1
|
Liang Q, Zhang C, Lv P, Huang Y, Zhao H, Jiang S, Xu W. The important role of the Wnt/β-catenin signaling pathway in small molecules mediated gingival mesenchymal stem cells transdifferentiate into neuron-like cells. Arch Oral Biol 2025; 169:106115. [PMID: 39488928 DOI: 10.1016/j.archoralbio.2024.106115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVE Given their neural crest origin, gingival mesenchymal stem cells (GMSCs) possess high neurogenic potential, which makes them suitable for cell replacement therapy against neurodegenerative diseases. This study investigated whether GMSCs can be transdifferentiated into neurons in vitro using a protocol involving small molecules VCRFY (VPA, CHIR99021, Repsox, Forskolin, and Y-27632). The regulatory mechanisms of key signaling pathways were also investigated. METHODS Neuronal induction of GMSCs was conducted using a small molecules-based protocol over 7 days, which included the evaluation of cell morphology, proliferation, expressions of neurogenic markers, and intracellular calcium oscillation. The activation of canonical the Wnt signaling pathway was assessed by examining the protein content and subcellular localization of β-catenin. RESULTS Small molecules-treated GMSCs displayed neuronal morphology and increased expression of neurogenic markers, including class III beta-tubulin (TUJ1), neuron-specific enolase (NSE), microtube-associated protein 2 (MAP2), and neurofilament medium (NFM), verified through RT-qPCR, western blotting, and immunocytochemistry. Based on the results of Fluo-4 AM calcium flux assay, small molecules-treated GMSCs exhibited enhanced electrophysiological activity. GMSC proliferation halted after 2 days of treatment. Among the small molecules, CHIR99021 exhibited the highest neuronal induction efficiency. Furthermore, activation of the Wnt/β-catenin signaling pathway augmented neuronal differentiation. CONCLUSIONS Small molecule-based cellular reprogramming can efficiently generate neurons from GMSCs, with Wnt/β-catenin signaling to play a critical role in neuronal induction.
Collapse
Affiliation(s)
- Qiuying Liang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Chuhan Zhang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Peiyi Lv
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Yongmao Huang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Hang Zhao
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China
| | - Shan Jiang
- Department of Periodontics and Oral Medicine, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China.
| | - Wenan Xu
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangdong, China; Department of Pediatric Dentistry, Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Guangdong, China.
| |
Collapse
|
2
|
Visintin PV, Zampieri BL, Griesi-Oliveira K. Chemical transdifferentiation of somatic cells to neural cells: a systematic review. EINSTEIN-SAO PAULO 2024; 22:eRW0423. [PMID: 39661857 PMCID: PMC11634374 DOI: 10.31744/einstein_journal/2024rw0423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/21/2024] [Indexed: 12/13/2024] Open
Abstract
INTRODUCTION Transdifferentiation is the conversion of a specific somatic cell into another cell type, bypassing a transient pluripotent state. This implies a faster method to generate cells of interest with the additional benefit of reduced tumorigenic risk for clinical use. OBJECTIVE We describe protocols that use small molecules as direct conversion inducers, without the need for exogenous factors, to evaluate the potential of cell transdifferentiation for pharmacological and clinical applications. METHODS In this systematic review, using PRISMA guidelines, we conducted a personalized search strategy in four databases (PubMed, Scopus, Embase, and Web Of Science), looking for experimental works that used exclusively small molecules for transdifferentiation of non-neural cell types into neural lineage cells. RESULTS We explored the main biological mechanisms involved in direct cell conversion induced by different small molecules used in 33 experimental in vitro and in vitro transdifferentiation protocols. We also summarize the main characteristics of these protocols, such as the chemical cocktails used, time for transdifferentiation, and conversion efficiency. CONCLUSION Small molecules-based protocols for neuronal transdifferentiation are reasonably safe, economical, accessible, and are a promising alternative for future use in regenerative medicine and pharmacology.
Collapse
Affiliation(s)
- Paulo Victor Visintin
- Hospital Israelita Albert EinsteinSão PauloSPBrazilHospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Bruna Lancia Zampieri
- Hospital Israelita Albert EinsteinSão PauloSPBrazilHospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Karina Griesi-Oliveira
- Hospital Israelita Albert EinsteinSão PauloSPBrazilHospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
3
|
Inchingolo AM, Inchingolo AD, Nardelli P, Latini G, Trilli I, Ferrante L, Malcangi G, Palermo A, Inchingolo F, Dipalma G. Stem Cells: Present Understanding and Prospects for Regenerative Dentistry. J Funct Biomater 2024; 15:308. [PMID: 39452606 PMCID: PMC11508604 DOI: 10.3390/jfb15100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Regenerative medicine in dentistry focuses on repairing damaged oral tissues using advanced tools like stem cells, biomaterials, and tissue engineering (TE). Mesenchymal stem cells (MSCs) from dental sources, such as dental pulp and periodontal ligament, show significant potential for tissue regeneration due to their proliferative and differentiative abilities. This systematic review, following PRISMA guidelines, evaluated fifteen studies and identified effective strategies for improving dental, periodontal, and bone tissue regeneration through scaffolds, secretomes, and bioengineering methods. Key advancements include the use of dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) to boost cell viability and manage inflammation. Additionally, pharmacological agents like matrine and surface modifications on biomaterials improve stem cell adhesion and promote osteogenic differentiation. By integrating these approaches, regenerative medicine and TE can optimize dental therapies and enhance patient outcomes. This review highlights the potential and challenges in this field, providing a critical assessment of current research and future directions.
Collapse
Affiliation(s)
- Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Paola Nardelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giulia Latini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Irma Trilli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| |
Collapse
|
4
|
Jiang S, Yuan C, Zou T, Koh JH, Basabrain M, Chen Q, Liu J, Heng BC, Lim LW, Wang P, Zhang C. An Injectable Hydrogel Loaded with GMSCs-Derived Neural Lineage Cells Promotes Recovery after Stroke. Tissue Eng Part A 2024; 30:563-576. [PMID: 38756085 DOI: 10.1089/ten.tea.2023.0330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
Ischemic stroke is a devastating medical condition with poor prognosis due to the lack of effective treatment modalities. Transplantation of human neural stem cells or primary neural cells is a promising treatment approach, but this is hindered by limited suitable cell sources and low in vitro expansion capacity. This study aimed (1) use small molecules (SM) to reprogram gingival mesenchymal stem cells (GMSCs) commitment to the neural lineage cells in vitro, and (2) use hyaluronic acid (HA) hydrogel scaffolds seeded with GMSCs-derived neural lineage cells to treat ischemic stroke in vivo. Neural induction was carried out with a SM cocktail-based one-step culture protocol over a period of 24 h. The induced cells were analyzed for expression of neural markers with immunocytochemistry and quantitative real-time polymerase chain reaction (qRT-PCR). The Sprague-Dawley (SD) rats (n = 100) were subjected to the middle cerebral artery occlusion (MCAO) reperfusion ischemic stroke model. Then, after 8 days post-MCAO, the modeled rats were randomly assigned to six study groups (n = 12 per group): (1) GMSCs, (2) GMSCs-derived neural lineage cells, (3) HA and GMSCs-derived neural lineage cells, (4) HA, (5) PBS, and (6) sham transplantation control, and received their respective transplantation. Evaluation of post-stroke recovery were performed by behavioral tests and histological assessments. The morphologically altered nature of neural lineages has been observed of the GMSCs treated with SMs compared to the untreated controls. As shown by the qRT-PCR and immunocytochemistry, SMs further significantly enhanced the expression level of neural markers of GMSCs as compared with the untreated controls (all p < 0.05). Intracerebral injection of self-assembling HA hydrogel carrying GMSCs-derived neural lineage cells promoted the recovery of neural function and reduced ischemic damage in rats with ischemic stroke, as demonstrated by histological examination and behavioral assessments (all p < 0.05). In conclusion, the SM cocktail significantly enhanced the differentiation of GMSCs into neural lineage cells. The HA hydrogel was found to facilitate the proliferation and differentiation of GMSCs-derived neural lineage cells. Furthermore, HA hydrogel seeded with GMSCs-derived neural lineage cells could promote tissue repair and functional recovery in rats with ischemic stroke and may be a promising alternative treatment modality for stroke.
Collapse
Affiliation(s)
- Shan Jiang
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Changyong Yuan
- The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ting Zou
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Jun Hao Koh
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
| | - Mohammed Basabrain
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
| | - Qixin Chen
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
| | - Junqing Liu
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
| | | | - Lee Wei Lim
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Penglai Wang
- The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chengfei Zhang
- Faculty of Dentistry, Restorative Dental Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
5
|
Koh J, Liu J, Poon CH, Kang J, Basabrain MS, Lim LW, Zhang C. Transplantation of Neural Progenitor Cells Derived from Stem Cells from Apical Papilla Through Small-Molecule Induction in a Rat Model of Sciatic Nerve Injury. Tissue Eng Regen Med 2024; 21:867-879. [PMID: 38904732 PMCID: PMC11286922 DOI: 10.1007/s13770-024-00648-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Stem cell-based transplantation therapy holds promise for peripheral nerve injury treatment, but adult availability is limited. A cell culture protocol utilizing a small-molecule cocktail effectively reprogrammed stem cells from apical papilla (SCAPs) into neural progenitor cells, subsequently differentiating into neuron-like cells. This study aims to evaluate neural-induced SCAPs, with and without small-molecule cocktail, for sciatic nerve repair potential. METHODS A scaffold-free cell sheet technique was used to construct a three-dimensional cell sheet. Subsequently, this cell sheet was carefully rolled into a tube and seamlessly inserted into a collagen conduit, which was then transplanted into a 5 mm sciatic nerve injury rat model. Functional sciatic nerve regeneration was evaluated via toe spread test, walking track analysis and gastrocnemius muscle weight. Additionally, degree of sciatic nerve regeneration was determined based on total amount of myelinated fibers. RESULTS Small-molecule cocktail induced SCAPs enhanced motor function recovery, evident in improved sciatic function index and gastrocnemius muscle retention. We also observed better host myelinated fiber retention than undifferentiated SCAPs or neural-induced SCAPs without small-molecule cocktail. However, clusters of neuron-like cell bodies (surrounded by sparse myelinated fibers) were found in all cell sheet-implanted groups in the implantation region. This suggests that while the implanted cells likely survived transplantation, integration was poor and would likely hinder long-term recovery by occupying the space needed for host nerve fibers to project through. CONCLUSION Neural-induced SCAPs with small-molecule cocktail demonstrated promising benefits for nerve repair; further research is needed to improve its integration and optimize its potential for long-term recovery.
Collapse
Affiliation(s)
- Junhao Koh
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Junqing Liu
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Chi Him Poon
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jun Kang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Mohammed S Basabrain
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
- Restorative Dental Sciences, Faculty of Dentistry, Umm Al-Qura, University, Makkah, Saudi Arabia
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
6
|
Raghavan A, Ghosh S. Influence of Graphene-Based Nanocomposites in Neurogenesis and Neuritogenesis: A Brief Summary. ACS APPLIED BIO MATERIALS 2024; 7:711-726. [PMID: 38265040 DOI: 10.1021/acsabm.3c00852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Graphene is a prospective candidate for various biomedical applications, including drug transporters, bioimaging agents, and scaffolds for tissue engineering, thanks to its superior electrical conductivity and biocompatibility. The clinical issue of nerve regeneration and rehabilitation still has a major influence on people's lives. Nanomaterials based on graphene have been exploited extensively to promote nerve cell differentiation and proliferation. Their high electrical conductivity and mechanical robustness make them appropriate for nerve tissue engineering. Combining graphene with other substances, such as biopolymers, may transmit biochemical signals that support brain cell division, proliferation, and regeneration. The utilization of nanocomposites based on graphene in neurogenesis and neuritogenesis is the primary emphasis of this review. Here are some examples of the many synthetic strategies used. For neuritogenesis and neurogenesis, it has also been explored to combine electrical stimulation with graphene-based materials.
Collapse
Affiliation(s)
- Akshaya Raghavan
- Polymers & Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sutapa Ghosh
- Polymers & Functional Materials Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
7
|
Basabrain MS, Zhong J, Liu J, Zhang Y, Abdalla MM, Zhang C. Interactions of Neuronally Induced Stem Cells from Apical Papilla Spheres, Stems Cells from Apical Papilla, and Human Umbilical Vascular Endothelial Cells on Vasculogenesis and Neurogenesis. J Endod 2024; 50:64-73.e4. [PMID: 37866800 DOI: 10.1016/j.joen.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/24/2023]
Abstract
INTRODUCTION Stem cell-based dental pulp regeneration has been extensively studied, mainly focusing on exploiting dental stem cells' osteogenic and angiogenic potentials. Dental stem cells' neurogenic role is often overlooked. Stem cells from apical papilla (SCAPs), originating from the neural crest and capable of sphere formation, display potent neurogenic capacity. This study aimed to investigate the interactions of neuronally induced stem cells from apical papilla (iSCAP) spheres, SCAPs, and human umbilical vascular endothelial cells (HUVECs) on vasculogenesis and neurogenesis. METHODS SCAPs were isolated and characterized using flow cytometry and multilineage differentiation assays. SCAP monolayer culture and spheres were neuronally induced by a small molecule neural induction medium, and the neural gene expression and neurite formation at days 0, 3, and 7 were evaluated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and using phase-contrast light and fluorescence microscopy. Direct coculture or pulp-on-chip was used to investigate iSCAP sphere interaction with SCAPs and HUVECs. RT-qPCR, fluorescence microscopy, and immunostaining with β-tubulin III, alpha-smooth muscle actin, and CD31 were used to study neural gene expression, neurite formation, and neurovascular cell interactions. RESULTS Neural induction medium with small molecules rapidly induced SCAP differentiation toward neural-like cells. Gene expression of Nestin, β-tubulin III, microtubule-associated protein 2, neuron-specific enolase, and NeuN was higher in iSCAP spheres than in iSCAPs. iSCAP spheres formed more and longer neurites compared with iSCAPs. iSCAP sphere, HUVEC, and SCAP direct coculture significantly enhanced vessel formation along with up-regulated VEGF (P < .001) and multiple neural markers, such as Nestin (P < .01), microtubule-associated protein 2 (P < .001), S100 (P < .001), and NG2 (P < .001). iSCAP spheres, SCAPs, and HUVECs cultured in a pulp-on-chip system promoted endothelial and neural cell migration toward each other and alpha-smooth muscle actin-positive and CD31-positive cells assembling for the vascular constitution. CONCLUSIONS iSCAP-formed spheres interact with SCAPs and HUVECs, promoting vasculogenesis and neurogenesis.
Collapse
Affiliation(s)
- Mohammed S Basabrain
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China; Restorative Dental Sciences, Faculty of Dentistry, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jialin Zhong
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Junqing Liu
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yuchen Zhang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Mohamed Mahmoud Abdalla
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China; Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, P.R. China.
| |
Collapse
|
8
|
Hanna S, Eldeen GN, Alfayate RP, Aly R. The regenerative potential of Tideglusib and CHIR99021 small molecules as potent odontogenic differentiation enhancers of human dental pulp stem cells. Clin Oral Investig 2023; 28:48. [PMID: 38153556 DOI: 10.1007/s00784-023-05452-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVES To assess the effect of Tideglusib and CHIR99021 small molecules on the odontogenic differentiation potential of human dental pulp stem cells (hDPSCs) via Wnt/β-catenin pathway activation. METHODOLOGY hDPSCs were isolated from impacted third molars indicated for extraction and were characterized by flow cytometry. hDPSCs were then induced to differentiate into odontogenic lineage in the presence of Tideglusib and CHIR99021. Odontogenic differentiation was evaluated using Alizarin Red stain and RT-PCR for expression of odontogenic specific differentiation markers: DSPP, DMP1, ALP, OPN, and RUNX2 in relation to undifferentiated cells. RT-PCR was also conducted to assess the expression of Wnt/β-catenin pathway activation marker (AXIN2). One-way ANOVA Kruskal-Wallis test was used for statistical analysis. RESULTS Wnt/β-catenin pathway was successfully activated by Tideglusib and CHIR99021 in hDPSCs where AXIN2 was significantly upregulated. Successful odontogenic differentiation was confirmed by Alizarin Red staining of calcified nodules. RT-PCR for odontogenic differentiation markers DSPP, DMP1, and RUNX expression by hDPSCs induced by CHIR99021 was higher than that expressed by hDPSCs induced by Tideglusib, whereas expression of OPN and ALP was higher in Tideglusib-induced cells than in CHIR99021-induced cells. CONCLUSIONS Both small molecules successfully induced odontogenic differentiation of hDPSCs through Wnt/β-catenin pathway activation. CLINICAL RELEVANCE These findings suggest that Tideglusib and CHIR99021 can be applied clinically in pulp regeneration to improve strategies for vital pulp regeneration and to promote dentine repair.
Collapse
Affiliation(s)
- Samer Hanna
- Endodontics Department, Universidad Europea De Madrid (UEM), Madrid, Spain
| | - Ghada Nour Eldeen
- Molecular Genetics & Enzymology Department, Human Genetic & Genome Research Institute, National Research Centre, Dokki, Giza, Egypt
| | | | - Riham Aly
- Basic Dental Science Department, Oral Medicine & Dentistry Research Institute, National Research Centre, Dokki, Giza, Egypt.
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt.
| |
Collapse
|
9
|
Prateeksha P, Naidu P, Das M, Barthels D, Das H. KLF2 Regulates Neural Differentiation of Dental Pulp-derived Stem Cells by Modulating Autophagy and Mitophagy. Stem Cell Rev Rep 2023; 19:2886-2900. [PMID: 37642902 DOI: 10.1007/s12015-023-10607-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Transplantation of stem cells for treating neurodegenerative disorders is a promising future therapeutic approach. However, the molecular mechanism underlying the neuronal differentiation of dental pulp-derived stem cells (DPSC) remains inadequately explored. The current study aims to define the regulatory role of KLF2 (Kruppel-like factor 2) during the neural differentiation (ND) of DPSC. METHODS We first investigated the transcriptional and translational expression of KLF2, autophagy, and mitophagy-associated markers during the ND of DPSC by using quantitative RT-PCR and western blot methods. After that, we applied the chemical-mediated loss- and gain-of-function approaches using KLF2 inhibitor, GGPP (geranylgeranyl pyrophosphate), and KLF2 activator, GGTI-298 (geranylgeranyl transferase inhibitor-298) to delineate the role of KLF2 during ND of DPSC. The western blot, qRT-PCR, and immunocytochemistry were performed to determine the molecular changes during ND after KLF2 deficiency and KLF2 sufficiency. We also analyzed the oxygen consumption rate (OCR) and the extracellular acidification rate (ECAR) using the Seahorse XFe24 analyzer. RESULTS Our study demonstrated that the expression level of KLF2, autophagy, and mitophagy-associated markers were significantly elevated during the ND of DPSC. Next, we found that the KLF2 inhibitor, GGPP significantly reduced the ND of DPSC. Inversely, KLF2 overexpression accelerated the molecular phenomenon of DPSC's commitment towards ND, indicating the crucial role of KLF2 in neurogenesis. Moreover, we found that the KLF2 positively regulated autophagy, mitophagy, and the Wnt5a signaling pathway during neurogenesis. Seahorse XFe24 analysis revealed that the ECAR and OCR parameters were significantly increased during ND, and inhibition of KLF2 marginally reversed them towards DPSC's cellular bioenergetics. However, KLF2 overexpression shifted the cellular energy metabolism toward the quiescent stage. CONCLUSION Collectively, our findings provide the first evidence that the KLF2 critically regulates the neurogenesis of DPSC by inducing autophagy and mitophagy.
Collapse
Affiliation(s)
- Prateeksha Prateeksha
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Prathyusha Naidu
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Manjusri Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Derek Barthels
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA.
| |
Collapse
|
10
|
Fujii Y, Hatori A, Chikazu D, Ogasawara T. Application of Dental Pulp Stem Cells for Bone and Neural Tissue Regeneration in Oral and Maxillofacial Region. Stem Cells Int 2023; 2023:2026572. [PMID: 37035445 PMCID: PMC10076122 DOI: 10.1155/2023/2026572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 10/21/2022] [Accepted: 03/18/2023] [Indexed: 03/31/2023] Open
Abstract
In the oral and maxillofacial region, the treatment of severe bone defects, caused by fractures, cancers, congenital abnormalities, etc., remains a great challenge. In addition, neurological disorders are frequently accompanied by these bone defects or the treatments for them. Therefore, novel bone regenerative techniques and methods to repair nerve injury are eagerly sought. Among them, strategies using dental pulp stem cells (DPSCs) are promising options. Human DPSCs can be collected easily from extracted teeth and are now considered a type of mesenchymal stem cell with higher clonogenic and proliferative potential. DPSCs have been getting attention as a cell source for bone and nerve regeneration. In this article, we reviewed the latest studies on osteogenic or neural differentiation of DPSCs as well as bone or neural regeneration methods using DPSCs and discussed the potential of DPSCs for bone and nerve tissue regeneration.
Collapse
|
11
|
Song WP, Jin LY, Zhu MD, Wang H, Xia DS. Clinical trials using dental stem cells: 2022 update. World J Stem Cells 2023; 15:31-51. [PMID: 37007456 PMCID: PMC10052340 DOI: 10.4252/wjsc.v15.i3.31] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/20/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
For nearly 20 years, dental stem cells (DSCs) have been successfully isolated from mature/immature teeth and surrounding tissue, including dental pulp of permanent teeth and exfoliated deciduous teeth, periodontal ligaments, dental follicles, and gingival and apical papilla. They have several properties (such as self-renewal, multidirectional differentiation, and immunomodulation) and exhibit enormous potential for clinical applications. To date, many clinical articles and clinical trials using DSCs have reported the treatment of pulpitis, periapical lesions, periodontitis, cleft lip and palate, acute ischemic stroke, and so on, and DSC-based therapies obtained satisfactory effects in most clinical trials. In these studies, no adverse events were reported, which suggested the safety of DSC-based therapy. In this review, we outline the characteristics of DSCs and summarize clinical trials and their safety as DSC-based therapies. Meanwhile, we also present the current limitations and perspectives of DSC-based therapy (such as harvesting DSCs from inflamed tissue, applying DSC-conditioned medium/DSC-derived extracellular vesicles, and expanding-free strategies) to provide a theoretical basis for their clinical applications.
Collapse
Affiliation(s)
- Wen-Peng Song
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Lu-Yuan Jin
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Meng-Di Zhu
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Hao Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Deng-Sheng Xia
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
12
|
Small Molecule GSK-3 Inhibitors Safely Promote the Proliferation and Viability of Human Dental Pulp Stem Cells-In Vitro. Biomedicines 2023; 11:biomedicines11020542. [PMID: 36831078 PMCID: PMC9953089 DOI: 10.3390/biomedicines11020542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Small molecules have demonstrated promising results as successful alternatives to growth factors. In this study, focus was drawn to CHIR99021 and tideglusib as GSK-3 inhibitors known for their anti-inflammatory and regenerative potential. The effect of both tideglusib and CHIR99021 on the proliferation, viability, and stemness of human dental pulp stem cells (hDPSCs) was investigated to assess their possible role in regenerative dentistry. Briefly, hDPSCs were isolated from sound premolars extracted for orthodontic purposes. Cytotoxicity and proliferation assessment were performed via cell counting kit-8 followed by flow cytometric analysis of apoptotic marker ANNEXIN V. The effect of both small molecules on the stemness of hDPSCs was analyzed by qRT-PCR. Both tideglusib and CHIR99021 were proven to be safe on hDPSCs. The tideglusib concentration that resulted in higher viable cells was 100 nM, while the concentration for CHIR99021 was 5 nM. Both small molecules successfully induced cellular proliferation and demonstrated minimal expression of ANNEXIN V, indicative of the absence of cellular apoptosis and further confirming their positive effect on proliferation. Finally, both small molecules enhanced stemness markers expression as evidenced by qRT-PCR, which, again, highlighted the positive effect of both tideglusib and CHIR99021 on safely promoting the proliferation of hDPSCs while maintaining their stemness.
Collapse
|
13
|
Li X, Heng BC, Bai Y, Wang Q, Gao M, He Y, Zhang X, Deng X, Zhang X. Electrical charge on ferroelectric nanocomposite membranes enhances SHED neural differentiation. Bioact Mater 2023; 20:81-92. [PMID: 35633875 PMCID: PMC9131252 DOI: 10.1016/j.bioactmat.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Stem cells from human exfoliated deciduous teeth (SHED) uniquely exhibit high proliferative and neurogenic potential. Charged biomaterials have been demonstrated to promote neural differentiation of stem cells, but the dose-response effect of electrical stimuli from these materials on neural differentiation of SHED remains to be elucidated. Here, by utilizing different annealing temperatures prior to corona poling treatment, BaTiO3/P(VDF-TrFE) ferroelectric nanocomposite membranes with varying charge polarization intensity (d33 ≈ 0, 4, 12 and 19 pC N−1) were fabricated. Enhanced expression of neural markers, increased cell elongation and more prominent neurite outgrowths were observed with increasing surface charge of the nanocomposite membrane indicating a dose-response effect of surface electrical charge on SHED neural differentiation. Further investigations of the underlying molecular mechanisms revealed that intracellular calcium influx, focal adhesion formation, FAK-ERK mechanosensing pathway and neurogenic-related ErbB signaling pathway were implicated in the enhancement of SHED neural differentiation by surface electrical charge. Hence, this study confirms the dose-response effect of biomaterial surface charge on SHED neural differentiation and provides preliminary insights into the molecular mechanisms and signaling pathways involved. Membrane surface charge can be precisely controlled by adjusting annealing temperature and corona poling parameters. Both earlier and later neurogenic differentiation of SHED appear to be dose-dependently enhanced by surface charge. Underlying molecular mechanisms may involve intracellular Ca2+ influx, focal adhesion formation, FAK-ERK and ErbB signaling.
Collapse
Affiliation(s)
- Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Qianqian Wang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Min Gao
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Xinwen Zhang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, 110002, PR China
- Corresponding author.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Corresponding author. Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- Corresponding author. Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| |
Collapse
|
14
|
Sramkó B, Földes A, Kádár K, Varga G, Zsembery Á, Pircs K. The Wisdom in Teeth: Neuronal Differentiation of Dental Pulp Cells. Cell Reprogram 2023; 25:32-44. [PMID: 36719998 PMCID: PMC9963504 DOI: 10.1089/cell.2022.0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are found in almost all postnatal organs. Under appropriate environmental cues, multipotency enables MSCs to serve as progenitors for several lineage-specific, differentiated cell types. In vitro expansion and differentiation of MSCs give the opportunity to obtain hardly available somatic cells, such as neurons. The neurogenic potential of MSCs makes them a promising, autologous source to restore damaged tissue and as such, they have received much attention in the field of regenerative medicine. Several stem cell pool candidates have been studied thus far, but only a few of them showed neurogenic differentiation potential. Due to their embryonic ontology, stem cells residing in the stroma of the dental pulp chamber are an exciting source for in vitro neural cell differentiation. In this study, we review the key properties of dental pulp stem cells (DPSCs), with a particular focus on their neurogenic potential. Moreover, we summarize the various presently available methods used for neural differentiation of human DPSCs also emphasizing the difficulties in reproducibly high production of such cells. We postulate that because DPSCs are stem cells with very close ontology to neurogenic lineages, they may serve as excellent targets for neuronal differentiation in vitro and even for direct reprogramming.
Collapse
Affiliation(s)
- Bendegúz Sramkó
- HCEMM-SU Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary.,Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Földes
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Kristóf Kádár
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Gábor Varga
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Ákos Zsembery
- Department of Oral Biology, Faculty of Dentistry, Semmelweis University, Budapest, Hungary
| | - Karolina Pircs
- HCEMM-SU Neurobiology and Neurodegenerative Diseases Research Group, Budapest, Hungary.,Institute of Translational Medicine, Semmelweis University, Budapest, Hungary.,Laboratory of Molecular Neurogenetics, Department of Experimental Medical Science, Wallenberg Neuroscience Center and Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Mitchell J, Lo KWH. Small molecule-mediated regenerative engineering for craniofacial and dentoalveolar bone. Front Bioeng Biotechnol 2022; 10:1003936. [PMID: 36406208 PMCID: PMC9667056 DOI: 10.3389/fbioe.2022.1003936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 09/29/2023] Open
Abstract
The comprehensive reconstruction of extensive craniofacial and dentoalveolar defects remains a major clinical challenge to this day, especially in complex medical cases involving cancer, cranioplasty, and traumatic injury. Currently, osteogenic small molecule-based compounds have been explored extensively to repair and regenerate bone tissue because of their unique advantages. Over the past few years, a number of small molecules with the potential of craniofacial and periodontal bone tissue regeneration have been reported in literature. In this review, we discuss current progress using small molecules to regulate cranial and periodontal bone regeneration. Future directions of craniofacial bone regenerative engineering using the small molecule-based compounds will be discussed as well.
Collapse
Affiliation(s)
- Juan Mitchell
- School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Kevin W. H. Lo
- School of Medicine, Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health Center, Farmington, CT, United States
- Department of Medicine, Division of Endocrinology, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
- Department of Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT, United States
- School of Engineering, Institute of Materials Science (IMS), University of Connecticut, Storrs, CT, United States
| |
Collapse
|
16
|
Basabrain MS, Zhong J, Luo H, Liu J, Yi B, Zaeneldin A, Koh J, Zou T, Zhang C. Formation of Three-Dimensional Spheres Enhances the Neurogenic Potential of Stem Cells from Apical Papilla. Bioengineering (Basel) 2022; 9:604. [PMID: 36354515 PMCID: PMC9687952 DOI: 10.3390/bioengineering9110604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/18/2022] [Accepted: 10/21/2022] [Indexed: 10/24/2023] Open
Abstract
UNLABELLED Cell-based neural regeneration is challenging due to the difficulty in obtaining sufficient neural stem cells with clinical applicability. Stem cells from apical papilla (SCAPs) originating from embryonic neural crests with high neurogenic potential could be a promising cell source for neural regeneration. This study aimed to investigate whether the formation of 3D spheres can promote SCAPs' neurogenic potential. MATERIAL AND METHODS Three-dimensional SCAP spheres were first generated in a 256-well agarose microtissue mold. The spheres and single cells were individually cultured on collagen I-coated μ-slides. Cell morphological changes, neural marker expression, and neurite outgrowth were evaluated by confocal microscope, ELISA, and RT-qPCR. RESULTS Pronounced morphological changes were noticed in a time-dependent manner. The migrating cells' morphology changed from fibroblast-like cells to neuron-like cells. Compared to the 2D culture, neurite length, number, and the expression of multiple progenitors, immature and mature neural markers were significantly higher in the 3D spheres. BDNF and NGF-β may play a significant role in the neural differentiation of SCAP spheres. CONCLUSION The formation of 3D spheres enhanced the neurogenic potential of SCAPs, suggesting the advantage of using the 3D spheres of SCAPs for treating neural diseases.
Collapse
Affiliation(s)
- Mohammed S. Basabrain
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Jialin Zhong
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Haiyun Luo
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
- Stomatological Hospital, Southern Medical University, 366 Jiangnan Avenue South, Guangzhou 510280, China
| | - Junqing Liu
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Baicheng Yi
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Ahmed Zaeneldin
- Restorative Dental Sciences, Cariology, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Junhao Koh
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Ting Zou
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Simonovic J, Toljic B, Lazarevic M, Markovic MM, Peric M, Vujin J, Panajotovic R, Milasin J. The Effect of Liquid-Phase Exfoliated Graphene Film on Neurodifferentiation of Stem Cells from Apical Papilla. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12183116. [PMID: 36144905 PMCID: PMC9502655 DOI: 10.3390/nano12183116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 05/29/2023]
Abstract
BACKGROUND Dental stem cells, which originate from the neural crest, due to their easy accessibility might be good candidates in neuro-regenerative procedures, along with graphene-based nanomaterials shown to promote neurogenesis in vitro. We aimed to explore the potential of liquid-phase exfoliated graphene (LPEG) film to stimulate the neuro-differentiation of stem cells from apical papilla (SCAP). METHODS The experimental procedure was structured as follows: (1) fabrication of graphene film; (2) isolation, cultivation and SCAP stemness characterization by flowcytometry, multilineage differentiation (osteo, chondro and adipo) and quantitative PCR (qPCR); (3) SCAP neuro-induction by cultivation on polyethylene terephthalate (PET) coated with graphene film; (4) evaluation of neural differentiation by means of several microscopy techniques (light, confocal, atomic force and scanning electron microscopy), followed by neural marker gene expression analysis using qPCR. RESULTS SCAP demonstrated exceptional stemness, as judged by mesenchymal markers' expression (CD73, CD90 and CD105), and by multilineage differentiation capacity (osteo, chondro and adipo-differentiation). Neuro-induction of SCAP grown on PET coated with graphene film resulted in neuron-like cellular phenotype observed under different microscopes. This was corroborated by the high gene expression of all examined key neuronal markers (Ngn2, NF-M, Nestin, MAP2, MASH1). CONCLUSIONS The ability of SCAPs to differentiate toward neural lineages was markedly enhanced by graphene film.
Collapse
Affiliation(s)
- Jelena Simonovic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Bosko Toljic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milos Lazarevic
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Mina Peric
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Jasna Vujin
- Graphene Laboratory, Center for Solid State Physics and New Materials, Institute of Physics, University of Belgrade, 11000 Belgrade, Serbia
| | - Radmila Panajotovic
- Graphene Laboratory, Center for Solid State Physics and New Materials, Institute of Physics, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Milasin
- School of Dental Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
18
|
Luo H, Basabrain MS, Zhong J, Liu J, Zhang Y, Qi Y, Zou T, Zhang C. Neuroregenerative potential of SCAP-derived neuronal cell spheroids regulated by SCAPs under various microenvironments in a pulp-on-chip system. J Endod 2022. [DOI: 10.1016/j.joen.2022.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
19
|
Liu J, Zou T, Zhang Y, Koh J, Li H, Wang Y, Zhao Y, Zhang C. Three-dimensional electroconductive carbon nanotube-based hydrogel scaffolds enhance neural differentiation of stem cells from apical papilla. BIOMATERIALS ADVANCES 2022; 138:212868. [PMID: 35913250 DOI: 10.1016/j.bioadv.2022.212868] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/30/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
The radical treatment of neurological impairments remains a major clinical challenge. Stem cells with high neural differentiation ability delivered by electroconductive hydrogel scaffolds have demonstrated promising applications in neural tissue regeneration. However, there are still challenges in designing bioactive scaffolds with good biocompatibility, appropriate electrical conductivity, and neurogenic niche. Herein, a three-dimensional (3D) electroconductive gelatin methacryloyl-multi-walled carbon nanotube/cobalt (GelMA-MWCNTs/Co) hydrogel scaffold was fabricated by incorporating MWCNTs/Co composites into a GelMA hydrogel matrix. The surface morphology, pore size, elastic modulus, swelling ratio, and conductivity of the hydrogels were measured. GelMA-MWCNTs/Co exhibited higher electrical conductivity than GelMA-MWCNTs. Live/dead and CCK8 assays demonstrated the good biocompatibility of the hydrogel for stem cells from apical papilla (SCAP) growth and differentiation. The cells encapsulated in the GelMA-MWCNTs and GelMA-MWCNTs/Co hydrogel scaffolds exhibited significant neuronal cell-like changes and a notable level of neuronal-specific marker expression after the electrical stimulation (ES) for 7 days, compared to that in the hydrogels without ES. Notably, the neurite spreading and Tuj1 fluorescent intensity of the SCAP in the electrically conductive GelMA-MWCNTs/Co hydrogel were more prominent compared to those of the other two groups. In addition, the 3D conductive hydrogel scaffolds advanced the neural differentiation of SCAP to an earlier time point. Considering these aspects, the novel electroconductive GelMA-MWCNTs/Co hydrogel synergized with ES greatly promotes SCAP neuronal differentiation.
Collapse
Affiliation(s)
- Junqing Liu
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, Guangdong, China
| | - Ting Zou
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, Guangdong, China
| | - Yuchen Zhang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, Guangdong, China
| | - Junhao Koh
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Hongwen Li
- Department of Dentistry, Longgang ENT Hospital, Shenzhen, Guangdong, China; Shenzhen Longgang Institute of Stomatology, Longgang, Shenzhen, Guangdong, China
| | - Yan Wang
- Department of VIP Center, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, China
| | - Yi Zhao
- Straits Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China; Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, Guangdong, China..
| |
Collapse
|
20
|
Wang X, Wu J, Wang W, Zhang Y, He D, Xiao B, Zhang H, Song A, Xing Y, Li B. Reprogramming of Rat Fibroblasts into Induced Neurons by Small-Molecule Compounds In Vitro and In Vivo. ACS Chem Neurosci 2022; 13:2099-2109. [PMID: 35723446 DOI: 10.1021/acschemneuro.2c00078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cell replacement is a promising approach for neurodegenerative disease treatment. Somatic cells such as fibroblasts can be induced to differentiate into neurons by specific transcription factors; however, the potential of viral vectors used for reprogramming to integrate into the genome raises concerns about the potential clinical applications of this approach. Here, we directly reprogrammed rat embryonic skin fibroblasts into induced neurons (iNs) via six small-molecule compounds (SMs) (VPA, CHIR99021, forskolin, Y-27632, Repsox, and P7C3-A20). iNs exhibit typical neuronal morphology, and immunofluorescence showed that more than 96% of the iNs expressed the early neuronal marker class III beta-tubulin (TUJ1) and that more than 91% of iNs expressed the mature neuronal marker neuron-specific enolase (NSE) after 10 days of reprogramming. Quantitative real-time polymerase chain reaction also showed that most iNs expressed the dopaminergic neuron marker tyrosine hydroxylase, the neural marker Nur correlation factor 1, the (γ-aminobutyric acid, GABA) GABAergic neuronal marker GABA, and the cholinergic neuron marker choline acetyltransferase. In addition, we found that cell proliferation decreased during reprogramming and that protein synthesis increased initially and then decreased. SMs were mixed with hydrogels, and the hydrogels were implanted subcutaneously into the backs of rats. After 7 days, the TUJ1 and NSE proteins were expressed in surrounding tissues, indicating that SMs caused reprogramming in vivo. In summary, rat skin fibroblasts can be efficiently reprogrammed into iNs by SMs in vitro and in vivo.
Collapse
Affiliation(s)
- Xueyun Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Jing Wu
- Department of Paediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001 Henan, P.R. China
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Yuanwang Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Dixin He
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Boying Xiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Haohao Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Anqi Song
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Bo Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| |
Collapse
|
21
|
Guo M, Liu F, Wang W, Liu Z, Zhu Z, Liu Y, Huang Z. Naringin Promotes Osteogenic/Odontogenic Differentiation of Dental Pulp Stem Cells via Wnt/ β-Catenin. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4505471. [PMID: 35677363 PMCID: PMC9168102 DOI: 10.1155/2022/4505471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022]
Abstract
Purpose This investigation intended to unravel the effect and mechanism of naringin on the proliferation and osteogenic differentiation of human dental pulp stem cells (hDPSCs). Methods hDPSCs were induced to differentiate, and the degree of cell differentiation was observed by alizarin red staining, Oil Red O staining, and Alcian blue staining. hDPSCs were treated with 0, 20, 40, and 80 μmol/L naringin for 48 h, respectively. The proliferation rate and chemotaxis of the cells were measured by MTT and transwell assay, alkaline phosphatase (ALP) activity and osteogenic differentiation degree by ALP staining and alizarin red staining, and gene expression of osteogenic markers by qRT-PCR. Additionally, western blot was performed to test the levels of Wnt/β-catenin signaling-related proteins in hDPSCs. Results The isolated hDPSCs with spindle-shaped morphology had good differentiation capability. Further experiments confirmed naringin-caused increases in the proliferation rate and migration ability of hDPSCs. In addition, compared with the control group, naringin-treated cells had strong ALP activity and ossification levels and higher expression of Runx2, OPN, DSPP, and DMP1. The western blot results showed that naringin significantly activated Wnt/β-catenin signaling in hDPSCs. Conclusion Taken together, naringin enhances the proliferation, migration, and osteogenesis of hDPSCs through stimulating Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Meiling Guo
- Department of General Dentistry, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
| | - Fen Liu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Wenjuan Wang
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Zhirong Liu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Zhipeng Zhu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Yiyu Liu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Zhen Huang
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| |
Collapse
|
22
|
Srikawnawan W, Songsaad A, Gonmanee T, Thonabulsombat C, Phruksaniyom C, White KL, Ruangsawasdi N. Rho kinase inhibitor induced human dental pulp stem cells to differentiate into neurons. Life Sci 2022; 300:120566. [DOI: 10.1016/j.lfs.2022.120566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 10/18/2022]
|
23
|
Cui Y, Bai M, Guo D, Yang Y, Chen H, Sun J, Xie J, Zhou X. Insulin-like growth factor 1 promotes neural differentiation of human stem cells from the apical papilla. Arch Oral Biol 2021; 131:105264. [PMID: 34598025 DOI: 10.1016/j.archoralbio.2021.105264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Insulin-like growth factor 1 (IGF1) is one of the vital factors in regenerative endodontics. Previous studies have focused on the role of IGF1 in the mineralization of dental tissues. However, the role of IGF1 in the neural differentiation of dental stem cells was little discussed. DESIGN IGF1 was overexpressed in human stem cells from the apical papilla (hSCAPs) by lentivirus and knocked down in hSCAPs by small interfering RNA. The neural differentiation level of hSCAPs was investigated histologically by HE staining and Nissl staining after neural induction for 3 days. The expression of proteins was examined by western blot and immunofluorescence. RESULTS IGF1 promoted neural differentiation of hSCAPs, more cell processes and Nissl-positive body stained cells. IGF1 overexpression could both promote glial differentiation in hSCAPs, characterized by the increase of S100β and GFAP proteins, and neuronal differentiation, characterized by the increase of βIII-tubulin and functional GAD67/vGLUT1 proteins. Conversely, IGF1 knockdown suppressed both glial and neuronal differentiation. IGF1 activated AKT to regulate the early neural differentiation of hSCAPs. CONCLUSIONS The results indicate IGF1 could promote neural differentiation of hSCAPs by activating AKT signaling and provide a cue for the candidate of induced neural seeding cells in regenerative endodontics.
Collapse
Affiliation(s)
- Yujia Cui
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mingru Bai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Daimo Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yueyi Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Haoran Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jianxun Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
24
|
Zou T, Jiang S, Yi B, Chen Q, Heng BC, Zhang C. Gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor enhances small molecule-induced neurogenic differentiation of stem cells from apical papilla. J Biomed Mater Res A 2021; 110:623-634. [PMID: 34590393 DOI: 10.1002/jbm.a.37315] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
The limited neurogenic potential of adult stem cells and their non-specific lineage differentiation pose major challenges in cell-replacement therapy for neurological disorders. In our previous study, we demonstrated that the neurogenic potential of stem cells from apical papilla (SCAPs) was significantly improved upon induction with a small molecule cocktail. This study attempted to investigate whether neuronal differentiation of SCAPs induced by a small molecule cocktail can be further enhanced in a three-dimensional gelatin methacrylate hydrogel loaded with brain-derived neurotrophic factor (BDNF-GelMA). The physiological properties and neural differentiation of SCAPs treated with a combination of small molecules and BDNF-GelMA were evaluated by CCK8, Live/Dead assay, quantitative reverse transcription-polymerase chain reaction, western blot and immunocytochemistry. SCAPs embedded in BDNF-GelMA displayed superior morphological characteristics when induced by a small molecule cocktail, similar to neuronal phenotypes as compared to pure GelMA. There was significant upregulation of neural markers including Tuj1 and MAP2 by SCAPs embedded in BDNF-GelMA, as compared to pure GelMA. Hence, GelMA hydrogel loaded with a potent neurotrophic factor (BDNF) provides a conducive scaffold that can further enhance the differentiation of small molecule-treated SCAPs into neuronal-like cells, which may provide a therapeutic platform for the management of neurological disorders.
Collapse
Affiliation(s)
- Ting Zou
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shan Jiang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Baicheng Yi
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Qixin Chen
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | | | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
25
|
Kim D, Lee AE, Xu Q, Zhang Q, Le AD. Gingiva-Derived Mesenchymal Stem Cells: Potential Application in Tissue Engineering and Regenerative Medicine - A Comprehensive Review. Front Immunol 2021; 12:667221. [PMID: 33936109 PMCID: PMC8085523 DOI: 10.3389/fimmu.2021.667221] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/30/2021] [Indexed: 12/15/2022] Open
Abstract
A unique subpopulation of mesenchymal stem cells (MSCs) has been isolated and characterized from human gingival tissues (GMSCs). Similar to MSCs derived from other sources of tissues, e.g. bone marrow, adipose or umbilical cord, GMSCs also possess multipotent differentiation capacities and potent immunomodulatory effects on both innate and adaptive immune cells through the secretion of various types of bioactive factors with immunosuppressive and anti-inflammatory functions. Uniquely, GMSCs are highly proliferative and have the propensity to differentiate into neural cell lineages due to the neural crest-origin. These properties have endowed GMSCs with potent regenerative and therapeutic potentials in various preclinical models of human disorders, particularly, some inflammatory and autoimmune diseases, skin diseases, oral and maxillofacial disorders, and peripheral nerve injuries. All types of cells release extracellular vesicles (EVs), including exosomes, that play critical roles in cell-cell communication through their cargos containing a variety of bioactive molecules, such as proteins, nucleic acids, and lipids. Like EVs released by other sources of MSCs, GMSC-derived EVs have been shown to possess similar biological functions and therapeutic effects on several preclinical diseases models as GMSCs, thus representing a promising cell-free platform for regenerative therapy. Taken together, due to the easily accessibility and less morbidity of harvesting gingival tissues as well as the potent immunomodulatory and anti-inflammatory functions, GMSCs represent a unique source of MSCs of a neural crest-origin for potential application in tissue engineering and regenerative therapy.
Collapse
Affiliation(s)
- Dane Kim
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alisa E Lee
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Qilin Xu
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Qunzhou Zhang
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anh D Le
- Department of Oral & Maxillofacial Surgery & Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center of Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States.,Department of Oral & Maxillofacial Surgery, Penn Medicine Hospital of the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
26
|
Chen Q, Yuan C, Jiang S, Heng BC, Zou T, Shen Z, Wang P, Zhang C. Small molecules efficiently reprogram apical papilla stem cells into neuron-like cells. Exp Ther Med 2021; 21:546. [PMID: 33850518 DOI: 10.3892/etm.2021.9978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
Stem cell-based therapy may provide a novel approach for neural tissue regeneration. A small molecule cocktail-based culture protocol was previously shown to enhance neurogenic differentiation of stem cells from dental tissues. The present study aimed to investigate the early phase of small molecule-induced neurogenic differentiation of stem cells from the apical papilla (SCAP). SCAP were cultured in neural-induction medium or neural-induction medium with small molecules (NIMS-SCAP) and examined for their cell morphologies. Expression levels of neural progenitor cell-related markers, including Nestin, paired-box gene 6 (Pax6) and Sry-related HMG box 2 (Sox2), were examined using western blotting and immunocytofluorescence. Expression of differentiated neuron-related markers, including neurofilament protein (NFM), neuron-specific nuclear protein (NeuN) and microtubule-associated protein (MAP)-2, were also examined using western blotting, while NFM and MAP2 gene expression and cell proliferation were assessed using reverse transcription-quantitative (RT-q)PCR and Cell Counting Kit (CCK)-8 assays, respectively. SCAP morphology was affected by small molecules after as little as 30 min. Specifically, Nestin, Pax6 and Sox2 expression detected using western blotting was increased by day 3 but then decreased over the course of 7 days with neural induction, while immunocytofluorescence revealed expression of all three markers in NIMS-SCAP. The protein levels of NFM, NeuN and MAP2 on day 7 were significantly upregulated in NIMS-SCAP, as detected using western blotting, while NFM and MAP2 gene expression levels detected using RT-qPCR were significantly increased on days 5 and 7. Proliferation of NIMS-SCAP ceased after 5 days. Electrophysiological analysis showed that only SCAP cultured in NIMS had the functional activity of neuronal cells. Thus, small molecules reprogrammed SCAP into neural progenitor cells within the first 3 days, followed by further differentiation into neuron-like cells.
Collapse
Affiliation(s)
- Qixin Chen
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China.,Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Changyong Yuan
- Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Shan Jiang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Ting Zou
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| | - Zhongshan Shen
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Penglai Wang
- Department of Implant Dentistry, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong 999077, SAR, P.R. China
| |
Collapse
|
27
|
Sharma Y, Shobha K, Sundeep M, Pinnelli VB, Parveen S, Dhanushkodi A. Neural Basis of Dental Pulp Stem Cells and its Potential Application in Parkinson's disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:62-76. [PMID: 33719979 DOI: 10.2174/1871527320666210311122921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/16/2021] [Accepted: 01/29/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease. Though significant insights into the molecular-biochemical-cellular-behavioral basis of PD have been understood, there is no appreciable treatment available till date. Current therapies provide symptomatic relief without any influence on the progression of the disease. Stem cell therapy has been vigorously explored to treat PD. In this comprehensive review, we analyze various stem cell candidates for treating PD and discuss the possible mechanisms. We advocate the advantage of using neural crest originated dental pulp stem cells (DPSC) due to their predisposition towards neural differentiation and their potential to regenerate neurons far better than commonly used bone marrow derived mesenchymal stem cells (BM-MSCs). Eventually, we highlight the current challenges in the field and the strategies which may be used for overcoming the impediments.
Collapse
Affiliation(s)
- Yogita Sharma
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, Karnataka. India
| | - Shobha K
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, Karnataka. India
| | - Mata Sundeep
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, Karnataka. India
| | | | - Shagufta Parveen
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, Karnataka. India
| | - Anandh Dhanushkodi
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, Karnataka. India
| |
Collapse
|
28
|
De A, Beligala DH, Sharma VP, Burgos CA, Lee AM, Geusz ME. Cancer stem cell generation during epithelial-mesenchymal transition is temporally gated by intrinsic circadian clocks. Clin Exp Metastasis 2020; 37:617-635. [PMID: 32816185 DOI: 10.1007/s10585-020-10051-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a key event preceding tumor cell metastasis that increases cell invasiveness and cancer stem cell (CSC) populations. Studies suggest that genes used in generating circadian rhythms also serve in regulating EMT. To test the role of circadian clocks in cellular EMT events two cancer cell lines were compared, one that has a well-established circadian clock, C6 from rat glioma, and one that does not, MCF-7 from human breast tumor. MCF-7 tumorsphere cultures were tested for evidence of circadian rhythms because of previously reported circadian rhythm enhancement in C6 tumorspheres shown by elevated rhythm amplitude and increased expression of circadian clock gene Per2. Bioluminescence imaging of Per2 gene expression in MCF-7 tumorspheres revealed a previously unconfirmed circadian clock in this important cancer research model. Inducing CSC generation through EMT in C6 and MCF-7 monolayer cultures revealed circadian oscillations in the size of the post-EMT CSC population, confirming that circadian rhythms are additional processes controlling this stage of cancer progression. EMT was verified by distinct cellular morphological changes and expression of stem cell proteins OCT4, nestin, MSI1, and CD133 along with EMT-related proteins ZEB1, vimentin, and TWIST. Quantifying single-cell events and behaviors through time-lapse imaging indicated the post-EMT population size was determined largely by circadian rhythms in epithelial-like cancer cells undergoing EMT. We then identified a specific phase of the circadian rhythm in Per2 gene activation as a potential target for therapeutic treatments that may suppress EMT, minimize CSCs, and limit metastasis.
Collapse
Affiliation(s)
- Arpan De
- Department of Biological Sciences, Bowling Green State University, 217 Life Science Bldg., Bowling Green, OH, 43403, USA
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dilshan H Beligala
- Department of Biological Sciences, Bowling Green State University, 217 Life Science Bldg., Bowling Green, OH, 43403, USA
- Department of Molecular Biology and Biotechnology, Faculty of Science, University of Peradeniya, Peradeniya, 20400, Sri Lanka
| | - Vishal P Sharma
- Department of Biological Sciences, Bowling Green State University, 217 Life Science Bldg., Bowling Green, OH, 43403, USA
- Celsee, Inc., Ann Arbor, MI, 48108, USA
| | - Christian A Burgos
- Department of Biological Sciences, Bowling Green State University, 217 Life Science Bldg., Bowling Green, OH, 43403, USA
| | - Angelia M Lee
- Department of Biological Sciences, Bowling Green State University, 217 Life Science Bldg., Bowling Green, OH, 43403, USA
| | - Michael E Geusz
- Department of Biological Sciences, Bowling Green State University, 217 Life Science Bldg., Bowling Green, OH, 43403, USA.
| |
Collapse
|