joint Surgery British volume, Vol. 91, No. 8, 2009, pp. 987-996, https://doi.org/10.1302/0301-620X.91B8.22546.[12] W. J. Berghoff, W. S. Pietrzak, R. D. Rhodes, Platelet-rich Plasma Application During Closure Following Total Knee Arthroplasty, Orthopedics, Vol. 29, No. 7, 2006, pp. 590-598, https://doi.org/10.3928/01477447-20060701-11.[13] D. R. Knighton, K. F. Ciresi, V. D. Fiegel, L. L. Austin, E. L. Butler, Classification and Treatment of Chronic Nonhealing Wounds. Successful Treatment with Autologous Platelet-derived Wound Healing Factors (PDWHF), Annals of Surgery, Vol. 204, No. 3, 1986, pp. 322-330, https://doi.org/10.1097/00000658-198609000-00011.[14] N. N. Tuan, N. T. Dung, Studying the Effects of Platelet-rich Plasma and Adipose Tissue-derived Stem cells on Chronic Wound Treatment, Journal of Disaster Medicine and Burn Injuries, Vol. 5, 2018 (in Vietnamese).[15] D. M. Dohan Ehrenfest, T. Bielecki, A. Mishra, P. Borzini, F. Inchingolo, G. Sammartino et al., in Search of A Consensus Terminology in The Field of Platelet Concentrates for Surgical Use: Platelet-rich Plasma (PRP), Platelet-rich Fibrin (PRF), Fibrin Gel Polymerization and Leukocytes, Current Pharmaceutical Biotechnology, Vol. 13, No. 7, 2012, pp. 1131-1137, https://doi.org/10.2174/138920112800624328.[16] D. H. Whitman, R. L. Berry, D. M. Green, Platelet Gel: An Autologous Alternative to Fibrin Glue with Applications in Oral and Maxillofacial Surgery, Journal of Oral Maxillofacial Surgery, Vol. 55, No. 11, 1997, pp. 1294-1299, https://doi.org/10.1016/s0278-2391(97)90187-7.[17] P. A. Mooar, M. J. Gardner, P. R. Klepchick, The Efficacy of Autologous Platelet Gel in Total Knee Arthroplasty: An Analysis of Range of Motion, Hemoblobin, and Narcotic Requirements, In: 67th Annual Meeting. American Academy of Orthopaedic Surgeons, 2000.[18] T. M. Bielecki, T. S. Gazdzik, J. Arendt, T. Szczepanski, W. Krol, T. Wielkoszynski, Antibacterial Effect of Autologous Platelet Gel Enriched with Growth Factors and Other Active Substances: An in Vitro Study, The Journal of Bone and Joint Surgery British volume, Vol. 89, No. 3, 2007, pp. 417-420, https://doi.org/10.1302/0301-620X.89B3.18491.[19] D. J. Moojen, P. A. Everts, R. M. Schure, E. P. Overdevest, A. V. Zundert, J. T. Knape et al., Antimicrobial Activity of Platelet-leukocyte Gel Against Staphylococcus Aureus, Journal of Orthopaedic Research, Vol. 26, No. 3, 2008, pp. 404-410, https://doi.org/10.1002/jor.20519.[20] D. M. D. Ehrenfest, L. Rasmusson, T. Albrektsson, Classification of Platelet Concentrates: from Pure Platelet-rich Plasma (P-PRP) to Leucocyte- and Platelet-rich Fibrin (L-PRF), Trends in Biotechnology, Vol. 27, No. 3, 2009, pp. 158-167, https://doi.org/10.1016/j.tibtech.2008.11.009.[21] R. E. Marx, E. R. Carlson, R. M. Eichstaedt, S. R. Schimmele, J. E. Strauss, K. R. Georgeff, Platelet-rich Plasma: Growth Factor Enhancement for Bone Grafts, Oral Surgery Oral Medicine Oral Pathology Oral Radiology, Vol. 85, No. 6, 1998, pp. 638-646, https://doi.org/10.1016/s1079-2104(98)90029-4.[22] R. Alissa, M. Esposito, K. Horner, R. Oliver, The Influence of Platelet-rich Plasma on The Healing of Extraction Sockets: An Explorative Randomised Clinical Trial, European Journal of Oral Implantology, Vol. 3, No. 2, 2010, pp. 121-134.[23] R. C. Mariano, W. M. D. Melo, C. C. Avelino, Comparative Radiographic Evaluation of Alveolar Bone Healing Associated with Autologous Platelet-rich Plasma After Impacted Mandibular Third Molar Surgery, Journal of Oral and Maxillofacial Surgery, Vol. 70, No. 1, 2012, pp. 19-24, https://doi.org/10.1016/j.joms.2011.03.028.[24] C. Bacci, M. Maglione, L. Favero, A. Perini, R. D. Lenarda, M. Berengo et al., Management of Dental Extraction in Patients Undergoing Anticoagulant Treatment. Results From A Large, Multicentre, Prospective, Case-control Study, Journal of Thrombosis and Haemostasis, Vol. 104, No. 5, 2010, pp. 972-975, https://doi.org/10.1160/TH10-02-0139.[25] E. Anitua, M. Sanchez, A. T. Nurden, M. Zalduendo, M. D. L. Fuente, J. Azofra et al., Reciprocal Actions of Platelet-secreted TGF-beta1 on The Production of VEGF and HGF by Human Tendon Cells, Plastic and Reconstructive Surgery, Vol. 119, No. 3, 2007, pp. 950-959, https://doi.org/10.1097/01.prs.0000255543.43695.1d.[26] B. O'Connell, N. M. Wragg, S. L. Wilson, The Use of PRP Injections in The Management of Knee Osteoarthritis, Cell and Tissue Research, Vol. 376, No. 2, 2019, pp. 143-152, https://doi.org/10.1007/s00441-019-02996-x.[27] M. A. S. Mendia, J. F. V. Cavazos, H. G. M. Rodriguez, Platelet-rich Plasma in Knee Osteoarthritis Treatment, Cirugia y Cirujanos, Vol. 83, No. 4, 2015, pp. 352-358, https://doi.org/10.1016/j.circir.2014.06.001.[28] H. H. E. Hadmed, R. F. Castillo, Cosmeceuticals: Peptides, Proteins, and Growth Factors, Journal of Cosmetic Dermatology, Vol. 15, No. 4, 2016, pp. 514-519, https://doi.org/10.1111/jocd.12229.[29] D. H. Kim, Y. J. Je, C. D. Kim, Y. H. Lee, Y. J. Seo, J. H. Lee et al., Can Platelet-rich Plasma Be Used for Skin Rejuvenation? Evaluation of Effects of Platelet-rich Plasma on Human Dermal Fibroblast, Annals of Dermatology, Vol. 23, No. 4, 2011, pp. 424-431, https://doi.org/10.5021/ad.2011.23.4.424.[30] N. Cameli, M. Mariano, I. Cordone, E. Abril, S. Masi, M. L. Foddai, Autologous Pure Platelet-Rich Plasma Dermal Injections for Facial Skin Rejuvenation: Clinical, Instrumental, and Flow Cytometry Assessment, Dermatologic Surgery, Vol. 43, No. 6, 2017, pp. 826-835, https://doi.org/10.1097/DSS.0000000000001083.[31] O. K. Abuaf, H. Yildiz, H. Baloglu, M. E. Bilgili, H. A. Simsek, B. Dogan, Histologic Evidence of New Collagen Formulation Using Platelet Rich Plasma in Skin Rejuvenation: A Prospective Controlled Clinical Study, Annals of Dermatology, Vol. 28, No. 6, 2016, pp. 718-724, https://doi.org/10.5021/ad.2016.28.6.718.[32] N. Y. Elnehrawy, Z. A. Ibrahim, A. M. Eltoukhy, H. M. Nagy, Assessment of the Efficacy and Safety of Single Platelet-rich Plasma Injection on Different Types and Grades of Facial Wrinkles, Journal of Cosmetic Dermatology, Vol. 16, No. 1, 2017, pp. 103-111, https://doi.org/10.1111/jocd.12258.[33] B. Hersant, M. S. Mezi, J. Niddam, S. L. Padula, W. Noel, K. Ezzedine et al., Efficacy of Autologous Platelet-rich Plasma Combined with Hyaluronic Acid on Skin Facial Rejuvenation: A Prospective Study, Journal of the American Academy of Dermatology, Vol. 77, No. 3, 2017, pp. 584-586, https://doi.org/10.1016/j.jaad.2017.05.022.[34] Z. A. Ibrahim, R. A. E. Tatawy, M. A. E. Samongy, D. A. Ali, Comparison Between The Efficacy and Safety of Platelet-rich Plasma vs. Microdermabrasion in the Treatment of Striae Distensae: Clinical and Histopathological Study, Journal of Cosmetic Dermatology, Vol. 14, No. 4, 2015, pp. 336-346, https://doi.org/10.1111/jocd.12160.[35] A. Tuknayat, G. P. Thami, M. Bhalla, J. K. Sandhu, Autologous Intralesional Platelet-rich Plasma Improves Melasma, Dermatologic Therapy, Vol. 34, No. 2, 2021, pp. e14881, https://doi.org/10.1111/dth.14881.[36] V. Cervelli, F. Nicoli, D. Spallone, S. Verardi, R. Sorge, M. Nicoli et al., Treatment of Traumatic Scars Using Fat Grafts Mixed with Platelet-rich Plasma, and Resurfacing of Skin with The 1540 nm Nonablative Laser, Clinical and Experimental Dermatology, Vol. 37, No. 1, 2012, pp. 55-61, https://doi.org/10.1111/j.13652230.2011.04199.x.[37] A. C. Nita, O. A. Orzan, M. Filipescu, D. Jianu, Fat Graft, Laser CO(2) and Platelet-rich Plasma Synergy in Scars Treatment, Journal of Medicine and Life, Vol. 6, No. 4, 2013, pp. 430-433.[38] R. James, R. Chetry, V. Subramanian, A. Ashtekar, N. Srikruthi, S. Ramachandran et al., Efficacy of Activated 3X Platelet-Rich Plasma in the Treatment of Androgenic Alopecia, Journal of Stem Cells, Vol. 11, No. 4, 2016, pp. 191-199.[39] R. Alves, R. Grimalt, Randomized Placebo-Controlled, Double-Blind, Half-Head Study to Assess the Efficacy of Platelet-rich Plasma on the Treatment of Androgenetic Alopecia, Dermatologic Surgery, Vol. 42, No. 4, 2016, pp. 491-497, https://doi.org/10.1097/DSS.0000000000000665.[40] C. E. Finch, The Menopause and Aging, A Comparative Perspective, The Journal of Steroid Biochemistry and Molecular Biology, Vol. 142, No., 2014, pp. 132-141, https://doi.org/10.1016/j.jsbmb.2013.03.010.[41] Y. Chang, J. Li, Y. Chen, L. Wei, X. Yang, Y. Shi et al., Autologous Platelet-rich Plasma Promotes Endometrial Growth and Improves Pregnancy Outcome During In Vitro Fertilization, International Journal of Clinical and Experimental Medicine, Vol. 8, No. 1, 2015, pp. 1286-1290.[42] S. Zadehmodarres, S. Salehpour, N. Saharkhiz, L. Nazari, Treatment of Thin Endometrium with Autologous Platelet-rich Plasma: A Pilot Study, JBRA Assisted Reproduction, Vol. 21, No. 1, 2017, pp. 54-56, https://doi.org/10.5935/1518-0557.20170013.[43] Y. Chang, J. Li, L. N. Wei, J. Pang, J. Chen, X. Liang, Autologous Platelet-rich Plasma Infusion Improves Clinical Pregnancy Rate in Frozen Embryo Transfer Cycles for Women with Thin Endometrium, Medicine (Baltimore), Vol. 98, No. 3, 2019, https://doi.org/10.1097/MD.0000000000014062.[44] S. R. Tandulwadkar, M. V. Naralkar, A. D. Surana, M. Selvakarthick, A. H. Kharat, Autologous Intrauterine Platelet-Rich Plasma Instillation for Suboptimal Endometrium in Frozen Embryo Transfer Cycles: A Pilot Study, Journal of Human Reproductive Sciences, Vol. 10, No. 3, 2017, pp. 208-212, https://doi.org/10.4103/jhrs.JHRS_28_17.[45] A. Molina, J. Sanchez, W. Sanchez, V. Vielma, Platelet-rich Plasma As An Adjuvant in the Endometrial Preparation of Patients with Refractory Endometrium, JBRA Assisted Reproduction, Vol. 22, No. 1, 2018, pp. 42-48, https://doi.org/10.5935/1518-0557.20180009.[46] K. Pantos, N. Nitsos, G. Kokkali, T. Vaxevanoglou, C. Markomichali, A. Pantou, Ovarian Rejuvenation and Folliculogenesis Reactivation in Peri-menopausal Women After Autologous Platelet-rich Plasma Treatment, in Proceedings of the 32nd Annual Meeting of ESHRE, Helsinki, Finland, 2016.[47] K. Pantos, M. Simopoulou, A. Pantou, A. Rapani, P. Tsioulou, N. Nitsos, et al., A Case Series on Natural Conceptions Resulting in Ongoing Pregnancies in Menopausal and Prematurely Menopausal Women Following Platelet-rich Plasma Treatment, Cell Transplant, Vol. 28, No. 9-10, 2019, pp. 1333-1340, https://doi.org/10.1177/0963689719859539.[48] J. Fanning, L. Murrain, R. Flora, T. Hutchings, J. M. Johnson, B. W. Fenton, Phase I/II Prospective Trial of Autologous Platelet Tissue Graft in Gynecologic Surgery, Journal of Minimally Invasive Gynecology, Vol. 14, No. 5, 2007, pp. 633-637, https://doi.org/10.1016/j.jmig.2007.05.014.[49] X. Hua, Y. Zeng, R. Zhang, H. Wang, J. Diao, P. Zhang, Using Platelet-rich Plasma for the Treatment of Symptomatic Cervical Ectopy, International Journal of Gynaecology & Obstetrics, Vol. 119, No. 1, 2012, pp. 26-29, https://doi.org/10.1016/j.ijgo.2012.05.029.[50] F. B. Willison, N. R. Pour, B. Mohamadi, N. Willison, M. Rock, I. W. Holten et al., Use of Platelet-rich Plasma for Vulvovaginal Autoimmune Conditions Like Lichen Sclerosus, Plastic and Reconstructive Surgery Global Open, Vol. 4, No. 11, 2016, pp. e1124, https://doi.org/10.1097/GOX.0000000000001124.[51] M. Morelli, M. L. Rocca, R. Venturella, A. D. Cello, S. D. Negro, M. Condorelli et al., Adjuvant Use of Platelet Gel for Wound Breakdown Prevention in Advanced Vulvar Cancer Surgery: A Retrospective Study, International Journal of Gynecological Cancer, Vol. 23, No. 8, 2013, pp. 1490-1494, https://doi.org/10.1097/IGC.0b013e3182a50517.[52] B. B. Adler, E. Hanzal, E. Pablik, H. Koelbl, K. Bodner, Management of Vesicovaginal Fistulas (VVFs) in Women Following Benign Gynaecologic Surgery: A Systematic Review and Meta-analysis, PLoS One, Vol. 12, No. 2, 2017, pp. e0171554, https://doi.org/10.1371/journal.pone.0171554.[53] A. S. Plachokova, J. V. D. Dolder, P. J. Stoelinga, J. A. Jansen, The Bone Regenerative Effect of Platelet-rich Plasma in Combination with An Osteoconductive Material in Rat Cranial Defects, Clinical Oral Implants Research, Vol. 17, No. 3, 2006, pp. 305-311, https://doi.org/10.1111/j.1600-0501.2005.01208.x.[54] A. S. Plachokova, J. V. D. Dolder, P. J. Stoelinga, J. A. Jansen, Early Effect of Platelet-rich Plasma on Bone Healing in Combination with An Osteoconductive Material in Rat Cranial Defects, Clinical Oral Implants Research, Vol. 18, No. 2, 2007, pp. 244-251, https://doi.org/10.1111/j.1600-0501.2006.01327.x.[55] B. H. Choi, C. J. Im, J. Y. Huh, J. J. Suh, S. H. Lee, Effect of Platelet-rich Plasma on Bone Regeneration in Autogenous Bone Graft, International Journal of Oral Maxillofacial Surgery, Vol. 33, No. 1, 2004, pp. 56-59, https://doi.org/10.1054/ijom.2003.0466.[56] B. L. Walters, D. A. Porter, S. J. Hobart, B. B. Bedford, D. E. Hogan, M. M. McHugh et al., Effect of Intraoperative Platelet-Rich Plasma Treatment on Postoperative Donor Site Knee Pain in Patellar Tendon Autograft Anterior Cruciate Ligament Reconstruction: A Double-Blind Randomized Controlled Trial, The American Journal of Sports Medicine, Vol. 46, No. 8, 2018, pp. 1827-1835, https://doi.org/10.1177/0363546518769295.[57] G. E. Niazi, M. S. Hassan, D. M. Elfawy, Ultrasound-guided Injection of Platelet-rich Plasma (PRP) in Rotator Cuff Tendinopathy: Effect on Patients’ Symptoms and Supraspinatus Tendon Thickness, Egyptian Journal of Radiology and Nuclear Medicine, Vol. 51, No. 1, 2020, https://doi.org/ 10.1186/s43055-020-00221-2.[58] L. D. Sante, C. Villani, V. Santilli, M. Valeo, E. Bologna, L. Imparato et al., Intra-articular Hyaluronic Acid vs Platelet-rich Plasma in the Treatment of Hip Osteoarthritis, Medical Ultrasonography, Vol. 18, No. 4, 2016, pp. 463-468, https://doi.org/10.11152/mu-874.[59] J. Wiltfang, K. A. Schlegel, S. S. Mosgau, E. Nkenke, R. Zimmermann, P. Kessler, Sinus Floor Augmentation with Beta-tricalciumphosphate (beta-TCP): Does Platelet-rich Plasma Promote its Osseous Integration and Degradation?, Clinical Oral Implants Research, Vol. 14, No. 2, 2003, pp. 213-218, https://doi.org/10.1034/j.16000501.2003.140212.x.[60] J. H. Wang, X. Nirmala, Application of Tendon Stem/Progenitor Cells and Platelet-Rich Plasma to Treat Tendon Injuries, Operative Techniques in Orthopaedics, Vol. 26, No. 2, 2016, pp. 68-72, https://doi.org/10.1053/j.oto.2015.12.008.[61] H. Toumi, T. M. Best, The Inflammatory Response: Friend or Enemy for Muscle Injury?, British Journal of Sports Medicine, Vol. 37, No. 4, 2003, pp. 284-286, https://doi.org/10.1136/bjsm.37.4.284.[62] C. Smith, M. J. Kruger, R. M. Smith, K. H. Myburgh, The Inflammatory Response to Skeletal Muscle Injury: Illuminating Complexities, Sports Medicine, Vol. 38, No. 11, 2008, pp. 947-969, https://doi.org/10.2165/00007256-200838110-00005.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography