51
|
Deferoxamine but Not Dimethyloxalylglycine, L-Mimosine, or Cobalt Dichloride Can Interfere with the MTT Assay. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5872865. [PMID: 30581861 PMCID: PMC6276496 DOI: 10.1155/2018/5872865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/11/2018] [Accepted: 09/26/2018] [Indexed: 11/17/2022]
Abstract
Hypoxia mimetic agents (HMAs) have been shown to have a positive influence on cellular functions in a multitude of tissue regenerative strategies. Novel experimental approaches use biomaterials as carriers for controlled delivery of these HMAs. Here, the cytotoxic aspects of biocompatibility are of key relevance. The MTT assay is widely used to evaluate cytotoxicity and proliferation. Based on the implications from the proceeding research we hypothesized that specific HMAs such as deferoxamine at high concentrations can interfere with the MTT assay. Thus, the aim of this study was to test the repercussions of the HMAs dimethyloxalylglycine, deferoxamine, L-mimosine, and CoCl2 on the validity of the MTT assay. Murine MC3T3-E1 cells were cultured in serum-free alphaMEM and in alphaMEM supplemented with 10 % fetal bovine serum with the HMAs dimethyloxalylglycine, deferoxamine, L-mimosine, and CoCl2, respectively, at 3 mM-0.3 mM for 24 h (experimental groups). Cells without HMAs served as control (control groups). The same experiments were performed with medium and phosphate buffered saline (PBS) without cells. In all settings MTT solution was added to PBS-washed or unwashed culture plates for the last two hours of the incubation period. Then MTT solution was removed and dimethyl sulfoxide was added to dissolve the formazan crystals and absorption was measured. Our data show that the presence of deferoxamine can interfere with the MTT assay if not removed before the addition of MTT. This is particularly important when evaluating cell viability in setups where deferoxamine-loaded biomaterials are used.
Collapse
|
52
|
Nada OA, El Backly RM. Stem Cells From the Apical Papilla (SCAP) as a Tool for Endogenous Tissue Regeneration. Front Bioeng Biotechnol 2018; 6:103. [PMID: 30087893 PMCID: PMC6066565 DOI: 10.3389/fbioe.2018.00103] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022] Open
Abstract
Stem cells extracted from developing tissues possibly exhibit not only unique but also superior traits against their developed counterparts. Indeed, stem cells from the apical papilla (SCAP); a unique group of dental stem cells related to developing roots have been shown to be a promising tool for regenerative endodontic procedures and regeneration in general. Studies have characterized the phenotypic traits as well as other regenerative potentials of these cells. Specific sub-populations have been highlighted as well as their neurogenic and angiogenic properties. Nevertheless, in light of the previously discussed features and potential applications of SCAP, there is still much to understand and a lot of information to unravel. The current review will discuss the role of specific markers for detection of different functional populations of SCAP; including CD146 and STRO-1, as well as their true multilineage differentiation potential. In particular, the role of the secretome in association with paracrine signaling in inflammatory microenvironments is also tackled. Additionally, the role of SCAP both in vitro and in vivo during regenerative approaches and in response to different growth factors and biologic scaffolds is highlighted. Finally, this review will shed light on current knowledge regarding the clinical translational potential of SCAP and elucidate possible areas for future research applications.
Collapse
Affiliation(s)
- Ola A Nada
- Oral Biology Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.,Tissue Engineering Laboratories, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| | - Rania M El Backly
- Tissue Engineering Laboratories, Faculty of Dentistry, Alexandria University, Alexandria, Egypt.,Endodontics, Conservative Dentistry Department, Faculty of Dentistry, Alexandria University, Alexandria, Egypt
| |
Collapse
|
53
|
Diogenes A, Hargreaves KM. Microbial Modulation of Stem Cells and Future Directions in Regenerative Endodontics. J Endod 2018; 43:S95-S101. [PMID: 28844309 DOI: 10.1016/j.joen.2017.07.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Regenerative endodontic procedures (REPs) have been shown to promote the resolution of signs and symptoms of disease and increase survival compared with traditional treatment procedures. However, there is still variable predictability of continued root development and evidence that the tissues formed do not recapitulate the native pulp-dentin complex. There is growing evidence that the apical papilla is capable of surviving prolonged endodontic infection and apical periodontitis and that it represents a rich source of undifferentiated mesenchymal stem cells in REPs. The survival and proper differentiation of stem cells transferred into infected root canals are fraught with challenges. Residual antigens, such as lipopolysaccharides, have been shown to be present in dentin even after adequate chemomechanical debridement. These antigens have a profound effect on stem cell fate by modulating their proliferative capacity and postdifferentiation phenotype. Thus, root canals must be detoxified in addition to disinfection. There is a strong need for translational studies that incorporate all aspects of tissue engineering in endodontics in models that include an existing infection to promote further advancement of the field. This is particularly important to make REPs more predictable when treating immature teeth in young patients. Importantly, regenerative procedures could eventually promote tooth longevity in our aging population. Lessons learned from translational studies that best mimic the clinical challenges could be evaluated in pragmatic clinical trials to determine the effectiveness of these procedures to promote desirable patient-centered outcomes.
Collapse
Affiliation(s)
- Anibal Diogenes
- Department of Endodontics at University of Texas Health Science Center at San Antonio, San Antonio, Texas.
| | - Kenneth M Hargreaves
- Department of Endodontics at University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
54
|
Wang P, Zhu S, Yuan C, Wang L, Xu J, Liu Z. Shear stress promotes differentiation of stem cells from human exfoliated deciduous teeth into endothelial cells via the downstream pathway of VEGF-Notch signaling. Int J Mol Med 2018; 42:1827-1836. [PMID: 30015843 PMCID: PMC6108868 DOI: 10.3892/ijmm.2018.3761] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/21/2018] [Indexed: 12/31/2022] Open
Abstract
Effects of shear stress on endotheliaxl differentiation of stem cells from human exfoliated deciduous teeth (SHEDs) were investigated. SHEDs were treated with shear stress, then reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to analyse the mRNA expression of arterial markers and western blot analysis was performed to analyse protein expression of angiogenic markers. Additionally, in vitro matrigel angiogenesis assay was performed to evaluate vascular-like structure formation. The secreted protein expression levels of the vascular endothelial growth factor (VEGF) of SHEDs after shear stress was also quantified using corresponding ELISA kits. Untreated SHEDs seeded on Matrigel cannot form vessel-like structures at any time points, whereas groups treated with shear stress formed a few vessel-like structures at 4, 8 and 12 h. When SHEDs were treated with EphrinB2-siRNA for 24, the capability of vessel-like structure formation was suppressed. After being treated with shear stress, the expression of VEGF, VEGFR2, DLL4, Notch1, EphrinB2, Hey1 and Hey2 (arterial markers) gene expression was significantly upregulated, moreover, the protein levels of VEGFR2, EphrinB2, CD31, Notch1, DLL4, Hey1, and Hey2 were also significantly up-regulated. Both the mRNA and protein expression levels of EphB4 (venous marker) were downregulated. The average VEGF protein concentration in supernatants secreted by shear stress treated SHEDs groups increased significantly. In conclusion, shear stress was able to induce arterial endothelial differentiation of stem cells from human exfoliated deciduous teeth, and VEGF-DLL4/Notch‑EphrinB2 signaling was involved in this process.
Collapse
Affiliation(s)
- Penglai Wang
- Dental Implant Center, Xuzhou Stomatological Hospital, Xuzhou, Jiangsu, P.R. China
| | - Shaoyue Zhu
- Department of Orthodontics, Xuzhou Stomatological Hospital, Xuzhou, Jiangsu, P.R. China
| | - Changyong Yuan
- Dental Implant Center, Xuzhou Stomatological Hospital, Xuzhou, Jiangsu, P.R. China
| | - Lei Wang
- Department of Periodontics, Xuzhou Stomatological Hospital, Xuzhou, Jiangsu, P.R. China
| | - Jianguang Xu
- The Discipline of Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Zongxiang Liu
- Department of ExperDignosis, Xuzhou Stomatological Hospital, Xuzhou, Jiangsu, P.R. China
| |
Collapse
|
55
|
Monteiro N, He W, Franca CM, Athirasala A, Bertassoni LE. Engineering Microvascular Networks in LED Light-Cured Cell-Laden Hydrogels. ACS Biomater Sci Eng 2018; 4:2563-2570. [PMID: 33435119 DOI: 10.1021/acsbiomaterials.8b00502] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The success of tissue engineering inevitably depends on the fabrication of tissue constructs that can be vascularized and that anastomose with the host vasculature. In this report, we studied the effects of light-emitting diode (LED) photopolymerized gelatin methacryloyl hydrogels (GelMA), encapsulated with stem cells from the apical papilla (SCAP) and human umbilical vein endothelial cells (HUVECs), in promoting vasculature network formation as a function of hydrogel physical and mechanical properties, as well as total cell density. Lithium acylphosphinate (LAP) was used as the photoinitiator in concentrations of 0.05, 0.075, 0.1% (w/v). GelMA hydrogel precursors of 5% (w/v) were encapsulated with cocultures of SCAPs and HUVECs at different cell densities (1×, 5×, and 10 × 106 cells/mL) and photo-cross-linked for 5 s. Results suggested that the compressive modulus of GelMA hydrogels increased as a function of LAP concentration, and had a maximum stiffness of 3.2 kPa. GelMA hydrogels photopolymerized using 0.05 or 0.075% LAP, which had an average of 1.5 and 1.6 kPa of elastic modulus respectively, had the most efficient vasculature formation after 5 days, and these results were further enhanced when the highest cell density (10 × 106 cells/mL) was used. Immunofluorescence images showed that SCAP cells spread in close contact with endothelial networks and expressed alpha smooth muscle actin (αSMA), which is suggestive of their differentiation into pericyte-like cells. αSMA expression was also apparently higher in hydrogels polymerized with 0.05% LAP and 10 × 106 cells/mLl. In conclusion, photopolymerization of GelMA hydrogels using an LED-light source can be an effective method for potential chair-side/in situ procedures for engineering of vascularized tissue constructs in regenerative medicine.
Collapse
Affiliation(s)
- Nelson Monteiro
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 Southwest Moody Avenue, Portland, Oregon 97201, United States
| | - Wenting He
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 Southwest Moody Avenue, Portland, Oregon 97201, United States
| | - Cristiane Miranda Franca
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 Southwest Moody Avenue, Portland, Oregon 97201, United States
| | - Avathamsa Athirasala
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 Southwest Moody Avenue, Portland, Oregon 97201, United States
| | - Luiz E Bertassoni
- Division of Biomaterials and Biomechanics, Department of Restorative Dentistry, OHSU School of Dentistry, 2730 Southwest Moody Avenue, Portland, Oregon 97201, United States
| |
Collapse
|
56
|
Wei ZZ, Zhu YB, Zhang JY, McCrary MR, Wang S, Zhang YB, Yu SP, Wei L. Priming of the Cells: Hypoxic Preconditioning for Stem Cell Therapy. Chin Med J (Engl) 2018; 130:2361-2374. [PMID: 28937044 PMCID: PMC5634089 DOI: 10.4103/0366-6999.215324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: Stem cell-based therapies are promising in regenerative medicine for protecting and repairing damaged brain tissues after injury or in the context of chronic diseases. Hypoxia can induce physiological and pathological responses. A hypoxic insult might act as a double-edged sword, it induces cell death and brain damage, but on the other hand, sublethal hypoxia can trigger an adaptation response called hypoxic preconditioning or hypoxic tolerance that is of immense importance for the survival of cells and tissues. Data Sources: This review was based on articles published in PubMed databases up to August 16, 2017, with the following keywords: “stem cells,” “hypoxic preconditioning,” “ischemic preconditioning,” and “cell transplantation.” Study Selection: Original articles and critical reviews on the topics were selected. Results: Hypoxic preconditioning has been investigated as a primary endogenous protective mechanism and possible treatment against ischemic injuries. Many cellular and molecular mechanisms underlying the protective effects of hypoxic preconditioning have been identified. Conclusions: In cell transplantation therapy, hypoxic pretreatment of stem cells and neural progenitors markedly increases the survival and regenerative capabilities of these cells in the host environment, leading to enhanced therapeutic effects in various disease models. Regenerative treatments can mobilize endogenous stem cells for neurogenesis and angiogenesis in the adult brain. Furthermore, transplantation of stem cells/neural progenitors achieves therapeutic benefits via cell replacement and/or increased trophic support. Combinatorial approaches of cell-based therapy with additional strategies such as neuroprotective protocols, anti-inflammatory treatment, and rehabilitation therapy can significantly improve therapeutic benefits. In this review, we will discuss the recent progress regarding cell types and applications in regenerative medicine as well as future applications.
Collapse
Affiliation(s)
- Zheng Z Wei
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yan-Bing Zhu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - James Y Zhang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Myles R McCrary
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Song Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Yong-Bo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shan-Ping Yu
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Ling Wei
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University; Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
57
|
Soares DG, Zhang Z, Mohamed F, Eyster TW, de Souza Costa CA, Ma PX. Simvastatin and nanofibrous poly(l-lactic acid) scaffolds to promote the odontogenic potential of dental pulp cells in an inflammatory environment. Acta Biomater 2018; 68:190-203. [PMID: 29294374 DOI: 10.1016/j.actbio.2017.12.037] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022]
Abstract
In this study, we investigated the anti-inflammatory, odontogenic and pro-angiogenic effects of integrating simvastatin and nanofibrous poly(l-lactic acid) (NF-PLLA) scaffolds on dental pulp cells (DPCs). Highly porous NF-PLLA scaffolds that mimic the nanofibrous architecture of extracellular matrix were first fabricated, then seeded with human DPCs and cultured with 0.1 μM simvastatin and/or 10 μg/mL pro-inflammatory stimulator lipopolysaccharide (LPS). The gene expression of pro-inflammatory mediators (TNF-α, IL-1β and MMP-9 mRNA) and odontoblastic markers (ALP activity, calcium content, DSPP, DMP-1 and BMP-2 mRNA) were quantified after long-term culture in vitro. In addition, we evaluated the scaffold's pro-angiogenic potential after 24 h of in vitro co-culture with endothelial cells. Finally, we assessed the combined effects of simvastatin and NF-PLLA scaffolds in vivo using a subcutaneous implantation mouse model. The in vitro studies demonstrated that, compared with the DPC/NF-PLLA scaffold constructs cultured only with pro-inflammatory stimulator LPS, adding simvastatin significantly repress the expression of pro-inflammatory mediators. Treating LPS+ DPC/NF-PLLA constructs with simvastatin also reverted the negative effects of LPS on expression of odontoblastic markers in vitro and in vivo. Western blot analysis demonstrated that these effects were related to a reduction in NFkBp65 phosphorylation and up-regulation of PPARγ expression, as well as to increased phosphorylation of pERK1/2 and pSmad1, mediated by simvastatin on LPS-stimulated DPCs. The DPC/NF-PLLA constructs treated with LPS/simvastatin also led to an increase in vessel-like structures, correlated with increased VEGF expression in both DPSCs and endothelial cells. Therefore, the combination of low dosage simvastatin and NF-PLLA scaffolds appears to be a promising strategy for dentin regeneration with inflamed dental pulp tissue, by minimizing the inflammatory reaction and increasing the regenerative potential of resident stem cells. STATEMENT OF SIGNIFICANCE The regeneration potential of stem cells is dependent on their microenvironment. In this study, we investigated the effect of the microenvironment of dental pulp stem cells (DPSCs), including 3D structure of a macroporous and nanofibrous scaffold, the inflammatory stimulus lipopolysaccharide (LPS) and a biological molecule simvastatin, on their regenerative potential of mineralized dentin tissue. The results demonstrated that LPS upregulated inflammatory mediators and suppressed the odontogenic potential of DPSCs. Known as a lipid-lowing agent, simvastatin was excitingly found to repress the expression of pro-inflammatory mediators, up-regulate odontoblastic markers, and exert a pro-angiogenic effect on endothelial cells, resulting in enhanced vascularization and mineralized dentin tissue regeneration in a biomimetic 3D tissue engineering scaffold. This novel finding is significant for the fields of stem cells, inflammation and dental tissue regeneration.
Collapse
|
58
|
Xu JG, Gong T, Wang YY, Zou T, Heng BC, Yang YQ, Zhang CF. Inhibition of TGF-β Signaling in SHED Enhances Endothelial Differentiation. J Dent Res 2017; 97:218-225. [PMID: 28972822 DOI: 10.1177/0022034517733741] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Low efficiency of deriving endothelial cells (ECs) from adult stem cells hampers their utilization in tissue engineering studies. The purpose of this study was to investigate whether suppression of transforming growth factor beta (TGF-β) signaling could enhance the differentiation efficiency of dental pulp-derived stem cells into ECs. We initially used vascular endothelial growth factor A (VEGF-A) to stimulate 2 dental pulp-derived stem cells (dental pulp stem cells and stem cells from human exfoliated deciduous teeth [SHED]) and compared their differentiation capacity into ECs. We further evaluated whether the vascular endothelial growth factor receptor I (VEGF-RI)-specific ligand placental growth factor-1 (PlGF-1) could mediate endothelial differentiation. Finally, we investigated whether the TGF-β signaling inhibitor SB-431542 could enhance the inductive effect of VEGF-A on endothelial differentiation, as well as the underlying mechanisms involved. ECs differentiated from dental pulp-derived stem cells exhibited the typical phenotypes of primary ECs, with SHED possessing a higher endothelial differentiation potential than dental pulp stem cells. VEGFR1-specific ligand-PLGF exerted a negligible effect on SHED-ECs differentiation. Compared with VEGF-A alone, the combination of VEGF-A and SB-431542 significantly enhanced the endothelial differentiation of SHED. The presence of SB-431542 inhibited the phosphorylation of Suppressor of Mothers Against Decapentaplegic 2/3 (SMAD2/3), allowing for VEGF-A-dependent phosphorylation and upregulation of VEGFR2. Our results indicate that the combination of VEGF-A and SB-431542 could enhance the differentiation of dental pulp-derived stem cells into endothelial cells, and this process is mediated through enhancement of VEGF-A-VEGFR2 signaling and concomitant inhibition of TGF-β-SMAD2/3 signaling.
Collapse
Affiliation(s)
- J G Xu
- 1 Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - T Gong
- 1 Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.,2 HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - Y Y Wang
- 3 Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, China
| | - T Zou
- 1 Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.,2 HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - B C Heng
- 1 Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.,2 HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - Y Q Yang
- 4 Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - C F Zhang
- 1 Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.,2 HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| |
Collapse
|
59
|
The Angiogenic Potential of DPSCs and SCAPs in an In Vivo Model of Dental Pulp Regeneration. Stem Cells Int 2017; 2017:2582080. [PMID: 29018483 PMCID: PMC5605798 DOI: 10.1155/2017/2582080] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/04/2017] [Accepted: 07/13/2017] [Indexed: 12/22/2022] Open
Abstract
Adequate vascularization, a restricting factor for the survival of engineered tissues, is often promoted by the addition of stem cells or the appropriate angiogenic growth factors. In this study, human dental pulp stem cells (DPSCs) and stem cells from the apical papilla (SCAPs) were applied in an in vivo model of dental pulp regeneration in order to compare their regenerative potential and confirm their previously demonstrated paracrine angiogenic properties. 3D-printed hydroxyapatite scaffolds containing DPSCs and/or SCAPs were subcutaneously transplanted into immunocompromised mice. After twelve weeks, histological and ultrastructural analysis demonstrated the regeneration of vascularized pulp-like tissue as well as mineralized tissue formation in all stem cell constructs. Despite the secretion of vascular endothelial growth factor in vitro, the stem cell constructs did not display a higher vascularization rate in comparison to control conditions. Similar results were found after eight weeks, which suggests both osteogenic/odontogenic differentiation of the transplanted stem cells and the promotion of angiogenesis in this particular setting. In conclusion, this is the first study to demonstrate the successful formation of vascularized pulp-like tissue in 3D-printed scaffolds containing dental stem cells, emphasizing the promising role of this approach in dental tissue engineering.
Collapse
|
60
|
Chaddad H, Kuchler-Bopp S, Fuhrmann G, Gegout H, Ubeaud-Sequier G, Schwinté P, Bornert F, Benkirane-Jessel N, Idoux-Gillet Y. Combining 2D angiogenesis and 3D osteosarcoma microtissues to improve vascularization. Exp Cell Res 2017; 360:138-145. [PMID: 28867479 DOI: 10.1016/j.yexcr.2017.08.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/12/2022]
Abstract
Angiogenesis is now well known for being involved in tumor progression, aggressiveness, emergence of metastases, and also resistance to cancer therapies. In this study, to better mimic tumor angiogenesis encountered in vivo, we used 3D culture of osteosarcoma cells (MG-63) that we deposited on 2D endothelial cells (HUVEC) grown in monolayer. We report that endothelial cells combined with tumor cells were able to form a well-organized network, and that tubule-like structures corresponding to new vessels infiltrate tumor spheroids. These vessels presented a lumen and expressed specific markers as CD31 and collagen IV. The combination of 2D endothelial cells and 3D microtissues of tumor cells also increased expression of angiogenic factors as VEGF, CXCR4 and ICAM1. The cell environment is the key point to develop tumor vascularization in vitro and to be closer to tumor encountered in vivo.
Collapse
Affiliation(s)
- Hassan Chaddad
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, UMR CNRS 7213, EA7293, Faculté de Pharmacie, route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Sabine Kuchler-Bopp
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg F-67000, France
| | - Guy Fuhrmann
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, UMR CNRS 7213, EA7293, Faculté de Pharmacie, route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Hervé Gegout
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg F-67000, France
| | - Geneviève Ubeaud-Sequier
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, UMR CNRS 7213, EA7293, Faculté de Pharmacie, route du Rhin, 67401 Illkirch-Graffenstaden, France
| | - Pascale Schwinté
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg F-67000, France
| | - Fabien Bornert
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg F-67000, France
| | - Nadia Benkirane-Jessel
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg F-67000, France.
| | - Ysia Idoux-Gillet
- INSERM, UMR 1109, Osteoarticular and Dental Regenerative NanoMedicine Laboratory, FMTS, 11 rue Humann, Strasbourg, France; Université de Strasbourg, Faculté de Chirurgie Dentaire, Hôpitaux Universitaires de Strasbourg (HUS), Strasbourg F-67000, France.
| |
Collapse
|
61
|
Müller AS, Janjić K, Lilaj B, Edelmayer M, Agis H. Hypoxia-based strategies for regenerative dentistry—Views from the different dental fields. Arch Oral Biol 2017; 81:121-130. [DOI: 10.1016/j.archoralbio.2017.04.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/24/2017] [Accepted: 04/25/2017] [Indexed: 12/20/2022]
|
62
|
Tudor D, Nenu I, Filip GA, Olteanu D, Cenariu M, Tabaran F, Ion RM, Gligor L, Baldea I. Combined regimen of photodynamic therapy mediated by Gallium phthalocyanine chloride and Metformin enhances anti-melanoma efficacy. PLoS One 2017; 12:e0173241. [PMID: 28278159 PMCID: PMC5344368 DOI: 10.1371/journal.pone.0173241] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 02/18/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Melanoma therapy is challenging, especially in advanced cases, due to multiple developed tumor defense mechanisms. Photodynamic therapy (PDT) might represent an adjuvant treatment, because of its bimodal action: tumor destruction and immune system awakening. In this study, a combination of PDT mediated by a metal substituted phthalocyanine-Gallium phthalocyanine chloride (GaPc) and Metformin was used against melanoma. The study aimed to: (1) find the anti-melanoma efficacy of GaPc-PDT, (2) assess possible beneficial effects of Metformin addition to PDT, (3) uncover some of the mechanisms underlining cell killing and anti-angiogenic effects. METHODS Two human lightly pigmented melanoma cell lines: WM35 and M1/15 subjected to previous Metformin exposure were treated by GaPc-PDT. Cell viability, death mechanism, cytoskeleton alterations, oxidative damage, were assessed by means of colorimetry, flowcytometry, confocal microscopy, spectrophotometry, ELISA, Western Blotting. RESULTS GaPc proved an efficient photosensitizer. Metformin addition enhanced cell killing by mechanisms dependent on the cell line, namely apoptosis in the metastatic M1/15 and necrosis in the radial growth phase, WM35. Cell death mechanism relied on the inhibition of nuclear transcription factor (NF)-κB activation and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) sensitization, leading to TRAIL and TNF-α induced apoptosis. Metformin diminished the anti-angiogenic effect of PDT. CONCLUSIONS Metformin addition to GaPc-PDT increased tumor cell killing through enhanced oxidative damage and induction of proapoptotic mechanisms, but altered PDT anti-angiogenic effects. GENERAL SIGNIFICANCE Combination of Metformin and PDT might represent a solution to enhance the efficacy, leading to a potential adjuvant role of PDT in melanoma therapy.
Collapse
Affiliation(s)
- Diana Tudor
- Department of Physiology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Iuliana Nenu
- Department of Physiology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Diana Olteanu
- Department of Physiology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai Cenariu
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Flaviu Tabaran
- Department of Pathology University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Rodica Mariana Ion
- Nanomedicine Research Group, National Institute for Research & Development in Chemistry and Petrochemistry - ICECHIM, Bucharest, Romania
| | - Lucian Gligor
- OSRAM Opto Semiconductors, OSRAM Romania, Global City Business Park, Voluntari, Ilfov, Romania
| | - Ioana Baldea
- Department of Physiology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
63
|
Semaphorin 4D Enhances Angiogenic Potential and Suppresses Osteo-/Odontogenic Differentiation of Human Dental Pulp Stem Cells. J Endod 2017; 43:297-305. [DOI: 10.1016/j.joen.2016.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/20/2016] [Accepted: 10/11/2016] [Indexed: 01/09/2023]
|
64
|
Xu JG, Zhu SY, Heng BC, Dissanayaka WL, Zhang CF. TGF-β1-induced differentiation of SHED into functional smooth muscle cells. Stem Cell Res Ther 2017; 8:10. [PMID: 28114966 PMCID: PMC5260045 DOI: 10.1186/s13287-016-0459-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/02/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs. Obtaining the appropriate building blocks for vascular tissue engineering (i.e. endothelial and mural cells) is a challenging task for tissue neovascularization. Hence, we investigated whether stem cells from human exfoliated deciduous teeth (SHED) could be induced to differentiate into functional vascular smooth muscle cells (vSMCs). METHODS We utilized two cytokines of the TGF-β family, transforming growth factor beta 1 (TGF-β1) and bone morphogenetic protein 4 (BMP4), to induce SHED differentiation into SMCs. Quantitative real-time polymerase chain reaction (RT-qPCR) was used to assess mRNA expression, and protein expression was analyzed using flow cytometry, western blot and immunostaining. Additionally, to examine whether these SHED-derived SMCs possess the same function as primary SMCs, in vitro Matrigel angiogenesis assay, fibrin gel bead assay, and functional contraction study were used here. RESULTS By analyzing the expression of specific markers of SMCs (α-SMA, SM22α, Calponin, and SM-MHC), we confirmed that TGF-β1, and not BMP4, could induce SHED differentiation into SMCs. The differentiation efficiency was relatively high (α-SMA+ 86.1%, SM22α+ 93.9%, Calponin+ 56.8%, and SM-MHC+ 88.2%) as assessed by flow cytometry. In vitro Matrigel angiogenesis assay showed that the vascular structures generated by SHED-derived SMCs and human umbilical vein endothelial cells (HUVECs) were comparable to primary SMCs and HUVECs in terms of vessel stability. Fibrin gel bead assay showed that SHED-derived SMCs had a stronger capacity for promoting vessel formation compared with primary SMCs. Further analyses of protein expression in fibrin gel showed that cultures containing SHED-derived SMCs exhibited higher expression levels of Fibronectin than the primary SMCs group. Additionally, it was also confirmed that SHED-derived SMCs exhibited functional contractility. When SB-431542, a specific inhibitor of ALK5 was administered, TGF-β1 stimulation could not induce SHED into SMCs, indicating that the differentiation of SHED into SMCs is somehow related to the TGF-β1-ALK5 signaling pathway. CONCLUSIONS SHED could be successfully induced into functional SMCs for vascular tissue engineering, and this course could be regulated through the ALK5 signaling pathway. Hence, SHED appear to be a promising candidate cell type for vascular tissue engineering.
Collapse
Affiliation(s)
- Jian Guang Xu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Shao Yue Zhu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Boon Chin Heng
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Waruna Lakmal Dissanayaka
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - Cheng Fei Zhang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| |
Collapse
|
65
|
Stem Cells of Dental Origin: Current Research Trends and Key Milestones towards Clinical Application. Stem Cells Int 2016; 2016:4209891. [PMID: 27818690 PMCID: PMC5081960 DOI: 10.1155/2016/4209891] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/14/2016] [Indexed: 12/17/2022] Open
Abstract
Dental Mesenchymal Stem Cells (MSCs), including Dental Pulp Stem Cells (DPSCs), Stem Cells from Human Exfoliated Deciduous teeth (SHED), and Stem Cells From Apical Papilla (SCAP), have been extensively studied using highly sophisticated in vitro and in vivo systems, yielding substantially improved understanding of their intriguing biological properties. Their capacity to reconstitute various dental and nondental tissues and the inherent angiogenic, neurogenic, and immunomodulatory properties of their secretome have been a subject of meticulous and costly research by various groups over the past decade. Key milestone achievements have exemplified their clinical utility in Regenerative Dentistry, as surrogate therapeutic modules for conventional biomaterial-based approaches, offering regeneration of damaged oral tissues instead of simply “filling the gaps.” Thus, the essential next step to validate these immense advances is the implementation of well-designed clinical trials paving the way for exploiting these fascinating research achievements for patient well-being: the ultimate aim of this ground breaking technology. This review paper presents a concise overview of the major biological properties of the human dental MSCs, critical for the translational pathway “from bench to clinic.”
Collapse
|
66
|
Rombouts C, Giraud T, Jeanneau C, About I. Pulp Vascularization during Tooth Development, Regeneration, and Therapy. J Dent Res 2016; 96:137-144. [PMID: 28106505 DOI: 10.1177/0022034516671688] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The pulp is a highly vascularized tissue situated in an inextensible environment surrounded by rigid dentin walls, with the apical foramina being the only access. The pulp vascular system is not only responsible for nutrient supply and waste removal but also contributes actively to the pulp inflammatory response and subsequent regeneration. This review discusses the underlying mechanisms of pulp vascularization during tooth development, regeneration, and therapeutic procedures, such as tissue engineering and tooth transplantation. Whereas the pulp vascular system is established by vasculogenesis during embryonic development, sprouting angiogenesis is the predominant process during regeneration and therapeutic processes. Hypoxia can be considered a common driving force. Dental pulp cells under hypoxic stress release proangiogenic factors, with vascular endothelial growth factor being one of the most potent. The benefit of exogenous vascular endothelial growth factor application in tissue engineering has been well demonstrated. Interestingly, dental pulp stem cells have an important role in pulp revascularization. Indeed, recent studies show that dental pulp stem cell secretome possesses angiogenic potential that actively contributes to the angiogenic process by guiding endothelial cells and even by differentiating themselves into the endothelial lineage. Although considerable insight has been obtained in the processes underlying pulp vascularization, many questions remain relating to the signaling pathways, timing, and influence of various stress conditions.
Collapse
Affiliation(s)
- C Rombouts
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| | - T Giraud
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France.,2 Service d'Odontologie, Hôpital Timone, APHM, Marseille, France
| | - C Jeanneau
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| | - I About
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| |
Collapse
|
67
|
The Neurovascular Properties of Dental Stem Cells and Their Importance in Dental Tissue Engineering. Stem Cells Int 2016; 2016:9762871. [PMID: 27688777 PMCID: PMC5027319 DOI: 10.1155/2016/9762871] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022] Open
Abstract
Within the field of tissue engineering, natural tissues are reconstructed by combining growth factors, stem cells, and different biomaterials to serve as a scaffold for novel tissue growth. As adequate vascularization and innervation are essential components for the viability of regenerated tissues, there is a high need for easily accessible stem cells that are capable of supporting these functions. Within the human tooth and its surrounding tissues, different stem cell populations can be distinguished, such as dental pulp stem cells, stem cells from human deciduous teeth, stem cells from the apical papilla, dental follicle stem cells, and periodontal ligament stem cells. Given their straightforward and relatively easy isolation from extracted third molars, dental stem cells (DSCs) have become an attractive source of mesenchymal-like stem cells. Over the past decade, there have been numerous studies supporting the angiogenic, neuroprotective, and neurotrophic effects of the DSC secretome. Together with their ability to differentiate into endothelial cells and neural cell types, this makes DSCs suitable candidates for dental tissue engineering and nerve injury repair.
Collapse
|
68
|
Werle SB, Chagastelles P, Pranke P, Casagrande L. The effects of hypoxia on in vitro culture of dental-derived stem cells. Arch Oral Biol 2016; 68:13-20. [DOI: 10.1016/j.archoralbio.2016.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 03/18/2016] [Accepted: 03/20/2016] [Indexed: 12/19/2022]
|
69
|
Yuan C, Wang P, Zhu S, Zou T, Wang S, Xu J, Heng BC, Diogenes A, Zhang C. EphrinB2 Stabilizes Vascularlike Structures Generated by Endothelial Cells and Stem Cells from Apical Papilla. J Endod 2016; 42:1362-70. [PMID: 27451120 DOI: 10.1016/j.joen.2016.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/26/2016] [Accepted: 05/14/2016] [Indexed: 12/26/2022]
Abstract
INTRODUCTION This study aimed to investigate the roles of ephrinB2 in stabilizing vascularlike structures generated by stem cells from apical papilla (SCAPs) and human umbilical vein endothelial cells (HUVECs). METHODS HUVECs were seeded alone or with SCAPs concurrently or 12 hours later. Angiogenesis and ephrinB2 phosphorylation were assayed at different time points. Additionally, ephrinB2 expression in SCAPs and HUVECs was silenced with small interfering RNA, and vascularlike structure formation within coculture was assessed; 1 × 10(5) HUVECs were seeded in transwell inserts, and 6 × 10(5) SCAPs were plated in lower wells with or without ephrinB2-Fc. Migratory cells were stained and counted. Delayed addition of ephrinB2-Fc to the coculture of HUVECs and SCAPs was performed to evaluate the role of ephrinB2 on the stabilization of vascularlike structures. RESULTS Concurrent coculture of SCAPs and HUVECs yielded significantly longer tubule lengths at 4, 8, and 12 hours (P < .05). Delayed addition of SCAPs to coculture with HUVECs resulted in vascularlike structures persisting longer than the HUVEC monoculture. Western blot confirmed that ephrinB2 phosphorylation was initiated at 0.5 hours of coculture and peaked at 1 hour. Silencing ephrinB2 expression in SCAPs and HUVECs resulted in the absence of vascularlike structures. Enhanced migration of HUVECs by SCAPs could be inhibited by ephrinB2-Fc. When ephrinB2-Fc was added at 3 hours of coculture, the vascularlike structures were stabilized for more than 12 hours as compared with 9 hours in the control group. CONCLUSIONS EphrinB2 plays an important role in the stabilization of vascularlike structures generated by HUVECs and SCAPs.
Collapse
Affiliation(s)
- Changyong Yuan
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Penglai Wang
- Dental Implant Center, Xuzhou Stomatological Hospital, Xuzhou, China
| | - Shaoyue Zhu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China; Dental Implant Center, Xuzhou Stomatological Hospital, Xuzhou, China
| | - Ting Zou
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shuai Wang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jianguang Xu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Boon Chin Heng
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Anibal Diogenes
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chengfei Zhang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
70
|
Current Advance and Future Prospects of Tissue Engineering Approach to Dentin/Pulp Regenerative Therapy. Stem Cells Int 2016; 2016:9204574. [PMID: 27069484 PMCID: PMC4812497 DOI: 10.1155/2016/9204574] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/25/2016] [Accepted: 02/17/2016] [Indexed: 01/09/2023] Open
Abstract
Recent advances in biomaterial science and tissue engineering technology have greatly spurred the development of regenerative endodontics. This has led to a paradigm shift in endodontic treatment from simply filling the root canal systems with biologically inert materials to restoring the infected dental pulp with functional replacement tissues. Currently, cell transplantation has gained increasing attention as a scientifically valid method for dentin-pulp complex regeneration. This multidisciplinary approach which involves the interplay of three key elements of tissue engineering—stem cells, scaffolds, and signaling molecules—has produced an impressive number of favorable outcomes in preclinical animal studies. Nevertheless, many practical hurdles need to be overcome prior to its application in clinical settings. Apart from the potential health risks of immunological rejection and pathogenic transmission, the lack of a well-established banking system for the isolation and storage of dental-derived stem cells is the most pressing issue that awaits resolution and the properties of supportive scaffold materials vary across different studies and remain inconsistent. This review critically examines the classic triad of tissue engineering utilized in current regenerative endodontics and summarizes the possible techniques developed for dentin/pulp regeneration.
Collapse
|
71
|
|
72
|
Gorin C, Rochefort GY, Bascetin R, Ying H, Lesieur J, Sadoine J, Beckouche N, Berndt S, Novais A, Lesage M, Hosten B, Vercellino L, Merlet P, Le-Denmat D, Marchiol C, Letourneur D, Nicoletti A, Vital SO, Poliard A, Salmon B, Muller L, Chaussain C, Germain S. Priming Dental Pulp Stem Cells With Fibroblast Growth Factor-2 Increases Angiogenesis of Implanted Tissue-Engineered Constructs Through Hepatocyte Growth Factor and Vascular Endothelial Growth Factor Secretion. Stem Cells Transl Med 2016; 5:392-404. [PMID: 26798059 DOI: 10.5966/sctm.2015-0166] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/07/2015] [Indexed: 12/18/2022] Open
Abstract
Tissue engineering strategies based on implanting cellularized biomaterials are promising therapeutic approaches for the reconstruction of large tissue defects. A major hurdle for the reliable establishment of such therapeutic approaches is the lack of rapid blood perfusion of the tissue construct to provide oxygen and nutrients. Numerous sources of mesenchymal stem cells (MSCs) displaying angiogenic potential have been characterized in the past years, including the adult dental pulp. Establishment of efficient strategies for improving angiogenesis in tissue constructs is nevertheless still an important challenge. Hypoxia was proposed as a priming treatment owing to its capacity to enhance the angiogenic potential of stem cells through vascular endothelial growth factor (VEGF) release. The present study aimed to characterize additional key factors regulating the angiogenic capacity of such MSCs, namely, dental pulp stem cells derived from deciduous teeth (SHED). We identified fibroblast growth factor-2 (FGF-2) as a potent inducer of the release of VEGF and hepatocyte growth factor (HGF) by SHED. We found that FGF-2 limited hypoxia-induced downregulation of HGF release. Using three-dimensional culture models of angiogenesis, we demonstrated that VEGF and HGF were both responsible for the high angiogenic potential of SHED through direct targeting of endothelial cells. In addition, FGF-2 treatment increased the fraction of Stro-1+/CD146+ progenitor cells. We then applied in vitro FGF-2 priming to SHED before encapsulation in hydrogels and in vivo subcutaneous implantation. Our results showed that FGF-2 priming is more efficient than hypoxia at increasing SHED-induced vascularization compared with nonprimed controls. Altogether, these data demonstrate that FGF-2 priming enhances the angiogenic potential of SHED through the secretion of both HGF and VEGF.
Collapse
Affiliation(s)
- Caroline Gorin
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Gael Y Rochefort
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Rumeyza Bascetin
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Hanru Ying
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Julie Lesieur
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Jérémy Sadoine
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Nathan Beckouche
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Sarah Berndt
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Anita Novais
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Matthieu Lesage
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Benoit Hosten
- INSERM UMR-S1144, Université Paris Descartes-Paris Diderot Sorbonne Paris Cité, AP-HP, Hôpital St. Louis, Unité Claude Kellershohn, Paris, France
| | - Laetitia Vercellino
- Université Paris Diderot, AP-HP, Hôpital St. Louis, Unité Claude Kellershohn, Paris, France
| | - Pascal Merlet
- Université Paris Diderot, AP-HP, Hôpital St. Louis, Unité Claude Kellershohn, Paris, France
| | - Dominique Le-Denmat
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Carmen Marchiol
- Institut Cochin, Plateforme Imagerie du vivant, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Didier Letourneur
- INSERM U1148, Laboratory of Vascular Translational Science, Université Paris Diderot Sorbonne Paris Cité, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, and Département Hospitalo-Universitaire Fibrosis, Inflammation, and Remodeling, Paris, France
| | - Antonino Nicoletti
- INSERM U1148, Laboratory of Vascular Translational Science, Université Paris Diderot Sorbonne Paris Cité, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, and Département Hospitalo-Universitaire Fibrosis, Inflammation, and Remodeling, Paris, France
| | - Sibylle Opsahl Vital
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Anne Poliard
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France
| | - Benjamin Salmon
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| | - Catherine Chaussain
- EA 2496 Pathologies, Imagerie et Biothérapies orofaciales et Plateforme Imagerie du Vivant, Dental School, Université Paris Descartes Sorbonne Paris Cité, Montrouge, France Assistance Publique des Hôpitaux de Paris (AP-HP) Département d'Odontologie, Hôpitaux Universitaires PNVS, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France Inserm U1050, Paris, France CNRS UMRS 7241, Paris, France
| |
Collapse
|
73
|
Cryopreservation and Banking of Dental Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 951:199-235. [DOI: 10.1007/978-3-319-45457-3_17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
74
|
Peters OA, Galicia J, Arias A, Tolar M, Ng E, Shin SJ. Effects of two calcium silicate cements on cell viability, angiogenic growth factor release and related gene expression in stem cells from the apical papilla. Int Endod J 2015; 49:1132-1140. [DOI: 10.1111/iej.12571] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/30/2015] [Indexed: 12/30/2022]
Affiliation(s)
- O. A. Peters
- Department of Endodontics; University of the Pacific Arthur A. Dugonic School of Dentistry; San Francisco CA USA
| | - J. Galicia
- Department of Endodontics; University of the Pacific Arthur A. Dugonic School of Dentistry; San Francisco CA USA
| | - A. Arias
- Department of Endodontics; University of the Pacific Arthur A. Dugonic School of Dentistry; San Francisco CA USA
| | - M. Tolar
- Department of Orthodontics; University of the Pacific Arthur A. Dugonic School of Dentistry; San Francisco CA USA
| | - E. Ng
- Department of Endodontics; University of the Pacific Arthur A. Dugonic School of Dentistry; San Francisco CA USA
| | - S. J. Shin
- Department of Endodontics; University of the Pacific Arthur A. Dugonic School of Dentistry; San Francisco CA USA
- Department of Conservative Dentistry and Oral Science Research Center; College of Dentistry; Gangnam Severance Hospital; Yonsei University; Seoul Korea
| |
Collapse
|
75
|
Lowenthal J, Gerecht S. Stem cell-derived vasculature: A potent and multidimensional technology for basic research, disease modeling, and tissue engineering. Biochem Biophys Res Commun 2015; 473:733-42. [PMID: 26427871 DOI: 10.1016/j.bbrc.2015.09.127] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/23/2015] [Indexed: 02/08/2023]
Abstract
Proper blood vessel networks are necessary for constructing and re-constructing tissues, promoting wound healing, and delivering metabolic necessities throughout the body. Conversely, an understanding of vascular dysfunction has provided insight into the pathogenesis and progression of diseases both common and rare. Recent advances in stem cell-based regenerative medicine - including advances in stem cell technologies and related progress in bioscaffold design and complex tissue engineering - have allowed rapid advances in the field of vascular biology, leading in turn to more advanced modeling of vascular pathophysiology and improved engineering of vascularized tissue constructs. In this review we examine recent advances in the field of stem cell-derived vasculature, providing an overview of stem cell technologies as a source for vascular cell types and then focusing on their use in three primary areas: studies of vascular development and angiogenesis, improved disease modeling, and the engineering of vascularized constructs for tissue-level modeling and cell-based therapies.
Collapse
Affiliation(s)
- Justin Lowenthal
- Medical Scientist Training Program, School of Medicine, Johns Hopkins University, Baltimore, MD, United States; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
76
|
Bakopoulou A, Kritis A, Andreadis D, Papachristou E, Leyhausen G, Koidis P, Geurtsen W, Tsiftsoglou A. Angiogenic Potential and Secretome of Human Apical Papilla Mesenchymal Stem Cells in Various Stress Microenvironments. Stem Cells Dev 2015. [PMID: 26203919 DOI: 10.1089/scd.2015.0197] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stem cells from the apical papilla (SCAP) of human adult teeth are considered an accessible source of cells with angiogenic properties. The aims of this study were to investigate the endothelial transdifferentiation of SCAP, the secretion of pro- and antiangiogenic factors from SCAP, and the paracrine effects of SCAP when exposed to environmental stress to stimulate tissue damage. SCAP were exposed to serum deprivation (SD), glucose deprivation (GD), and oxygen deprivation/hypoxia (OD) conditions, individually or in combination. Endothelial transdifferentiation was evaluated by in vitro capillary-like formation assays, real-time polymerase chain reaction, western blot, and flow cytometric analyses of angiogenesis-related markers; secretome by antibody arrays and enzyme-linked immunosorbent assays (ELISA); and paracrine impact on human umbilical vein endothelial cells (HUVECs) by in vitro transwell migration and capillary-like formation assays. The short-term exposure of SCAP to glucose/oxygen deprivation (GOD) in the presence, but mainly in deprivation, of serum (SGOD) elicited a proangiogenesis effect indicated by expression of angiogenesis-related genes involved in vascular endothelial growth factor (VEGF)/VEGFR and angiopoietins/Tie pathways. This effect was unachievable under SD in normoxia, suggesting that the critical microenvironmental condition inducing rapid endothelial shift of SCAP is the combination of SGOD. Interestingly, SCAP showed high adaptability to these adverse conditions, retaining cell viability and acquiring a capillary-forming phenotype. SCAP secreted higher numbers and amounts of pro- (angiogenin, IGFBP-3, VEGF) and lower amounts of antiangiogenic factors (serpin-E1, TIMP-1, TSP-1) under SGOD compared with SOD or SD alone. Finally, secretome obtained under SGOD was most effective in inducing migration and capillary-like formation by HUVECs. These data provide new evidence on the microenvironmental factors favoring endothelial transdifferentiation of SCAP, uncovering the molecular mechanisms regulating their fate. They also validate the angiogenic properties of their secretome giving insights into preconditioning strategies enhancing their therapeutic potential.
Collapse
Affiliation(s)
- Athina Bakopoulou
- 1 Department of Fixed Prosthesis and Implant Prosthodontics, School of Dentistry, Aristotle University of Thessaloniki (A.U.TH.) , Thessaloniki, Greece .,2 Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH) , Hannover, Germany
| | - Aristeidis Kritis
- 3 Department of Physiology and Pharmacology, School of Medicine, Aristotle University of Thessaloniki (A.U.TH.) , Thessaloniki, Greece
| | - Dimitrios Andreadis
- 4 Department of Oral Medicine and Pathology, School of Dentistry, Aristotle University of Thessaloniki (A.U.TH.) , Thessaloniki, Greece
| | - Eleni Papachristou
- 1 Department of Fixed Prosthesis and Implant Prosthodontics, School of Dentistry, Aristotle University of Thessaloniki (A.U.TH.) , Thessaloniki, Greece
| | - Gabriele Leyhausen
- 2 Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH) , Hannover, Germany
| | - Petros Koidis
- 1 Department of Fixed Prosthesis and Implant Prosthodontics, School of Dentistry, Aristotle University of Thessaloniki (A.U.TH.) , Thessaloniki, Greece
| | - Werner Geurtsen
- 2 Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH) , Hannover, Germany
| | - Asterios Tsiftsoglou
- 5 Laboratory of Pharmacology, School of Pharmaceutical Sciences, Aristotle University of Thessaloniki (A.U.TH.) , Thessaloniki, Greece
| |
Collapse
|
77
|
Hypoxia-Inducible Factor-1 in Physiological and Pathophysiological Angiogenesis: Applications and Therapies. BIOMED RESEARCH INTERNATIONAL 2015; 2015:549412. [PMID: 26146622 PMCID: PMC4471260 DOI: 10.1155/2015/549412] [Citation(s) in RCA: 404] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/10/2015] [Accepted: 04/17/2015] [Indexed: 02/07/2023]
Abstract
The cardiovascular system ensures the delivery of oxygen and nutrients to all cells, tissues, and organs. Under extended exposure to reduced oxygen levels, cells are able to survive through the transcriptional activation of a series of genes that participate in angiogenesis, glucose metabolism, and cell proliferation. The oxygen-sensitive transcriptional activator HIF-1 (hypoxia-inducible factor-1) is a key transcriptional mediator of the response to hypoxic conditions. The HIF-1 pathway was found to be a master regulator of angiogenesis. Whether the process is physiological or pathological, HIF-1 seems to participate in vasculature formation by synergistic correlations with other proangiogenic factors such as VEGF (vascular endothelial growth factor), PlGF (placental growth factor), or angiopoietins. Considering the important contributions of HIF-1 in angiogenesis and vasculogenesis, it should be considered a promising target for treating ischaemic diseases or cancer. In this review, we discuss the roles of HIF-1 in both physiological/pathophysiological angiogenesis and potential strategies for clinical therapy.
Collapse
|
78
|
Bhoj M, Zhang C, Green DW. A First Step in De Novo Synthesis of a Living Pulp Tissue Replacement Using Dental Pulp MSCs and Tissue Growth Factors, Encapsulated within a Bioinspired Alginate Hydrogel. J Endod 2015; 41:1100-7. [PMID: 25958179 DOI: 10.1016/j.joen.2015.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 01/15/2015] [Accepted: 03/08/2015] [Indexed: 01/17/2023]
Abstract
INTRODUCTION A living, self-supporting pulp tissue replacement in vitro and for transplantation is an attractive yet unmet bioengineering challenge. Our aim is to create 3-dimensional alginate-based microenvironments that replicate the shape of gutta-percha and comprise key elements for the proliferation of progenitor cells and the release of growth factors. METHODS An RGD-bearing alginate framework was used to encapsulate dental pulp stem cells and human umbilical vein endothelial cells in a ratio of 1:1. The alginate hydrogel also retained and delivered 2 key growth factors, vascular endothelial growth factor-121 and fibroblast growth factor, in a sufficient amount to induce proliferation. A method was then devised to replicate the shape of gutta-percha using RGD alginate within a custom-made mold of thermoresponsive N-isopropylacrylamide. Plugs of alginate containing different permutations of growth factor-based encapsulates were tested and evaluated for viability, proliferation, and release kinetics between 1 and 14 days. RESULTS According to scanning electron microscopic and confocal microscopic observations, the encapsulated human endothelial cells and dental pulp stem cell distribution were frequent and extensive throughout the length of the construct. There were also high levels of viability in all test environments. Furthermore, cell proliferation was higher in the growth factor-based groups. Growth factor release kinetics also showed significant differences between them. Interestingly, the combination of vascular endothelial growth factor and fibroblast growth factor synergize to significantly up-regulate cell proliferation. CONCLUSIONS RGD-alginate scaffolds can be fabricated into shapes to fill the pulp space by simple templating. The addition of dual growth factors to cocultures of stem cells within RGD-alginate scaffolds led to the creation of microenvironments that significantly enhance the proliferation of dental pulp stem cell/human umbilical vein endothelial cell combinations.
Collapse
Affiliation(s)
- Manasi Bhoj
- Oral Biosciences, Faculty of Dentistry, The University of Hong Kong Hospital, Sai Ying Pun, Hong Kong
| | - Chengfei Zhang
- Comprehensive Dental Care, Faculty of Dentistry, The University of Hong Kong Hospital, Sai Ying Pun, Hong Kong.
| | - David W Green
- Oral Biosciences, Faculty of Dentistry, The University of Hong Kong Hospital, Sai Ying Pun, Hong Kong.
| |
Collapse
|