SciDoc Publishers | Open Access | Science Journals | Media Partners


International Journal of Dentistry and Oral Science (IJDOS)  /  IJDOS-2377-8075-08-5056

Regenerative Capacity Of Resolvin D2 On Stem Cells Of Apical Papilla


Manish Ranjan1*, Rukhsaar Akbar Gulzar2

1 Associate Professor, Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
2 Post Graduate Student, Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.


*Corresponding Author

Manish Ranjan,
Associate Professor, Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
E-mail: manish@gmail.com

Received: April 28, 2021; Accepted: May 28, 2021; Published: May 30, 2021

Citation: Manish Ranjan, Rukhsaar Akbar Gulzar. Regenerative Capacity Of Resolvin D2 On Stem Cells Of Apical Papilla. Int J Dentistry Oral Sci. 2021;08(05):2637-2642. doi: dx.doi.org/10.19070/2377-8075-21000516

Copyright: Manish Ranjan©2021. 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.



Abstract

Introduction: Regenerative endodontics is based on the triad of tissue engineering which has three components; stem cells, biomimetic scaffold and biomimetic growth factors which are introduced in to the canal space so as to induce pulp tissue regeneration and root maturation. Resolvins are lipid mediators that are released during the resolution phase of inflammation and regulate tissue repair. Thus the present study was conducted with the aim to evaluate whether resolvin D2 (RvD2) is capable of inducing hard tissue formation by its action on stem cells of apical papilla and compare it to concentrated growth factor (CGF).
Materials and Methods: The root apical papilla tissues were carefully isolated from the root apex. Enzymatic separation was used for the cells of the primary apical papilla. The cells were subjected to the three groups namely RvD2, CGF and a combination of CGF and RvD2. Mineralized nodule formation was analyzed by alizarin red staining and dentin matrix protein 1 secretion was analyzed using ELISA test.
Result: The combination of RvD2 and CGF had the highest values for mineralized nodule formation and DMP1 secretion followed by CGF. These were statistically more significant than the control group. However the results for RvD2 alone did not exceed that of CGF.
Conclusion: Although RvD2 possesses regenerative potential and is capable of inducing stem cells of apical papilla for formation of mineralized hard tissue, its potential by itself does not surpass that of CGF. However the combination could be a promising new strategy in the management of immature necrotic permanent teeth.



1.Keywords
2.Introduction
3.Materials and Methods
4.Results
5.Discussion
6.Conclusion
7.References


Keywords

Concentrated Growth Factor; Lipid Mediators; Regeneration; Resolvin D2; Stem Cells.


Introduction

Traditionally, immature permanent teeth with necrotic pulp were treated by apexification using calcium hydroxide which helped in generating a hard tissue barrier or by placing an apical plug using MTA, biodentine or calcium hydroxide followed by root canal filling [1, 2]. However, apexification does not restore the vitality of the tooth and neither does it help in maturation of the root by thickening of the walls or closure of the apex. It was in 2001 that Iwaya et al introduced the term revascularisation [3]. Regenerative endodontics is based on the triad of tissue engineering which has three components; stem cells, biomimetic scaffold and biomimetic growth factors which are introduced into the canal space so as to induce pulp tissue regeneration and root maturation [4].

Stem cells from the apical papilla (SCAP) are related to developing roots and have been shown to be a promising tool with the ability for self-renewal, multilineage differentiation and regenerative potential [5]. They play a pivotal role in regenerative endodontics which is currently based on the cell homing approach [6]. Growth factors are proteins that bind to the receptors present on the target cells and induce the migration, proliferation and differentiation of stem cells [7]. Scaffold is a three dimensional matrix that helps in cell adhesion, interaction and deposition of the extracellular matrix. These scaffolds can either be biological such as Platelet derived factors or artificially synthesized [8].

Resolvins are lipid mediators that are released during the resolution phase of inflammation and regulate tissue repair. Resolvin D2 (RvD2) has been shown to enhance post ischaemic revascularization while resolving inflammation by promoting apoptosis of polymorphonuclear neutrophils (PMSs), controlling bacterial sepsis as well as promoting arteriogenesis [9]. It has also been shown to have defensive properties against P. gingivalis which causes periodontal bone loss. Studies have shown that RvD2 is capable of regulating the RANKL/OPG ratio [10]. RvD2 is also known to be capable of inhibiting transient receptor potential channels present in sensory neurons and is thus capable of reducing postoperative pain [11]. Siddiqui et al in a study in 2019 showed that RvD2 induced resolution of periapical inflammation and promoted periapical healing in rats. Further, calcified canals apices were observed in RvD2 treated canals with apex closure [12]. However in their study the regenerative capacity of RvD2 was observed against a placebo control.

Previously our team has a rich experience in working on various research projects across multiple disciplines [13-27]. Now the growing trend in this area motivated us to pursue this project.

Thus the present study was conducted with the aim to evaluate whether resolvin D2 is capable of inducing hard tissue formation by its action on stem cells of apical papilla and to compare it to the regenerative potential of concentrated growth factor (CGF) and to evaluate whether the combination of concentrated growth factor and RvD2 has any synergistic effect.


Materials and Methods

Chemicals

The test drug RvD2 was purchased from Santa Cruz BioTech and CGF was obtained commercially. All media, broth and other chemicals required for the project were obtained from Himedia Laboratories (Anna Nagar, chennai).

Stem Cell Separation

The root apical papilla tissues were carefully isolated from the root apex. Enzymatic separation was used for the cells of the primary apical papilla. SCAPs were seeded on 100mm plates at 20,000 cells/cm2. The medium was changed to a growth medium after analyzing that it reached the 90% confluence. After six days, the cells were washed five times with phosphate-buffered saline (PBS) and cultured in a serum-free medium for 24 hours. The media was collected after centrifugation at 1,000rpm for 10 mins to remove the cellular debris and filtered through a 0.22µm filter. The samples were concentrated, air-dried, re-dissolved in triethylammonium bicarbonate, and reduced with dithiothreitol at 55°C for one hour. Next, iodoacetamide was added to the samples, which were maintained for one hour at room temperature in the dark. The protein concentration was determined.

Cell Culturing

The separated cells were cultured in Dulbecco’s modified Eagle’s medium (Himedia Laboratories), supplemented with 10% fetal bovine serum and 100 U/ml penicillin-G and streptomycin in a humidified atmosphere of 5% CO2 at 37 °C. The characterization of SCAPs was analyzed for other assays.

Grouping

The SCAPs were treated with different drugs.

Group 1: Cells treated with concentrated growth factor (1µg/ml).

Group 2: Cells treated with Resolvin D2 100nM (1µg/ml).

Group 3: Cells treated with the combination of concentrated growth factor and resolvin D2 100 nM.

Group 4: Negative control group (NC).

For alizarin red staining to check the mineralized nodule formation and to quantify the DMP-1 levels the cells were incubated with inducing medium for three weeks.

Alizarin Red Staining (ARS)

The culture medium was removed from each well and gently washed the cells three times with 1xPBS. The cells were fixed in 4% formaldehyde for 15 minutes at room temperature. The fixative was removed and the cells were washed three times with distilled water. The distilled water was removed completely and 1mL of 40 mM Alizarin red stain was added per well. It was incubated at room temperature for 20-30 min with gentle shaking followed by removal of the dye and washing of the cells five times with distilled water. The test plates were stored at -20°C prior to dye extraction. 800µL of 10% acetic acid was added to each well of a 6-well plate and incubated at room temperature for 30 minutes with shaking (200µL per well for a 24-well plate). The cells were collected using a cell scraper and transferred in 10% acetic acid to a 1.5-mL microcentrifuge tube and vortexed for one min. The samples were sealed with parafilm heated at exactly 85°C for 10 minutes and then incubated on ice for five minutes. The centrifugation was done at 20,000g for 15 minutes. After centrifugation, 500µL of the supernatant was transferred to a new tube and 200µL of 10% ammonium hydroxide was added to neutralize the acid. 150µL of the sample was added per well in a 96-well plate and the absorbance was read at 405 nm with a plate reader.

Quantification Of Dentin Matrix Protein -1 (DMP 1)

The adherent cells were detached with trypsin and then collected by centrifugation and washed three times in PBS. Cells were resuspended in PBS and subjected to ultrasonication for three times. Alternatively, freezing and thawing the cells was done and repeated for three times. Centrifugation was done at 1000×g (or 3000rpm) for 15 minutes at 2-8°C to remove cellular debris. The cell were coated into the wells at a density of 2x104 and then 100µL of standards or samples was added to the appropriate well in the antibody pre-coated Microtiter Plate. Next 100µL of PBS (pH 7.0-7.2) was added in the blank control well and 100µl of primary Ab (DMP-1) 1:1000 dilution was added to each well and after 1-1.5 hours the primary antibody was removed, and the wells were washed 3 times with PBS. Then 100µL of secondary antibody (anti-rabbit horseradish protein) was added to each well and incubated for 1-1.5 hours. The secondary antibody was discarded and washed three times with PBS. After washing for 5 times, 50 µL of tetramethylbenzidine (TMB) substrate was added to each well including blank control well, subsequently. It was covered and incubated for 10-15 minutes at 20-25°C followed by addition of 50µL of 1M sulphuric acid Solution to each well including blank control well. The Optical Density (O.D.) at 450nm was read using a microplate reader immediately.

Statistical Analysis

Results were expressed as mean ± standard deviation (SD). Statistical significance was determined by one-way analysis of variance (ANOVA) and post hoc least-significant difference test. Intercomparison groups analysis was done using the SPSS software version 22.0.


Results

It was observed that RvD2 induced the stem cells for the deposition of mineralized nodules (Table 1 and fig 1 and 2) (0.34 ± 0.027) and DMP-1 secretion (Table 2 and Fig 2) (8.267 ± 1.305). This was significantly higher than the negative control group. However, the highest values were observed for the group of cells treated by the combination of CGF and RvD2 followed by the group of cells treated only by CGF. These values were significantly higher when compared to the RvD2 treated group. However, there was no significant difference seen between the CGF group and the group of cells treated by the combination of two.



Table 1. Quantification of Alizarin red staining expressed as mean and standard deviation.



Table 2. Quantification of DMP1 secretion by SCAP expressed as mean and standard deviation.



Figure 1. ARS quantification of SCAPs cells in control and experimental groups. The results expressed as Mean ± SEM (n = 3). ***p<0.001 statistically significant as compared with NC. RvD2 is statistically significant (ap<0.01) as compared with the CGF treated group. CGF+RvD2 statistically significant (#p<0.05) as compared with RvD2.



Figure 2. DMP-1 quantification of SCAPs cells in control and experimental groups. The results expressed as Mean ± SEM (n = 3). *P<0.05;**P<0.01; ***P<0.001 statistically significant as compared with NC. RvD2 is statistically significant (ap<0.05) as compared with the CGF treated group. CGF+RvD2 statistically significant (#P<0.05) as compared with RvD2.



Figure 3. Alizarin red staining of SCAP cells.


Discussion

Our institution is passionate about high quality evidence based research and has excelled in various fields [17, 28-37].

In the present study, the regenerative capacity of RvD2 was tested by identifying the ability to form mineralized hard tissue. This was demonstrated by alizarin red staining and DMP1 secretion by stem cells of apical papilla.

Dentin matrix protein 1 is involved in maturation of odontoblasts and osteoblasts, biomineralization of bone and dentin as well as hydroxyapatite formation [38, 39]. DMP1 is a key protein that induces odontogenesis. Over expression of DMP1 by pluripotent cells acts as a signal for differentiation of cells [40]. Hence DMP1 expression by SCAP was analysed. The concentration of RvD2 was set at 100nM based on the results by siddiqui et al. The results of the present study are in accordance to the study conducted by siddique et al where it was demonstrated that RvD2 increased expression of DMP1 in dental pulp stem cells. However in the present study, CGF showed better results than RvD2 and the two of them had a synergistic effect. Similarly observations were made with respect to mineralized tissue formation [12]. This could be due to the diversity of growth factors released by CGF. CGF has a complex three?dimensional structure with a high density of platelets and nucleated cells and factors such PDGF?BB, IGF?1, Transforming growth factor TGF?ß1, fibroblast growth factor bFGF and vascular endothelial growth factor VEGF [41].

Derived from omega 3 fatty acids namely, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) as well as docosapentaenoic acid (DPA); resolvins are specialised pro resolving mediators which restore normal cellular function after tissue injury [42]. It was from the self resolving exudates of murine during the resolution phase of acute inflammation, that RvD2 was first isolated [43]. The biosynthesis involves 17-lipoxygenation of DHA to 17S-hydroperoxy-DHA (17S-HpDHA). This is then further transformed to a 7(8) epoxide-containing intermediate in leukocytes via 5-lipoxygenase (LOX) enzymatically, followed by enzymatic hydrolysis to form RvD2. RvD2 can be endogenously found in human serum, plasma [44], adipose tissue [45], placenta [46], breast milk [47], sepsis patients [48] and lung [49].

Resolution at cellular level involves cessation of PMN entry in to the tissue and elevated phagocytosis of apoptotic PMN by macrophages. In human macrophages, RvD2 stimulates phagocytosis and efferocytosis in a DRV2-dependent manner. Resolvin acts by binding to a specific G protein coupled receptor GPR18/DRV2 which activates the cyclic AMP-PKA pathway and phosphorylation of STAT3 and increases phagocytosis mediated bacterial clearance [50].

Mizraji et al found that RvD2 prevented alveolar bone loss in murine periodontitis and prevented destructive immunity. It prevented osteoblast-mediated and T-cell-mediated signaling of osteoclast formation by RANKL leading to alveolar bone loss [10]. Proresolving mediators have shown promising results in the healing of apical periodontitis with reduction in the size of the periapical lesion and recalcification of bone [12, 51]. Resolvins can be administered in active inflammatory lesions without any deleterious effects, the bacterial load is reduced and there is no increase in disease activity [52].

One of the possible reasons for the regenerative potential of RvD2 is because of sustained vitality of migrated cells that could be a direct effect of reduction in inflammation, control of bacterial sepsis and stimulation of angiogenesis. The present study leaves a future scope for experimenting with higher concentrations of RvD2 to analyse if there is any increase in the regenerative potential and to determine weather RvD2 could be incorporated in to a scaffold which would induce vascularized tissue formation in immature necrotic permanent teeth.

In 2019, shi et al customised a polycaprolactone PCL graft with aspirin triggered RvD1 by electrospinning for vascular regeneration [53]. A similar concept can be adopted for generating synthetic scaffolds for regenerative endodontics that incorporate RvD2.


Conclusion

Although resolvin D2 possesses regenerative potential and is capable of inducing stem cells of apical papilla for formation of mineralized hard tissue, its potential by itself does not surpass that of CGF. However, owing to its good antimicrobial effect and anti-inflammatory properties, RvD2 can be used in combination with CGF or as a component of a synthetic scaffold in regenerative endodontics to enhance their bioactivity. This combination could be a promising new strategy in the management of immature necrotic permanent teeth if it translates from preclinical studies in to successful clinical trials in future.


References

  1. Rafter M. Apexification: a review. Dent Traumatol. 2005 Feb;21(1):1-8. Pubmed PMID: 15660748.
  2. Frank AL. Therapy for the divergent pulpless tooth by continued apical formation. J Am Dent Assoc. 1966 Jan;72(1):87-93. Pubmed PMID: 5215726.
  3. Iwaya SI, Ikawa M, Kubota M. Revascularization of an immature permanent tooth with apical periodontitis and sinus tract. Dent Traumatol. 2001 Aug;17(4):185-7. Pubmed PMID: 11585146.
  4. Nakashima M, Akamine A. The application of tissue engineering to regeneration of pulp and dentin in endodontics. J Endod. 2005 Oct;31(10):711- 8. Pubmed PMID: 16186748.
  5. Nada OA, El Backly RM. Stem Cells From the Apical Papilla (SCAP) as a Tool for Endogenous Tissue Regeneration. Front Bioeng Biotechnol. 2018 Jul 24;6:103. Pubmed PMID: 30087893.
  6. Kim SG, Malek M, Sigurdsson A, Lin LM, Kahler B. Regenerative endodontics: a comprehensive review. Int Endod J. 2018 Dec;51(12):1367-1388. Pubmed PMID: 29777616.
  7. Sharmin F, McDermott CC, Khan YM. Regenerative engineering: role of scaffolds, cells, and growth factors. Injectable hydrogels for regenerative engineering. 2016:1-32.
  8. Gathani KM, Raghavendra SS. Scaffolds in regenerative endodontics: A review. Dent Res J (Isfahan). 2016 Sep;13(5):379-386. Pubmed PMID: 27857762.
  9. Zhang MJ, Sansbury BE, Hellmann J, Baker JF, Guo L, Parmer CM, et al. Resolvin D2 Enhances Postischemic Revascularization While Resolving Inflammation. Circulation. 2016 Aug 30;134(9):666-680. Pubmed PMID: 27507404.
  10. Mizraji G, Heyman O, Van Dyke TE, Wilensky A. Resolvin D2 Restrains Th1 Immunity and Prevents Alveolar Bone Loss in Murine Periodontitis. Front Immunol. 2018 Apr 25;9:785. Pubmed PMID: 29922275.
  11. Park CK, Xu ZZ, Liu T, Lü N, Serhan CN, Ji RR. Resolvin D2 is a potent endogenous inhibitor for transient receptor potential subtype V1/A1, inflammatory pain, and spinal cord synaptic plasticity in mice: distinct roles of resolvin D1, D2, and E1. J Neurosci. 2011 Dec 14;31(50):18433-8. Pubmed PMID: 22171045.
  12. Siddiqui YD, Omori K, Ito T, Yamashiro K, Nakamura S, Okamoto K, et al. Resolvin D2 Induces Resolution of Periapical Inflammation and Promotes Healing of Periapical Lesions in Rat Periapical Periodontitis. Front Immunol. 2019 Feb 26;10:307. Pubmed PMID: 30863409.
  13. Govindaraju L, Gurunathan D. Effectiveness of Chewable Tooth Brush in Children-A Prospective Clinical Study. J Clin Diagn Res. 2017 Mar;11(3):ZC31-ZC34. Pubmed PMID: 28511505.
  14. Christabel A, Anantanarayanan P, Subash P, Soh CL, Ramanathan M, Muthusekhar MR, et al. Comparison of pterygomaxillary dysjunction with tuberosity separation in isolated Le Fort I osteotomies: a prospective, multi-centre, triple-blind, randomized controlled trial. Int J Oral Maxillofac Surg. 2016 Feb;45(2):180-5. Pubmed PMID: 26338075.
  15. Soh CL, Narayanan V. Quality of life assessment in patients with dentofacial deformity undergoing orthognathic surgery--a systematic review. Int J Oral Maxillofac Surg. 2013 Aug;42(8):974-80. Pubmed PMID: 23702370.
  16. Mehta M, Deeksha, Tewari D, Gupta G, Awasthi R, Singh H, et al. Oligonucleotide therapy: An emerging focus area for drug delivery in chronic inflammatory respiratory diseases. Chem Biol Interact. 2019 Aug 1;308:206- 215. Pubmed PMID: 31136735.
  17. Ezhilarasan D, Apoorva VS, Ashok Vardhan N. Syzygium cumini extract induced reactive oxygen species-mediated apoptosis in human oral squamous carcinoma cells. J Oral Pathol Med. 2019 Feb;48(2):115-121. Pubmed PMID: 30451321.
  18. Campeau PM, Kasperaviciute D, Lu JT, Burrage LC, Kim C, Hori M, et al. The genetic basis of DOORS syndrome: an exome-sequencing study. Lancet Neurol. 2014 Jan;13(1):44-58. Pubmed PMID: 24291220.
  19. Kumar S, Sneha S. Knowledge and awareness regarding antibiotic prophylaxis for infective endocarditis among undergraduate dental students. Asian Journal of Pharmaceutical and Clinical Research. 2016;154.
  20. Christabel SL, Gurunathan D. Prevalence of type of frenal attachment and morphology of frenum in children, Chennai, Tamil Nadu. World J Dent. 2015 Oct;6(4):203-7.
  21. Kumar S, Rahman RE. Knowledge, awareness, and practices regarding biomedical waste management among undergraduate dental students. Asian Journal of Pharmaceutical and Clinical Research. 2017;10(8):341.
  22. Sridharan G, Ramani P, Patankar S. Serum metabolomics in oral leukoplakia and oral squamous cell carcinoma. J Cancer Res Ther. 2017 Jul- Sep;13(3):556-561. Pubmed PMID: 28862226.
  23. Ramesh A, Varghese SS, Doraiswamy JN, Malaiappan S. Herbs as an antioxidant arsenal for periodontal diseases. J Intercult Ethnopharmacol. 2016 Jan 27;5(1):92-6. Pubmed PMID: 27069730.
  24. Thamaraiselvan M, Elavarasu S, Thangakumaran S, Gadagi JS, Arthie T. Comparative clinical evaluation of coronally advanced flap with or without platelet rich fibrin membrane in the treatment of isolated gingival recession. J Indian Soc Periodontol. 2015 Jan-Feb;19(1):66-71. Pubmed PMID: 25810596.
  25. Thangaraj SV, Shyamsundar V, Krishnamurthy A, Ramani P, Ganesan K, Muthuswami M, et al. Molecular Portrait of Oral Tongue Squamous Cell Carcinoma Shown by Integrative Meta-Analysis of Expression Profiles with Validations. PLoS One. 2016 Jun 9;11(6):e0156582. Pubmed PMID: 27280700.
  26. Ponnulakshmi R, Shyamaladevi B, Vijayalakshmi P, Selvaraj J. In silico and in vivo analysis to identify the antidiabetic activity of beta sitosterol in adipose tissue of high fat diet and sucrose induced type-2 diabetic experimental rats. Toxicol Mech Methods. 2019 May;29(4):276-290. Pubmed PMID: 30461321.
  27. Mahesh R, Masitah M. Fluoride, fluoridated toothpaste efficacy and its safety in children. International Journal of Pharmaceutical Research. 2018;10(4):109-14.
  28. Vijayashree Priyadharsini J. In silico validation of the non-antibiotic drugs acetaminophen and ibuprofen as antibacterial agents against red complex pathogens. J Periodontol. 2019 Dec;90(12):1441-1448. Pubmed PMID: 31257588.
  29. J PC, Marimuthu T, C K, Devadoss P, Kumar SM. Prevalence and measurement of anterior loop of the mandibular canal using CBCT: A cross sectional study. Clin Implant Dent Relat Res. 2018 Aug;20(4):531-534. Pubmed PMID: 29624863.
  30. Ramesh A, Varghese S, Jayakumar ND, Malaiappan S. Comparative estimation of sulfiredoxin levels between chronic periodontitis and healthy patients - A case-control study. J Periodontol. 2018 Oct;89(10):1241-1248. Pubmed PMID: 30044495.
  31. Ramadurai N, Gurunathan D, Samuel AV, Subramanian E, Rodrigues SJL. Effectiveness of 2% Articaine as an anesthetic agent in children: randomized controlled trial. Clin Oral Investig. 2019 Sep;23(9):3543-3550. Pubmed PMID: 30552590.
  32. Sridharan G, Ramani P, Patankar S, Vijayaraghavan R. Evaluation of salivary metabolomics in oral leukoplakia and oral squamous cell carcinoma. J Oral Pathol Med. 2019 Apr;48(4):299-306. Pubmed PMID: 30714209.
  33. Mathew MG, Samuel SR, Soni AJ, Roopa KB. Evaluation of adhesion of Streptococcus mutans, plaque accumulation on zirconia and stainless steel crowns, and surrounding gingival inflammation in primary molars: randomized controlled trial. Clin Oral Investig. 2020 Sep;24(9):3275-3280. Pubmed PMID: 31955271.
  34. Samuel SR. Can 5-year-olds sensibly self-report the impact of developmental enamel defects on their quality of life? Int J Paediatr Dent. 2021 Mar;31(2):285-286. Pubmed PMID: 32416620.
  35. R H, Ramani P, Ramanathan A, R JM, S G, Ramasubramanian A, et al. CYP2 C9 polymorphism among patients with oral squamous cell carcinoma and its role in altering the metabolism of benzo[a]pyrene. Oral Surg Oral Med Oral Pathol Oral Radiol. 2020 Sep;130(3):306-312. Pubmed PMID: 32773350.
  36. Chandrasekar R, Chandrasekhar S, Sundari KKS, Ravi P. Development and validation of a formula for objective assessment of cervical vertebral bone age. Prog Orthod. 2020 Oct 12;21(1):38. Pubmed PMID: 33043408.
  37. Vijayashree Priyadharsini J, Smiline Girija AS, Paramasivam A. In silico analysis of virulence genes in an emerging dental pathogen A. baumannii and related species. Arch Oral Biol. 2018 Oct;94:93-98. Pubmed PMID: 30015217.
  38. Qin C, D'Souza R, Feng JQ. Dentin matrix protein 1 (DMP1): new and important roles for biomineralization and phosphate homeostasis. J Dent Res. 2007 Dec;86(12):1134-41. Pubmed PMID: 18037646.
  39. He G, Dahl T, Veis A, George A. Dentin matrix protein 1 initiates hydroxyapatite formation in vitro. Connect Tissue Res. 2003;44 Suppl 1:240- 5. Pubmed PMID: 12952204.
  40. Narayanan K, Srinivas R, Ramachandran A, Hao J, Quinn B, George A. Differentiation of embryonic mesenchymal cells to odontoblast-like cells by overexpression of dentin matrix protein 1. Proc Natl Acad Sci U S A. 2001 Apr 10;98(8):4516-21. Pubmed PMID: 11287660.
  41. Hong S, Li L, Cai W, Jiang B. The potential application of concentrated growth factor in regenerative endodontics. Int Endod J. 2019 May;52(5):646-655. Pubmed PMID: 30471228.
  42. Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014 Jun 5;510(7503):92-101. Pubmed PMID: 24899309.
  43. Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick G, et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med. 2002 Oct 21;196(8):1025-37. Pubmed PMID: 12391014.
  44. Colas RA, Shinohara M, Dalli J, Chiang N, Serhan CN. Identification and signature profiles for pro-resolving and inflammatory lipid mediators in human tissue. Am J Physiol Cell Physiol. 2014 Jul 1;307(1):C39-54. Pubmed PMID: 24696140.
  45. Clària J, Dalli J, Yacoubian S, Gao F, Serhan CN. Resolvin D1 and resolvin D2 govern local inflammatory tone in obese fat. J Immunol. 2012 Sep 1;189(5):2597-605. Pubmed PMID: 22844113.
  46. Keelan JA, Mas E, D'Vaz N, Dunstan JA, Li S, Barden AE, et al. Effects of maternal n-3 fatty acid supplementation on placental cytokines, pro-resolving lipid mediators and their precursors. Reproduction. 2015 Feb;149(2):171-8. Pubmed PMID: 25504868.
  47. Arnardottir H, Orr SK, Dalli J, Serhan CN. Human milk proresolving mediators stimulate resolution of acute inflammation. Mucosal Immunol. 2016 May;9(3):757-766. Pubmed PMID: 26462421.
  48. Dalli J, Colas RA, Quintana C, Barragan-Bradford D, Hurwitz S, Levy BD, et al. Human Sepsis Eicosanoid and Proresolving Lipid Mediator Temporal Profiles: Correlations With Survival and Clinical Outcomes. Crit Care Med. 2017 Jan;45(1):58-68. Pubmed PMID: 27632672.
  49. Frediani JK, Jones DP, Tukvadze N, Uppal K, Sanikidze E, Kipiani M, et al. Plasma metabolomics in human pulmonary tuberculosis disease: a pilot study. PLoS One. 2014 Oct 15;9(10):e108854. Pubmed PMID: 25329995.
  50. Chiang N, de la Rosa X, Libreros S, Serhan CN. Novel Resolvin D2 Receptor Axis in Infectious Inflammation. J Immunol. 2017 Jan 15;198(2):842- 851. Pubmed PMID: 27994074.
  51. Cotti E, Ideo F, Pedrazzini A, Bardini G, Musu D, Kantarci A. Proresolving Mediators in Endodontics: A Systematic Review. J Endod. 2021 May;47(5):711-720. Pubmed PMID: 33548330.
  52. Spite M, Norling LV, Summers L, Yang R, Cooper D, Petasis NA, et al. Resolvin D2 is a potent regulator of leukocytes and controls microbial sepsis. Nature. 2009 Oct 29;461(7268):1287-91. Pubmed PMID: 19865173.
  53. Shi J, Zhang X, Jiang L, Zhang L, Dong Y, Midgley AC, et al. Regulation of the inflammatory response by vascular grafts modified with Aspirin-Triggered Resolvin D1 promotes blood vessel regeneration. Acta Biomater. 2019 Oct 1;97:360-373. Pubmed PMID: 31351251.

         Indexed in

pubhub  CGS  indexcoop  
j-gate  DOAJ  Google_Scholar_logo

       Total Visitors

SciDoc Counter

Get in Touch

SciDoc Publishers
16192 Coastal Highway
Lewes, Delaware 19958
Tel :+1-(302)-703-1005
Fax :+1-(302)-351-7355
Email: contact.scidoc@scidoc.org


porn