7 research outputs found

    Oral plaque from Type 2 diabetic patients reduces the clonogenic capacity of dental pulp-derived mesenchymal stem cells

    Get PDF
    Type 2 diabetes (T2D) is a major metabolic disease and a key epigenetic risk factor for the development of additional clinical complications. Among them, periodontitis (PD), a severe inflammatory disease ascribable to a dysregulated physiology and composition of the oral microbiota, represents one of the most relevant complications. Periodontitis can impact the structure of the tooth and likely the stem and progenitor cell pool, which actively contributes to the periodontal microenvironment and homeostasis. Modifications of the oral plaque play a key role in the etiopathogenesis of PD caused by T2D. However, to what extent the biology of the progenitor pool is affected has still to be elucidated. In this short report, we aimed to explore the biological effects of oral plaque derived from T2D patients with PD in comparison to non-diabetic patients with PD. Oral plaque samples were isolated from T2D and non-diabetic subjects with PD. Dental pulp stem cells (DPSCs), derived from the premolar tooth, were conditioned for 21 days with oral plaque samples and tested for their clonogenic ability. Cultures were also induced to differentiate towards the osteogenic lineage, and ALP and osteocalcin gene expression levels were evaluated by real-time qPCR. Results have shown that the number of clones generated by DPSCs exposed to T2D oral plaque was significantly lower compared to controls (ctl). The multivariate analysis confirmed that the decreased clonogenesis was significantly correlated only with T2D diagnosis. Moreover, the effect of T2D oral plaque was specific to DPSCs. Indicators of osteogenic differentiation were not significantly affected. This study provides a new biological insight into the effects ascribable to T2D in PD

    The Effects of Bone Morphogenic Protein 2 on the Viability and Proliferation of Dental Pulp Stem Cell Isolates

    Full text link
    Growth factor is an umbrella term used to describe a signaling molecule present in organisms that serves the purpose of influencing a stimulatory or inhibitory response from the target cell it acts on. Growth factors are categorized and classified by the types of tissues they act on and the cellular responses they illicit. For example, there are growth factors that act primarily in connective tissue on fibroblasts and they are aptly classified as the fibroblast growth factor family. Similarly, there is another family of growth factors that act on bone forming cells, among others, and they are known as bone morphogenic proteins. One member from this group of growth factors, BMP-2, is the focus of the current research. Bone Morphogenic Proteins are part of the Transforming Growth Factor-beta superfamily of growth factors. Recently they have been implicated in a variety of pathological processes including colon cancer and Barrettโ€™s esophagus. BMP-2 is known for its ability to stimulate bone formation and is the most successful bone modulator in the family. Its involvement in cellular differentiation into bone forming cells has been shown in many studies. BMP-2 acts on cell surface receptors which, through a series of reactions, enhances the likelihood of differentiating into a bone forming cell. This induction can be demonstrated in a variety of cell types. Those of particular interest are stem cells. There are three main types of stem cells including: embryonic stem cells, adult non- embryonic stem cells, induced pluripotent stem cells. Adult stem cells are the most abundant and are less controversial than embryonic stem cells. Currently adult stem cells are being isolated from a variety of tissues including those of mesenchymal tissue origin. The cell type in the current study are dental pulp stem cells (DPSC) which have been shown to display phenotypic changes in response to various growth factors. The use of multiple growth factors concomitantly on DPSC has not been studied in great depth, and thus it is not known if multiple stimulatory growth factors will act synergistically or antagonistically with one another. The data from the two following studies provide evidence that BMP-2 provides a stimulating influence on at least one dental pulp stem cell subset. When used concomitantly, BMP-2 and Vascular Endothelial Growth Factor (VEGF) enhances sDT DPSC phenotype (viability and growth) beyond that of either growth factor independently. Future studies may be needed to evaluate the potential for BMP-2 and other growth factors to induce DPSC differentiation and lineage-specific phenotypic changes for bioengineering applications or tissue regeneration

    ZIF-8 Modified Polypropylene Membrane: A Biomimetic Cell Culture Platform with a View to the Improvement of Guided Bone Regeneration.

    Full text link
    PurposeDespite the significant advances in modeling of biomechanical aspects of cell microenvironment, it remains a major challenge to precisely mimic the physiological condition of the particular cell niche. Here, the metal-organic frameworks (MOFs) have been introduced as a feasible platform for multifactorial control of cell-substrate interaction, given the wide range of physical and mechanical properties of MOF materials and their structural flexibility.ResultsIn situ crystallization of zeolitic imidazolate framework-8 (ZIF-8) on the polydopamine (PDA)-modified membrane significantly raised surface energy, wettability, roughness, and stiffness of the substrate. This modulation led to an almost twofold increment in the primary attachment of dental pulp stem cells (DPSCs) compare to conventional plastic culture dishes. The findings indicate that polypropylene (PP) membrane modified by PDA/ZIF-8 coating effectively supports the growth and proliferation of DPSCs at a substantial rate. Further analysis also displayed the exaggerated multilineage differentiation of DPSCs with amplified level of autocrine cell fate determination signals, like BSP1, BMP2, PPARG, FABP4, ACAN, and COL2A. Notably, osteogenic markers were dramatically overexpressed (more than 100-folds rather than tissue culture plate) in response to biomechanical characteristics of the ZIF-8 layer.ConclusionHence, surface modification of cell culture platforms with MOF nanostructures proposed as a powerful nanomedical approach for selectively guiding stem cells for tissue regeneration. In particular, PP/PDA/ZIF-8 membrane presented ideal characteristics for using as a barrier membrane for guided bone regeneration (GBR) in periodontal tissue engineering

    ์‚ฌ๋žŒ์น˜์•„ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ์—์„œ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค๋ฅผ ์ด์šฉํ•œ ์ƒ์•„์งˆ-ํŽ„ํ”„ ์žฌ์ƒ

    Get PDF
    ํ•™์œ„๋…ผ๋ฌธ (๋ฐ•์‚ฌ) -- ์„œ์šธ๋Œ€ํ•™๊ต ๋Œ€ํ•™์› : ์น˜์˜ํ•™๋Œ€ํ•™์› ์น˜์˜๊ณผํ•™๊ณผ, 2020. 8. ์ •ํ•„ํ›ˆ.Background and Purpose of the Study Xenogeneic serum is widely used as a growth supplement for cell culture medium; however, animal-borne pathogens increase the risk of transmitting infectious agents. Human autologous supplements, including platelet derivatives such as platelet-rich fibrin (PRF), have been assessed in clinical studies as a possible replacement for fetal bovine serum. However, concentrated leukocytes may affect catabolic gene expression in tendons and ligaments, and concentrated red blood cells (RBCs) cause inflammation due to numerous pro-inflammatory interleukins. It was hypothesized that minimizing the number of leukocytes would contribute to the differentiation activity of human dental pulp stem cells (hDPSCs). This study aimed to explore the ability of autologous fibrin (AF) serum prepared after leukocyte removal to induce the differentiation of hDPSCs in vitro and in vivo. Materials and Methods Human DPSCs were obtained from extracted third molars. To characterize the immunophenotype of the hDPSCs, the expression of mesenchymal stem cell-associated surface markers at passage 5 was analyzed by flow cytometry. Next, AF was prepared by high-speed centrifugation and most of the leukocytes were removed. The AF serum was then subjected to three freeze-thaw cycles and used in vitro for the cell culture. hDPSCs were treated with AF serum, and then odontogenic-associated markers were investigated by real-time polymerase chain reaction (PCR). For the in vivo experiments, autologous fibrin matrix (AFM) was fabricated on a plate and hDPSCs cultured on the AFM formed a cell-fibrin complex. Then, the potency of the differentiation induction of the hDPSCs in the cell-fibrin complex was evaluated via real-time PCR and Western blot. The hDPSCs mixed with a cell-fibrin complex were subcutaneously transplanted into nude mice and allowed to grow for 8 weeks. Then, the cell-fibrin complex was examined how induced odontoblastic differentiation and dentin formation of the hDPSCs in the mice by immunohistochemical (IHC) analysis. Results Flow cytometric analysis showed that approximately 90% or more of the hPDLSCs expressed CD13, CD90, and CD146, and 1.58% expressed CD34. The multi-lineage differentiation capacity of hDPSCs in vitro in osteogenic, chondrogenic, and adipogenic medium was observed. Scanning electron microscopy (SEM) images verified that leukocytes were almost absent from the fibrin matrix after high-speed centrifugation. A cytokine array showed that AF serum released sufficient cytokines to induce the differentiation of hDPSCs. AF serum accelerated mineral nodule formation in vitro and increased the expression of odontoblast-associated genes, such as alkaline phosphatase (ALP), bone sialoprotein (BSP), dentin matrix protein 1 (DMP1), dentin sialophosphoprotein (DSPP), runt-related transcription factor 2 (Runx2), and vascular endothelial growth factor (VEGF). In addition, the cell-fibrin complex also was observed that it significantly up-regulated the levels of the proteins DMP1, osteopontin (OPN), and DSPP during odontogenic differentiation. Furthermore, IHC staining revealed proteins were expressed after the transplantation of the hDPSCs and the cell-fibrin complex induced the hDPSCs to differentiate into odontoblasts-like cells and to regenerate dentin-pulp tissue in vivo. Conclusions The present study investigated the potential safe methods to expand autologous cells in a risk-free environment, and the cell-fibrin complex scaffold could be generated without the introduction of foreign scaffold materials. The results of experiments confirmed that 1. AF serum prepared after removal of leukocytes to could be used for the proliferation and promotion of hDPSCs odontogenic differentiation under animal serum-free conditions. 2. Human DPSCs cultured on the AFM formed a cell-fibrin complex which became stable and secreted their own matrix to form an autogenous 3D structure. 3. AFM can be effectively used as a carrier for successful regeneration of dentin-pulp complexes, which has been demonstrated by the in vivo subcutaneous transplantation in nude mice. 4. As a clinical implication, AFM possesses good bio-compatibility and cell infiltration and is suitable for dental tissue regeneration in hDPSCs.1. ์—ฐ๊ตฌ๋ชฉ์  ์‚ฌ๋žŒ์น˜์•„์ค„๊ธฐ์„ธํฌ๋ฅผ ๋ฐฐ์–‘ ์‹œ ์ด์ข…์ฒจ๊ฐ€์ œ๋ฅผ ์ฃผ๋กœ ์‚ฌ์šฉํ•˜์—ฌ ์™”๋Š”๋ฐ ์ด๋Š” ์ด์ข… ๊ธฐ์›์ด๊ธฐ ๋•Œ๋ฌธ์— ๋™๋ฌผ์„ฑ ๋ณ‘์›์ฒด ๋“ฑ์˜ ๊ฐ์—ผ์›์„ ์ง€๋‹ ๊ฐ€๋Šฅ์„ฑ์ด ์žˆ๋‹ค. ์ตœ๊ทผ fetal bovine serum (FBS)๋ฅผ ๋Œ€์ฒดํ•˜๊ธฐ ์œ„ํ•œ ์ž„์ƒ ์—ฐ๊ตฌ์—์„œ ํ˜ˆ์†ŒํŒ ํ’๋ถ€ ํ”ผ๋ธŒ๋ฆฐ (Platelet rich fibrin; PRF)๊ณผ ๊ฐ™์€ ํ˜ˆ์†ŒํŒ ์œ ๋„์ฒด๋ฅผ ํฌํ•จํ•œ ์ธ๊ฐ„ ์ž๊ฐ€ ๋ณด์ถฉ์ œ๊ฐ€ ๋งŽ์ด ์—ฐ๊ตฌ๋˜์—ˆ๋‹ค. ๊ทธ๋Ÿฌ๋‚˜ PRF๋Š” ๋†์ถ•๋œ ๋ฐฑํ˜ˆ๊ตฌ์™€ ์ ํ˜ˆ๊ตฌ๋ฅผ ํ•จ์œ ํ•˜๊ณ  ์žˆ์œผ๋ฉฐ, ๊ทธ์ค‘ ๋ฐฑํ˜ˆ๊ตฌ๋Š” ๊ฑด(tendon)๊ณผ ์ธ๋Œ€(ligament)์—์„œ ์œ ์ „์ž ๋ฐœํ˜„์— ์˜ํ–ฅ์„ ์ค„ ์ˆ˜ ์žˆ๊ณ , ์ ํ˜ˆ๊ตฌ๋Š” ์—ผ์ฆ๋ฐ˜์‘์„ ์ด‰์ง„ํ•˜๋Š” ์ธํ„ฐ๋ฃจํ‚จ์„ ๋‹ค๋Ÿ‰ ํ•จ์œ ํ•˜๊ธฐ ๋•Œ๋ฌธ์— ์˜๋„์น˜ ์•Š์€ ์—ผ์ฆ์ด ์œ ๋ฐœ๋  ์ˆ˜ ์žˆ๋‹ค๊ณ  ๋ณด๊ณ ๋˜์–ด์™”๋‹ค. ํ•˜์—ฌ ๋ฐฑํ˜ˆ๊ตฌ ์ˆ˜๋ฅผ ์ตœ์†Œํ™”ํ•˜๋ฉด ์ธ๊ฐ„์น˜์•„ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ (human dental pulp stem cells; hDPSCs)์˜ ๋ถ„ํ™”์— ๊ธฐ์—ฌํ•  ๊ฒƒ์ด๋ผ๊ณ  ๊ฐ€์„ค์„ ์„ธ์› ๋‹ค. ์ด์— ๋ณธ ์—ฐ๊ตฌ๋ฅผ ํ†ตํ•ด ๋ฐฑํ˜ˆ๊ตฌ๋ฅผ ์ œ๊ฑฐํ•œ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ์œผ๋กœ ์ฒด์™ธ ๋ฐ ์ƒ์ฒด ๋‚ด์—์„œ ์‚ฌ๋žŒ์น˜์•„ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ ์˜ ๋ถ„ํ™”๋ฅผ ์œ ๋„ํ•˜๋Š” ๋Šฅ๋ ฅ์— ๋Œ€ํ•ด ์•Œ์•„๋ณด๊ณ ์ž ํ•œ๋‹ค. 2. ์—ฐ๊ตฌ๋ฐฉ๋ฒ• ๋ฐœ๊ฑฐ๋œ ์ œ3๋Œ€๊ตฌ์น˜์˜ ์น˜์ˆ˜์กฐ์ง์—์„œ ์‚ฌ๋žŒ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ๋ฅผ ์ถ”์ถœํ•˜์˜€์œผ๋ฉฐ, ์ด๋ฅผ ๋Œ€์ƒ์œผ๋กœ ์ค„๊ธฐ์„ธํฌ ํŠน์„ฑ์„ ์กฐ์‚ฌํ•˜์˜€๋‹ค. ์‚ฌ๋žŒ ํ˜ˆ์•ก์„ ๊ณ ์† ์›์‹ฌ ๋ถ„๋ฆฌํ•˜์—ฌ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ์„ ๋งŒ๋“ค์—ˆ๊ณ  ๊ทธ ํ”ผ๋ธŒ๋ฆฐ ์ถ”์ถœ๋ฌผ๋กœ in vitro ์‹คํ—˜์„ ์œ„ํ•œ ์„ธํฌ ๋ฐฐ์–‘์— ์‚ฌ์šฉ๋˜์—ˆ์œผ๋ฉฐ, ๋˜ํ•œ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค๋ฅผ ์‚ฌ์šฉํ•˜์—ฌ in vivo ์‹คํ—˜์„ ์‹œํ–‰ํ•˜์˜€๋‹ค. ์šฐ์„ , ํ˜ˆ์•ก์„ ์›์‹ฌ ๋ถ„๋ฆฌํ•˜์—ฌ ์–ป์€ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค์—์„œ ๋ฐฑํ˜ˆ๊ตฌ๊ฐ€ ์ œ๊ฑฐ๋˜์—ˆ๋Š”์ง€ ํ™•์ธํ•˜๊ธฐ ์œ„ํ•˜์—ฌ ์กฐ์งํ•™์  ๋ถ„์„ ๋ฐ ์ฃผ์‚ฌ์ „์žํ˜„๋ฏธ๊ฒฝ(scanning electron microscopy; SEM) ์ดฌ์˜์„ ํ†ตํ•ด ํ™•์ธํ•˜์˜€๊ณ , ์‚ฌ๋žŒ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ์— ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ์ถ”์ถœ๋ฌผ์„ ์ฒ˜๋ฆฌํ•œ ๋’ค qRT-PCR (quantitative real-time polymerase chain reaction)์„ ํ†ตํ•ด ์น˜์„ฑ ๊ด€๋ จ ์œ ์ „์ž ๋ฐœํ˜„์„ ์กฐ์‚ฌํ•˜์˜€๋‹ค. ๊ทธ๋ฆฌ๊ณ  ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ๋ฅผ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค ์œ„์—์„œ ๋ฐฐ์–‘ํ•˜์—ฌ ์ค„๊ธฐ์„ธํฌ-ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค ํ˜ผํ•ฉ์ฒด๋ฅผ ์–ป์–ด ๊ทธ ํšจ๋Šฅ์„ Western blot์„ ํ†ตํ•ด ํ‰๊ฐ€ํ•˜์˜€๋‹ค. in vivo ํ™˜๊ฒฝ์—์„œ์˜ ์˜ํ–ฅ์„ ํ™•์ธํ•˜๊ธฐ ์œ„ํ•ด ๋ˆ„๋“œ๋งˆ์šฐ์Šค๋ฅผ ๋Œ€์ƒ์œผ๋กœ, ์‹คํ—˜๊ตฐ์œผ๋กœ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ์™€ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค ํ˜ผํ•ฉ์ฒด๋ฅผ ํ•œ ์ชฝ ํ”ผํ•˜์— ์ด์‹ํ•˜์˜€๊ณ , ๋Œ€์กฐ๊ตฐ์œผ๋กœ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค๋ฅผ ์‹คํ—˜๊ตฐ ๋ฐ˜๋Œ€ํŽธ์— ์ด์‹ํ•˜์˜€๋‹ค. 8์ฃผ ํ›„ ์ด์‹ํ•œ ์กฐ์ง์„ ์ฑ„์ทจํ•˜์—ฌ ์ƒ์•„์งˆ๋ชจ์„ธํฌ(odontoblast) ๋ถ„ํ™”์™€ ์ƒ์•„์งˆ๋ชจ์„ธํฌ์— ์˜ํ•œ ์ƒ์•„์งˆ ์ƒ์„ฑ ๋ฐ ํŠน์ •๋œ ๋‹จ๋ฐฑ์งˆ ๋ฐœํ˜„์„ immunohistochemistry๋กœ ๋น„๊ต ํ™•์ธํ•˜์˜€๋‹ค. 3. ์—ฐ๊ตฌ๊ฒฐ๊ณผ ์œ ์„ธํฌ ๋ถ„์„ ๊ฒฐ๊ณผ ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ ํ‘œ์ง€์ธ์ž์ธ CD13, CD90, CD146์ด ๋ฐœํ˜„ ๋˜์—ˆ์œผ๋ฉฐ, ๋ถ„ํ™”๋ฅผ ํ†ตํ•ด ๋‹ค์ค‘ ๋ถ„ํ™” ๋Šฅ๋ ฅ์ด ์žˆ๋Š” ์ค„๊ธฐ์„ธํฌ์˜ ํŠน์ง•์„ ๊ฐ€์ง€๊ณ  ์žˆ์Œ์„ ํ™•์ธํ•˜์˜€๋‹ค. ์กฐ์งํ•™์  ๋ถ„์„๊ณผ SEM์œผ๋กœ ํ™•์ธํ•œ ๊ฒฐ๊ณผ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค์—์„œ ๋ฐฑํ˜ˆ๊ตฌ๋Š” ๊ฑฐ์˜ ํ™•์ธ๋˜์ง€ ์•Š์•˜๋‹ค. In vitro์—์„œ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ์ถ”์ถœ๋ฌผ์€ ๊ฒฝ์กฐ์ง ๋ถ„ํ™” ๋งˆ์ปค์ธ alkaline phosphatase (ALP), bone sialoprotein (BSP), dentin matrix protein 1 (DMP1), dentin sialophosphoprotein (DSPP), runt-related transcription factor 2 (Runx2), vascular endothelial growth factor (VEGF)์˜ ๋ฐœํ˜„์ด ์ฆ๊ฐ€ํ•œ ๊ฒƒ์„ ํ™•์ธํ•˜์˜€๋‹ค. ๋˜ํ•œ ์ค„๊ธฐ์„ธํฌ-๋งคํŠธ๋ฆญ์Šค ํ˜ผํ•ฉ์ฒด๋Š” ์ƒ์•„์งˆ๋ชจ์„ธํฌ์˜ ๋ถ„ํ™”๊ณผ์ •์—์„œ DMP1, osteopontin (OPN) ๋ฐ DSPP์˜ ๋‹จ๋ฐฑ์งˆ ๋ฐœํ˜„์„ ์œ ์˜ํ•˜๊ฒŒ ์ด‰์ง„ํ•จ์„ ํ™•์ธํ•  ์ˆ˜ ์žˆ์—ˆ๋‹ค. ๋ˆ„๋“œ๋งˆ์šฐ์Šค์˜ ํ”ผํ•˜์— ์ด์‹ํ•œ ๋งค์‹์ฒด์˜ ์กฐ์งํ•™์  ๋ถ„์„๊ฒฐ๊ณผ ๋Œ€์กฐ๊ตฐ์— ๋น„ํ•ด ์น˜์ˆ˜์ค„๊ธฐ์„ธํฌ์™€ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค ํ˜ผํ•ฉ์ฒด์˜ ์ด์‹๊ตฐ์—์„œ ์ƒ์•„์งˆ ํŠน์ • ๋‹จ๋ฐฑ์งˆ์ด ๋”์šฑ ๊ฐ•ํ•˜๊ฒŒ ์—ผ์ƒ‰๋˜์—ˆ์œผ๋ฉฐ, ์ƒ์•„์งˆ-ํŽ„ํ”„ ์กฐ์ง์ด ์žฌ์ƒ๋˜์–ด ์žˆ์Œ์„ ํ™•์ธํ•˜์˜€๋‹ค. 4. ๊ฒฐ๋ก  ์ด ์—ฐ๊ตฌ๋Š” ๋ฌด์œ„ํ—˜ ํ™˜๊ฒฝ์—์„œ ์ž๊ฐ€ ์„ธํฌ๋ฅผ ํ™•์žฅ์‹œํ‚ค๋Š” ์ž ์žฌ์ ์ธ ์•ˆ์ „ํ•œ ๋ฐฉ๋ฒ•์„ ์กฐ์‚ฌํ–ˆ์œผ๋ฉฐ, ์„ธํฌ ์Šค์บํด๋“œ ๋ณตํ•ฉ์ฒด๋Š” ์™ธ์ธ์„ฑ ๋ฌผ์งˆ์˜ ๋„์ž…์—†์ด ์น˜์•„ ์กฐ์ง์ด ์ƒ์„ฑ ๋  ์ˆ˜ ์žˆ๋‹ค. 1. ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ์ถ”์ถœ๋ฌผ์€ ๋ฐฑํ˜ˆ๊ตฌ๋ฅผ ์ œ๊ฑฐํ•œํ›„ ์ œ์กฐ๋˜์—ˆ์œผ๋ฉฐ ๋™๋ฌผ ๋ฌด ํ˜ˆ์ฒญ ์กฐ๊ฑดํ•˜์— ์น˜์•„์ค„๊ธฐ์„ธํฌ์˜ ์ฆ์‹๊ณผ ๋ถ„ํ™”์— ์œ ์šฉํ•˜๊ฒŒ ์ด์šฉ๊ฐ€๋Šฅํ•˜๋‹ค. 2. ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค์—์„œ ๋ฐฐ์–‘๋œ ์น˜์•„์ค„๊ธฐ์„ธํฌ๋Š” ์„ธํฌ-ํ”ผ๋ธŒ๋ฆฐ ๋ณตํ•ฉ์ฒด๋ฅผ ํ˜•์„ฑํ•˜์˜€์œผ๋ฉฐ, ์ด๋Š” ์•ˆ์ •์ ์ด๊ณ  3์ฐจ์›์ ์ธ ์ž๊ฐ€ ๋งคํŠธ๋ฆญ์Šค ๊ตฌ์กฐ๋ฅผ ํ˜•์„ฑํ•˜์˜€๋‹ค. 3. ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค๋Š” ์ƒ์ฒด ๋‚ด ๋ˆ„๋“œ๋งˆ์šฐ์Šค ํ”ผํ•˜์— ์ด์‹ํ•˜์—ฌ ์ƒ์•„์งˆ-ํŽ„ํ”„ ๋ณตํ•ฉ์ฒด์˜ ์„ฑ๊ณต์ ์ธ ์žฌ์ƒ์— ์šฐ์ˆ˜ํ•œ ์šด๋ฐ˜์ฒด๋กœ์„œ ์œ ์šฉํ•˜๊ฒŒ ์ด์šฉํ•  ์ˆ˜ ์žˆ์Œ์ด ์ž…์ฆ๋˜์—ˆ๋‹ค. 4. ์ž„์ƒ์  ์˜๋ฏธ์—์„œ ์ž๊ฐ€ ํ”ผ๋ธŒ๋ฆฐ ๋งคํŠธ๋ฆญ์Šค๋Š” ์šฐ์ˆ˜ํ•œ ์ƒ์ฒด ์ ํ•ฉ์„ฑ ๋ฐ ์„ธํฌ ์นจ์œค์„ ๋ณด์œ ํ•˜๋ฉฐ ์น˜์•„์ค„๊ธฐ์„ธํฌ์—์„œ ์น˜์•„์กฐ์ง ์žฌ์ƒ์— ์œ ์šฉํ•˜๋‹ค๋Š” ๊ฒƒ์„ ์•Œ ์ˆ˜ ์žˆ๋‹ค.โ… . INTRODUCT 1 โ…ก. MATERIALS AND METHODS 5 1. Samples 5 2. Fabrication of AF and AF serum 5 3. Histological analysis of AF and PRF 6 4. Scanning electron microscopy (SEM) analysis 7 5. Human cytokine antibody array 8 6. Primary cell culture 9 7. Proliferation assay 10 8. Flow cytometry analysis 10 9. Multi-differentiation of hDPSCs 11 10. Gene expression of the hDPSCs 12 11. Fabrication of autologous fibrin matrix (AFM) and dental pulp stem cells culture on the AFM (cell-fibrin complex) 13 12. The invasion of the hDPSCs into the AFM 14 13. Immunofluorescence analysis of cell adhesion 15 14. Evaluation of the differentiation of hDPSCs on the AFM by quantitative real-time PCR 15 15. Western blot analysis 16 16. Transfection of AFM into a rat calvarial defect 17 17. Micro-computed tomography imaging analysis 18 18. Preparation of human dentin matrix 18 19. Transplantation 19 20. Histological analysis 20 21. IHC analysis 21 22. Statistical analysis 22 โ…ข. RESULTS 1. Characterization of AF 23 2. Detection of cytokines in the AF serum 24 3. Characterization of hDPSCs in the AF serum 25 4. Effects of the AF serum on the hDPSCs in vitro 26 5. Optimal thickness of the AFM for the hDPSCs culture 27 6. Infiltration and morphological observation of the hDPSCs on AFM 28 7. The odontogenic influences of hDPSCs on the AFM 28 8. Effects of AFM on the rat calvarial defect model 29 9. Histological analysis of human dentin matrix surface in vivo 30 โ…ฃ. DISCUSSION 32 โ…ค. CONCLUSIONS 43 โ…ฅ. REFERENCES 44 โ…ฆ. FIGURES AND TABLE 54 โ…ง. ABSTRACT IN KOREAN 77 LIST OF FIGURES Figure 1. Fabrication of an AF and AF serum 54 Figure 2. AF compared with PRF 55 Figure 3. SEM evaluation of the AF 56 Figure 4. Evaluation of a human cytokine/chemokine array for AF serum 57 Figure 5. Characterization of hDPSCs at passage 5 by flow cytometry assay 58 Figure 6. Characterization of hDPSCs 59 Figure 7. Osteogenic differentiation of hDPSCs in vitro 60 Figure 8. Morphology of hDPSCs cultured in AF serum compared with FBS 61 Figure 9. Effects of AF serum on the proliferation of hDPSCs compared with 10% FBS in vitro 62 Figure 10. Effects of AF serum on the odontogenic influence of hDPSCs in vitro 63 Figure 11. Images of hDPSCs cultured on the AFM (cell-fibrin complex) 64 Figure 12. Optimal AFM thickness for hDPSCs culture 65 Figure 13. Infiltration of DPSCs on AFM 66 Figure 14. Morphological observation of hDPSCs on AFM 67 Figure 15. Evaluation of the odontogenic differentiation of hDPSCs cultured on the AFM 68 Figure 16. Investigation of odontogenic-specific protein expression by Western blot assay 69 Figure 17. Effects of AFM on the rat calvarial defect model 70 Figure 18. Morphology of the human dentin matrix 71 Figure 19. Image of individual components of the transplantation composite and schematic of the final construct 72 Figure 20. Effects on the AFM surface in vivo 73 Figure 21. Immunohistochemical analysis showing the effect of the hDPSCs cultured on the AFM in vivo (low magnification) 74 Figure 22. Immunohistochemical analysis showing the effect of the hDPSCs cultured on the AFM in vivo (high magnification) 75Docto

    Dental Mesenchymal Stem Cell-Based Translational Regenerative Dentistry: From Artificial to Biological Replacement

    No full text
    Dentistry is a continuously changing field that has witnessed much advancement in the past century. Prosthodontics is that branch of dentistry that deals with replacing missing teeth using either fixed or removable appliances in an attempt to simulate natural tooth function. Although such โ€œreplacement therapiesโ€ appear to be easy and economic they fall short of ever coming close to their natural counterparts. Complications that arise often lead to failures and frequent repairs of such devices which seldom allow true physiological function of dental and oral-maxillofacial tissues. Such factors can critically affect the quality of life of an individual. The market for dental implants is continuously growing with huge economic revenues. Unfortunately, such treatments are again associated with frequent problems such as peri-implantitis resulting in an eventual loss or replacement of implants. This is particularly influential for patients having co-morbid diseases such as diabetes or osteoporosis and in association with smoking and other conditions that undoubtedly affect the final treatment outcome. The advent of tissue engineering and regenerative medicine therapies along with the enormous strides taken in their associated interdisciplinary fields such as stem cell therapy, biomaterial development, and others may open arenas to enhancing tissue regeneration via designing and construction of patient-specific biological and/or biomimetic substitutes. This review will overview current strategies in regenerative dentistry while overviewing key roles of dental mesenchymal stem cells particularly those of the dental pulp, until paving the way to precision/translational regenerative medicine therapies for future clinical use
    corecore